Next Article in Journal
Intrauterine Transfusion for Rhesus Alloimmunization: A Historical Retrospective Cohort from A Single Reference Center in Brazil
Next Article in Special Issue
Coping Strategies and Inflammatory Bowel Disease: A Narrative Review
Previous Article in Journal
The Uricosuric Effect of SGLT2 Inhibitors Is Maintained in the Long Term in Patients with Chronic Kidney Disease and Type 2 Diabetes Mellitus
Previous Article in Special Issue
Intestinal Microbiome Changes and Clinical Outcomes of Patients with Ulcerative Colitis after Fecal Microbiota Transplantation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Interaction between Stress and Inflammatory Bowel Disease in Pediatric and Adult Patients

by
Oana Belei
1,2,
Diana-Georgiana Basaca
1,2,*,
Laura Olariu
2,
Manuela Pantea
3,
Daiana Bozgan
4,
Anda Nanu
5,
Iuliana Sîrbu
5,
Otilia Mărginean
1,2 and
Ileana Enătescu
3
1
First Pediatric Clinic, Disturbances of Growth and Development on Children Research Center, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
2
Department of Pediatrics, First Pediatric Clinic, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
3
Twelfth Department, Neonatology Clinic, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
4
Clinic of Neonatology, “Pius Brânzeu” County Emergency Clinical Hospital, 300723 Timișoara, Romania
5
Third Pediatric Clinic, “Louis Țurcanu” Emergency Children Hospital, 300011 Timișoara, Romania
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2024, 13(5), 1361; https://doi.org/10.3390/jcm13051361
Submission received: 27 January 2024 / Revised: 23 February 2024 / Accepted: 25 February 2024 / Published: 27 February 2024
(This article belongs to the Special Issue Multidisciplinary Approaches to Inflammatory Bowel Disease Management)

Abstract

:
Background: Inflammatory bowel diseases (IBDs) have seen an exponential increase in incidence, particularly among pediatric patients. Psychological stress is a significant risk factor influencing the disease course. This review assesses the interaction between stress and disease progression, focusing on articles that quantified inflammatory markers in IBD patients exposed to varying degrees of psychological stress. Methods: A systematic narrative literature review was conducted, focusing on the interaction between IBD and stress among adult and pediatric patients, as well as animal subjects. The research involved searching PubMed, Scopus, Medline, and Cochrane Library databases from 2000 to December 2023. Results: The interplay between the intestinal immunity response, the nervous system, and psychological disorders, known as the gut–brain axis, plays a major role in IBD pathophysiology. Various types of stressors alter gut mucosal integrity through different pathways, increasing gut mucosa permeability and promoting bacterial translocation. A denser microbial load in the gut wall emphasizes cytokine production, worsening the disease course. The risk of developing depression and anxiety is higher in IBD patients compared with the general population, and stress is a significant trigger for inducing acute flares of the disease. Conclusions: Further large studies should be conducted to assess the relationship between stressors, psychological disorders, and their impact on the course of IBD. Clinicians involved in the medical care of IBD patients should aim to implement stress reduction practices in addition to pharmacological therapies.

Graphical Abstract

1. Introduction

1.1. Overview of the Epidemiology and Pathophysiology of Inflammatory Bowel Disease

The economic and financial cost of inflammatory bowel disease (IBD) is recognized as a severe, global public health problem. IBD is a broad category of complex, protracted bowel inflammation characterized by a variety of variables, including emotional distress, autonomic dysfunction, dysbiosis of the gut microbiota, and immunological modulations related to disease activity. The characterization of local and systemic immune responses in IBD and the pathways via which inflammation enters the central nervous system (CNS) as well as their effects on brain-resident immune and glial cells have become more and more the focus of recent research [1].
Significant attention should be given to the gradual increase in the prevalence of IBD in both children and adults. Currently, there remains a lack of a complete understanding regarding the underlying mechanisms of IBD. It is believed that the development of IBD involves complex interplays between genetics, environmental factors, and gut microbiota. However, the fluctuating nature of IBD, characterized by periods of relapse and remission, emphasizes the significance of additional factors, including psychological stress [2].
In this review, the authors will present up-to-date evidence discussing the impact of stress on IBD across different stages, encompassing both children and adults. IBD, encompassing ulcerative colitis (UC) and Crohn’s disease (CD), is a chronic inflammatory disorder of the intestines that affects a substantial number of individuals globally, with a relapsing and remitting course [2]. It should be noted that in the twenty-first century, IBD is quickly expanding in prevalence in rising industrial nations and is progressively becoming a global illness [3]. Even though IBD can develop at any age, it is diagnosed in 25% of individuals before the age of 20 [4]. IBD in children is more common in some nations than others, although the global trend is rising. The incidence ranges from 0.1 to 13.9/100,000 for CD, from 0.3 to 15/100,000 for UC, and between 0.5 and 23/100,000 for IBD [4,5]. Children may present with particular signs, such as poor growth and delayed puberty, in addition to the typical gastrointestinal (GI) symptoms (abdominal discomfort, diarrhea, hematochezia, and weight loss) identical to those of adults [6]. Periods of heightened symptomatology known as active phases alternate with remission phases throughout the disease, which is typically unpredictable. CD is characterized by non-continuous inflammation that extends through the entire wall of the intestine, primarily affecting the terminal ileum, caecum, colon, and perianal area. In contrast, UC is distinguished by continuous inflammation and ulceration limited to the colon and rectum [7].
IBD is acknowledged as an immune-mediated disorder of the intestines, stemming from multifaceted interactions among genetics, environmental factors, and the gut microbiota [2]. According to studies conducted in the last few decades, several factors, including genetic transmission, intestinal immune disruption, gut microbiota disruption, diet, infection, lifestyle, psychological stress, sleep disorders, smoking, and early-life antibiotic exposure, can affect the development of IBD [8,9]. The exact underlying mechanism responsible for IBD’s pathophysiology remains poorly understood.
Immunologically, IBD is characterized by dysregulation of the mucosal immune system, involving the loss of immune tolerance and the emergence of uncontrolled immune responses to antigens derived from the normal gut microbiota [10]. More than 240 susceptibility loci were found by extensive genome-wide association studies, many of which involve genes encoding proteins that trigger adaptive and effector immune activities [11]. IBD patients deal with a heavy disease load, have difficulty completing everyday tasks, and run the risk of developing anxiety and depressive disorders [12].

1.2. Stress Definition

The definition of “stress” in medicine was initially provided more than 80 years ago by the Hungarian endocrinologist Hans Hugo Bruno Selye as the physiological adaptive reactions of organisms to harmful threats (stressors), whether endogenous or external, physical or psychological, or actual or perceived [13]. The stress system, which organisms have developed to preserve homeostasis under threat, incorporates physiological and behavioral adaptations via appropriate central and peripheral neuroendocrine responses and is incredibly complicated. The organisms may enter a condition known as cacostasis, in which many crucial physiological activities are compromised, and may develop various acute and chronic diseases when exposed to long-term or severe stress [8].
The flooding of adaptive capacities, which results in an excessive vital overflow beyond normal self-regulatory potential, is one of psychological stress’s principal impacts. Psychological stress is a specific kind of relationship between an individual and the environment [14]. The association between the stress factor and the emergence of several pathologies, including cardiovascular diseases [15,16], coronary disorders [16,17], high blood pressure [18,19], strokes [20], and brain damage [21,22], has been extensively researched by many different authors. Additionally, it is obvious that stress negatively affects a variety of illnesses [15,23]. Similar to other diseases, the research at hand appears to support the hypothesis that, among other things, excessive levels of stress have a role in the etiology, course, and responsiveness to the therapy of digestive problems [13,24]. In the current competitive environment, stress is unavoidable, and chronic stress is linked to negative consequences on physical health, including the development of IBD.
Stress is broadly described as a threat to a steady state of homeostasis in a person’s life, in contrast to psychiatric disorders like sadness and anxiety. It involves both a stressor (i.e., an environmental demand) and a person’s physiological and emotional reaction to the stressor [25]. The majority of Americans—60%—report experiencing stress in their daily lives [25]. In contrast, in a patient with anxiety, stress perception may be disproportionate to environmental demand. An exaggerated response to a stressor may be functionally disruptive and develop into a psychiatric disease. It is becoming more widely understood that significant psychiatric comorbidities have a negative impact on IBD patients’ disease activity and use of healthcare services, but it is less known how daily stressors and perceived stress affect patients’ IBD outcomes and disease courses [26]. Through its effects on the immunological, endocrine, and neurological systems, stress has been shown to negatively affect GI function and increase gut permeability.

2. Objectives of This Study

This review aims to assess the influence of psychological stress on the prevalence and outcomes of intestinal inflammation among pediatric and adult patients. The onset, degree of activity, and response to treatment among children and adults with IBD are incompletely understood and are still the subject of current research. There are a lot of factors influencing IBD activity (environmental, genetic factors, neuropsychological or intestinal microbiota imbalance, and intestinal immunity impairment). This paper aims to analyze the interplay between these factors and the course of the disease. Furthermore, this review sets out to quantify if a reduction in stressful factors decreases the incidence of IBD relapses and prolongs the remission time of the disease.

3. Materials and Methods

3.1. Search Strategy

The authors performed narrative literature research centered on the interaction between IBD and stress among adult and pediatric patients and also in animal subjects. This research was conducted by electronically searching PubMed, Scopus, Medline, and Cochrane Library databases from 2000 to December 2023.

3.2. Study Selection

All publications focusing on stress and pathogenic, clinic, and diagnostic aspects and therapeutic interventions in pediatric and adult patients with IBD were assessed. Also, in this narrative review, we included studies conducted on animals. The inclusion criteria used to extract relevant information included the following: clinical and preclinical/laboratory studies published in the English language in the last 23 years, with a sample size comprising more than 15 subjects. The most important data were summarized in this narrative review.

4. Stress and Gut Microbiota Brain Axis

The hypothalamic–pituitary–adrenal axis (HPA) represents the most important neuroendocrine system involved in the stress response of the host. The link between the perceived stressful stimuli and the physiological reaction to different types of stress is encoded by the HPA. The HPA, the autonomic nervous system (ANS), the CNS, the stress response, the GI corticotropin-releasing factor system (CRF), and the intestinal response (including the intestinal barrier, the luminal microbiota, and the intestinal immune response) are among the neural components that interact in the brain–gut axis [27].
The cholinergic anti-inflammatory system modulates the innate immunity action against different types of tissue alterations induced by infectious pathogens or hypoxia. It represents the efferent pathway of the inflammatory response, which is the neural feedback that regulates the inflammatory reaction. In the cholinergic anti-inflammatory system, through an anti-TNF-α, the action of the efferent vagus nerve may be a therapeutic target in IBD by a pharmacological, dietary, or neurostimulation strategy, according to animal studies [28,29]. The psychological requirements of patients with IBD are also highlighted by the psychophysiological susceptibility of these patients due to the potential presence of any mood disorders, distress, increased perceived stress, or maladaptive coping mechanisms. There is growing evidence that stress or other negative psychological traits may have an impact on the disease course; thus, clinicians need to talk to patients about these difficulties.
An organism’s reaction to a request from the environment is stress. Stress is a physiological response that can turn pathologic when there is an imbalance between the body’s ability to adapt and what it needs from its environment. This can cause functional, metabolic, and even lesion-related illnesses [30]. The typical mechanism by which stress results in an adaptation is the hypothalamic–pituitary–adrenal (HPA) axis. The main neuromediator of stress, CRF, when delivered directly into the brain, mimics the general endocrine, behavioral, autonomic, and visceral alterations brought on by stress in experimental animals [31].
Recent research [31,32,33,34] indicates the gut microbiota’s involvement in IBD. The neurological system and commensal, pathogenic, and probiotic microorganisms can communicate in both directions. Bacteria are capable of passing through the epithelial barrier during times of stress, triggering the mucosal immune response, and they can go to secondary lymphoid organs [32], activating the innate immune system. Mice exposed to a social stressor experience changes in the intestinal microbiota and an increase in the amount of cytokines in their bloodstreams; antibiotics counteract these outcomes [33]. Intestinal microbiota changes decrease resistance to intestinal pathogen-induced infection [34]. The results of these research studies demonstrate the method by which stress, the gut microbiota, and the immune response interrelate. The proliferation of bacteria is stimulated by the sympathetic nervous system’s release of catecholamines (norepinephrine) [35]. Stress-mediated changes might influence host susceptibility to infection and modify microbial colonization patterns on the mucosal surface. These modifications to host–microbe interactions could cause an impact on neuronal activity in stress-sensitive areas of the brain [36]. The microbiota brain–gut axis, which connects the gut and the brain, may be mediated by the intestinal bacteria. This is illustrated in Figure 1.
The ANS, alongside the HPA axis, modulates the efficiency with which the GI tract works. To control mucosal immune responses and other intestinal activities, such as nutrition absorption [37], the ANS is responsible for causing efferent signals to be transmitted from the CNS (brain and spinal cord) to the intestinal wall [38]. Afferent signals from the lumen of the intestine are also known to control behavior, sleep, and stress responses through enteric, spinal, and vagal nerve cells [39]. The enteric nervous system (ENS, the “second brain”), a component of the peripheral nervous system, primarily communicates with the CNS in a bidirectional pattern upon receiving inputs from the diet and gut bacteria [40]. The ENS can, however, also inherently innervate the gut in an autonomous way [41].
For the gut microbiota, the GI tract acts as a dynamic, local ecosystem. The gut microbiota is made up of around 35,000 different bacterial species, which are frequently divided into two major groups: Bacteroidetes and Firmicutes [42,43]. It is necessary for modulating processes associated with barrier function against pathogenic microorganism colonization, such as mucosal integrity [44], immunomodulation [45], and pathogen protection, in addition to its role in metabolism [46]. In recent times, preclinical, translational, and clinical studies [47,48] have indicated that modifications to the microbiome’s structural composition or function may play a crucial role in the occurrence of mental illness, including depressive-like behavior. According to some studies, the development of multifactorial chronic inflammatory illnesses, including IBD, has been strongly correlated with changes in the gut microbiota, indicating that dysbiosis is a significant component in both GI and mental health [49,50].
Short-chain fatty acids (SCFAs), such as butyric acid, propionic acid, and acetic acid, which are commonly observed to be reduced in mucosa and feces of individuals with IBD, are also produced by the gut microbiota through the fermentation of dietary fibers [51]. These metabolic products have been demonstrated to have a significant role in increasing epithelial cell proliferation [52], barrier function [53], and cellular metabolism, as thoroughly reviewed by Parada Venegas et al. [54,55]. By simulating G-protein coupled receptor signaling pathways, SCFAs have also been linked to the regulation of intestinal homeostasis and the prevention of pathogen colonization [56]. SCFAs are also known to have neuroprotective effects, which is pertinent. Gamma-aminobutyric acid, for instance, can influence behavior since it is an inhibitory neurotransmitter that has a role in anxiety and sadness [57]. Modifications in bacterial neurometabolites or bacterial cell wall carbohydrates are two more ways that the intestinal microbiota influences neuronal responses. These substances either directly affect primary afferent axons or cause the release of chemicals by epithelial cells that control neural signaling in the ENS [58].
Together, the complex interactions between the gut, microbiota, and brain enable intestinal and extraintestinal homeostasis, which regulates higher cognitive and affective processes as well as GI functions.
Early life stress can be related to admission to the Neonatal Intensive Care Unit (NICU). Many preterm infants are exposed to stress, pain, and complications of the GI system. One of the most dangerous GI neonatal emergencies is necrotizing enterocolitis (NEC), which is related to multiple risk factors. The most important is prematurity, which comes together with hypoxia, sepsis, abnormal colonization of the bowel, and the release of inflammatory mediators. These inflammatory mediators are set off by a triggering event linked to stress [59]. Early life stress can leave lasting effects. Some stressors may enhance growth and adaptation, but others may be innate to alter future health trajectories. For many preterm infants, early life exposure to persistent and intense stressors may become potentially toxic [60]. Recent studies have linked stress to the gut microbiome, leading to dysbiosis and suppressing the activation of the innate immune system in response to stress [61,62]. The gut colonization process is dynamic, depending on environmental factors. Preterm infants admitted to the NICU experience dysbiosis with an abundance of Gram-negative bacteria. There is a need to determine if stress is related to prematurity and antibiotic usage and, therefore, create an appropriate environment for bacteria abundance [60]. To optimize the management of NEC, it is necessary to identify critical diagnostic methods and their ability to determine the existence of future inflammatory bowel disease. Over the years, several studies have aimed to identify specific biomarkers [63,64,65,66]. A literature review was performed to update data on NEC biomarkers, which concluded that various proteins and products of metabolism can be used to determine NEC with modern technology. Nevertheless, future research is needed to determine non-invasive panels of high-value and diagnostic algorithms [67]. Long-term implications for NEC survivors include short bowel syndrome, cholestatic liver disease, and impaired neurodevelopment [68]. Since modern technology has arisen over the years, the link between NEC and the possibility of IBD development in the future has followed. Tremblay et al. used deep sequencing (RNA-Seq) to determine the gene expression profile in preterm infants diagnosed with NEC and non-NEC conditions. Analysis of the data indicated that the relevant functional pathways in preterm infants with NEC were associated with immune functions, such as altered T and B cell signaling, B cell development, and the role of pattern recognition receptors for bacteria and viruses. Genes strongly modulated in NEC neonates are significantly similar to those reported in CD, which is a chronic inflammatory bowel disease. The results of this cited study confirm that a large proportion of the significant functional pathways and phenotypes are common between NEC and CD and that some of the biomarkers used for diagnosing CD can be used for predicting NEC development in intensive care units [69]. This approach can be used as a point-of-care tool for diagnosing NEC or bowel inflammation, as demonstrated for lipocalin 2 and calprotectin [70,71,72].
Neonatal early-life stress is predominantly related to admission to the NICU. The NICU uses a patient-centered approach driven by protocols; notwithstanding, preterm newborns experience a significant level of stress from their surroundings since they must endure complex life-saving medical operations, protracted absence from their parents, and continuous and extreme stressors [60]. The vulnerability of newborns allows for the stress and complications associated with prematurity to also affect neurological development [73,74].
As stated before, the link between stress and gut microbiome has been proven. Questions have arisen regarding how these two can impact neurobehavioral development in preterm infants during NICU hospitalization. The gut microbiome is involved in the regulation of neurological, behavioral, and cognitive development [75,76,77]. Early alleviation and treatment of neurobehavioral abnormalities in preterm newborns can be made more accessible by identifying possible pathogens and understanding the pathogenic process of gut microbiota involved in neurobehavioral development.
Neurobehavioral outcomes are assessed using the NICU Network Neurobehavioral Scale (NNNS), which is a standardized score and includes, among others, stress, handling, and quality of movement [78]. It is shown that infants who had less acute stressful events during hospitalization had improved neurobehavioral outcomes [73,79]. Several gut bacteria were studied in terms of their link with compromised neurodevelopmental outcomes. Chen J et al. conducted a longitudinal study and identified eight gut microbiome bacterial communities associated with neurobehavioral profiles in early life. The vast amount of Enterobacteriaceae is linked to an increased NSTRESS score [79]. Another study suggested that Klebsiella overgrowth was associated with brain injury involving immunological alterations [75]. These findings suggest that targeted interventions positively impact developmental outcomes in infants.
The GI health of a newborn is an area of great interest; therefore, interventions are needed to establish a well-desired, healthy GI system. For digestion and nutrient absorption, newborns require the GI tract to mature structurally and functionally. A term newborn will have a normal neonatal adaptation with appropriate nutritional requirements. Preterm newborns are characterized by an immature GI system, which limits the utilization of enteral nutrition. In addition, factors such as stress, infections, and antibiotics contribute to impaired bowel function and gut–brain axis [80].
Antibiotic use in neonates admitted to the NICU is a common practice, which includes the most prescribed drugs in the NICU [81]. The gut microbiome is influenced by antibiotics and their associated side effects, which influence the early establishment of intestinal microflora [82,83]. The use of antibiotics within the first 2 weeks of life for preterm newborns is linked to an increased risk of late-onset sepsis, NEC, or death [84]. The randomized study REASON [85] was conducted to determine if antibiotics should be used in the first 48 h after birth and their effects on gut microbiome and inflammatory status. The results suggest that using antibiotics in the first 48 h of life has no long-term effect on the microbiome. Moreover, the microbiome diversity is recoverable. The REASON study also suggests that Bifidobacteria may influence GABA signaling in the brain. Thus, antibiotic use can interfere with the gut–brain axis [85].
Postpartum Gi colonization is influenced by additional factors such as delivery mode and feeding regime. Newborns delivered by cesarean section have a reduced diversity of gut microbiome compared with those vaginally delivered [86,87]. Delivery mode has long-term effects on dysbiosis, leading to autoimmune and metabolic disorders [88]. A factor of debate in neonatal care is the use of prebiotics and probiotics to regulate the microbiome. Dermyshi et al. conducted a systematic review and concluded that probiotic use was significant in preventing severe NEC, late-onset sepsis, and all-cause death in infants with very low birth weight [89]. There are some arguments against probiotic use based on knowledge of some conditions of preterm newborns, such as extremely low birth weight, the immaturity of the immune system, susceptibility to infections, and similarly, the conjecture of probiotic formulas and doses [90,91]. Bifidobacterium species are major colonizers of the infant gut, comprising about 70% of the gut microbial population while breastfeeding [92]. These species have become the standard bearers for probiotic formulations, considering their unique abilities to metabolize complex carbohydrates from human milk—human milk oligosaccharides (HMOs) [93]. A recently published review highlights that infants treated with probiotics have bountiful Bifidobacterium spp. independent of the probiotic formulation and reduction in potentially pathogenic bacteria [94]. Probiotics containing both Bifidobacterium and Lactobacillaceae can influence the preterm gut microbiome configuration while promoting the development of a microbiome that is more typical of term newborns [95].
After delivery, the gut microbiome is influenced by interactions between the mother, the newborn, and environmental factors. The feeding regime is one essential variable. Breastmilk is the optimal nutrition for a newborn. Nevertheless, not all newborns can be fed with their own mother’s milk. Cesarean section can delay the breastfeeding process [96,97]. In addition, the immature GI system of preterm newborns will delay the feeding process. Human milk composition influences intestinal immunological processes and digestion, including GI colonization. Human milk microbiota is the second source of microorganisms for an infant. Thus, early gut colonization with human milk is essential for developing the immune system [98]. Breast milk is to be recognized as the gold standard for neonatal nutrition. However, in some circumstances, breast milk is unavailable, and formula feeding is required. The modernization of milk formula is a continuous process to create the best alternative regime [99]. HMOs are now artificially synthesized to be added to milk formula. These complex carbohydrates abound in human milk, modulate microbial composition, impede pathogenic invasion, and influence the immune response [100]. The benefits of human milk extend to premature newborns through its potent trophic effect on the immature gut. This valuable effect leads to earlier full feeding and prevention of late-onset sepsis and NEC. A good feeding practice is one key component to decreasing the burden of prematurity [101]. The GI health of a newborn plays a crucial role in the short term as well as in the long term. As discussed, there are some potential interventions to alleviate the effects of early life stress on the GI environment. Notwithstanding, further research is needed to provide a relevant understanding of the complex interplay between early life stress and neonatal outcomes.
Early childhood has a relatively complex and unstable gut microbiota; thus, any change is likely to influence the intestinal immune system and predispose people to IBD. Antibiotics, birth control, and non-steroidal anti-inflammatory drugs (NSAIDs), for example, have been shown to increase the risk of IBD, most likely by changing the commensal flora and/or intestinal barrier [9]. More specifically, a meta-analysis revealed that antibiotics were more strongly linked to an elevated risk of newly developing CD than UC [102]. Accordingly, a search across numerous databases showed that, compared with people who were not exposed to the medicine, those who used oral contraceptives had a 24% and 30% higher chance of getting CD and UC, respectively [103]. Similarly, long-term use of large dosages of NSAIDs [104] led to the worsening of IBD [105], possibly by non-selective inhibition of the cyclo-oxygenase [106].
Table 1 presents the studies conducted on humans and animals that investigated the link between stress and gut microbiota.
As shown by these studies, appropriate physiological responses to stress and/or immunity are necessary for survival. As such, aberrant responsiveness can be detrimental to the host, leading to the development of chronic disorders, including IBD [132] and brain disorders [135].

5. Stress-Induced Alterations/Inflammation in the Gastrointestinal Mucosa

In individuals with inactive IBD, unfavorable life events, depression, and chronic stress increase the risk of relapse, according to recent research. It has long been suggested that psychological stress enhances disease activity in IBD. According to recent studies, stress-related changes in GI inflammation may be mediated by altered bacterial–mucosal interactions, altered HPA axis function, mucosal mast cells, and mediators like CRF.
The hypothalamus, amygdala, and hippocampus are three particularly intertwined brain areas involved in the complex integration of the stress response. Higher cortical structures as well as visceral and somatic afferents provide input to this network. The HPA axis and the ANS are two interrelated effector pathways by which they control the neuroendocrine stress response. Adrenocorticotrophic hormone (ACTH) is released from the anterior pituitary gland in response to the stimulation of CRF release from the hypothalamus by stress. Cortisol, the main glucocorticoid, is then secreted from the adrenal cortex as a result [136,137].
The hypothalamus’s direct descending neuronal routes to the pontomedullary nuclei, which regulate the autonomic response, are activated by stress. The adrenal medulla releases adrenaline and noradrenaline in reaction to stress by stimulating the sympathetic nervous system. The vagus and sacral nerves offer parasympathetic input to the upper gut and the distal colon and rectum, respectively, while sympathetic neurons of the sympathetic ANS also directly supply the entire gut. The ENS, the gut’s abundant nerve supply, and the efferent and afferent neurons of the sympathetic and parasympathetic ANSs communicate with one another. This network is known as the brain–gut axis. The 100 million neurons that make up the ENS control the GI tract’s motility, exocrine, endocrine, and microcirculation functions [136,138].
The lymph nodes, the mucosa-associated lymphoid tissue, bone marrow, spleen, and thymus all have intimate effector junctions formed by nerve fibers of the ANS with lymphocytes and macrophages. At the neuron–immune cell junction, some neurotransmitters, contained in the neurons of the ENS and ANS, including catecholamines, vasoactive intestinal peptide, angiotensin II, neurotensin, somatostatin, and substance P (SP), can affect lymphocytes, macrophages, neutrophils, and other inflammatory cells [136,138].
Glucocorticoids, which are released from the adrenal cortex in response to ACTH from the pituitary gland, primarily depress the immune system when present in high amounts. They promote the production of anti-inflammatory proteins like IL-10, IL-1 receptor antagonists, and lipocortin 1 [139]. Glucocorticoids have an inhibitory effect on the transcription factors AP-1 and nuclear factor-kB, which in turn inhibits the production of several inflammatory chemokines and cytokines, including TNF-α, IL-1, and IL-6. T cells and eosinophils are two examples of inflammatory cell types that glucocorticoids also encourage to undergo apoptosis. On the other hand, cortisol has an immunostimulatory impact in lower amounts [138,140,141,142].
The complexity of psychological stress’s effects on the body’s immune and inflammatory systems depends on the stressor’s length and degree. Affected systemic immunological and inflammatory function is linked to both acute and chronic stress, which may be relevant to the pathophysiology of IBD [143].
Chronic persistent stress, such as that brought on by unfavorable life circumstances, results in a protracted rise in cortisol over several days, which is typically accompanied by immunosuppression. Reductions in macrophages, CD8+ lymphocytes, and NK cells have all been linked to divorce, bereavement, and depression [143,144,145,146]. Chronic psychological stress, however, has also been linked to subclinical increases in inflammation, in addition to immunosuppression. Serum C reactive protein levels have been reported to be elevated in individuals with depression as well as middle-aged and elderly patients with lower heart rate variability, a marker of chronic stress, and increased sympathetic tone [147,148].
Experimental stress tests and real acute stress both stimulate the sympathetic nervous system acutely and result in an almost immediate rise in adrenaline and noradrenaline levels. Cortisol levels then increase, but these shifts only last for a short time. This method of stimulating the stress axes has been linked to improved immunity. Inflammatory cytokines, which are known to play a significant role in the pathophysiology of IBD, are produced by whole blood in greater quantities and at higher serum levels. Additionally, it has been demonstrated that acute stress can quickly redistribute the lymphocyte population and result in leucocytosis in both healthy individuals and people with UC in the remission phase. Also, there is an increase in the proportion of NK cells and CD8+ cytotoxic T cells and a commensurate rise in their cytolytic activity [136,143,149].
Experimental stress has been demonstrated to promote platelet activation, as measured by aggregation and production of inflammatory mediators and platelet-dependent thrombin generation in healthy participants, as well as in patients with UC in remission. Patients with IBD have increased levels of platelet activation in their blood, which may play a role in pathogenesis by promoting the production of thrombi and microinfarcts as a result of microvascular ischemia. Instead of aspirin, beta-blockers may be able to suppress the stress-induced activation of platelets, indicating that sympathetic stimulation is an important factor in the process. Acute experimental psychological stress also causes platelet–leucocyte aggregation formation; this factor is elevated in IBD patients and may make it easier for leucocytes to extravasate to specific regions of inflammation [136,150,151,152].
It has been suggested that abnormal mucosal immunity to the intestinal microbiota is the cause of IBD. The growth and maintenance of secretory cells depend on the transcription factor X-box-binding protein-1 (XBP1), which is associated with JNK activation and an essential part of the endoplasmic reticulum (ER) stress response.
It was demonstrated that XBP1 deletion in intestinal epithelial cells (IECs) causes spontaneous enteritis and enhanced vulnerability to produce colitis as a result of both Paneth cell insufficiency and IEC hyperactive responses to the IBD-inducing agents TNF-α and flagellin [153,154]. As a result, intestinal inflammation can only result from XBP1 aberrations in IEC, establishing a link between the development of organ-specific inflammation and cell-specific ER stress.
Inflammation may be brought on by a stressful external setting in cells with high secretory activity. If true, cell-specific XBP1 deletion that induces ER stress in vivo may result in organ-specific inflammation and offer a molecular explanation for the onset of proinflammatory illnesses. The intestinal epithelium contains four highly secretory cell lineages that are exposed to high concentrations of exogenous antigens: goblet and Paneth cells, absorptive epithelium, and enteroendocrine cells, that descended from a common, continuously regenerating intestinal epithelial stem cell [154,155].
The studies showed that spontaneous enteritis results from ER stress induction in intestinal epithelium caused by tissue (and cell type)-specific disruption of XBP1 because XBP1-deficient IECs are unable to generate antimicrobial activity and respond suitably to inflammatory signals in the local environment. The XBP1 gene locus on chromosome 22q12.1 contains some single nucleotide polymorphisms that increase the risk for CD and UC, establishing the ER stress pathway as a common genetic contribution to IBD [154,155].

6. The Role of the Nervous System as an Immune Modulator in Patients with IBD

The brain–gut axis is an intricate, bidirectional system that includes several connections between the GI tract, the autonomous nervous system, and neuroendocrine pathways. This network may have an impact on the emergence of functional GI disorders like irritable bowel syndrome (IBS), gastroesophageal reflux disease, and IBD [8,156,157].
The pathogenesis of IBD is thought to be influenced by psycho–neuro–endocrine–immune regulation via the brain–gut axis. The stress system (the HPA axis), the ANS, the CNS, the (GI) CRF system, and the intestinal response (which includes the intestinal barrier, luminal microbiota, and the intestinal immune response) are among the neural components that interact to form the brain–gut axis [157,158].
It is crucial to investigate both the drivers of the immunoinflammatory response as well as the peripheral mediators of inflammation (cellular components and their byproducts) to comprehend the etiology of IBD. For IBD, the gut microbiome has recently drawn more attention as an essential component of this process. To date, IBD therapy has focused on treating the phenotypic manifestation of IBD; however, psycho-neuroimmune modulation may be the platform that connects the human experience, mental state, gut microbiome, and immune response [158,159].
It is known that the brain system regulates immune function, and this ability could be used to inhibit the immune system in IBD. Since vagus nerve stimulation (VNS) was demonstrated to diminish local and systemic inflammation in animal models of endotoxemia, arthritis, and colitis, the vagus nerve and its primary neurotransmitter acetylcholine have received particular attention [160,161,162,163,164].
VNS decreases inflammation, but the exact mechanism by which it does so is still being investigated. It is unclear if the vagal efferent nerves genuinely innervate mucosal cells, despite the intestine being highly innervated. This might point to a function for different kinds of nerves. It has also been shown that a wide range of neurotransmitters, including ACh, as well as nitric oxide, adrenaline, norepinephrine (NE), and a huge number of neuropeptides that function as immune modulators, may act directly on a variety of immune cells in the gut [162,163,164].
The autonomic nerve system, which operates autonomously in that its actions are not directly under conscious control, regulates important digestive tract functions like motility, secretion, and vasoregulation. Based on anatomy and neurotransmitter activity, it is categorized as sympathetic and parasympathetic branches and reflects the extrinsic regulation of the gut. The intrinsic neurons of the ENS are found within the wall of the GI tract, and the sympathetic and parasympathetic systems have their origins in the CNS (with cell bodies in the brainstem and spinal cord). The ENS is a unique component of the CNS that can function on its own or in reaction to external signals coming from sympathetic and parasympathetic nerves. The enteric ganglia, which are clusters of nerve cells that form the ENS, generate nerve fibers that innervate effector tissues like the gastroenteropancreatic endocrine cells, blood vessels, and the intestine’s muscular layer. The ENS comprises the myenteric plexus and the submucosal plexus [160].
ACh is one of the neurotransmitters found in the ENS, which is thought to control gut immunity. Neuropeptides, acting as crucial mediators between the nervous system and neurons or other cell types in the effector tissues, are also a part of the crosstalk between the gut and nervous system. These tiny proteins play a crucial role in multimodal neural communication, including SP, vasoactive intestinal polypeptide, calcitonin-gene-related peptide, neuropeptide Y, somatostatin, serotonin, and CRF.
When the cholinergic anti-inflammatory pathway was originally discovered, the impact of neurons on gut inflammation first came to light. According to one theory, the neurological system uses this pathway as a reflex mechanism to regulate abnormal, increased immune responses [165,166].
Since inflammation of the gut also affects the nerves and their adrenergic activity, there is a reciprocal relationship between the SNS and the inflamed intestine. In the inflamed colonic mucosa of IBD patients as well as in several colitis animal (mouse) models, sympathetic innervation is noticeably reduced. A loss of tyrosine hydroxylase (TH) nerve fiber was shown in patients with IBD, where TH is the rate-limiting enzyme for the synthesis of epinephrine and NE. Additionally, it was discovered that proinflammatory SP+ fibers predominated noticeably [167].
It makes sense that a decrease in sympathetic neurotransmitter levels would follow the loss of sympathetic nerves in inflamed tissue. CD patients had markedly lower NE levels than healthy controls. This is corroborated by the observation that in inflamed tissue, the release of NE from sympathetic nerve terminals is constrained. As NE-negative immune regulation is reduced, inflammation-induced inhibition may increase the chronicity of the inflammation. Furthermore, the primary function of sympathetic nerves is vasoregulation, which is distinct from the SNS’s anti-inflammatory function [168,169].
When these nerves are lost, blood flow is hampered, which may contribute to maintaining the inflammatory environment. According to the concentration of neurotransmitters and neuropeptides (which is dependent on their release and the presence of sympathetic nerves), the quantity and accessibility of receptors, the receptor affinity, and the timing of sympathetic activity, the SNS has opposing proinflammatory and anti-inflammatory functions. There is disagreement over how the SNS and the inflammatory environment contribute to the persistence of inflammatory processes [168,169]

7. Current Publications on the Interplay between Stress and Psychosocial Disorders in Children and Adults with IBD

Previously conducted systematic reviews have confirmed that individuals diagnosed with IBD exhibit a decreased quality of life in comparison with the general population [170]. This decline in quality of life is more pronounced during active phases of IBD rather than inactive periods, and it is particularly notable in individuals with CD as opposed to UC [171]. However, the quality of life tends to improve as the disease progresses over time. Anxiety and depression commonly co-occur as comorbidities in individuals with IBD and have a reciprocal relationship with the disease [172]. Nevertheless, the precise nature of this relationship between anxiety, depression, and IBD has yet to be established due to the limited availability of prospective study designs [173]. A recent meta-analysis investigating the potential causal link between anxiety, depression, and the exacerbation of symptoms in IBD yielded inconclusive findings [174]. Psychological interventions have only demonstrated modest beneficial effects on the quality of life and depression experienced by individuals with IBD, and further clinical trials are necessary to ascertain their impact on disease activity [175]. Consequently, there remains a critical need for an enhanced understanding of the psychological factors influencing individuals diagnosed with IBD.
According to Lazarus and Folkman, stress is defined as a complex interaction between an individual and their environment, where the individual perceives the demands and challenges as overwhelming, surpassing their available resources, and posing a threat to their overall well-being [176]. This relationship is influenced by various factors, including personal and environmental factors, the nature of the stressor itself, how it is perceived and evaluated, and its immediate and long-term effects. Emotions play a significant role in the appraisal of stress, as recognized in both psychological [177] and physiological [178] models, leading some researchers to use the terms stress and distress interchangeably.
Stress is believed to activate the HPA axis, which has been implicated in the inflammation of the GI system [136]. Changes in this axis, such as alterations in the neuroendocrine-immune system, can contribute to the likelihood of disease exacerbation [179]. Additionally, stress can lead to behavioral changes, including nonadherence to medication [180], poor dietary choices [181], and alcohol consumption [182], all of which increase the risk of disease flare-ups. Measures of perceived stress may be valuable in identifying individuals at risk of relapse and indicating the need for intervention when used alongside disease activity indices.

8. Anxiety and Depression in Children and Adults with IBD

Psychiatric disorders have a significant negative impact on various aspects of IBD. Depression has been linked to an elevated risk of disease relapse and poorer treatment response [183,184,185,186]. Similarly, anxiety is associated with a higher likelihood of undergoing surgery, lower adherence to medication, and a diminished quality of life [183,187,188]. In a comprehensive study involving multiple institutions, after accounting for confounding factors, the presence of comorbid depression and/or anxiety was associated with a 28% increased risk of surgery in individuals with CD. Moreover, it was correlated with a higher number of colonoscopies and an increased probability of utilizing immunomodulators as part of the treatment regimen [189].
Although psychiatric disorders have been observed to manifest after an individual is diagnosed with IBD [190], there is evidence suggesting that these disorders may predate the diagnosis of IBD by several years [191,192]. Most studies investigating this relationship have primarily focused on depression or anxiety, placing greater emphasis on the prevalence rather than the incidence of psychiatric disorders. However, understanding the incidence of these disorders is crucial for unraveling their etiology. Considering the detrimental impact of comorbid psychiatric disorders on the progression of IBD, it is vital to thoroughly examine the burden of psychiatric comorbidity in individuals with IBD.
The highest occurrence of CD is typically observed during the third decade of life, while UC incidence begins to rise in the same decade and maintains more consistent incidence rates across different age groups [193]. However, IBD typically affects individuals during the crucial period of social and career development [194]. With an increased occurrence of psychiatric disorders following an IBD diagnosis, there is potential for an enduring burden of mental health issues in individuals with IBD. Limited research has been conducted specifically on the management of mental health disorders in individuals with IBD [195]. Considering the potential significance of the brain–gut axis in the pathobiology of IBD [157], it is possible that the inflammatory state of the gut can influence brain function and mental well-being. Therefore, assuming that treatments for psychiatric disorders in the general population will be equally effective for individuals with IBD would be an oversimplification. While the mental health effects of corticosteroids are well-known, there is limited information regarding the potential harms or benefits of mental health associated with various effective biological therapies used for IBD. Consequently, there is a need for extensive research to optimize therapy for individuals with IBD who develop a psychiatric disorder.

9. Mendelian Randomization Studies Evaluating the Causal Associations between IBD and Psychological Conditions

Compared with the general population’s prevalence (3.4%), anxiety is a common comorbidity in people with IBD, ranging from 19.1% to 35.1% [172,196,197]. Because of the potential pathophysiological pathways mediated by the gut–brain axis, there has been a lot of interest in the relationship between anxiety and IBD, or vice versa.
A few observational studies have looked into the temporal relationship between anxiety and IBD and theorized a bidirectional relationship between the two conditions. Anxiety may be more common in IBD patients than in people who only seek medical check-ups [198,199,200].
Also, throughout a 10-year follow-up period, newly diagnosed IBD patients experienced an increased prevalence of anxiety (incidence rate ratio: 1.39) in comparison with matched control persons [199]. Observational studies have demonstrated that those with anxiety are more likely to have IBD, but the reason is unknown [200]. During 6.7 years of follow-up, cohort research revealed a greater prevalence of IBD in patients with newly diagnosed anxiety than in control persons [201].
Using bidirectional Mendelian randomization methodology, He Y and collaborators conducted a new study to examine the causal connection between IBD and anxiety. MR is a genetic technique that determines the causal relationship between an exposure and an outcome via genetic variants found in genome-wide association studies, typically utilizing single nucleotide polymorphisms (SNPs) [196]. The findings indicated that a higher incidence of anxiety could be related to a genetic vulnerability to UC (odds ratio: 1.071 (95% confidence interval: 1.009–1.135), p = 0.023). However, anxiety was not related to a genetic predisposition to CD. But neither UC nor CD have been related to a genetic predisposition to anxiety. This cited study demonstrated the strong relationship between anxiety and genetic susceptibility to UC, emphasizing the value of early screening and effective treatment for anxiety in UC patients [196].
It is still unclear how exactly UC or CD causes anxiety from a physiologic standpoint. Some research suggests that the inflammation-regulated gut–brain axis can impact neuronal development and ensuing behavioral traits [156,202].
The blood–brain barrier allows for circulating leukocytes and cytokines to enter the brain, where they can cause neuropsychiatric diseases [156]. The exact biochemical mechanisms by which UC influences the development of anxiety are still unknown, even though He Y and collaborators’ investigation examined the causal association between IBD and anxiety [196]. For example, it is unclear how the gut–brain axis plays a part in this process. Therefore, to fully understand the molecular mechanisms, additional basic and clinical research is required to identify important regulators and pathways.

10. Quality of Life among Pediatric and Adult Patients with IBD

IBD significantly impacts quality of life and comes with personal, emotional, and social burdens. Numerous studies have shown that health-related quality of life (HRQOL) is impaired in patients with IBD compared with the general population. While disease activity and severity impact physical and psychological HRQOL, patients may also suffer from psychological problems during clinical remission. Impaired quality of life can affect the employment, family planning, and personal life goals of people with IBD. Improving quality of life requires a multidisciplinary approach that, together with adaptive coping mechanisms, helps to manage illness perceptions and reduce psychosocial burden. HRQOL is a concept that encloses those aspects of overall quality of life that have a demonstrable impact on physical or mental health. Measuring HRQOL can help improve connections between medical management and personal life and guide strategic plans [203]. Measuring and assessing the disabilities associated with IBD and the impact on the quality of life of those affected is critical to understanding the often-hidden burden that this disease places on those affected and society. This section of this review describes the impact of IBD on patients’ quality of life.
Several studies showed that persons diagnosed with IBD have significantly lower HRQOL in comparison with the general population, with an emphasis on severe disease activity [204,205,206,207,208]. Wilburn et al. conducted a study focused on patients’ appraisal of the disease rather than symptoms and medical management. It emphasized that IBD affects the daily routine and the need for fulfillment–nutrition, hygiene, self-esteem, attractiveness, and intimacy. The respondents indicated that their diets were restricted, and small portions replaced large meals. They reported hygiene concerns. It was common for them to carry deodorants, spare clothing, and wet wipes. Suffering from the disease meant they had to orient their lives according to the location of toilets. Finding the nearest public toilet became the first task when arriving at an unfamiliar location. Respondents reported being cautious about getting emotionally close to people because they found it difficult to initiate relationships [209]. IBD influences the need for fulfillment by affecting self-image, attitude toward life, and well-being [210]. This includes traveling as well. Foreign travel for people with IBD increases the risk of morbidity due to exacerbations, infectious diseases, and a lack of healthcare providers abroad. It is recommended that travelers seek thorough pre-trip counseling and vaccinations to ensure they are equipped with the appropriate information and resources to stay healthy during their journey [211,212]. A diagnosis of IBD limits travel for many patients; however, they should be counseled about travel behavior and seek medical advice before traveling abroad [213].
IBD is a chronic disease that alters individuals’ quality of life due to symptoms, medical management, depreciation of body image, psychological disorders, and long-term treatment. The sexual functioning of patients with IBD is a constant concern. The first report of sexual dysfunction in IBD dates back to 1978, which reported less sexual intercourse or even ceasing sexual intercourse [214]. Since then, several studies have emphasized that sexually dysfunctional rates are higher in patients with IBD than in the general population, affecting women more than men [215,216]. A meta-analysis regarding sexual dysfunction in IBD was performed in 2019, which confirmed that prevalence is higher in patients with IBD, reporting a relative risk of 1.41 for SD in men and 1.76 for SD in women [217]. Sexual dysfunction is a common yet little-known problem in patients with IBD. The control of IBD activity, together with mental and sexual health, is relevant to the well-being of individuals with IBD. For these patients, it is recommended to perform an early investigation of sexual dysfunction to improve their quality of life [218].
HRQOL is a key indicator of adjustment in adolescents with chronic diseases and has been used as an outcome in clinical trials and as a marker of treatment efficacy. HRQOL in adolescents with IBD has recently received considerable attention, given the physical and emotional demands of treating the disease. The treatment of IBD can include dietary changes, medication management, and surgical interventions [219]. Although medical interventions can relieve disease-related symptoms, they can also lead to adverse side effects impacting an individual’s HRQOL. In addition, the treatment of IBD and its symptoms can lead to increased self-consciousness and affect school and social life [220,221]. Youth with IBD are at risk for lower HRQOL than typically developing youth, including a higher risk for psychosocial, physical, and academic impairments. Kunz et al. conducted a study that compared youth and parent-proxy reports of HRQOL among youth with IBD to published comparison group data. It emphasized that youth with IBD reported lower psychosocial functioning than the healthy comparison group, higher physical and social functioning than the chronically ill group, and lower school functioning than all published comparison groups. Specific factors of IBD that may interfere with school functioning include frequent use of the bathroom, limited participation in physical classes, and embarrassing cosmetic side effects associated with having the disease or taking certain medications. Interestingly, even though more than half of this sample had an inactive illness, adolescents still reported deficits in academic performance compared with adolescents with an acute illness, suggesting that impairments in academic performance may persist even after symptoms of the illness have resolved. Even when the illness is dormant, the social stigma associated with the disease is perpetuated by frequent toilet use, visits to the nurse, and the need to take medication during the school day [222]. In addition, family stress levels can also affect the HRQOL of the youth. The study conducted by Gray et al. showed that high levels of parental stress were associated with lower HRQOL among adolescents with IBD [223]. The burden of IBD also takes its toll on the family. Knez et al. reported in their study that parents of children with IBD reported lower psychological and physical health than parents of healthy children [224].
Understanding how IBD impacts an individual’s day-to-day life means defining disability as a limitation in the ability to engage in usual activities. Work capacity was the most common IBD-related criterion for disability. However, more than this metric is needed because it does not capture all important aspects of the burden of this disease. In addition, in a relapsing–remitting disease such as IBD, the inability to work may be temporary and difficult to resolve. Although studies have demonstrated increased unemployment, sick leave, and disability retirement in IBD patients, most patients can work for many years after a diagnosis of IBD. They have lower productivity and fewer working hours than healthy controls, resulting in economic losses to individuals and society. However, the limitations are building interpersonal relationships, life activities, and mental well-being [225,226].

11. Lifestyle Factors and Psychological Stress in IBD Patients during the Coronavirus Pandemic Period

The coronavirus disease (COVID-19) is an infectious disease caused by severe acute respiratory syndrome Coronavirus-2, first reported in Wuhan, China, and soon spreading worldwide, affecting millions of people. Therefore, the interest of this review was also to examine the effect of the COVID-19 pandemic on lifestyle factors and psychological stress in patients with IBD.
For instance, Yu Nishida et al. performed a retrospective study of patients with IBD that compared the lives of 451 patients with UC or CD before and during lockdown. As illustrated in Figure 2, some aspects of their lives were affected. It appears that exercise time, walking time, and working time decreased while sleep time increased. The pandemic has created, among many other problems, anxiety, depression, and stress. In terms of psychological stress, IBD patients experienced a significant increase in stress from being at home and from being unable to exercise, while IBD itself remained equally stressful [227].
Regarding age, elderly patients, compared with younger, patients experienced less stress during the pandemic, and in terms of gender differences, men were found to be more affected [227,228]. Moreover, the pandemic has affected eating habits, leading to involuntary weight gain in both children and adults with IBD [228].
As a chronic and debilitating disease, IBD requires regular check-ups, but during the pandemic, hospitals were ordered to care for patients with COVID-19 rather than IBD patients [228], yet treatment adherence did not decrease [227]. On the other hand, limited access to medication and specialized treatment led to an increased risk of exacerbations [229].
Moreover, Conti C et al. conducted a cross-sectional study to determine the extent to which the quality of life of IBD patients was affected. The study included two groups of IBD patients, one recruited before the pandemic and the other during the COVID-19 outbreak, with a total of 902 people enrolled. Disease activity, somatization, and quality of life were investigated. Almost half of the patients reported symptoms, predominantly UC patients, and, as expected, a higher level of anxiety, depression, somatization, and implicitly a lower quality of life [229].
The fear of being admitted to a hospital during the pandemic has led to delays in the diagnosis and treatment of IBD flares, thereby increasing the need for urgent surgery and the risk of medical treatment failure [230].
The prevalence of COVID-19 infection in patients with IBD appears to be lower than in the general population [231]. Children with IBD are less affected by COVID-19 than adults with IBD, but they play a significant role in the spread of the virus. Therefore, strict hand hygiene remains essential to prevent infection in any category of patients [232].
Regarding IBD patients infected with COVID-19, a study by Sansotta N. et al. in Lombardy, the region of Italy that was most affected by COVID-19, points out that children with IBD under immunosuppressive therapy are not at greater risk of contracting the virus. Even if they do become infected, the symptoms are mild, so they do not require treatment adjustment or hospitalization to control IBD. However, IBD patients with comorbidities or treated with corticosteroids may develop a severe form of COVID-19. Still, no concrete evidence supports a direct link between them [233].
A study by Turner et al., using the Surveillance Epidemiology of Coronavirus Under Research Exclusion for Inflammatory Bowel Disease (SECURE-IBD), examined the first eight pediatric patients with IBD and COVID-19. These patients showed only mild symptoms such as fatigue, low-grade fever, and cough. Despite undergoing immunomodulatory therapy, none of the patients required hospital admission, and there were no reported deaths [234].
A group of 209 children from 23 countries with IBD who contracted COVID-19 was analyzed by Brenner J. et al. According to the study, only 7% of the children required hospitalization, which was due to a combination of pre-existing medical conditions, active IBD disease, GI symptoms, or the use of certain medications such as sulfasalazine/mesalamine and steroids. Two children developed a secondary infection and multisystem inflammatory syndrome and were admitted to the pediatric intensive care unit (PICU), where they required mechanical respiratory support. However, with appropriate treatment, the prognosis was good. Moreover, the study found that TNF-α was linked to a lower risk of hospitalization in these children [235].
Patients undergoing IBD treatment may experience lowered immunity as a side effect. This has raised concerns about whether the treatment should be modified. However, studies have shown that despite the increased risk of infections caused by lowered immunity, it is not recommended to interrupt treatment because of the high risk of disease activation [232,233,234]. Additionally, biological treatment may help prevent the cytokine storm associated with SARS-CoV-2, thereby having a protective effect and reducing the likelihood of patients contracting the disease by up to 5 times [236]. Nevertheless, some precautions should be taken with corticosteroid therapy. It is recommended to avoid starting a new steroid therapy and to decrease the dose of prednisone if it exceeds 20 mg/day. In children with CD, thiopurines may be used as an alternative to corticosteroids. Still, they are not without adverse effects as they increase the risk of viral infections including varicella-zoster virus, cytomegalovirus, and Epstein–Barr virus [231,237].
According to the British Society of Gastroenterology, vaccination against COVID-19 is safe, although there is a possibility of a lower immunological response in patients undergoing immunosuppressive therapy [231].
The negative effects of the pandemic on patients with IBD persisted even after its end. This condition is known as long COVID and is recognized as a health issue with a detrimental impact on patients. The most prevalent symptom is asthenia, and women seem to be more susceptible to this condition [238].
Therefore, the COVID-19 pandemic has certainly been a burden on the healthcare system, with still important consequences, especially for patients with IBD.

12. Association between Psychological Stress and IBD Outcomes/Relapses

Several studies have used their resources to examine the relationship between psychological stress and IBD outcomes/relapses. It turns out that stress is a trigger of the HPA axis, which, as mentioned before, affects the GI tract.
A systematic review by Black J. et al. demonstrates a link between stress and IBD disease activity. Although different subtypes of stress have been analyzed, perceived stress seems to be the key factor involved in exacerbating IBD. Therefore, measuring perceived stress using the Perceived Stress Scale as an assessment tool may predict an exacerbation episode [239].
A prospective population-based study reported that a major stressful event can cause an activation of the disease in the following 3 months. The most stressful situations turned out to be related to family, followed by work, school, and financial stress. It also established a strong inverse association between perceived stress and disease outcome [240]. Regarding UC, it has been observed that long-term perceived stress can triple the risk of UC flares [241].
Moreover, three prospective observational studies claimed that there is a psychological basis behind the course of the disease. This argument was supported by the fact that patients with UC in clinical and endoscopic remission, following repeated stressful events, triggered a relapse of the disease [242]. For instance, Jennifer L et al. showed that a better psychological adjustment can reduce perceived stress and therefore decrease the number of hospitalizations due to IBD flares [243].
Furthermore, Bonaz and Bernstein strongly suggested that there is a bidirectional relationship between active disease and stress, as being stressed can trigger symptomatic disease, and being symptomatic can exacerbate or even encourage the state of stress [157].
In addition, Sunavcky A. et al. aimed to study the psychosocial mediators underlying the relationship between illness severity and perceived stress. Thus, as shown in Figure 3, four psychosocial variables (catastrophizing, illness stigma, illness uncertainty, and illness shame) were investigated. Except for illness stigma, all the other variables were equally strong positive mediators between IBD severity and perceived stress [244].
Chronic psychological stress accelerates the progression of IBD and also leads to behavioral comorbidities such as anxiety and depression [245]. It is known that quality of life is affected in patients suffering from a chronic disease and that stress can even cause behavioral changes such as alcohol consumption, non-adherence to treatment, or a poor diet, thereby increasing the risk of IBD relapses [239].
Therefore, high levels of stress and its effect on IBD progression is still a topic of great interest, as it is difficult to determine whether psychological stress is a predisposing and contributing risk factor to the disease or the result of a chronic debilitating disorder such as IBD [246].

13. Interventions That Contribute to Stress Reduction in IBD

IBD is a chronic, relapsing pathology of unknown etiology, which carries the burden of affecting millions of people worldwide [247]. The pathophysiology of IBD is multifactorial, relying on an interaction between genetic factors, the microbiome, the immune system, gut mucosal integrity, and environmental triggers. The link between the immune system, nervous system, and psychological processes plays an essential role in IBD. Psychological stressors affect the gut through the increased production of proinflammatory cytokines, activation of macrophages, and TNF-α via the HPA axis [181,248]. IBD and psychological disorders share common proinflammatory pathways, which explains the association between flare-ups of IBD in individuals with depression and the poorer outcomes of IBD patients diagnosed with mental illnesses [249]. In addition to this is the gut–brain axis, defined as a bidirectional network between the nervous system and the intestines. The main component of this axis is the ANS, in which the vagal nerve plays the main role. It is well known that stress inhibits the stimuli of the vagal nerve, and, therefore, its anti-inflammatory properties result in negative effects on the GI tract [250]. Multiple studies showed that stress, anxiety, and depression are trigger factors for relapsing in IBD, and additionally, IBD patients are at higher risk of developing depression than healthy individuals [8,172,186,193,251].
Recent research suggests that psychological interventions can improve the treatment effect of GI diseases, thus improving patients’ quality of life [252,253,254,255]. Cognitive behavioral therapy (CBT) is considered the most effective psychotherapy for an IBD multidisciplinary approach among various psychological interventions. It can reduce the rate of psychological disorders and improve the quality of life of IBD patients [256,257,258,259]. Furthermore, Jordan et al. emphasized in their study that CBT significantly reduced anxiety and low mood scores and increased quality of life scores. It also reduced disease activity in the specific group of IBD patients who also experienced anxiety and low mood [257]. Similarly, treatment with CBT was associated with significantly greater improvement in depressive severity in the overall sample of young people with depression and CD and correlated with a significantly greater improvement in pediatric CD activity in the subgroup with active IBD [260]. However, a recent systematic review has shown that the positive effects of CBT regarding improving the mental state of IBD patients are not long-term. There are insufficient data to determine if CBT improves disease activity and reduces inflammation [261].
Another type of psychological intervention to reduce stress is mindfulness activities. This intervention is shown to have a positive role in stress level reduction and improving the quality of life in patients with IBD [262,263,264]. Additionally, mindfulness-based therapy reduces inflammation related to IBD by decreasing the levels of inflammatory biomarkers, such as interleukin-6, fecal calprotectin, and C reactive protein [264,265,266].
The multidisciplinary approach to IBD includes various psychotherapy interventions, such as gut-directed hypnotherapy, breath–body–mind workshops, advanced combination treatment, and psychological counseling. Gut-directed hypnotherapy is associated with improved GI function and may work through immune-mediated pathways in chronic diseases. It is demonstrated to have a significant effect of psychosocial therapy on extending clinical remission in patients with UC [267]. A recent randomized controlled trial showed that hypnotherapy was not superior to standard medical therapy in patients with IBD in remission with IBS-type symptoms [268]. Gut-directed hypnotherapy is an increasingly used therapy for patients with IBS and IBD. However, it remains to be investigated whether it can be used for first-choice adjuvant therapy [269]. Combining educational and psychological counseling with CBT improves a patient’s quality of life in both emotional and social functions [270]. Gerbarg et al. demonstrate that breath–body–mind workshops for patients with IBD are linked to significant enhancement in psychological and physical symptoms and higher quality of life scores [271]. Mind–body interventions can include participating in yoga classes related to reducing stress and increasing the ability to manage symptoms [272,273,274].
Potential adjunctive therapy involves combining psychotherapy with antidepressant medication. It was demonstrated that using serotonin–noradrenaline reuptake inhibitors, selective serotonin reuptake inhibitors, and tricyclic antidepressants has a positive influence by alleviating psychological and physical symptoms in patients with IBD and disease activity. Additionally, antidepressants improve sleep quality and chronic pain [275,276,277]. Despite the potential adverse effects of the medication, evidence suggests that antidepressant medication has a significant role in improving patients’ quality of life and mental health, notwithstanding the management of the disease [201,278].

14. Future Perspectives Regarding the Interaction between Stress and Inflammation

14.1. miR-129-5p—A Significant Controller of Different Pathways

In the past few years, research teams have extensively investigated the role of RNA molecules, particularly microRNAs (miRNAs), in the oncogenic processes related to digestion [279,280]. Cellular apoptosis exhibits tissue specificity, while miRNAs emerge as genuine biomarkers of the tumorigenesis phenomenon [279,281]. The importance of miRNAs in pathology is notable, given their responsiveness to stress and altered expression patterns as diseases progress. Extensive research has focused on understanding the roles and modifications of miRNAs in cancer development and progression, with potential implications for significant clinical and therapeutic advancements in molecular research. Additionally, in vitro studies highlight miRNAs as promising candidates for molecular replacement therapy, offering potential avenues to hinder cancer progression, prevent lymph node metastasis, and induce tumor cell apoptosis [282,283]. Furthermore, every investigation into miRNAs advances our understanding of the molecular mechanisms underlying oncogenesis and neurodegenerative disorders. Recent research underscores the significance of exploring miRNAs and their target genes for their prospective contributions to personalized treatment strategies across diverse diseases [279,282,283].
Given that circulating miRNAs serve as significant regulators in intercellular communication and exhibit stable expression patterns in both tissues and biological fluids, their clinical significance deserves highlighting. MiRNAs, non-coding molecules comprising 21 to 23 nucleotides, have garnered attention due to their presence in body fluids. Recent research underscores their potential as disease biomarkers for screening, monitoring disease progression, and predicting therapeutic outcomes [279]. Recent research [279,284] has underscored the significance of miRNAs in the pathogenesis of numerous diseases. It has been observed that a single miRNA can regulate multiple genes, while conversely, a single gene can be targeted by multiple miRNAs [284].
Post-transcriptional mechanisms allow for a specific miRNA to regulate up to roughly 60% of the protein-coding genes. Moreover, miRNAs are involved in vital cellular functions such as cell growth, homeostasis, apoptosis, and cell migration. In the realm of brain development, a solitary miRNA may participate in multiple processes, including synaptic formation and neural development. Contemporary molecular research is centered on identifying clinical alterations resulting from miRNA dysregulation to tailor targeted treatments for various diseases [279,280,282,284]. Research indicates that miRNAs play a pivotal role in neurological disorders by modulating both the inflammatory response and nerve injury [279,285]. This offers fresh perspectives for understanding degenerative diseases and neural damage. For instance, recent studies have focused on the detrimental aspects of Alzheimer’s disease (AD) [279,286], the primary cause of dementia. AD is characterized by the accumulation of beta-amyloid aggregates, leading to neuroinflammation and brain damage. Currently, the molecular alterations in AD are not fully understood, and recent research has aimed to elucidate the role of miRNAs and their implications in tau hyperphosphorylation, which contributes to the formation of neurofibrillary tangles and amyloid plaques [286].
Given the escalating incidence of neurodegenerative diseases, there is a demand for novel biomarkers to facilitate early diagnosis and innovative treatment avenues. MiRNAs present an opportunity to uncover molecular alterations in neurodegenerative disorders, underscoring the necessity for additional research to identify promising biomarkers for early detection. Furthermore, exploring the involved pathways and targeted genes is imperative in advancing this field.
A recent review article [279] underscored the pivotal role of miR-129-5p as a central regulator across diverse disease pathways. Notably, significant pathways implicated include WNT and PI3K/AKT/mTOR, whose aberrations contribute to digestive neoplasia and neurodegenerative disorders. Furthermore, miR-129-5p has been implicated in modulating transmembrane metalloproteinases, integrins, and high-mobility group protein B1 (HMGB1), with its heightened expression providing a shield against cell proliferation and metastasis in digestive cancer cases. The review also elucidates how acute and chronic stress contribute to the dysregulation of this miRNA, culminating in neuroinflammation, neural apoptosis, and the emergence of depression [279]. Given that both acute and chronic stress can potentially lead to the dysregulation of miR-129-5p, the authors observed a correlation between molecular regulation in the brain and the digestive tract [279]. The authors inferred that stress-induced downregulation of this miRNA could lead to the suppression of HMGB1 inhibition, consequently promoting tumor proliferation [279]. The heightened expression of miR-129-5p disrupts pathways involving various targeted genes, offering protective and therapeutic potential against numerous diseases. Moreover, while miR-129-5p has been extensively researched and assessed in various gastrointestinal cancers, it also serves as a significant indicator in brain development, neurodegenerative diseases, depression, and responses to both chronic and acute stress [287].

14.2. Fecal Microbiota Transplantation in Inflammatory Bowel Disease

In recent years, fecal microbiota transplantation (FMT), a treatment centered on the microbiome, has garnered significant attention across scientific, clinical, and public audiences [288]. Using a range of techniques and methods, FMT is a complex procedure aimed at reinstating a harmonious intestinal flora. It involves infusing feces from healthy donors into the GI of individuals with certain conditions to promote recovery. FMT has demonstrated effectiveness in treating various non-GI and GI diseases, including idiopathic constipation, recurrent Clostridioides difficile infection, IBD, and IBS [289,290].
FMT is not a novel therapeutic concept; however, it has garnered heightened interest in recent years due to advancements in methodology and expanding clinical applications [291].
The severity of IBD and its complications are positively correlated with an overgrowth of pathogenic bacteria, including Coprobacillus, Clostridium ramosum, and Clostridium hathewayi. Conversely, a decrease in beneficial anti-inflammatory bacteria like Faecalibacterium prausnitzii is observed, which typically regulates the host’s immune system within the gut. These protective bacteria play a crucial role in immunosuppression, thereby preventing the induction of cytokines and potential intestinal damage [292]. Among patients with CD, their microbiota exhibited a predominance of Actinomyces spp., along with elevated levels of Veillonella spp., E. coli, and Intestinibacter spp. Conversely, in patients with UC, the gut microbiota showed an augmentation of Eubacterium rectum, E. coli, and Ruminococcus gnavus, which are microbes known to sustain and trigger cellular inflammation [292,293].
FMT is emerging as a groundbreaking approach for managing severe cases of IBD, demonstrating a significant success rate. This procedure involves delivering a healthy fecal solution into the recipient’s intestinal tract. Studies have shown that autologous FMT (a-FMT) yields comparable benefits to heterologous FMT (h-FMT) [289,292].
The objective of a-FMT is to restore the disrupted gut microbial community by utilizing one’s feces in a healthy condition. Meanwhile, h-FMT involves transplanting feces from a healthy donor into the affected individual to address conditions like IBD and other infectious diseases. It is generally preferred to opt for a-FMT over h-FMT to mitigate the risk of infectious complications [294,295]; nonetheless, it is crucial to identify stool samples that are functionally optimal to mitigate complications associated with inflammation in IBD. Enhancing the efficacy of FMT in IBD necessitates consideration of various factors, including donor selection criteria, the recipient’s current disease status, and the standardization of processing protocols [296].
The ideal timing for conducting the transplant continues to be a topic of debate, with doctors lacking consensus on this matter. While greater effectiveness of transplantation has been noted in severe cases of IBD, the procedure is also advocated for individuals newly diagnosed with the condition. FMT seems to offer both safety and success in averting recurrent infections among individuals with IBD [297]. Due to the inclusion of various factors and the absence of established protocols, conducting studies on FMT can be challenging. Despite its promise and the new avenues it has opened in research, it will likely be a considerable amount of time before this treatment method becomes standard practice in hospitals worldwide [298].

15. Discussion

CD and UC are long-lasting debilitating conditions linked to psychological and social complications. Adolescents are particularly susceptible to the additional stress of dealing with IBD while navigating important developmental stages. Psychological and social factors, including catastrophizing, the stigma surrounding the illness, uncertainty about the condition, and feelings of shame associated with the illness, frequently contribute to perceived stress in chronic illnesses. Nonetheless, the impact of these variables on perceived stress among adolescents with IBD remains unexplored [244].
Recent clinical research studies have also shown that stress is linked to gastrointestinal health and problems with digestion [133]. A phylogenetic microarray, for instance, was used in one study to demonstrate that stress exposure during pregnancy led to abnormal microbiota colonization patterns in children, which probably enhanced inflammation and gastrointestinal complaints [118]. According to these findings, enhanced bacterial translocation was also linked to stress-related psychiatric diseases including depression, which in turn activated immune responses against commensal bacteria [134]. While the data suggest a significant impact of stress on the intestinal microbiota, it is important to note that stress is a subjective phenomenon. This subjectivity poses a challenge when attempting to objectively assess the effects of stress. Consequently, additional studies involving human participants are necessary to confirm whether stress indeed leads to dysbiosis of the gut microbiota.
IBD was categorized as a psychosomatic condition in the 1950s after numerous early research studies revealed a connection between IBD and mental diagnoses [123]. In numerous IBD treatment trials [24,25,26], placebo response rates might still reach 30–40%, so these results support the idea that changes in one’s psychological state can alter disease activity. Acute psychological stress has an impact on water and ion secretion as well as gastrointestinal motility. Acute short-term stress, such as stressful interviews, dichotomous listening exams, and painful stimuli, increases colonic motility in healthy human volunteers and enhances the secretion of salt, chloride, and jejunal water. Although these are non-inflammatory changes, stress-related elevations in IBD patients’ symptomatology may be a result of them [299,300,301].
Acute psychological stress also has an impact on mucosal inflammation. It has been demonstrated that the central release of neuropeptide SP from afferent neurons plays a critical role in modulating stress-induced gastrointestinal hyperalgesia. Peripheral release of SP from the ENS may contribute to stress-related increases in mucosal inflammation in addition to its central effects. Mast cells, a cell type regarded as crucial in mediating stress-induced permeability changes, are found in close connection with SP-containing neurons, even though there are no published data that show an increase in mucosal SP in response to stress. It has been demonstrated that SP causes IBD patients’ mucosal mast cells to release more histamine. Finally, SP can operate as a neurotransmitter as well as an independent inflammatory cytokine, increasing cytokine production and promoting the migration of inflammatory cells. Additionally, it stimulates the expression of CD11b on neutrophils and leucocyte adhesion molecules on microvascular endothelium, promoting leucocyte adherence at inflammatory locations [153,302].
Stress has been linked to increased disease activity [303] and decreased quality of life [304] in individuals with IBD. Since the 1930s [305], it has been suspected to contribute to the onset of the disease and serve as a potential trigger for disease flares [240,306]. Previous reviews have found a significant association between stress and IBD disease activity in the majority of studies reviewed. However, concerns have been raised regarding the heterogeneity in study design, participant samples, and measures of disease activity and stress, which have hindered the establishment of a clear relationship between stress and disease activity in IBD [174,307,308].
Given the recent promotion of collaborative care models for IBD [309], it is crucial to have a comprehensive understanding of the burden of psychiatric comorbidity in IBD to allocate appropriate resources. Further research is necessary to uncover the underlying causes of these associations and to optimize the treatment of mental disorders in individuals with IBD. Clinicians should remain highly vigilant in identifying and treating these psychiatric associations, as it can benefit individuals with IBD both in terms of their mental health and potentially impact the course of their disease.
IBD often occurs at younger ages, thus interfering with one’s education, career, or daily routine. IBD negatively influences quality of life, with its restrictions on activities, interpersonal relationships, and well-being. The burden of IBD begins with symptoms and extends to mental well-being. IBD uniquely impacts an individual’s life, but the influence crosses borders and affects the entire family and society [204].

16. Conclusions

A lot of contributing factors can influence the course of the disease over time for a patient diagnosed with IBD. Psychological stress and psychosocial impairment conditions can exacerbate the intestinal inflammatory response, increasing the likelihood of being diagnosed with IBD in the following years. Current research has shown that exposure to high levels of stress can increase relapse rates in patients with quiescent forms of IBD. Reducing exposure to stress can reduce the severity of symptoms in patients diagnosed with IBD, improving their quality of life. The early and correct approach to the course of somatic and psychological conditions can improve the analysis of their interdependence on the disease state.
It is well known that stress is a factor that accelerates the progression of IBD. Therefore, psychological interventions can improve the treatment effect of gastrointestinal diseases, thus improving patients’ quality of life. There are multiple interdisciplinary approaches to IBD such as cognitive behavioral therapy, mindfulness-based therapy, breath–body–mind workshops, or gut-directed hypnotherapy. Management of these chronic diseases (CD and UC) should be designed individually, respecting medical interventions. Areas of adjuvant therapies have arisen to treat comorbidities and to increase patient quality of life. Thus, the need for future research into preventing IBD and healthcare innovations to manage these complex and costly diseases is highlighted.

17. The Limitations and Strengths of this Work

A limitation of this work is related to the fact that this review relies heavily on animal studies and preclinical research to support its arguments. While animal studies can provide valuable insights, their relevance to human physiology and pathology may be limited. However, this is one of the few published reviews describing the interplay between stress and inflammatory bowel diseases not only in adult patients but also in children. This study also addresses the role of early-life stress and its potential impact on neonatal gastrointestinal health, with an emphasis on NEC and its association with stress in preterm infants.

Author Contributions

O.B., O.M. and I.E. contributed to the conception and design of this study. A.N. organized the database. I.S. performed the statistical analysis. O.B. wrote the first draft of the manuscript. O.B., D.-G.B., L.O., M.P. and D.B. wrote sections of this manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available upon request from the corresponding author.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Ho, S.M.; Lewis, J.D.; Mayer, E.A.; Plevy, S.E.; Chuang, E.; Rappaport, S.M.; Croitoru, K.; Korzenik, J.R.; Krischer, J.; Hyams, J.S.; et al. Challenges in IBD Research: Environmental Triggers. Inflamm. Bowel Dis. 2019, 25 (Suppl. S2), S13–S23. [Google Scholar] [CrossRef] [PubMed]
  2. Yeshi, K.; Ruscher, R.; Hunter, L.; Daly, N.L.; Loukas, A.; Wangchuk, P. Revisiting Inflammatory Bowel Disease: Pathology, Treatments, Challenges and Emerging Therapeutics Including Drug Leads from Natural Products. J. Clin. Med. 2020, 9, 1273. [Google Scholar] [CrossRef] [PubMed]
  3. Agrawal, M.; Jess, T. Implications of the Changing Epidemiology of Inflammatory Bowel Disease in a Changing World. United Eur. Gastroenterol. J. 2022, 10, 1113–1120. [Google Scholar] [CrossRef]
  4. Kaplan, G.G.; Windsor, J.W. The Four Epidemiological Stages in the Global Evolution of Inflammatory Bowel Disease. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 56–66. [Google Scholar] [CrossRef] [PubMed]
  5. Mak, W.Y.; Zhao, M.; Ng, S.C.; Burisch, J. The Epidemiology of Inflammatory Bowel Disease: East Meets West. J. Gastroenterol. Hepatol. 2020, 35, 380–389. [Google Scholar] [CrossRef] [PubMed]
  6. Kuenzig, M.E.; Fung, S.G.; Marderfeld, L.; Mak, J.W.Y.; Kaplan, G.G.; Ng, S.C.; Wilson, D.C.; Cameron, F.; Henderson, P.; Kotze, P.G.; et al. Twenty-First Century Trends in the Global Epidemiology of Pediatric-Onset Inflammatory Bowel Disease: Systematic Review. Gastroenterology 2022, 162, 1147–1159.e4. [Google Scholar] [CrossRef]
  7. Yu, Y.R.; Rodriguez, J.R. Clinical Presentation of Crohn’s, Ulcerative Colitis, and Indeterminate Colitis: Symptoms, Extraintestinal Manifestations, and Disease Phenotypes. Semin. Pediatr. Surg. 2017, 26, 349–355. [Google Scholar] [CrossRef]
  8. Sun, Y.; Li, L.; Xie, R.; Wang, B.; Jiang, K.; Cao, H. Stress Triggers Flare of Inflammatory Bowel Disease in Children and Adults. Front. Pediatr. 2019, 7, 432. [Google Scholar] [CrossRef]
  9. Piovani, D.; Danese, S.; Peyrin-Biroulet, L.; Nikolopoulos, G.K.; Lytras, T.; Bonovas, S. Environmental Risk Factors for Inflammatory Bowel Diseases: An Umbrella Review of Meta-Analyses. Gastroenterology 2019, 157, 647–659.e4. [Google Scholar] [CrossRef]
  10. Stolzer, I.; Kaden-Volynets, V.; Ruder, B.; Letizia, M.; Bittel, M.; Rausch, P.; Basic, M.; Bleich, A.; Baines, J.F.; Neurath, M.F.; et al. Environmental Microbial Factors Determine the Pattern of Inflammatory Lesions in a Murine Model of Crohn’s Disease-Like Inflammation. Inflamm. Bowel Dis. 2020, 26, 66–79. [Google Scholar] [CrossRef]
  11. De Lange, K.M.; Moutsianas, L.; Lee, J.C.; Lamb, C.A.; Luo, Y.; Kennedy, N.A.; Jostins, L.; Rice, D.L.; Gutierrez-Achury, J.; Ji, S.G.; et al. Genome-Wide Association Study Implicates Immune Activation of Multiple Integrin Genes in Inflammatory Bowel Disease. Nat. Genet. 2017, 49, 256–261. [Google Scholar] [CrossRef] [PubMed]
  12. Trindade, I.A.; Pereira, J.; Galhardo, A.; Ferreira, N.B.; Lucena-Santos, P.; Carvalho, S.A.; Oliveira, S.; Skvarc, D.; Rocha, B.S.; Portela, F.; et al. The LIFE with IBD Intervention: Study Protocol for a Randomized Controlled Trial of a Face-to-Face Acceptance and Commitment Therapy and Compassion-Based Intervention Tailored to People with Inflammatory Bowel Disease. Front. Psychiatry 2021, 12, 699367. [Google Scholar] [CrossRef] [PubMed]
  13. de Dios-Duarte, M.J.; Arias, A.; Durantez-Fernández, C.; Niño Martín, V.; Olea, E.; Barba-Pérez, M.Á.; Pérez-Pérez, L.; Cárdaba-García, R.M.; Barrón, A. Flare-Ups in Crohn’s Disease: Influence of Stress and the External Locus of Control. Int. J. Environ. Res. Public Health 2022, 19, 13131. [Google Scholar] [CrossRef] [PubMed]
  14. Pinillos Díaz, J.L. Principios de Psicología—El Proceso Activo de Construir Conocimiento: Aprendizaje; Grupo Anaya Publicaciones Generales: Madrid, Spain, 2020. [Google Scholar]
  15. Kashani, M.; Eliasson, A.; Vernalis, M. Perceived Stress Correlates with Disturbed Sleep: A Link Connecting Stress and Cardiovascular Disease. Stress 2012, 15, 45–51. [Google Scholar] [CrossRef] [PubMed]
  16. Steptoe, A.; Kivimäki, M. Stress and Cardiovascular Disease. Nat. Rev. Cardiol. 2012, 9, 360–370. [Google Scholar] [CrossRef] [PubMed]
  17. Palmero Cantero, F.; Breva, A.; Landeta, Ó. Hostilidad Defensiva y Reactividad Cardiovascular En Una Situación de Estrés Real. Ansiedad y Estrés 2002, 8, 115–142. [Google Scholar]
  18. Rosenthal, T.; Alter, A. Occupational Stress and Hypertension. J. Am. Soc. Hypertens. 2012, 6, 2–22. [Google Scholar] [CrossRef] [PubMed]
  19. Sanz, J.; García-Vera, M.P.; Espinosa, R.; Inés, M.; Fortún, M.; Segura, J. Psychological Factors Associated with Poor Hypertension Control: Differences in Personality and Stress between Patients with Controlled and Uncontrolled Hypertension. Psychol. Rep. 2010, 107, 923–938. [Google Scholar] [CrossRef]
  20. Katan, M.; Nigro, N.; Fluri, F.; Schuetz, P.; Morgenthaler, N.G.; Jax, F.; Meckel, S.; Gass, A.; Bingisser, R.; Steck, A.; et al. Stress Hormones Predict Cerebrovascular Re-Events after Transient Ischemic Attacks. Neurology 2011, 76, 563–566. [Google Scholar] [CrossRef]
  21. Mayer, E.A. Does Stress Damage the Brain? In Understanding Trauma: Integrating Biological, Clinical, and Cultural Perspectives; Cambridge University Press: Cambridge, UK, 2007; pp. 118–141. [Google Scholar] [CrossRef]
  22. Leza, J.C. Mecanismos de Daño Cerebral Inducido Por Estrés. Ansiedad Estrés 2005, 11, 123–140. [Google Scholar]
  23. Del Carmen Real Pérez, M.; Alonso, S.R.L. Influencia de Los Factores Psicosociales En Adolescentes Con Diabetes Mellitus Tipo I. Cultura de los Cuidados 2017, 21, 190–198. [Google Scholar] [CrossRef]
  24. Bitton, A.; Dobkin, P.L.; Edwardes, M.D.; Sewitch, M.J.; Meddings, J.B.; Rawal, S.; Cohen, A.; Vermeire, S.; Dufresne, L.; Franchimont, D.; et al. Predicting Relapse in Crohn’s Disease: A Biopsychosocial Model. Gut 2008, 57, 1386–1392. [Google Scholar] [CrossRef] [PubMed]
  25. Bernstein, C.N. The Brain-Gut Axis and Stress in Inflammatory Bowel Disease. Gastroenterol. Clin. N. Am. 2017, 46, 839–846. [Google Scholar] [CrossRef] [PubMed]
  26. Nguyen, N.H.; Koola, J.; Dulai, P.S.; Prokop, L.J.; Sandborn, W.J.; Singh, S. Rate of Risk Factors for and Interventions to Reduce Hospital Readmission in Patients with Inflammatory Bowel Diseases. Clin. Gastroenterol. Hepatol. 2020, 18, 1939–1948.e7. [Google Scholar] [CrossRef] [PubMed]
  27. Mayer, E.A.; Tillisch, K. The Brain-Gut Axis in Abdominal Pain Syndromes. Annu. Rev. Med. 2011, 62, 381–396. [Google Scholar] [CrossRef] [PubMed]
  28. Galitovskiy, V.; Qian, J.; Chernyavsky, A.I.; Marchenko, S.; Gindi, V.; Edwards, R.A.; Grando, S.A. Cytokine-Induced Alterations of A7 Nicotinic Receptor in Colonic CD4 T-Cells Mediate Dichotomous Response to Nicotine in Murine Models of Th1/Th17 vs. Th2-Mediated Colitis. J. Immunol. 2011, 187, 2677. [Google Scholar] [CrossRef] [PubMed]
  29. Bercik, P.; Denou, E.; Collins, J.; Jackson, W.; Lu, J.; Jury, J.; Deng, Y.; Blennerhassett, P.; MacRi, J.; McCoy, K.D.; et al. The Intestinal Microbiota Affect Central Levels of Brain-Derived Neurotropic Factor and Behavior in Mice. Gastroenterology 2011, 141, 599–609.e3. [Google Scholar] [CrossRef]
  30. Chrousos, G.P. Stress and Disorders of the Stress System. Nat. Rev. Endocrinol. 2009, 5, 374–381. [Google Scholar] [CrossRef]
  31. Stengel, A.; Taché, Y. Corticotropin-Releasing Factor Signaling and Visceral Response to Stress. Exp. Biol. Med. 2010, 235, 1168–1178. [Google Scholar] [CrossRef]
  32. Bailey, M.T.; Engler, H.; Sheridan, J.F. Stress Induces the Translocation of Cutaneous and Gastrointestinal Microflora to Secondary Lymphoid Organs of C57BL/6 Mice. J. Neuroimmunol. 2006, 171, 29–37. [Google Scholar] [CrossRef]
  33. Bailey, M.T.; Dowd, S.E.; Galley, J.D.; Hufnagle, A.R.; Allen, R.G.; Lyte, M. Exposure to a Social Stressor Alters the Structure of the Intestinal Microbiota: Implications for Stressor-Induced Immunomodulation. Brain Behav. Immun. 2011, 25, 397–407. [Google Scholar] [CrossRef] [PubMed]
  34. Bailey, M.T.; Dowd, S.E.; Parry, N.M.A.; Galley, J.D.; Schauer, D.B.; Lyte, M. Stressor Exposure Disrupts Commensal Microbial Populations in the Intestines and Leads to Increased Colonization by Citrobacter Rodentium. Infect. Immun. 2010, 78, 1509–1519. [Google Scholar] [CrossRef] [PubMed]
  35. Lyte, M.; Vulchanova, L.; Brown, D.R. Stress at the Intestinal Surface: Catecholamines and Mucosa-Bacteria Interactions. Cell Tissue Res. 2011, 343, 23–32. [Google Scholar] [CrossRef] [PubMed]
  36. Sudo, N.; Chida, Y.; Aiba, Y.; Sonoda, J.; Oyama, N.; Yu, X.N.; Kubo, C.; Koga, Y. Postnatal Microbial Colonization Programs the Hypothalamic-Pituitary-Adrenal System for Stress Response in Mice. J. Physiol. 2004, 558 Pt 1, 263–275. [Google Scholar] [CrossRef] [PubMed]
  37. Mourad, F.H.; Saadé, N.E. Neural Regulation of Intestinal Nutrient Absorption. Prog. Neurobiol. 2011, 95, 149–162. [Google Scholar] [CrossRef] [PubMed]
  38. Cryan, J.F.; O’Mahony, S.M. The Microbiome-Gut-Brain Axis: From Bowel to Behavior. Neurogastroenterol. Motil. 2011, 23, 187–192. [Google Scholar] [CrossRef] [PubMed]
  39. Clapp, M.; Aurora, N.; Herrera, L.; Bhatia, M.; Wilen, E.; Wakefield, S. Gut Microbiota’s Effect on Mental Health: The Gut-Brain Axis. Clin. Pract. 2017, 7, 987. [Google Scholar] [CrossRef] [PubMed]
  40. Yoo, B.B.; Mazmanian, S.K. The Enteric Network: Interactions between the Immune and Nervous Systems of the Gut. Immunity 2017, 46, 910–926. [Google Scholar] [CrossRef]
  41. Schneider, S.; Wright, C.M.; Heuckeroth, R.O. Unexpected Roles for the Second Brain: Enteric Nervous System as Master Regulator of Bowel Function. Annu. Rev. Physiol. 2019, 81, 235–259. [Google Scholar] [CrossRef]
  42. Jandhyala, S.M.; Talukdar, R.; Subramanyam, C.; Vuyyuru, H.; Sasikala, M.; Reddy, D.N. Role of the Normal Gut Microbiota. World J. Gastroenterol. WJG 2015, 21, 8787. [Google Scholar] [CrossRef]
  43. Frank, D.N.; St. Amand, A.L.; Feldman, R.A.; Boedeker, E.C.; Harpaz, N.; Pace, N.R. Molecular-Phylogenetic Characterization of Microbial Community Imbalances in Human Inflammatory Bowel Diseases. Proc. Natl. Acad. Sci. USA 2007, 104, 13780–13785. [Google Scholar] [CrossRef] [PubMed]
  44. Haiser, H.J.; Turnbaugh, P.J. Developing a Metagenomic View of Xenobiotic Metabolism. Pharmacol. Res. 2013, 69, 21–31. [Google Scholar] [CrossRef] [PubMed]
  45. Donaldson, G.P.; Lee, S.M.; Mazmanian, S.K. Gut Biogeography of the Bacterial Microbiota. Nat. Rev. Microbiol. 2016, 14, 20–32. [Google Scholar] [CrossRef] [PubMed]
  46. Guida, F.; Turco, F.; Iannotta, M.; De Gregorio, D.; Palumbo, I.; Sarnelli, G.; Furiano, A.; Napolitano, F.; Boccella, S.; Luongo, L.; et al. Antibiotic-Induced Microbiota Perturbation Causes Gut Endocannabinoidome Changes, Hippocampal Neuroglial Reorganization and Depression in Mice. Brain Behav. Immun. 2018, 67, 230–245. [Google Scholar] [CrossRef] [PubMed]
  47. Mi, G.L.; Zhao, L.; Qiao, D.D.; Kang, W.Q.; Tang, M.Q.; Xu, J.K. Effectiveness of Lactobacillus reuteri in Infantile Colic and Colicky Induced Maternal Depression: A Prospective Single Blind Randomized Trial. Antonie Van Leeuwenhoek Int. J. Gen. Mol. Microbiol. 2015, 107, 1547–1553. [Google Scholar] [CrossRef] [PubMed]
  48. Ni, J.; Wu, G.D.; Albenberg, L.; Tomov, V.T. Gut Microbiota and IBD: Causation or Correlation? Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 573–584. [Google Scholar] [CrossRef] [PubMed]
  49. Zhang, M.; Sun, K.; Wu, Y.; Yang, Y.; Tso, P.; Wu, Z. Interactions between Intestinal Microbiota and Host Immune Response in Inflammatory Bowel Disease. Front. Immunol. 2017, 8, 942. [Google Scholar] [CrossRef] [PubMed]
  50. Zamani, M.; Alizadeh-Tabari, S.; Singh, S.; Loomba, R. Meta-Analysis: Prevalence of, and Risk Factors for, Non-Alcoholic Fatty Liver Disease in Patients with Inflammatory Bowel Disease. Aliment. Pharmacol. Ther. 2022, 55, 894–907. [Google Scholar] [CrossRef]
  51. Takahashi, K.; Nishida, A.; Fujimoto, T.; Fujii, M.; Shioya, M.; Imaeda, H.; Inatomi, O.; Bamba, S.; Andoh, A.; Sugimoto, M. Reduced Abundance of Butyrate-Producing Bacteria Species in the Fecal Microbial Community in Crohn’s Disease. Digestion 2016, 93, 59–65. [Google Scholar] [CrossRef]
  52. Park, J.H.; Kotani, T.; Konno, T.; Setiawan, J.; Kitamura, Y.; Imada, S.; Usui, Y.; Hatano, N.; Shinohara, M.; Saito, Y.; et al. Promotion of Intestinal Epithelial Cell Turnover by Commensal Bacteria: Role of Short-Chain Fatty Acids. PLoS ONE 2016, 11, e0156334. [Google Scholar] [CrossRef]
  53. Zheng, L.; Kelly, C.J.; Battista, K.D.; Schaefer, R.; Lanis, J.M.; Alexeev, E.E.; Wang, R.X.; Onyiah, J.C.; Kominsky, D.J.; Colgan, S.P. Microbial-Derived Butyrate Promotes Epithelial Barrier Function through IL-10 Receptor-Dependent Repression of Claudin-2. J. Immunol. 2017, 199, 2976–2984. [Google Scholar] [CrossRef] [PubMed]
  54. Venegas, D.P.; De La Fuente, M.K.; Landskron, G.; González, M.J.; Quera, R.; Dijkstra, G.; Harmsen, H.J.M.; Faber, K.N.; Hermoso, M.A. Short Chain Fatty Acids (SCFAs)Mediated Gut Epithelial and Immune Regulation and Its Relevance for Inflammatory Bowel Diseases. Front. Immunol. 2019, 10, 424615. [Google Scholar] [CrossRef]
  55. Donohoe, D.R.; Garge, N.; Zhang, X.; Sun, W.; O’Connell, T.M.; Bunger, M.K.; Bultman, S.J. The Microbiome and Butyrate Regulate Energy Metabolism and Autophagy in the Mammalian Colon. Cell Metab. 2011, 13, 517. [Google Scholar] [CrossRef] [PubMed]
  56. Macia, L.; Tan, J.; Vieira, A.T.; Leach, K.; Stanley, D.; Luong, S.; Maruya, M.; Ian McKenzie, C.; Hijikata, A.; Wong, C.; et al. Metabolite-Sensing Receptors GPR43 and GPR109A Facilitate Dietary Fibre-Induced Gut Homeostasis through Regulation of the Inflammasome. Nat. Commun. 2015, 6, 6734. [Google Scholar] [CrossRef] [PubMed]
  57. Lydiard, R.B. The Role of GABA in Anxiety Disorders. J. Clin. Psychiatry 2003, 64 (Suppl. S3), 21–27. [Google Scholar] [PubMed]
  58. Cryan, J.F.; Dinan, T.G. Mind-Altering Microorganisms: The Impact of the Gut Microbiota on Brain and Behaviour. Nat. Rev. Neurosci. 2012, 13, 701–712. [Google Scholar] [CrossRef] [PubMed]
  59. Gephart, S.M.; McGrath, J.M.; Effken, J.A.; Halpern, M.D. Necrotizing Enterocolitis Risk: State of the Science. Adv. Neonatal Care 2012, 12, 77–87. [Google Scholar] [CrossRef]
  60. D’Agata, A.L.; Wu, J.; Welandawe, M.K.V.; Dutra, S.V.O.; Kane, B.; Groer, M.W. Effects of Early Life NICU Stress on the Developing Gut Microbiome. Dev. Psychobiol. 2019, 61, 650–660. [Google Scholar] [CrossRef]
  61. Dantzer, R.; Cohen, S.; Russo, S.J.; Dinan, T.G. Resilience and Immunity. Brain Behav. Immun. 2018, 74, 28–42. [Google Scholar] [CrossRef]
  62. Karl, P.J.; Hatch, A.M.; Arcidiacono, S.M.; Pearce, S.C.; Pantoja-Feliciano, I.G.; Doherty, L.A.; Soares, J.W. Effects of Psychological, Environmental and Physical Stressors on the Gut Microbiota. Front. Microbiol. 2018, 9, 2013. [Google Scholar] [CrossRef]
  63. Oh, S.; Young, C.; Gravenstein, N.; Islam, S.; Neu, J. Monitoring Technologies in the Neonatal Intensive Care Unit: Implications for the Detection of Necrotizing Enterocolitis. J. Perinatol. 2010, 30, 701–708. [Google Scholar] [CrossRef] [PubMed]
  64. Thomaidou, A.; Chatziioannou, A.C.; Deda, O.; Benaki, D.; Gika, H.; Mikros, E.; Agakidis, C.; Raikos, N.; Theodoridis, G.; Sarafidis, K. A Pilot Case-Control Study of Urine Metabolomics in Preterm Neonates with Necrotizing Enterocolitis. J. Chromatogr. B Analyt Technol. Biomed. Life Sci. 2019, 1117, 10–21. [Google Scholar] [CrossRef]
  65. Sylvester, K.G.; Ling, X.B.; Liu, G.Y.; Kastenberg, Z.J.; Ji, J.; Hu, Z.; Peng, S.; Lau, K.; Abdullah, F.; Brandt, M.L.; et al. A Novel Urine Peptide Biomarker-Based Algorithm for the Prognosis of Necrotising Enterocolitis in Human Infants. Gut 2014, 63, 1284–1292. [Google Scholar] [CrossRef]
  66. Chatziioannou, A.C.; Wolters, J.C.; Sarafidis, K.; Thomaidou, A.; Agakidis, C.; Govorukhina, N.; Kuivenhoven, J.A.; Bischoff, R.; Theodoridis, G. Targeted LC-MS/MS for the Evaluation of Proteomics Biomarkers in the Blood of Neonates with Necrotizing Enterocolitis and Late-Onset Sepsis. Anal. Bioanal. Chem. 2018, 410, 7163–7175. [Google Scholar] [CrossRef] [PubMed]
  67. Agakidou, E.; Agakidis, C.; Gika, H.; Sarafidis, K. Emerging Biomarkers for Prediction and Early Diagnosis of Necrotizing Enterocolitis in the Era of Metabolomics and Proteomics. Front. Pediatr. 2020, 8, 602255. [Google Scholar] [CrossRef] [PubMed]
  68. Huda, S.; Chaudhery, S.; Ibrahim, H.; Pramanik, A. Neonatal Necrotizing Enterocolitis: Clinical Challenges, Pathophysiology and Management. Pathophysiology 2014, 21, 3–12. [Google Scholar] [CrossRef] [PubMed]
  69. Tremblay, É.; Thibault, M.P.; Ferretti, E.; Babakissa, C.; Bertelle, V.; Bettolli, M.; Burghardt, K.M.; Colombani, J.F.; Grynspan, D.; Levy, E.; et al. Gene Expression Profiling in Necrotizing Enterocolitis Reveals Pathways Common to Those Reported in Crohn’s Disease. BMC Med. Genom. 2016, 9, 6. [Google Scholar] [CrossRef]
  70. Chassaing, B.; Srinivasan, G.; Delgado, M.A.; Young, A.N.; Gewirtz, A.T.; Vijay-Kumar, M. Fecal Lipocalin 2, a Sensitive and Broadly Dynamic Non-Invasive Biomarker for Intestinal Inflammation. PLoS ONE 2012, 7, e44328. [Google Scholar] [CrossRef]
  71. Oikonomou, K.A.; Kapsoritakis, A.N.; Theodoridou, C.; Karangelis, D.; Germenis, A.; Stefanidis, I.; Potamianos, S.P. Neutrophil Gelatinase-Associated Lipocalin (NGAL) in Inflammatory Bowel Disease: Association with Pathophysiology of Inflammation, Established Markers, and Disease Activity. J. Gastroenterol. 2012, 47, 519–530. [Google Scholar] [CrossRef]
  72. Bin-Nun, A.; Booms, C.; Sabag, N.; Mevorach, R.; Algur, N.; Hammerman, C. Rapid Fecal Calprotectin (FC) Analysis: Point of Care Testing for Diagnosing Early Necrotizing Enterocolitis. Am. J. Perinatol. 2015, 32, 337–342. [Google Scholar] [CrossRef]
  73. Zhao, T.; Griffith, T.; Zhang, Y.; Li, H.; Hussain, N.; Lester, B.; Cong, X. Early-Life Factors Associated with Neurobehavioral Outcomes in Preterm Infants during NICU Hospitalization. Pediatr. Res. 2022, 92, 1695–1704. [Google Scholar] [CrossRef] [PubMed]
  74. McGowan, E.C.; Hofheimer, J.A.; O’Shea, T.M.; Kilbride, H.; Carter, B.S.; Check, J.; Helderman, J.; Neal, C.R.; Pastyrnak, S.; Smith, L.M.; et al. Analysis of Neonatal Neurobehavior and Developmental Outcomes Among Preterm Infants. JAMA Netw. Open 2022, 5, e2222249. [Google Scholar] [CrossRef] [PubMed]
  75. Seki, D.; Mayer, M.; Hausmann, B.; Pjevac, P.; Giordano, V.; Goeral, K.; Unterasinger, L.; Klebermaß-Schrehof, K.; De Paepe, K.; Van de Wiele, T.; et al. Aberrant Gut-Microbiota-Immune-Brain Axis Development in Premature Neonates with Brain Damage. Cell Host Microbe 2021, 29, 1558–1572.e6. [Google Scholar] [CrossRef]
  76. Rozé, J.C.; Ancel, P.Y.; Marchand-Martin, L.; Rousseau, C.; Montassier, E.; Monot, C.; Le Roux, K.; Butin, M.; Resche-Rigon, M.; Aires, J.; et al. Assessment of Neonatal Intensive Care Unit Practices and Preterm Newborn Gut Microbiota and 2-Year Neurodevelopmental Outcomes. JAMA Netw. Open 2020, 3, e2018119. [Google Scholar] [CrossRef] [PubMed]
  77. Vogel, S.C.; Brito, N.H.; Callaghan, B.L. Early Life Stress and the Development of the Infant Gut Microbiota: Implications for Mental Health and Neurocognitive Development. Curr. Psychiatry Rep. 2020, 22, 61. [Google Scholar] [CrossRef] [PubMed]
  78. Provenzi, L.; Olson, K.; Giusti, L.; Montirosso, R.; Desantis, A.; Tronick, E. NICU Network Neurobehavioral Scale: 1-Month Normative Data and Variation from Birth to 1 Month. Pediatr. Res. 2018, 83, 1104–1109. [Google Scholar] [CrossRef]
  79. Chen, J.; Li, H.; Zhao, T.; Chen, K.; Chen, M.H.; Sun, Z.; Xu, W.; Maas, K.; Lester, B.M.; Cong, X.S. The Impact of Early Life Experiences and Gut Microbiota on Neurobehavioral Development in Preterm Infants: A Longitudinal Cohort Study. Microorganisms 2023, 11, 814. [Google Scholar] [CrossRef] [PubMed]
  80. Indrio, F.; Neu, J.; Pettoello-Mantovani, M.; Marchese, F.; Martini, S.; Salatto, A.; Aceti, A. Development of the Gastrointestinal Tract in Newborns as a Challenge for an Appropriate Nutrition: A Narrative Review. Nutrients 2022, 14, 1405. [Google Scholar] [CrossRef]
  81. Al-Turkait, A.; Szatkowski, L.; Choonara, I.; Ojha, S. Review of Drug Utilization Studies in Neonatal Units: A Global Perspective. Int. J. Environ. Res. Public Health 2020, 17, 5669. [Google Scholar] [CrossRef]
  82. Sun, X.; Zhuan, C.; Xiao, J.; Yao, E.; Chen, L. Impact of Postnatal Exposure to Antibiotics on Intestinal Microbiome in Preterm Infants. Chin. J. Perinat. Med. 2018, 458–464. [Google Scholar] [CrossRef]
  83. Zou, Z.H.; Liu, D.; Li, H.D.; Zhu, D.P.; He, Y.; Hou, T.; Yu, J.L. Prenatal and Postnatal Antibiotic Exposure Influences the Gut Microbiota of Preterm Infants in Neonatal Intensive Care Units. Ann. Clin. Microbiol. Antimicrob. 2018, 17, 9. [Google Scholar] [CrossRef] [PubMed]
  84. Cantey, J.B.; Pyle, A.K.; Wozniak, P.S.; Hynan, L.S.; Sánchez, P.J. Early Antibiotic Exposure and Adverse Outcomes in Preterm, Very Low Birth Weight Infants. J. Pediatr. 2018, 203, 62–67. [Google Scholar] [CrossRef] [PubMed]
  85. Russell, J.T.; Lauren Ruoss, J.; de la Cruz, D.; Li, N.; Bazacliu, C.; Patton, L.; McKinley, K.L.; Garrett, T.J.; Polin, R.A.; Triplett, E.W.; et al. Antibiotics and the Developing Intestinal Microbiome, Metabolome and Inflammatory Environment in a Randomized Trial of Preterm Infants. Sci. Rep. 2021, 11, 1943. [Google Scholar] [CrossRef] [PubMed]
  86. Akagawa, S.; Tsuji, S.; Onuma, C.; Akagawa, Y.; Yamaguchi, T.; Yamagishi, M.; Yamanouchi, S.; Kimata, T.; Sekiya, S.I.; Ohashi, A.; et al. Effect of Delivery Mode and Nutrition on Gut Microbiota in Neonates. Ann. Nutr. Metab. 2019, 74, 132–139. [Google Scholar] [CrossRef] [PubMed]
  87. Korpela, K. Impact of Delivery Mode on Infant Gut Microbiota. Ann. Nutr. Metab. 2021, 77, 11–19. [Google Scholar] [CrossRef] [PubMed]
  88. Zhang, C.; Li, L.; Jin, B.; Xu, X.; Zuo, X.; Li, Y.; Li, Z. The Effects of Delivery Mode on the Gut Microbiota and Health: State of Art. Front. Microbiol. 2021, 12, 724449. [Google Scholar] [CrossRef]
  89. Dermyshi, E.; Wang, Y.; Yan, C.; Hong, W.; Qiu, G.; Gong, X.; Zhang, T. The “Golden Age” of Probiotics: A Systematic Review and Meta-Analysis of Randomized and Observational Studies in Preterm Infants. Neonatology 2017, 112, 9–23. [Google Scholar] [CrossRef] [PubMed]
  90. Van Den Akker, C.H.P.; Van Goudoever, J.B.; Shamir, R.; Domellöf, M.; Embleton, N.D.; Hojsak, I.; Lapillonne, A.; Mihatsch, W.A.; Berni Canani, R.; Bronsky, J.; et al. Probiotics and Preterm Infants: A Position Paper by the European Society for Paediatric Gastroenterology Hepatology and Nutrition Committee on Nutrition and the European Society for Paediatric Gastroenterology Hepatology and Nutrition Working Group for Probiotics and Prebiotics. J. Pediatr. Gastroenterol. Nutr. 2020, 70, 664–680. [Google Scholar] [CrossRef]
  91. Poindexter, B.; Cummings, J.; Hand, I.; Adams-Chapman, I.; Aucott, S.W.; Puopolo, K.M.; Goldsmith, J.P.; Kaufman, D.; Martin, C.; Mowitz, M. Use of Probiotics in Preterm Infants. Pediatrics 2021, 147, e2021051485. [Google Scholar] [CrossRef]
  92. Ojima, M.N.; Jiang, L.; Arzamasov, A.A.; Yoshida, K.; Odamaki, T.; Xiao, J.; Nakajima, A.; Kitaoka, M.; Hirose, J.; Urashima, T.; et al. Priority Effects Shape the Structure of Infant-Type Bifidobacterium Communities on Human Milk Oligosaccharides. ISME J. 2022, 16, 2265–2279. [Google Scholar] [CrossRef]
  93. Lawson, M.A.E.; O’Neill, I.J.; Kujawska, M.; Gowrinadh Javvadi, S.; Wijeyesekera, A.; Flegg, Z.; Chalklen, L.; Hall, L.J. Breast Milk-Derived Human Milk Oligosaccharides Promote Bifidobacterium Interactions within a Single Ecosystem. ISME J. 2020, 14, 635–648. [Google Scholar] [CrossRef] [PubMed]
  94. Mercer, E.M.; Arrieta, M.C. Probiotics to Improve the Gut Microbiome in Premature Infants: Are We There Yet? Gut Microbes 2023, 15, 2201160. [Google Scholar] [CrossRef] [PubMed]
  95. Alcon-Giner, C.; Dalby, M.J.; Caim, S.; Ketskemety, J.; Shaw, A.; Sim, K.; Lawson, M.A.E.; Kiu, R.; Leclaire, C.; Chalklen, L.; et al. Microbiota Supplementation with Bifidobacterium and Lactobacillus Modifies the Preterm Infant Gut Microbiota and Metabolome: An Observational Study. Cell Rep. Med. 2020, 1, 100077. [Google Scholar] [CrossRef] [PubMed]
  96. Hobbs, A.J.; Mannion, C.A.; McDonald, S.W.; Brockway, M.; Tough, S.C. The Impact of Caesarean Section on Breastfeeding Initiation, Duration and Difficulties in the First Four Months Postpartum. BMC Pregnancy Childbirth 2016, 16, 90. [Google Scholar] [CrossRef] [PubMed]
  97. Pivrncova, E.; Kotaskova, I.; Thon, V. Neonatal Diet and Gut Microbiome Development After C-Section During the First Three Months After Birth: A Systematic Review. Front. Nutr. 2022, 9, 941549. [Google Scholar] [CrossRef] [PubMed]
  98. Notarbartolo, V.; Giuffre, M.; Montante, C.; Corsello, G.; Carta, M. Composition of Human Breast Milk Microbiota and Its Role in Children’s Health. Pediatr. Gastroenterol. Hepatol. Nutr. 2022, 25, 194–210. [Google Scholar] [CrossRef] [PubMed]
  99. Chong, H.Y.; Tan, L.T.H.; Law, J.W.F.; Hong, K.W.; Ratnasingam, V.; Ab Mutalib, N.S.; Lee, L.H.; Letchumanan, V. Exploring the Potential of Human Milk and Formula Milk on Infants’ Gut and Health. Nutrients 2022, 14, 3554. [Google Scholar] [CrossRef]
  100. Zhang, S.; Li, T.; Xie, J.; Zhang, D.; Pi, C.; Zhou, L.; Yang, W. Gold Standard for Nutrition: A Review of Human Milk Oligosaccharide and Its Effects on Infant Gut Microbiota. Microb. Cell Fact. 2021, 20, 108. [Google Scholar] [CrossRef]
  101. Ziegler, E.E. Human Milk-a Valuable Tool in the Early Days of Life of Premature Infants. Front. Pediatr. 2019, 7, 411362. [Google Scholar] [CrossRef]
  102. Ungaro, R.; Bernstein, C.N.; Gearry, R.; Hviid, A.; Kolho, K.L.; Kronman, M.P.; Shaw, S.; Van Kruiningen, H.; Colombel, J.F.; Atreja, A. Antibiotics Associated with Increased Risk of New-Onset Crohn’s Disease but Not Ulcerative Colitis: A Meta-Analysis. Am. J. Gastroenterol. 2014, 109, 1728–1738. [Google Scholar] [CrossRef]
  103. Ortizo, R.; Lee, S.Y.; Nguyen, E.T.; Jamal, M.M.; Bechtold, M.M.; Nguyen, D.L. Exposure to Oral Contraceptives Increases the Risk for Development of Inflammatory Bowel Disease: A Meta-Analysis of Case-Controlled and Cohort Studies. Eur. J. Gastroenterol. Hepatol. 2017, 29, 1064–1070. [Google Scholar] [CrossRef] [PubMed]
  104. Felder, J.B.; Korelitz, B.I.; Rajapakse, R.; Schwarz, S.; Horatagis, A.P.; Gleim, G. Effects of Nonsteroidal Antiinflammatory Drugs on Inflammatory Bowel Disease: A Case-Control Study. Am. J. Gastroenterol. 2000, 95, 1949–1954. [Google Scholar] [CrossRef] [PubMed]
  105. Singh, S.; Graff, L.A.; Bernstein, C.N. Do NSAIDs, Antibiotics, Infections, or Stress Trigger Flares in IBD? Am. J. Gastroenterol. 2009, 104, 1298–1313. [Google Scholar] [CrossRef] [PubMed]
  106. Guslandi, M. Exacerbation of Inflammatory Bowel Disease by Nonsteroidal Anti-Inflammatory Drugs and Cyclooxygenase-2 Inhibitors: Fact or Fiction? World J. Gastroenterol. WJG 2006, 12, 1509. [Google Scholar] [CrossRef] [PubMed]
  107. Messaoudi, M.; Lalonde, R.; Violle, N.; Javelot, H.; Desor, D.; Nejdi, A.; Bisson, J.F.; Rougeot, C.; Pichelin, M.; Cazaubiel, M.; et al. Assessment of Psychotropic-like Properties of a Probiotic Formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in Rats and Human Subjects. Br. J. Nutr. 2011, 105, 755–764. [Google Scholar] [CrossRef] [PubMed]
  108. García-Ródenas, C.L.; Bergonzelli, G.E.; Nutten, S.; Schumann, A.; Cherbut, C.; Turini, M.; Ornstein, K.; Rochat, F.; Corthésy-Theulaz, I. Nutritional Approach to Restore Impaired Intestinal Barrier Function and Growth after Neonatal Stress in Rats. J. Pediatr. Gastroenterol. Nutr. 2006, 43, 16–24. [Google Scholar] [CrossRef] [PubMed]
  109. Zareie, M.; Johnson-Henry, K.; Jury, J.; Yang, P.C.; Ngan, B.Y.; McKay, D.M.; Soderholm, J.D.; Perdue, M.H.; Sherman, P.M. Probiotics Prevent Bacterial Translocation and Improve Intestinal Barrier Function in Rats Following Chronic Psychological Stress. Gut 2006, 55, 1553–1560. [Google Scholar] [CrossRef]
  110. Li, N.; Wang, Q.; Wang, Y.; Sun, A.; Lin, Y.; Jin, Y.; Li, X. Oral Probiotics Ameliorate the Behavioral Deficits Induced by Chronic Mild Stress in Mice via the Gut Microbiota-Inflammation Axis. Front. Behav. Neurosci. 2018, 12, 266. [Google Scholar] [CrossRef]
  111. Bravo, J.A.; Forsythe, P.; Chew, M.V.; Escaravage, E.; Savignac, H.M.; Dinan, T.G.; Bienenstock, J.; Cryan, J.F. Ingestion of Lactobacillus Strain Regulates Emotional Behavior and Central GABA Receptor Expression in a Mouse via the Vagus Nerve. Proc. Natl. Acad. Sci. USA 2011, 108, 16050–16055. [Google Scholar] [CrossRef]
  112. Rao, S.; Srinivasjois, R.; Patole, S. Prebiotic Supplementation in Full-Term Neonates. Arch. Pediatr. Adolesc. Med. 2009, 163, 755–764. [Google Scholar] [CrossRef]
  113. O’Mahony, S.M.; Marchesi, J.R.; Scully, P.; Codling, C.; Ceolho, A.M.; Quigley, E.M.M.; Cryan, J.F.; Dinan, T.G. Early Life Stress Alters Behavior, Immunity, and Microbiota in Rats: Implications for Irritable Bowel Syndrome and Psychiatric Illnesses. Biol. Psychiatry 2009, 65, 263–267. [Google Scholar] [CrossRef]
  114. Golubeva, A.V.; Crampton, S.; Desbonnet, L.; Edge, D.; O’Sullivan, O.; Lomasney, K.W.; Zhdanov, A.V.; Crispie, F.; Moloney, R.D.; Borre, Y.E.; et al. Prenatal Stress-Induced Alterations in Major Physiological Systems Correlate with Gut Microbiota Composition in Adulthood. Psychoneuroendocrinology 2015, 60, 58–74. [Google Scholar] [CrossRef] [PubMed]
  115. Jašarević, E.; Howerton, C.L.; Howard, C.D.; Bale, T.L. Alterations in the Vaginal Microbiome by Maternal Stress Are Associated with Metabolic Reprogramming of the Offspring Gut and Brain. Endocrinology 2015, 156, 3265–3276. [Google Scholar] [CrossRef] [PubMed]
  116. Xie, R.; Sun, Y.; Wu, J.; Huang, S.; Jin, G.; Guo, Z.; Zhang, Y.; Liu, T.; Liu, X.; Cao, X.; et al. Maternal High Fat Diet Alters Gut Microbiota of Offspring and Exacerbates Dss-Induced Colitis in Adulthood. Front. Immunol. 2018, 9, 2608. [Google Scholar] [CrossRef] [PubMed]
  117. Bailey, M.T.; Coe, C.L. Maternal Separation Disrupts the Integrity of the Intestinal Microflora in Infant Rhesus Monkeys. Dev. Psychobiol. 1999, 35, 146–155. [Google Scholar] [CrossRef]
  118. Zijlmans, M.A.C.; Korpela, K.; Riksen-Walraven, J.M.; de Vos, W.M.; de Weerth, C. Maternal Prenatal Stress Is Associated with the Infant Intestinal Microbiota. Psychoneuroendocrinology 2015, 53, 233–245. [Google Scholar] [CrossRef] [PubMed]
  119. Söderholm, J.D.; Yang, P.C.; Ceponis, P.; Vohra, A.; Riddell, R.; Sherman, P.M.; Perdue, M.H. Chronic Stress Induces Mast Cell-Dependent Bacterial Adherence and Initiates Mucosal Inflammation in Rat Intestine. Gastroenterology 2002, 123, 1099–1108. [Google Scholar] [CrossRef] [PubMed]
  120. Da Silva, S.; Robbe-Masselot, C.; Ait-Belgnaoui, A.; Mancuso, A.; Mercade-Loubière, M.; Salvador-Cartier, C.; Gillet, M.; Ferrier, L.; Loubière, P.; Dague, E.; et al. Stress Disrupts Intestinal Mucus Barrier in Rats via Mucin O-Glycosylation Shift: Prevention by a Probiotic Treatment. Am. J. Physiol. Gastrointest. Liver Physiol. 2014, 307, 420–429. [Google Scholar] [CrossRef]
  121. Meddings, J.B.; Swain, M.G. Environmental Stress-Induced Gastrointestinal Permeability Is Mediated by Endogenous Glucocorticoids in the Rat. Gastroenterology 2000, 119, 1019–1028. [Google Scholar] [CrossRef]
  122. Saunders, P.R.; Santos, J.; Hanssen, N.P.M.; Yates, D.; Groot, J.A.; Perdue, M.H. Physical and Psychological Stress in Rats Enhances Colonic Epithelial Permeability via Peripheral CRH. Dig. Dis. Sci. 2002, 47, 208–215. [Google Scholar] [CrossRef]
  123. Santos, J.; Yang, P.C.; Söderholm, J.D.; Benjamin, M.; Perdue, M.H. Role of Mast Cells in Chronic Stress Induced Colonic Epithelial Barrier Dysfunction in the Rat. Gut 2001, 48, 630–636. [Google Scholar] [CrossRef] [PubMed]
  124. Gao, X.; Cao, Q.; Cheng, Y.; Zhao, D.; Wang, Z.; Yang, H.; Wu, Q.; You, L.; Wang, Y.; Lin, Y.; et al. Chronic Stress Promotes Colitis by Disturbing the Gut Microbiota and Triggering Immune System Response. Proc. Natl. Acad. Sci. USA 2018, 115, E2960–E2969. [Google Scholar] [CrossRef] [PubMed]
  125. Neufeld, K.M.; Kang, N.; Bienenstock, J.; Foster, J.A. Reduced Anxiety-like Behavior and Central Neurochemical Change in Germ-Free Mice. Neurogastroenterol. Motil. 2011, 23, 255–265. [Google Scholar] [CrossRef] [PubMed]
  126. Donnet-Hughes, A.; Perez, P.F.; Doré, J.; Leclerc, M.; Levenez, F.; Benyacoub, J.; Serrant, P.; Segura-Roggero, I.; Schiffrin, E.J. Potential Role of the Intestinal Microbiota of the Mother in Neonatal Immune Education. Proc. Nutr. Soc. 2010, 69, 407–415. [Google Scholar] [CrossRef] [PubMed]
  127. Heijtz, R.D.; Wang, S.; Anuar, F.; Qian, Y.; Björkholm, B.; Samuelsson, A.; Hibberd, M.L.; Forssberg, H.; Pettersson, S. Normal Gut Microbiota Modulates Brain Development and Behavior. Proc. Natl. Acad. Sci. USA 2011, 108, 3047–3052. [Google Scholar] [CrossRef] [PubMed]
  128. Marin, I.A.; Goertz, J.E.; Ren, T.; Rich, S.S.; Onengut-Gumuscu, S.; Farber, E.; Wu, M.; Overall, C.C.; Kipnis, J.; Gaultier, A. Microbiota Alteration Is Associated with the Development of Stress-Induced Despair Behavior. Sci. Rep. 2017, 7, srep43859. [Google Scholar] [CrossRef] [PubMed]
  129. Bharwani, A.; Mian, M.F.; Foster, J.A.; Surette, M.G.; Bienenstock, J.; Forsythe, P. Structural and Functional Consequences of Chronic Psychosocial Stress on the Microbiome and Host; Elsevier Ltd.: Amsterdam, The Netherlands, 2016; Volume 63. [Google Scholar] [CrossRef]
  130. Galley, J.D.; Nelson, M.C.; Yu, Z.; Dowd, S.E.; Walter, J.; Kumar, P.S.; Lyte, M.; Bailey, M.T. Exposure to a Social Stressor Disrupts the Community Structure of the Colonic Mucosa-Associated Microbiota. BMC Microbiol. 2014, 14, 189. [Google Scholar] [CrossRef]
  131. Noguera, J.C.; Aira, M.; Pérez-Losada, M.; Domínguez, J.; Velando, A. Glucocorticoids Modulate Gastrointestinal Microbiome in a Wild Bird. R. Soc. Open Sci. 2018, 5, 171743. [Google Scholar] [CrossRef]
  132. Van Der Zaag-Loonen, H.J.; Grootenhuis, M.A.; Last, B.F.; Derkx, H.H.F. Coping Strategies and Quality of Life of Adolescents with Inflammatory Bowel Disease. Qual. Life Res. 2004, 13, 1011–1019. [Google Scholar] [CrossRef]
  133. Walker, L.S.; Garber, J.; Smith, C.A.; Van Slyke, D.A.; Claar, R.L. The Relation of Daily Stressors to Somatic and Emotional Symptoms in Children with and without Recurrent Abdominal Pain. J. Consult. Clin. Psychol. 2001, 69, 85–91. [Google Scholar] [CrossRef]
  134. Maes, M.; Kubera, M.; Leunis, J.C.; Berk, M. Increased IgA and IgM Responses against Gut Commensals in Chronic Depression: Further Evidence for Increased Bacterial Translocation or Leaky Gut. J. Affect. Disord. 2012, 141, 55–62. [Google Scholar] [CrossRef] [PubMed]
  135. McEwen, B.S.; Gianaros, P.J. Central Role of the Brain in Stress and Adaptation: Links to Socioeconomic Status, Health, and Disease. Ann. N. Y. Acad. Sci. 2010, 1186, 190–222. [Google Scholar] [CrossRef] [PubMed]
  136. Mawdsley, J.E.; Rampton, D.S. Psychological Stress in IBD: New Insights into Pathogenic and Therapeutic Implications. Gut 2005, 54, 1481–1491. [Google Scholar] [CrossRef] [PubMed]
  137. Carrasco, G.A.; Van De Kar, L.D. Neuroendocrine Pharmacology of Stress. Eur. J. Pharmacol. 2003, 463, 235–272. [Google Scholar] [CrossRef] [PubMed]
  138. Mayer, E.A. The Neurobiology of Stress and Gastrointestinal Disease. Gut 2000, 47, 861–869. [Google Scholar] [CrossRef] [PubMed]
  139. Barnes, P.J. Molecular Mechanisms of Corticosteroids in Allergic Diseases. Allergy 2001, 56, 928–936. [Google Scholar] [CrossRef]
  140. Joyce, D.A.; Gimblett, G.; Steer, J.H. Targets of Glucocorticoid Action on TNF-Alpha Release by Macrophages. Inflamm. Res. 2001, 50, 337–340. [Google Scholar] [CrossRef]
  141. Franchimont, D.; Kino, T.; Galon, J.; Meduri, G.U.; Chrousos, G. Glucocorticoids and Inflammation Revisited: The State of the Art. NIH Clinical Staff Conference. Neuroimmunomodulation 2002, 10, 247–260. [Google Scholar] [CrossRef]
  142. Amsterdam, A.; Tajima, K.; Sasson, R. Cell-Specific Regulation of Apoptosis by Glucocorticoids: Implication to Their Anti-Inflammatory Action. Biochem. Pharmacol. 2002, 64, 843–850. [Google Scholar] [CrossRef]
  143. Straub, R.H.; Dhabhar, F.S.; Bijlsma, J.W.J.; Cutolo, M. How Psychological Stress via Hormones and Nerve Fibers May Exacerbate Rheumatoid Arthritis. Arthritis Rheum. 2005, 52, 16–26. [Google Scholar] [CrossRef]
  144. Frank, M.G.; Wieseler Frank, J.L.; Hendricks, S.E.; Burke, W.J.; Johnson, D.R. Age at Onset of Major Depressive Disorder Predicts Reductions in NK Cell Number and Activity. J. Affect. Disord. 2002, 71, 159–167. [Google Scholar] [CrossRef] [PubMed]
  145. Fortes, C.; Farchi, S.; Forastiere, F.; Agabiti, N.; Pacifici, R.; Zuccaro, P.; Perucci, C.A.; Ebrahim, S. Depressive Symptoms Lead to Impaired Cellular Immune Response. Psychother. Psychosom. 2003, 72, 253–260. [Google Scholar] [CrossRef] [PubMed]
  146. Schleifer, S.J.; Bartlett, J.A.; Keller, S.E.; Eckholdt, H.M.; Shiflett, S.C.; Delaney, B.R. Immunity in Adolescents with Major Depression. J. Am. Acad. Child. Adolesc. Psychiatry 2002, 41, 1054–1060. [Google Scholar] [CrossRef] [PubMed]
  147. Danner, M.; Kasl, S.V.; Abramson, J.L.; Vaccarino, V. Association between Depression and Elevated C-Reactive Protein. Psychosom. Med. 2003, 65, 347–356. [Google Scholar] [CrossRef] [PubMed]
  148. Sajadieh, A.; Nielsen, O.W.; Rasmussen, V.; Hein, H.O.; Abedini, S.; Hansen, J.F. Increased Heart Rate and Reduced Heart-Rate Variability Are Associated with Subclinical Inflammation in Middle-Aged and Elderly Subjects with No Apparent Heart Disease. Eur. Heart J. 2004, 25, 363–370. [Google Scholar] [CrossRef] [PubMed]
  149. Schmid-Ott, G.; Jaeger, B.; Meyer, S.; Stephan, E.; Kapp, A.; Werfel, T. Different Expression of Cytokine and Membrane Molecules by Circulating Lymphocytes on Acute Mental Stress in Patients with Atopic Dermatitis in Comparison with Healthy Controls. J. Allergy Clin. Immunol. 2001, 108, 455–462. [Google Scholar] [CrossRef] [PubMed]
  150. Kawano, T.A.; Aoki, N.; Homori, M.; Kawano, K.; Maki, A.; Kimura, M.; Yanagisawa, A.; Ohsaki, T.; Takahashi, R.; Shiohara, T.; et al. Mental Stress and Physical Exercise Increase Platelet-Dependent Thrombin Generation. Heart Vessel. 2000, 15, 280–288. [Google Scholar] [CrossRef]
  151. Steptoe, A.; Magid, K.; Edwards, S.; Brydon, L.; Hong, Y.; Erusalimsky, J. The Influence of Psychological Stress and Socioeconomic Status on Platelet Activation in Men. Atherosclerosis 2003, 168, 57–63. [Google Scholar] [CrossRef]
  152. Irving, P.M.; Macey, M.G.; Shah, U.; Webb, L.; Langmead, L.; Rampton, D.S. Formation of Platelet-Leukocyte Aggregates in Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2004, 10, 361–372. [Google Scholar] [CrossRef]
  153. O’Connor, T.M.; O’Connell, J.; O’Brien, D.I.; Goode, T.; Bredin, C.P.; Shanahan, F. The Role of Substance P in Inflammatory Disease. J. Cell Physiol. 2004, 201, 167–180. [Google Scholar] [CrossRef]
  154. Barker, N.; Van Es, J.H.; Kuipers, J.; Kujala, P.; Van Den Born, M.; Cozijnsen, M.; Haegebarth, A.; Korving, J.; Begthel, H.; Peters, P.J.; et al. Identification of Stem Cells in Small Intestine and Colon by Marker Gene Lgr5. Nature 2007, 449, 1003–1007. [Google Scholar] [CrossRef] [PubMed]
  155. Kaser, A.; Lee, A.H.; Franke, A.; Glickman, J.N.; Zeissig, S.; Tilg, H.; Nieuwenhuis, E.E.S.; Higgins, D.E.; Schreiber, S.; Glimcher, L.H.; et al. XBP1 Links ER Stress to Intestinal Inflammation and Confers Genetic Risk for Human Inflammatory Bowel Disease. Cell 2008, 134, 743–756. [Google Scholar] [CrossRef] [PubMed]
  156. Gracie, D.J.; Hamlin, P.J.; Ford, A.C. The Influence of the Brain-Gut Axis in Inflammatory Bowel Disease and Possible Implications for Treatment. Lancet Gastroenterol. Hepatol. 2019, 4, 632–642. [Google Scholar] [CrossRef] [PubMed]
  157. Bonaz, B.L.; Bernstein, C.N. Brain-Gut Interactions in Inflammatory Bowel Disease. Gastroenterology 2013, 144, 36–49. [Google Scholar] [CrossRef] [PubMed]
  158. Shanahan, F. 99th Dahlem Conference on Infection, Inflammation and Chronic Inflammatory Disorders: Host–Microbe Interactions in the Gut: Target for Drug Therapy, Opportunity for Drug Discovery. Clin. Exp. Immunol. 2010, 160, 92. [Google Scholar] [CrossRef] [PubMed]
  159. Burgmann, T.; Clara, I.; Graff, L.; Walker, J.; Lix, L.; Rawsthorne, P.; McPhail, C.; Rogala, L.; Miller, N.; Bernstein, C.N. The Manitoba Inflammatory Bowel Disease Cohort Study: Prolonged Symptoms before Diagnosis—How Much Is Irritable Bowel Syndrome? Clin. Gastroenterol. Hepatol. 2006, 4, 614–620. [Google Scholar] [CrossRef]
  160. Brinkman, D.J.; Ten Hove, A.S.; Vervoordeldonk, M.J.; Luyer, M.D.; de Jonge, W.J. Neuroimmune Interactions in the Gut and Their Significance for Intestinal Immunity. Cells 2019, 8, 670. [Google Scholar] [CrossRef]
  161. Van Maanen, M.A.; Lebre, M.C.; Van Der Poll, T.; LaRosa, G.J.; Elbaum, D.; Vervoordeldonk, M.J.; Tak, P.P. Stimulation of Nicotinic Acetylcholine Receptors Attenuates Collagen-Induced Arthritis in Mice. Arthritis Rheum. 2009, 60, 114–122. [Google Scholar] [CrossRef]
  162. Ghia, J.E.; Blennerhassett, P.; Kumar-Ondiveeran, H.; Verdu, E.F.; Collins, S.M. The Vagus Nerve: A Tonic Inhibitory Influence Associated with Inflammatory Bowel Disease in a Murine Model. Gastroenterology 2006, 131, 1122–1130. [Google Scholar] [CrossRef]
  163. Borovikova, L.V.; Ivanova, S.; Zhang, M.; Yang, H.; Botchkina, G.I.; Watkins, L.R.; Wang, H.; Abumrad, N.; Eaton, J.W.; Tracey, K.J. Vagus Nerve Stimulation Attenuates the Systemic Inflammatory Response to Endotoxin. Nature 2000, 405, 458–462. [Google Scholar] [CrossRef]
  164. Meroni, E.; Stakenborg, N.; Gomez-Pinilla, P.J.; De Hertogh, G.; Goverse, G.; Matteoli, G.; Verheijden, S.; Boeckxstaens, G.E. Functional Characterization of Oxazolone-Induced Colitis and Survival Improvement by Vagus Nerve Stimulation. PLoS ONE 2018, 13, e0197487. [Google Scholar] [CrossRef] [PubMed]
  165. Verheijden, S.; Boeckxstaens, G.E. Neuroimmune Interaction and the Regulation of Intestinal Immune Homeostasis. Am. J. Physiol. Gastrointest. Liver Physiol. 2018, 314, G75–G80. [Google Scholar] [CrossRef] [PubMed]
  166. Tracey, K.J. The Inflammatory Reflex. Nature 2002, 420, 853–859. [Google Scholar] [CrossRef] [PubMed]
  167. Straub, R.H.; Grum, F.; Strauch, U.; Capellino, S.; Bataille, F.; Bleich, A.; Falk, W.; Schölmerich, J.; Obermeier, F. Anti-Inflammatory Role of Sympathetic Nerves in Chronic Intestinal Inflammation. Gut 2008, 57, 911–921. [Google Scholar] [CrossRef] [PubMed]
  168. Magro, F.; Vieira-Coelho, M.A.; Fraga, S.; Serräo, M.P.; Veloso, F.T.; Ribeiro, T.; Soares-da-Silva, P. Impaired Synthesis or Cellular Storage of Norepinephrine, Dopamine, and 5-Hydroxytryptamine in Human Inflammatory Bowel Disease. Dig. Dis. Sci. 2002, 47, 216–224. [Google Scholar] [CrossRef] [PubMed]
  169. Blandizzi, C.; Fornai, M.; Colucci, R.; Baschiera, F.; Barbara, G.; De Giorgio, R.; De Ponti, F.; Breschi, M.C.; Del Tacca, M. Altered Prejunctional Modulation of Intestinal Cholinergic and Noradrenergic Pathways by Alpha2-Adrenoceptors in the Presence of Experimental Colitis. Br. J. Pharmacol. 2003, 139, 309–320. [Google Scholar] [CrossRef] [PubMed]
  170. Knowles, S.R.; Graff, L.A.; Wilding, H.; Hewitt, C.; Keefer, L.; Mikocka-Walus, A. Quality of Life in Inflammatory Bowel Disease: A Systematic Review and Meta-Analyses—Part I. Inflamm. Bowel Dis. 2018, 24, 742–751. [Google Scholar] [CrossRef] [PubMed]
  171. Knowles, S.R.; Keefer, L.; Wilding, H.; Hewitt, C.; Graff, L.A.; Mikocka-Walus, A. Quality of Life in Inflammatory Bowel Disease: A Systematic Review and Meta-Analyses—Part II. Inflamm. Bowel Dis. 2018, 24, 966–976. [Google Scholar] [CrossRef]
  172. Barberio, B.; Zamani, M.; Black, C.J.; Savarino, E.V.; Ford, A.C. Prevalence of Symptoms of Anxiety and Depression in Patients with Inflammatory Bowel Disease: A Systematic Review and Meta-Analysis. Lancet Gastroenterol. Hepatol. 2021, 6, 359–370. [Google Scholar] [CrossRef]
  173. Mikocka-Walus, A.A.; Turnbull, D.A.; Moulding, N.T.; Wilson, I.G.; Andrews, J.M.; Holtmann, G.J. Controversies Surrounding the Comorbidity of Depression and Anxiety in Inflammatory Bowel Disease PatientsA Literature Review. Inflamm. Bowel Dis. 2007, 13, 225–234. [Google Scholar] [CrossRef]
  174. Schoultz, M.; Beattie, M.; Gorely, T.; Leung, J. Assessment of Causal Link between Psychological Factors and Symptom Exacerbation in Inflammatory Bowel Disease: A Systematic Review Utilising Bradford Hill Criteria and Meta-Analysis of Prospective Cohort Studies. Syst. Rev. 2020, 9, 169. [Google Scholar] [CrossRef] [PubMed]
  175. Gracie, D.J.; Irvine, A.J.; Sood, R.; Mikocka-Walus, A.; Hamlin, P.J.; Ford, A.C. Effect of Psychological Therapy on Disease Activity, Psychological Comorbidity, and Quality of Life in Inflammatory Bowel Disease: A Systematic Review and Meta-Analysis. Lancet Gastroenterol. Hepatol. 2017, 2, 189–199. [Google Scholar] [CrossRef] [PubMed]
  176. Folkman, S. Stress: Appraisal and Coping; Springer: New York, NY, USA, 2013. [Google Scholar] [CrossRef]
  177. Lazarus, R.S. Stress and Emotion: A New Synthesis; Springer Pub. Co., Ltd.: Berlin/Heidelberg, Germany, 2006. [Google Scholar]
  178. Goodhand, J.R.; Wahed, M.; Rampton, D.S. Management of Stress in Inflammatory Bowel Disease: A Therapeutic Option? Expert Rev. Gastroenterol. Hepatol. 2014, 3, 661–679. [Google Scholar] [CrossRef] [PubMed]
  179. Kuroki, T.; Ohta, A.; Sherriff-Tadano, R.; Matsuura, E.; Takashima, T.; Iwakiri, R.; Fujimoto, K. Imbalance in the Stress-Adaptation System in Patients with Inflammatory Bowel Disease. Biol. Res. Nurs. 2010, 13, 391–398. [Google Scholar] [CrossRef] [PubMed]
  180. Nahon, S.; Lahmek, P.; Saas, C.; Durance, C.; Olympie, A.; Lesgourgues, B.; Gendre, J.P. Socioeconomic and Psychological Factors Associated with Nonadherence to Treatment in Inflammatory Bowel Disease Patients: Results of the ISSEO Survey. Inflamm. Bowel Dis. 2011, 17, 1270–1276. [Google Scholar] [CrossRef] [PubMed]
  181. Ananthakrishnan, A.N. Epidemiology and Risk Factors for IBD. Nat. Rev. Gastroenterol. Hepatol. 2015, 12, 205–217. [Google Scholar] [CrossRef] [PubMed]
  182. Ramos, G.P.; Kane, S. Alcohol Use in Patients with Inflammatory Bowel Disease. Gastroenterol. Hepatol. 2021, 17, 211. [Google Scholar]
  183. Regueiro, M.; Greer, J.B.; Szigethy, E. Etiology and Treatment of Pain and Psychosocial Issues in Patients with Inflammatory Bowel Diseases. Gastroenterology 2017, 152, 430–439.e4. [Google Scholar] [CrossRef]
  184. Mittermaier, C.; Dejaco, C.; Waldhoer, T.; Oefferlbauer-Ernst, A.; Miehsler, W.; Beier, M.; Tillinger, W.; Gangl, A.; Moser, G. Impact of Depressive Mood on Relapse in Patients with Inflammatory Bowel Disease: A Prospective 18-Month Follow-Up Study. Psychosom. Med. 2004, 66, 79–84. [Google Scholar] [CrossRef]
  185. Persoons, P.; Vermeire, S.; Demyttenaere, K.; Fischler, B.; Vandenberghe, J.; Van Oudenhove, L.; Pierik, M.; Hlavaty, T.; Van Assche, G.; Noman, M.; et al. The Impact of Major Depressive Disorder on the Short- and Long-Term Outcome of Crohn’s Disease Treatment with Infliximab. Aliment. Pharmacol. Ther. 2005, 22, 101–110. [Google Scholar] [CrossRef]
  186. Mikocka-Walus, A.; Pittet, V.; Rossel, J.B.; von Känel, R. Symptoms of Depression and Anxiety Are Independently Associated with Clinical Recurrence of Inflammatory Bowel Disease. Clin. Gastroenterol. Hepatol. 2016, 14, 829–835.e1. [Google Scholar] [CrossRef] [PubMed]
  187. Graff, L.A.; Walker, J.R.; Bernstein, C.N. Depression and Anxiety in Inflammatory Bowel Disease: A Review of Comorbidity and Management. Inflamm. Bowel Dis. 2009, 15, 1105–1118. [Google Scholar] [CrossRef] [PubMed]
  188. Gray, W.N.; Denson, L.A.; Baldassano, R.N.; Hommel, K.A. Treatment Adherence in Adolescents with Inflammatory Bowel Disease: The Collective Impact of Barriers to Adherence and Anxiety/Depressive Symptoms. J. Pediatr. Psychol. 2012, 37, 282. [Google Scholar] [CrossRef] [PubMed]
  189. Ananthakrishnan, A.N.; Gainer, V.S.; Perez, R.G.; Cai, T.; Cheng, S.C.; Savova, G.; Chen, P.; Szolovits, P.; Xia, Z.; De Jager, P.L.; et al. Psychiatric Co-Morbidity Is Associated with Increased Risk of Surgery in Crohn’s Disease. Aliment. Pharmacol. Ther. 2013, 37, 445–454. [Google Scholar] [CrossRef] [PubMed]
  190. Fuller-Thomson, E.; Lateef, R.; Sulman, J. Robust Association Between Inflammatory Bowel Disease and Generalized Anxiety Disorder: Findings from a Nationally Representative Canadian Study. Inflamm. Bowel Dis. 2015, 21, 2341–2348. [Google Scholar] [CrossRef] [PubMed]
  191. Ananthakrishnan, A.N.; Khalili, H.; Pan, A.; Higuchi, L.M.; de Silva, P.; Richter, J.M.; Fuchs, C.S.; Chan, A.T. Association between Depressive Symptoms and Incidence of Crohn’s Disease and Ulcerative Colitis: Results from the Nurses’ Health Study. Clin. Gastroenterol. Hepatol. 2013, 11, 57–62. [Google Scholar] [CrossRef] [PubMed]
  192. Walker, J.R.; Ediger, J.P.; Graff, L.A.; Greenfeld, J.M.; Clara, I.; Lix, L.; Rawsthorne, P.; Miller, N.; Rogala, L.; McPhail, C.M.; et al. The Manitoba IBD Cohort Study: A Population-Based Study of the Prevalence of Lifetime and 12-Month Anxiety and Mood Disorders. Am. J. Gastroenterol. 2008, 103, 1989–1997. [Google Scholar] [CrossRef]
  193. Bernstein, C.N.; Hitchon, C.A.; Walld, R.; Bolton, J.M.; Sareen, J.; Walker, J.R.; Graff, L.A.; Patten, S.B.; Singer, A.; Lix, L.M.; et al. Increased Burden of Psychiatric Disorders in Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2019, 25, 360–368. [Google Scholar] [CrossRef]
  194. Bernstein, C.N.; Wajda, A.; Svenson, L.W.; MacKenzie, A.; Koehoorn, M.; Jackson, M.; Fedorak, R.; Israel, D.; Blanchard, J.F. The Epidemiology of Inflammatory Bowel Disease in Canada: A Population-Based Study. Am. J. Gastroenterol. 2006, 101, 1559–1568. [Google Scholar] [CrossRef]
  195. Fiest, K.M.; Bernstein, C.N.; Walker, J.R.; Graff, L.A.; Hitchon, C.A.; Peschken, C.A.; Zarychanski, R.; Abou-Setta, A.; Patten, S.B.; Sareen, J.; et al. Systematic Review of Interventions for Depression and Anxiety in Persons with Inflammatory Bowel Disease. BMC Res. Notes 2016, 9, 404. [Google Scholar] [CrossRef]
  196. He, Y.; Chen, C.L.; He, J.; De Liu, S. Causal Associations between Inflammatory Bowel Disease and Anxiety: A Bidirectional Mendelian Randomization Study. World J. Gastroenterol. 2023, 29, 5872–5881. [Google Scholar] [CrossRef] [PubMed]
  197. Bisgaard, T.H.; Allin, K.H.; Keefer, L.; Ananthakrishnan, A.N.; Jess, T. Depression and Anxiety in Inflammatory Bowel Disease: Epidemiology, Mechanisms and Treatment. Nat. Rev. Gastroenterol. Hepatol. 2022, 19, 717–726. [Google Scholar] [CrossRef] [PubMed]
  198. Cooney, R.; Tang, D.; Barrett, K.; Russell, R.K. Children and Young Adults with Inflammatory Bowel Disease Have an Increased Incidence and Risk of Developing Mental Health Conditions: A UK Population-Based Cohort Study. Inflamm. Bowel Dis. 2023. online ahead of print. [Google Scholar] [CrossRef] [PubMed]
  199. Luo, J.; Xu, Z.; Noordam, R.; Van Heemst, D.; Li-Gao, R. Depression and Inflammatory Bowel Disease: A Bidirectional Two-Sample Mendelian Randomization Study. J. Crohns Colitis 2022, 16, 633–642. [Google Scholar] [CrossRef] [PubMed]
  200. Fairbrass, K.M.; Lovatt, J.; Barberio, B.; Yuan, Y.; Gracie, D.J.; Ford, A.C. Bidirectional Brain–Gut Axis Effects Influence Mood and Prognosis in IBD: A Systematic Review and Meta-Analysis. Gut 2022, 71, 1773–1780. [Google Scholar] [CrossRef] [PubMed]
  201. Frolkis, A.D.; Vallerand, I.A.; Shaheen, A.A.; Lowerison, M.W.; Swain, M.G.; Barnabe, C.; Patten, S.B.; Kaplan, G.G. Depression Increases the Risk of Inflammatory Bowel Disease, Which May Be Mitigated by the Use of Antidepressants in the Treatment of Depression. Gut 2019, 68, 1606–1612. [Google Scholar] [CrossRef]
  202. Agirman, G.; Yu, K.B.; Hsiao, E.Y. Signaling Inflammation across the Gut-Brain Axis. Science 2021, 374, 1087–1092. [Google Scholar] [CrossRef]
  203. HRQOL Concepts|CDC. Available online: https://www.cdc.gov/hrqol/concept.htm (accessed on 5 July 2023).
  204. Jones, J.L.; Nguyen, G.C.; Benchimol, E.I.; Bernstein, C.N.; Bitton, A.; Kaplan, G.G.; Murthy, S.K.; Lee, K.; Cooke-Lauder, J.; Otley, A.R. The Impact of Inflammatory Bowel Disease in Canada 2018: Quality of Life. J. Can. Assoc. Gastroenterol. 2019, 2 (Suppl. S1), S42–S48. [Google Scholar] [CrossRef]
  205. Bernklev, T.; Jahnsen, J.; Lygren, I.; Henriksen, M.; Vatn, M.; Moum, B. Health-Related Quality of Life in Patients with Inflammatory Bowel Disease Measured with the Short Form-36: Psychometric Assessments and a Comparison with General Population Norms. Inflamm. Bowel Dis. 2005, 11, 909–918. [Google Scholar] [CrossRef]
  206. Hoivik, M.L.; Moum, B.; Solberg, I.C.; Cvancarova, M.; Hoie, O.; Vatn, M.H.; Bernklev, T. Health-Related Quality of Life in Patients with Ulcerative Colitis after a 10-Year Disease Course: Results from the IBSEN Study. Inflamm. Bowel Dis. 2012, 18, 1540–1549. [Google Scholar] [CrossRef]
  207. Høivik, M.L.; Bernklev, T.; Solberg, I.C.; Cvancarova, M.; Lygren, I.; Jahnsen, J.; Moum, B. Patients with Crohn’s Disease Experience Reduced General Health and Vitality in the Chronic Stage: Ten-Year Results from the IBSEN Study. J. Crohns Colitis 2012, 6, 441–453. [Google Scholar] [CrossRef] [PubMed]
  208. Casellas, F.; Arenas, J.I.; Baudet, J.S.; Fábregas, S.; García, N.; Gelabert, J.; Medina, C.; Ochotorena, I.; Papo, M.; Rodrigo, L.; et al. Impairment of Health-Related Quality of Life in Patients with Inflammatory Bowel Disease: A Spanish Multicenter Study. Inflamm. Bowel Dis. 2005, 11, 488–496. [Google Scholar] [CrossRef] [PubMed]
  209. Wilburn, J.; Twiss, J.; Kemp, K.; McKenna, S.P. A Qualitative Study of the Impact of Crohn’s Disease from a Patient’s Perspective. Front. Gastroenterol. 2017, 8, 68–73. [Google Scholar] [CrossRef] [PubMed]
  210. Pihl-Lesnovska, K.; Hjortswang, H.; Ek, A.C.; Frisman, G.H. Patients’ Perspective of Factors Influencing Quality of Life While Living with Crohn Disease. Gastroenterol. Nurs. 2010, 33, 37–44. [Google Scholar] [CrossRef] [PubMed]
  211. Rahier, J.F.; Magro, F.; Abreu, C.; Armuzzi, A.; Ben-Horin, S.; Chowers, Y.; Cottone, M.; de Ridder, L.; Doherty, G.; Ehehalt, R.; et al. Second European Evidence-Based Consensus on the Prevention, Diagnosis and Management of Opportunistic Infections in Inflammatory Bowel Disease. J. Crohns Colitis 2014, 8, 443–468. [Google Scholar] [CrossRef] [PubMed]
  212. Ben-Horin, S.; Bujanover, Y.; Goldstein, S.; Nadler, M.; Lang, A.; Kopylov, U.; Katz, L.; Lahat, A.; Schwartz, E.; Avidan, B. Travel-Associated Health Risks for Patients with Inflammatory Bowel Disease. Clin. Gastroenterol. Hepatol. 2012, 10, 160–165.e1. [Google Scholar] [CrossRef] [PubMed]
  213. Greveson, K.; Shepherd, T.; Mulligan, J.P.; Hamilton, M.; Woodward, S.; Norton, C.; Murray, C. Travel Health and Pretravel Preparation in the Patient with Inflammatory Bowel Disease. Front. Gastroenterol. 2016, 7, 60–65. [Google Scholar] [CrossRef]
  214. Libby, G.W.; Dawson, A.M. The Social Toll of Crohn’s Disease. Br. Med. J. 1978, 2, 1117–1119. [Google Scholar] [CrossRef]
  215. Rivière, P.; Zallot, C.; Desobry, P.; Sabaté, J.M.; Vergniol, J.; Zerbib, F.; Peyrin-Biroulet, L.; Laharie, D.; Poullenot, F. Frequency of and Factors Associated with Sexual Dysfunction in Patients with Inflammatory Bowel Disease. J. Crohns Colitis 2017, 11, 1347–1352. [Google Scholar] [CrossRef]
  216. Bel, L.G.J.; Vollebregt, A.M.; Van der Meulen-de Jong, A.E.; Fidder, H.H.; Ten Hove, W.R.; Vliet-Vlieland, C.W.; ter Kuile, M.M.; de Groot, H.E.; Both, S. Sexual Dysfunctions in Men and Women with Inflammatory Bowel Disease: The Influence of IBD-Related Clinical Factors and Depression on Sexual Function. J. Sex. Med. 2015, 12, 1557–1567. [Google Scholar] [CrossRef]
  217. Zhao, S.; Wang, J.; Liu, Y.; Luo, L.; Zhu, Z.; Li, E.; Luo, J.; Zhao, Z. Inflammatory Bowel Diseases Were Associated with Risk of Sexual Dysfunction in Both Sexes: A Meta-Analysis. Inflamm. Bowel Dis. 2019, 25, 699–707. [Google Scholar] [CrossRef] [PubMed]
  218. de Arce, E.P.; Quera, R.; Barros, J.R.; Sassaki, L.Y. Sexual Dysfunction in Inflammatory Bowel Disease: What the Specialist Should Know and Ask. Int. J. Gen. Med. 2021, 14, 2003–2015. [Google Scholar] [CrossRef]
  219. Büller, H.A. Objectives and Outcomes in the Conventional Treatment of Pediatric Crohn’s Disease. J. Pediatr. Gastroenterol. Nutr. 2001, 33 (Suppl. S1), S11–S18. [Google Scholar] [CrossRef] [PubMed]
  220. Mackner, L.M.; Crandall, W.V. Brief Report: Psychosocial Adjustment in Adolescents with Inflammatory Bowel Disease. J. Pediatr. Psychol. 2006, 31, 281–285. [Google Scholar] [CrossRef] [PubMed]
  221. Cunningham, C.; Drotar, D.; Palermo, T.M.; McGowan, K.; Arendt, R. Health-Related Quality of Life in Children and Adolescents with Inflammatory Bowel Disease. Child. Health Care 2007, 36, 29–43. [Google Scholar] [CrossRef]
  222. Kunz, J.H.; Hommel, K.A.; Greenley, R.N. Health-Related Quality of Life of Youth with Inflammatory Bowel Disease: A Comparison with Published Data Using the PedsQL 4.0 Generic Core Scales. Inflamm. Bowel Dis. 2010, 16, 939–946. [Google Scholar] [CrossRef] [PubMed]
  223. Gray, W.N.; Boyle, S.L.; Graef, D.M.; Janicke, D.M.; Jolley, C.D.; Denson, L.A.; Baldassano, R.N.; Hommel, K.A. Health-Related Quality of Life in Youth with Crohn Disease: Role of Disease Activity and Parenting Stress. J. Pediatr. Gastroenterol. Nutr. 2015, 60, 749–753. [Google Scholar] [CrossRef]
  224. Knez, R.; Francisković, T.; Samarin, R.M.; Niksić, M. Parental Quality of Life in the Framework of Paediatric Chronic Gastrointestinal Disease. Coll. Antropol. 2011, 35 (Suppl. S2), 275–280. [Google Scholar]
  225. Argyriou, K.; Kapsoritakis, A.; Oikonomou, K.; Manolakis, A.; Tsakiridou, E.; Potamianos, S. Disability in Patients with Inflammatory Bowel Disease: Correlations with Quality of Life and Patient’s Characteristics. Can. J. Gastroenterol. Hepatol. 2017, 2017, 6138105. [Google Scholar] [CrossRef]
  226. Lönnfors, S.; Vermeire, S.; Greco, M.; Hommes, D.; Bell, C.; Avedano, L. IBD and Health-Related Quality of Life -- Discovering the True Impact. J. Crohns Colitis 2014, 8, 1281–1286. [Google Scholar] [CrossRef]
  227. Nishida, Y.; Hosomi, S.; Fujimoto, K.; Nakata, R.; Itani, S.; Ohminami, M.; Nadatani, Y.; Fukunaga, S.; Otani, K.; Tanaka, F.; et al. Effect of the Coronavirus Disease 2019 Lockdown on Lifestyle Factors in Japanese Patients with Inflammatory Bowel Disease. Intern. Med. 2022, 61, 1329–1336. [Google Scholar] [CrossRef] [PubMed]
  228. Koletzko, L.; Klucker, E.; Le Thi, T.G.; Breiteneicher, S.; Rubio-acero, R.; Neuhaus, L.; Stark, R.G.; Standl, M.; Wieser, A.; Török, H.; et al. Following Pediatric and Adult Ibd Patients through the COVID-19 Pandemic: Changes in Psychosocial Burden and Perception of Infection Risk and Harm over Time. J. Clin. Med. 2021, 10, 4124. [Google Scholar] [CrossRef]
  229. Conti, C.; Rosa, I.; Zito, L.; Grossi, L.; Efthymakis, K.; Neri, M.; Porcelli, P. Influence of the COVID-19 Outbreak on Disease Activity and Quality of Life in Inflammatory Bowel Disease Patients. Front. Psychiatry 2021, 12, 664088. [Google Scholar] [CrossRef] [PubMed]
  230. Occhipinti, V.; Pastorelli, L. Challenges in the Care of IBD Patients during the COVID-19 Pandemic: Report from a “Red Zone” Area in Northern Italy. Inflamm. Bowel Dis. 2020, 26, 793–796. [Google Scholar] [CrossRef] [PubMed]
  231. Corrias, A.; Cortes, G.M.; Bardanzellu, F.; Marcialis, M.A.; Melis, A.; Fanos, V. Risk, Course, and Effect of SARS-CoV-2 Infection in Children and Adults with Chronic Inflammatory Bowel Diseases. Children 2021, 8, 753. [Google Scholar] [CrossRef] [PubMed]
  232. Fragoso, R.P.; Rodrigues, M. COVID-19 and Pediatric Inflammatory Bowel Disease: How to Manage It? Clinics 2020, 75, e1962. [Google Scholar] [CrossRef]
  233. Sansotta, N.; Norsa, L.; Zuin, G.; Panceri, R.; Dilillo, D.; Pozzi, E.; De Giacomo, C.; Moretti, C.; Celano, R.; Nuti, F.; et al. Children with Inflammatory Bowel Disease in the COVID-19 Main Endemic Focus: The Lombardy Experience. Front. Pediatr. 2021, 9, 607285. [Google Scholar] [CrossRef] [PubMed]
  234. Turner, D.; Huang, Y.; Martín-De-Carpi, J.; Aloi, M.; Focht, G.; Kang, B.; Zhou, Y.; Sanchez, C.; Kappelman, M.D.; Uhlig, H.H.; et al. Corona Virus Disease 2019 and Paediatric Inflammatory Bowel Diseases: Global Experience and Provisional Guidance (March 2020) from the Paediatric IBD Porto Group of European Society of Paediatric Gastroenterology, Hepatology, and Nutrition. J. Pediatr. Gastroenterol. Nutr. 2020, 70, 727–733. [Google Scholar] [CrossRef]
  235. Brenner, E.J.; Pigneur, B.; Focht, G.; Zhang, X.; Ungaro, R.C.; Colombel, J.F.; Turner, D.; Kappelman, M.D.; Ruemmele, F.M. Benign Evolution of SARS-CoV2 Infections in Children with Inflammatory Bowel Disease: Results From Two International Databases. Clin. Gastroenterol. Hepatol. 2021, 19, 394–396.e5. [Google Scholar] [CrossRef]
  236. Bezzio, C.; Pellegrini, L.; Manes, G.; Arena, I.; Picascia, D.; Della Corte, C.; Devani, M.; Schettino, M.; Saibeni, S. Biologic Therapies May Reduce the Risk of COVID-19 in Patients with Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2020, 26, E107–E109. [Google Scholar] [CrossRef]
  237. Arrigo, S.; Alvisi, P.; Banzato, C.; Bramuzzo, M.; Civitelli, F.; Corsello, A.; D’Arcangelo, G.; Dilillo, A.; Dipasquale, V.; Felici, E.; et al. Management of Paediatric IBD after the Peak of COVID-19 Pandemic in Italy: A Position Paper on Behalf of the SIGENP IBD Working Group. Dig. Liver Dis. 2021, 53, 183–189. [Google Scholar] [CrossRef] [PubMed]
  238. Salvatori, S.; Baldassarre, F.; Mossa, M.; Monteleone, G. Long COVID in Inflammatory Bowel Diseases. J. Clin. Med. 2021, 10, 5575. [Google Scholar] [CrossRef] [PubMed]
  239. Black, J.; Sweeney, L.; Yuan, Y.; Singh, H.; Norton, C.; Czuber-Dochan, W. Systematic Review: The Role of Psychological Stress in Inflammatory Bowel Disease. Aliment. Pharmacol. Ther. 2022, 56, 1235–1249. [Google Scholar] [CrossRef] [PubMed]
  240. Bernstein, C.N.; Singh, S.; Graff, L.A.; Walker, J.R.; Miller, N.; Cheang, M. A Prospective Population-Based Study of Triggers of Symptomatic Flares in IBD. Am. J. Gastroenterol. 2010, 105, 1994–2002. [Google Scholar] [CrossRef] [PubMed]
  241. Rozich, J.J.; Holmer, A.; Singh, S. Effect of Lifestyle Factors on Outcomes in Patients with Inflammatory Bowel Diseases. Am. J. Gastroenterol. 2020, 115, 832–840. [Google Scholar] [CrossRef] [PubMed]
  242. Peppas, S.; Pansieri, C.; Piovani, D.; Danese, S.; Peyrin-Biroulet, L.; Tsantes, A.G.; Brunetta, E.; Tsantes, A.E.; Bonovas, S. The Brain-Gut Axis: Psychological Functioning and Inflammatory Bowel Diseases. J. Clin. Med. 2021, 10, 377. [Google Scholar] [CrossRef] [PubMed]
  243. Kiebles, J.L.; Doerfler, B.; Keefer, L. Preliminary Evidence Supporting a Framework of Psychological Adjustment to Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2010, 16, 1685–1695. [Google Scholar] [CrossRef]
  244. Sunavsky, A.; Moreau, J.; Tripp, D.A. Understanding Perceived Stress in Adolescent Inflammatory Bowel Disease. J. Can. Assoc. Gastroenterol. 2022, 5, 79–85. [Google Scholar] [CrossRef]
  245. Ge, L.; Liu, S.; Li, S.; Yang, J.; Hu, G.; Xu, C.; Song, W. Psychological Stress in Inflammatory Bowel Disease: Psychoneuroimmunological Insights into Bidirectional Gut–Brain Communications. Front. Immunol. 2022, 13, 1016578. [Google Scholar] [CrossRef]
  246. Tavakoli, P.; Vollmer-Conna, U.; Hadzi-Pavlovic, D.; Grimm, M.C. A Review of Inflammatory Bowel Disease: A Model of Microbial, Immune and Neuropsychological Integration. Public Health Rev. 2021, 42, 1603990. [Google Scholar] [CrossRef]
  247. Jairath, V.; Feagan, B.G. Global Burden of Inflammatory Bowel Disease. Lancet Gastroenterol. Hepatol. 2020, 5, 2–3. [Google Scholar] [CrossRef] [PubMed]
  248. Abraham, B.P.; Ahmed, T.; Ali, T. Inflammatory Bowel Disease: Pathophysiology and Current Therapeutic Approaches. Handb. Exp. Pharmacol. 2017, 239, 115–146. [Google Scholar] [CrossRef] [PubMed]
  249. Martin-Subero, M.; Anderson, G.; Kanchanatawan, B.; Berk, M.; Maes, M. Comorbidity between Depression and Inflammatory Bowel Disease Explained by Immune-Inflammatory, Oxidative, and Nitrosative Stress; Tryptophan Catabolite; and Gut-Brain Pathways. CNS Spectr. 2016, 21, 184–198. [Google Scholar] [CrossRef] [PubMed]
  250. Bonaz, B.; Bazin, T.; Pellissier, S. The Vagus Nerve at the Interface of the Microbiota-Gut-Brain Axis. Front. Neurosci. 2018, 12, 336468. [Google Scholar] [CrossRef] [PubMed]
  251. Neuendorf, R.; Harding, A.; Stello, N.; Hanes, D.; Wahbeh, H. Depression and Anxiety in Patients with Inflammatory Bowel Disease: A Systematic Review. J. Psychosom. Res. 2016, 87, 70–80. [Google Scholar] [CrossRef] [PubMed]
  252. Black, C.J.; Thakur, E.R.; Houghton, L.A.; Quigley, E.M.M.; Moayyedi, P.; Ford, A.C. Efficacy of Psychological Therapies for Irritable Bowel Syndrome: Systematic Review and Network Meta-Analysis. Gut 2020, 69, 1441–1451. [Google Scholar] [CrossRef] [PubMed]
  253. Hanlon, I.; Hewitt, C.; Bell, K.; Phillips, A.; Mikocka-Walus, A. Systematic Review with Meta-Analysis: Online Psychological Interventions for Mental and Physical Health Outcomes in Gastrointestinal Disorders Including Irritable Bowel Syndrome and Inflammatory Bowel Disease. Aliment. Pharmacol. Ther. 2018, 48, 244–259. [Google Scholar] [CrossRef] [PubMed]
  254. Jiang, Y.; Shorey, S.; Seah, B.; Chan, W.X.; Tam, W.W.S.; Wang, W. The Effectiveness of Psychological Interventions on Self-Care, Psychological and Health Outcomes in Patients with Chronic Heart Failure—A Systematic Review and Meta-Analysis. Int. J. Nurs. Stud. 2018, 78, 16–25. [Google Scholar] [CrossRef]
  255. Wynne, B.; McHugh, L.; Gao, W.; Keegan, D.; Byrne, K.; Rowan, C.; Hartery, K.; Kirschbaum, C.; Doherty, G.; Cullen, G.; et al. Acceptance and Commitment Therapy Reduces Psychological Stress in Patients With Inflammatory Bowel Diseases. Gastroenterology 2019, 156, 935–945.e1. [Google Scholar] [CrossRef]
  256. Li, C.; Hou, Z.; Liu, Y.; Ji, Y.; Xie, L. Cognitive-Behavioural Therapy in Patients with Inflammatory Bowel Diseases: A Systematic Review and Meta-Analysis. Int. J. Nurs. Pract. 2019, 25, e12699. [Google Scholar] [CrossRef]
  257. Jordan, C.; Hayee, B.; Chalder, T. Cognitive Behaviour Therapy for Distress in People with Inflammatory Bowel Disease: A Benchmarking Study. Clin. Psychol. Psychother. 2019, 26, 14–23. [Google Scholar] [CrossRef] [PubMed]
  258. Hunt, M.G.; Loftus, P.; Accardo, M.; Keenan, M.; Cohen, L.; Osterman, M.T. Self-Help Cognitive Behavioral Therapy Improves Health-Related Quality of Life for Inflammatory Bowel Disease Patients: A Randomized Controlled Effectiveness Trial. J. Clin. Psychol. Med. Settings 2020, 27, 467–479. [Google Scholar] [CrossRef] [PubMed]
  259. Bennebroek Evertsz, F.; Sprangers, M.A.G.; Sitnikova, K.; Stokkers, P.C.F.; Ponsioen, C.Y.; Bartelsman, J.F.W.M.; Van Bodegraven, A.A.; Fischer, S.; Depla, A.C.T.M.; Mallant, R.C.; et al. Effectiveness of Cognitive-Behavioral Therapy on Quality of Life, Anxiety, and Depressive Symptoms among Patients with Inflammatory Bowel Disease: A Multicenter Randomized Controlled Trial. J. Consult. Clin. Psychol. 2017, 85, 918–925. [Google Scholar] [CrossRef] [PubMed]
  260. Szigethy, E.; Youk, A.O.; Gonzalez-Heydrich, J.; Bujoreanu, S.I.; Weisz, J.; Fairclough, D.; Ducharme, P.; Jones, N.; Lotrich, F.; Keljo, D.; et al. Effect of 2 Psychotherapies on Depression and Disease Activity in Pediatric Crohn’s Disease. Inflamm. Bowel Dis. 2015, 21, 1321–1328. [Google Scholar] [CrossRef] [PubMed]
  261. Chen, J.; Chen, X.; Sun, Y.; Xie, Y.; Wang, X.; Li, R.; Hesketh, T. The Physiological and Psychological Effects of Cognitive Behavior Therapy on Patients with Inflammatory Bowel Disease before COVID-19: A Systematic Review. BMC Gastroenterol. 2021, 21, 469. [Google Scholar] [CrossRef] [PubMed]
  262. Hood, M.M.; Jedel, S. Mindfulness-Based Interventions in Inflammatory Bowel Disease. Gastroenterol. Clin. N. Am. 2017, 46, 859–874. [Google Scholar] [CrossRef] [PubMed]
  263. Berrill, J.W.; Sadlier, M.; Hood, K.; Green, J.T. Mindfulness-Based Therapy for Inflammatory Bowel Disease Patients with Functional Abdominal Symptoms or High Perceived Stress Levels. J. Crohns Colitis 2014, 8, 945–955. [Google Scholar] [CrossRef] [PubMed]
  264. Ewais, T.; Begun, J.; Kenny, M.; Hay, K.; Houldin, E.; Chuang, K.H.; Tefay, M.; Kisely, S. Mindfulness Based Cognitive Therapy for Youth with Inflammatory Bowel Disease and Depression—Findings from a Pilot Randomised Controlled Trial. J. Psychosom. Res. 2021, 149. [Google Scholar] [CrossRef]
  265. González-Moret, R.; Cebolla, A.; Cortés, X.; Baños, R.M.; Navarrete, J.; de la Rubia, J.E.; Lisón, J.F.; Soria, J.M. The Effect of a Mindfulness-Based Therapy on Different Biomarkers among Patients with Inflammatory Bowel Disease: A Randomised Controlled Trial. Sci. Rep. 2020, 10, 6071. [Google Scholar] [CrossRef]
  266. Georgescu, D.; Iurciuc, M.S.; Petre, I.; Georgescu, L.A.; Szasz, F.; Ionita, I.; Ancusa, O.E.; Ionita, M.; Lighezan, D. Chronic Pelvic Pain and Irritable Bowel Syndrome: Is Subclinical Inflammation Bridging the Gap? Rev. Chim. 2019, 70, 3634–3637. [Google Scholar] [CrossRef]
  267. Keefer, L.; Taft, T.H.; Kiebles, J.L.; Martinovich, Z.; Barrett, T.A.; Palsson, O.S. Gut-Directed Hypnotherapy Significantly Augments Clinical Remission in Quiescent Ulcerative Colitis. Aliment. Pharmacol. Ther. 2013, 38, 761–771. [Google Scholar] [CrossRef] [PubMed]
  268. Hoekman, D.R.; Vlieger, A.M.; Stokkers, P.C.; Mahhmod, N.; Rietdijk, S.; De Boer, N.K.; De Meij, T.G.; Frankenhuis, C.; D’Haens, G.R.; Benninga, M.A. Hypnotherapy for Irritable Bowel Syndrome-Type Symptoms in Patients with Quiescent Inflammatory Bowel Disease: A Randomized, Controlled Trial. J. Crohns Colitis 2021, 15, 1106–1113. [Google Scholar] [CrossRef] [PubMed]
  269. Peters, S.L.; Muir, J.G.; Gibson, P.R. Review Article: Gut-Directed Hypnotherapy in the Management of Irritable Bowel Syndrome and Inflammatory Bowel Disease. Aliment. Pharmacol. Ther. 2015, 41, 1104–1115. [Google Scholar] [CrossRef] [PubMed]
  270. Gavrilescu, O.; Prelipcean, C.C.; Dranga, M.; Soponaru, C.; Mihai, C. The Specialized Educational and Psychological Counseling in Inflammatory Bowel Disease Patients—A Target or a Challenge? Turk. J. Gastroenterol. 2020, 31, 760–766. [Google Scholar] [CrossRef]
  271. Gerbarg, P.L.; Jacob, V.E.; Stevens, L.; Bosworth, B.P.; Chabouni, F.; Defilippis, E.M.; Warren, R.; Trivellas, M.; Patel, P.V.; Webb, C.D.; et al. The Effect of Breathing, Movement, and Meditation on Psychological and Physical Symptoms and Inflammatory Biomarkers in Inflammatory Bowel Disease: A Randomized Controlled Trial. Inflamm. Bowel Dis. 2015, 21, 2886–2896. [Google Scholar] [CrossRef] [PubMed]
  272. Arruda, J.M.; Bogetz, A.L.; Vellanki, S.; Wren, A.; Yeh, A.M. Yoga as Adjunct Therapy for Adolescents with Inflammatory Bowel Disease: A Pilot Clinical Trial. Complement. Ther. Med. 2018, 41, 99–104. [Google Scholar] [CrossRef] [PubMed]
  273. Wilke, E.; Reindl, W.; Thomann, P.A.; Ebert, M.P.; Wuestenberg, T.; Thomann, A.K. Effects of Yoga in Inflammatory Bowel Diseases and on Frequent IBD-Associated Extraintestinal Symptoms like Fatigue and Depression. Complement. Ther. Clin. Pract. 2021, 45, 101465. [Google Scholar] [CrossRef] [PubMed]
  274. Kavuri, V.; Selvan, P.; Tabesh, A.; Raghuram, N.; Selvan, S.R. Remedial Yoga Module Improves Symptoms of Irritable Bowel Syndrome: Replication in the Wait-List Group and Sustained Improvements at 6 Months. Eur. J. Integr. Med. 2015, 7, 609–616. [Google Scholar] [CrossRef]
  275. Daghaghzadeh, H.; Naji, F.; Afshar, H.; Sharbafchi, M.R.; Feizi, A.; Maroufi, M.; Tabatabaeeyan, M.; Adibi, P.; Tavakoli, H. Efficacy of Duloxetine Add on in Treatment of Inflammatory Bowel Disease Patients: A Double-Blind Controlled Study. J. Res. Med. Sci. 2015, 20, 595–601. [Google Scholar] [CrossRef]
  276. Iskandar, H.N.; Cassell, B.; Kanuri, N.; Gyawali, C.P.; Gutierrez, A.; Dassopoulos, T.; Ciorba, M.A.; Sayuk, G.S. Tricyclic Antidepressants for Management of Residual Symptoms in Inflammatory Bowel Disease. J. Clin. Gastroenterol. 2014, 48, 423–429. [Google Scholar] [CrossRef]
  277. Wichniak, A.; Wierzbicka, A.; Walęcka, M.; Jernajczyk, W. Effects of Antidepressants on Sleep. Curr. Psychiatry Rep. 2017, 19, 63. [Google Scholar] [CrossRef] [PubMed]
  278. Hall, B.J.; Hamlin, P.J.; Gracie, D.J.; Ford, A.C. The Effect of Antidepressants on the Course of Inflammatory Bowel Disease. Can. J. Gastroenterol. Hepatol. 2018, 2018, 2047242. [Google Scholar] [CrossRef] [PubMed]
  279. Boicean, A.; Birsan, S.; Ichim, C.; Boeras, I.; Roman-Filip, I.; Blanca, G.; Bacila, C.; Fleaca, R.S.; Dura, H.; Roman-Filip, C. Has-MiR-129-5p’s Involvement in Different Disorders, from Digestive Cancer to Neurodegenerative Diseases. Biomedicines 2023, 11, 2058. [Google Scholar] [CrossRef] [PubMed]
  280. Ho, V.; Baker, J.R.; Willison, K.R.; Barnes, P.J.; Donnelly, L.E.; Klug, D.R. Single Cell Quantification of MicroRNA from Small Numbers of Non-Invasively Sampled Primary Human Cells. Commun. Biol. 2023, 6, 458. [Google Scholar] [CrossRef] [PubMed]
  281. Kalita, A.; Sikora-Skrabaka, M.; Nowakowska-Zajdel, E. Role of Some MicroRNA/ADAM Proteins Axes in Gastrointestinal Cancers as a Novel Biomarkers and Potential Therapeutic Targets—A Review. Curr. Issues Mol. Biol. 2023, 45, 2917–2936. [Google Scholar] [CrossRef]
  282. Ishaq, Y.; Ikram, A.; Alzahrani, B.; Khurshid, S. The Role of MiRNAs, CircRNAs and Their Interactions in Development and Progression of Hepatocellular Carcinoma: An Insilico Approach. Genes 2023, 14, 13. [Google Scholar] [CrossRef] [PubMed]
  283. Balaskas, P.; Goljanek-Whysall, K.; Clegg, P.D.; Fang, Y.; Cremers, A.; Smagul, A.; Welting, T.J.M.; Peffers, M.J. MicroRNA Signatures in Cartilage Ageing and Osteoarthritis. Biomedicines 2023, 11, 1189. [Google Scholar] [CrossRef] [PubMed]
  284. Angele, P.; Zellner, J.; Pattappa, G.; Ho, P.T.B.; Clark, I.M.; Le, L.T.T. MicroRNA-Based Diagnosis and Therapy. Int. J. Mol. Sci. 2022, 23, 7167. [Google Scholar] [CrossRef]
  285. Chandrasekera, P.; Perfetto, M.; Lu, C.; Zhuo, M.; Bahudhanapati, H.; Li, J.; Chen, W.C.; Kulkarni, P.; Christian, L.; Liu, J.; et al. Metalloprotease ADAM9 Cleaves Ephrin-B Ligands and Differentially Regulates Wnt and MTOR Signaling Downstream of Akt Kinase in Colorectal Cancer Cells. J. Biol. Chem. 2022, 298, 102225. [Google Scholar] [CrossRef]
  286. Dobricic, V.; Schilling, M.; Farkas, I.; Gveric, D.O.; Ohlei, O.; Schulz, J.; Middleton, L.; Gentleman, S.M.; Parkkinen, L.; Bertram, L.; et al. Common Signatures of Differential MicroRNA Expression in Parkinson’s and Alzheimer’s Disease Brains. Brain Commun. 2022, 4, fcac274. [Google Scholar] [CrossRef]
  287. Jiang, H.; Zhang, Y.; Yue, J.; Shi, Y.; Xiao, B.; Xiao, W.; Luo, Z. Non-Coding RNAs: The Neuroinflammatory Regulators in Neurodegenerative Diseases. Front. Neurol. 2022, 13, 929290. [Google Scholar] [CrossRef] [PubMed]
  288. Boicean, A.; Birlutiu, V.; Ichim, C.; Anderco, P.; Birsan, S. Fecal Microbiota Transplantation in Inflammatory Bowel Disease. Biomedicines 2023, 11, 1016. [Google Scholar] [CrossRef] [PubMed]
  289. Glassner, K.L.; Abraham, B.P.; Quigley, E.M.M. The Microbiome and Inflammatory Bowel Disease. J. Allergy Clin. Immunol. 2020, 145, 16–27. [Google Scholar] [CrossRef] [PubMed]
  290. Qiu, P.; Ishimoto, T.; Fu, L.; Zhang, J.; Zhang, Z.; Liu, Y. The Gut Microbiota in Inflammatory Bowel Disease. Front. Cell Infect. Microbiol. 2022, 12, 733992. [Google Scholar] [CrossRef] [PubMed]
  291. Popa, D.; Neamtu, B.; Mihalache, M.; Boicean, A.; Banciu, A.; Banciu, D.D.; Moga, D.F.C.; Birlutiu, V. Fecal Microbiota Transplant in Severe and Non-Severe Clostridioides Difficile Infection. Is There a Role of FMT in Primary Severe CDI? J. Clin. Med. 2021, 10, 5822. [Google Scholar] [CrossRef] [PubMed]
  292. Zatorski, H.; Nakov, R. Faecal Microbiota Transplantation in Inflammatory Bowel Disease: Current Concepts and Future Challenges. Curr. Drug Targets 2020, 21, 1440–1447. [Google Scholar] [CrossRef] [PubMed]
  293. Waller, K.M.J.; Leong, R.W.; Paramsothy, S. An Update on Fecal Microbiota Transplantation for the Treatment of Gastrointestinal Diseases. J. Gastroenterol. Hepatol. 2022, 37, 246–255. [Google Scholar] [CrossRef] [PubMed]
  294. Aggarwala, V.; Mogno, I.; Li, Z.; Yang, C.; Britton, G.J.; Chen-Liaw, A.; Mitcham, J.; Bongers, G.; Gevers, D.; Clemente, J.C.; et al. Precise Quantification of Bacterial Strains after Fecal Microbiota Transplantation Delineates Long-Term Engraftment and Explains Outcomes. Nat. Microbiol. 2021, 6, 1309–1318. [Google Scholar] [CrossRef]
  295. Ianiro, G.; Mullish, B.H.; Kelly, C.R.; Sokol, H.; Kassam, Z.; Ng, S.; Fischer, M.; Allegretti, J.R.; Masucci, L.; Zhang, F.; et al. Screening of Faecal Microbiota Transplant Donors during the COVID-19 Outbreak: Suggestions for Urgent Updates from an International Expert Panel. Lancet Gastroenterol. Hepatol. 2020, 5, 430–432. [Google Scholar] [CrossRef]
  296. Kelly, C.R.; Yen, E.F.; Grinspan, A.M.; Kahn, S.A.; Atreja, A.; Lewis, J.D.; Moore, T.A.; Rubin, D.T.; Kim, A.M.; Serra, S.; et al. Fecal Microbiota Transplantation Is Highly Effective in Real-World Practice: Initial Results From the FMT National Registry. Gastroenterology 2021, 160, 183–192.e3. [Google Scholar] [CrossRef]
  297. Michailidis, L.; Currier, A.C.; Le, M.; Flomenhoft, D.R. Adverse Events of Fecal Microbiota Transplantation: A Meta-Analysis of High-Quality Studies. Ann. Gastroenterol. 2021, 34, 802. [Google Scholar] [CrossRef] [PubMed]
  298. Lopetuso, L.R.; Deleu, S.; Godny, L.; Petito, V.; Puca, P.; Facciotti, F.; Sokol, H.; Ianiro, G.; Masucci, L.; Abreu, M.; et al. The First International Rome Consensus Conference on Gut Microbiota and Faecal Microbiota Transplantation in Inflammatory Bowel Disease. Gut 2023, 72, 1642–1650. [Google Scholar] [CrossRef] [PubMed]
  299. Santos, J.; Saperas, E.; Nogueiras, C.; Mourelle, M.; Antolin, M.; Cadahia, A.; Malagelada, J.R. Release of Mast Cell Mediators into the Jejunum by Cold Pain Stress in Humans. Gastroenterology 1998, 114, 640–648. [Google Scholar] [CrossRef] [PubMed]
  300. Rao, S.S.C.; Hatfield, R.A.; Suls, J.M.; Chamberlain, M.J. Psychological and Physical Stress Induce Differential Effects on Human Colonic Motility. Am. J. Gastroenterol. 1998, 93, 985–990. [Google Scholar] [CrossRef] [PubMed]
  301. Barclay, G.R.; Turnberg, L.A. Effect of Cold-Induced Pain on Salt and Water Transport in the Human Jejunum. Gastroenterology 1988, 94, 994–998. [Google Scholar] [CrossRef] [PubMed]
  302. Greenwood-Van Meerveld, B.; Gibson, M.S.; Johnson, A.C.; Venkova, K.; Sutkowski-Markmann, D. NK1 Receptor-Mediated Mechanisms Regulate Colonic Hypersensitivity in the Guinea Pig. Pharmacol. Biochem. Behav. 2003, 74, 1005–1013. [Google Scholar] [CrossRef] [PubMed]
  303. Bernstein, M.T.; Targownik, L.E.; Sexton, K.A.; Graff, L.A.; Miller, N.; Walker, J.R. Assessing the Relationship between Sources of Stress and Symptom Changes among Persons with IBD over Time: A Prospective Study. Can. J. Gastroenterol. Hepatol. 2016, 2016. [Google Scholar] [CrossRef] [PubMed]
  304. Crumbock, S.C.; Loeb, S.J.; Fick, D.M. Physical Activity, Stress, Disease Activity, and Quality of Life in Adults with Crohn Disease. Gastroenterol. Nurs. 2009, 32, 188–195. [Google Scholar] [CrossRef]
  305. Sullivan, A.J. Psychogenic Factors in Ulcerative Colitis. Am. J. Dig. Dis. 1935, 2, 651–656. [Google Scholar] [CrossRef]
  306. Duffy, L.C.; Zielezny, M.A.; Marshall, J.R.; Byers, T.E.; Weiser, M.M.; Phillips, J.F.; Calkins, B.M.; Ogra, P.L.; Graham, S. Relevance of Major Stress Events as an Indicator of Disease Activity Prevalence in Inflammatory Bowel Disease. Behav. Med. 1991, 17, 101–110. [Google Scholar] [CrossRef]
  307. Cámara, R.J.A.; Ziegler, R.; Begré, S.; Schoepfer, A.M.; Von Känel, R. The Role of Psychological Stress in Inflammatory Bowel Disease: Quality Assessment of Methods of 18 Prospective Studies and Suggestions for Future Research. Digestion 2009, 80, 129–139. [Google Scholar] [CrossRef]
  308. Keefer, L.; Keshavarzian, A.; Mutlu, E. Reconsidering the Methodology of “Stress” Research in Inflammatory Bowel Disease. J. Crohns Colitis 2008, 2, 193–201. [Google Scholar] [CrossRef]
  309. Szigethy, E.M.; Allen, J.I.; Reiss, M.; Cohen, W.; Perera, L.P.; Brillstein, L.; Cross, R.K.; Schwartz, D.A.; Kosinski, L.R.; Colton, J.B.; et al. White Paper AGA: The Impact of Mental and Psychosocial Factors on the Care of Patients With Inflammatory Bowel Disease. Clin. Gastroenterol. Hepatol. 2017, 15, 986–997. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of the brain–gut axis.
Figure 1. Schematic representation of the brain–gut axis.
Jcm 13 01361 g001
Figure 2. Comparison of psychological stress and lifestyle factors before and during lockdown in IBD patients.
Figure 2. Comparison of psychological stress and lifestyle factors before and during lockdown in IBD patients.
Jcm 13 01361 g002
Figure 3. Psychosocial variables (catastrophizing, illness stigma, illness uncertainty, and illness shame). Legend: solid lines represent significant effects, dotted lines represent nonsignificant effects; green represents indirect effects, and blue represents direct effects.
Figure 3. Psychosocial variables (catastrophizing, illness stigma, illness uncertainty, and illness shame). Legend: solid lines represent significant effects, dotted lines represent nonsignificant effects; green represents indirect effects, and blue represents direct effects.
Jcm 13 01361 g003
Table 1. Research exploring the correlation between stress and the composition of the gut microbiota.
Table 1. Research exploring the correlation between stress and the composition of the gut microbiota.
FactorAuthor(s)Type of StudyNIntervention/MethodologyResult
Pro-/prebioticsMessaoudi, M. et al. [107]In vivo rat study36 ratsProbiotic formulationObserved anxiolytic-like effects in rat models
Garcia-Rodenas, CL.
et al. [108]
In vivo rat study84Maternal separation and
prebiotics/probiotics/LC-PUFA
Implementation of a nutritional intervention during the weaning period resulted in the normalization of gut permeability and the restoration of the growth rate
Zareie, M. et al. [109]In vivo rat studyFour to five rats per groupWAS and probioticsProbiotics exhibited a preventive effect on chronic stress-induced gastrointestinal abnormalities
Li, N. et al. [110]In vivo mouse studyEight mice per groupChronic mild stress and
probiotics
Reduced levels of pro-inflammatory cytokines and modified stress-induced behavioral patterns
Bravo, J.A. et al. [111]In vivo mouse study36Probiotic formulationHighlighted the significance of probiotics in the bidirectional communication between the gut and the brain in stress-related disorders
Messaoudi, M. et al. [107]Double-blind, placebo-controlled, randomized parallel group study66 individualsProbiotic formulationEvidenced favorable psychological outcomes in a cohort of healthy human volunteers
Rao, S. et al. [112]Systematic review11 RCTsPrebiotic supplementationDemonstrated transient advantageous effects on the composition of the intestinal microbiota in the short term
Prenatal/early life stressO’Mahony, S.M. et al. [113]In vivo rat study22Maternal separationThe effects of early life stress on the gut–brain axis led to modifications that contributed to the manifestation of symptoms in IBD
Golubeva, A.V. et al. [114]In vivo rat study6–10 per groupPrenatal stressPersistent modifications in the composition of the intestinal microbiota over an extended period
Jasarevic, E. et al. [115]In vivo mouse study21–23 mice per groupPrenatal stressChanges in the vaginal microbiota were implicated in the process of reprogramming the developing brain
Xie, R. et al. [116]In vivo mouse study6–20 mice per groupMaternal high-fat dietIntestinal dysbiosis and the presence of chronic low-grade inflammation in the gastrointestinal tract
Bailey, M.T. et al. [117]In vivo primate
study
20Maternal separationPsychological disturbances resulting from maternal separation led to modifications in the composition of the intestinal microflora
Zijlmans, M.A. et al. [118]Longitudinal clinical study192 childrenQuestionnaireThe presence of prenatal stress was correlated with specific microbial colonization patterns in infants
Chronic/social/environmental stressSoderholm, J.D. et al. [119]In vivo rat studySeven to eight rats per groupWASImpaired mucosal defenses against luminal bacteria lead to intestinal inflammation
Da Silva, S. et al. [120]In vivo rat study13–14 rats per groupWASModified composition of the intestinal mucus
Meddings, J.B. et al. [121]In vivo rat studyNot specifiedStress inductionElevated gastrointestinal permeability facilitates the passage of luminal constituents to the mucosal immune system
Saunders, P.R. et al. [122]In vivo rat study6Cold-restraint stress or WASIntensified intestinal inflammation resulting from enhanced uptake of immunogenic substances
Santos, J. et al. [123]In vivo rat studyFour rats per groupWASEpithelial mitochondrial damage triggered by stress and activation of mucosal mast cells
Gao, X. et al. [124]In vivo mouse studyFour to six mice per groupChronic restraint stressDisrupted gut microbiota followed by immune system activation resulted in the development of colitis
Neufeld, K.M. et al. [125]In vivo mouse study12 mice per groupGerm-free and specific-pathogen-freeThe presence of typical intestinal microbiota played a role in the development of behavior
Donnet-Hughes, A. et al. [126]In vivo mouse study10 mice per groupLactationCellular transfer of bacterial translocation took place in mice during pregnancy and lactation
Heijtz, R. et al. [127]In vivo mouse study7–14 mice per groupGerm-free and specific-pathogen-freeThe gut microbiota impacted the development of the mammalian brain and subsequent behavioral patterns in adulthood
Marin, I.A. et al. [128]In vivo mouse study10–12 (three independent experiments)Unpredictable chronic mild stressModified composition of the intestinal microbiota, particularly within the lactobacillus component
Bharwani, A. et al. [129]In vivo mouse study7–20 mice per groupChronic social defeatStress triggered intricate structural alterations in the gut microbiota
Sudo, N. et al. [36]In vivo mouse study18–24 mice per groupGerm-free and specific-pathogen-free; acute restraint stressThe commensal microbiota had the potential to influence the postnatal maturation of the hypothalamic–pituitary–adrenal (HPA) stress response
Galley, J.D. et al. [130]In vivo mouse studyFive mice per groupSDRAltered microbial populations that had a close association with the colonic mucosa
Bailey, M.T. et al. [33]In vivo mouse studyFive mice per groupSDRStress resulted in notable alterations in the colonization of the intestinal microbiota
Noguera, J.C. et al. [131]Field experiment in wild birds64Corticosterone implantModified gut microbiome in birds living in their natural habitat
Van der Zaag-Loonen, H.J. et al. [132]Clinical study65Coping style instrumentAdolescents with IBD exhibited a higher utilization of avoidant coping strategies in comparison with healthy individuals
Walker, L.S. et al. [133]Clinical study263Daily interview assessmentThere was an association between stress and the occurrence of digestive problems and disruptions in gastrointestinal health
Maes, M. et al. [134]Clinical study40DepressionElevated bacterial translocation and heightened immune responses targeting commensal bacteria
HPA = hypothalamic–pituitary–adrenal axis; LC-PUFAs = long-chain poly-unsaturated fatty acids; RCT = randomized controlled trial; SDR = social disruption; WAS = water-avoidance stress.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Belei, O.; Basaca, D.-G.; Olariu, L.; Pantea, M.; Bozgan, D.; Nanu, A.; Sîrbu, I.; Mărginean, O.; Enătescu, I. The Interaction between Stress and Inflammatory Bowel Disease in Pediatric and Adult Patients. J. Clin. Med. 2024, 13, 1361. https://doi.org/10.3390/jcm13051361

AMA Style

Belei O, Basaca D-G, Olariu L, Pantea M, Bozgan D, Nanu A, Sîrbu I, Mărginean O, Enătescu I. The Interaction between Stress and Inflammatory Bowel Disease in Pediatric and Adult Patients. Journal of Clinical Medicine. 2024; 13(5):1361. https://doi.org/10.3390/jcm13051361

Chicago/Turabian Style

Belei, Oana, Diana-Georgiana Basaca, Laura Olariu, Manuela Pantea, Daiana Bozgan, Anda Nanu, Iuliana Sîrbu, Otilia Mărginean, and Ileana Enătescu. 2024. "The Interaction between Stress and Inflammatory Bowel Disease in Pediatric and Adult Patients" Journal of Clinical Medicine 13, no. 5: 1361. https://doi.org/10.3390/jcm13051361

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop