Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (45)

Search Parameters:
Keywords = neuroendocrine-immune interaction

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
28 pages, 872 KB  
Review
The Pathobiological Underpinnings of Psychosis: From the Stress-Related Hypothesis to a Multisystemic Approach
by Evangelos Karanikas
NeuroSci 2025, 6(4), 99; https://doi.org/10.3390/neurosci6040099 - 3 Oct 2025
Viewed by 368
Abstract
Until recently, research on the pathobiological substrate of psychosis has been focused on neurotransmitter perturbations. However, this scope has expanded to include new fields, such as the immune/redox/metabolic/neuroendocrine/stress systems. Indeed, basic research in the stress field showed that the systems above can represent [...] Read more.
Until recently, research on the pathobiological substrate of psychosis has been focused on neurotransmitter perturbations. However, this scope has expanded to include new fields, such as the immune/redox/metabolic/neuroendocrine/stress systems. Indeed, basic research in the stress field showed that the systems above can represent components of a general inflammatory process as tightly interconnected as a Gordian knot. Based on the inflammatory hypothesis concerning the psychosis etiopathology, the findings from psychotic cohort studies on each one of the immune/redox/metabolic/neuroendocrine/stress systems have started to accumulate. The evidence favors the involvement of these systems in the formation of the pathobiological psychotic substrate, yet little is known concerning their interplay. This review attempts to establish a frame of reference for the evidence concerning intersystemic interactions, starting with the basic research on the stress field and expanding to clinical studies with psychosis cohorts, hoping to instigate new avenues of research. Full article
Show Figures

Figure 1

20 pages, 994 KB  
Perspective
Endocrinology and the Lung: Exploring the Bidirectional Axis and Future Directions
by Pedro Iglesias
J. Clin. Med. 2025, 14(19), 6985; https://doi.org/10.3390/jcm14196985 - 2 Oct 2025
Viewed by 417
Abstract
The lung is increasingly recognized as an organ with dual endocrine and respiratory roles, participating in a complex bidirectional crosstalk with systemic hormones and local/paracrine activity. Endocrine and paracrine pathways regulate lung development, ventilation, immunity, and repair, while pulmonary cells express hormone receptors [...] Read more.
The lung is increasingly recognized as an organ with dual endocrine and respiratory roles, participating in a complex bidirectional crosstalk with systemic hormones and local/paracrine activity. Endocrine and paracrine pathways regulate lung development, ventilation, immunity, and repair, while pulmonary cells express hormone receptors and secrete mediators with both local and systemic effects, defining the concept of the “endocrine lung”. This narrative review summarizes current evidence on the endocrine–pulmonary axis. Thyroid hormones, glucocorticoids, sex steroids, and metabolic hormones (e.g., insulin, leptin, adiponectin) critically influence alveologenesis, surfactant production, ventilatory drive, airway mechanics, and immune responses. Conversely, the lung produces mediators such as serotonin, calcitonin gene-related peptide, endothelin-1, leptin, and keratinocyte growth factor, which regulate vascular tone, alveolar homeostasis, and immune modulation. We also describe the respiratory manifestations of major endocrine diseases, including obstructive sleep apnea and lung volume alterations in acromegaly, immunosuppression and myopathy in Cushing’s syndrome, hypoventilation in hypothyroidism, restrictive “diabetic lung”, and obesity-related phenotypes. In parallel, chronic pulmonary diseases such as chronic obstructive pulmonary disease, interstitial lung disease, and sleep apnea profoundly affect endocrine axes, promoting insulin resistance, hypogonadism, GH/IGF-1 suppression, and bone metabolism alterations. Pulmonary neuroendocrine tumors further highlight the interface, frequently presenting with paraneoplastic endocrine syndromes. Finally, therapeutic interactions are discussed, including the risks of hypothalamic–pituitary–adrenal axis suppression with inhaled corticosteroids, immunotherapy-induced endocrinopathies, and inhaled insulin. Future perspectives emphasize mapping pulmonary hormone networks, endocrine phenotyping of chronic respiratory diseases, and developing hormone-based interventions. Full article
(This article belongs to the Section Endocrinology & Metabolism)
Show Figures

Figure 1

20 pages, 1362 KB  
Opinion
From Microbial Consortia to Ecosystem Resilience: The Integrative Roles of Holobionts in Stress Biology
by Maximino Manzanera
Biology 2025, 14(9), 1203; https://doi.org/10.3390/biology14091203 - 6 Sep 2025
Viewed by 490
Abstract
The holobiont paradigm, conceptualizing host–microbiome assemblages as functionally integrated entities, has fundamentally altered interpretations of adaptive responses to environmental pressures spanning multiple organizational levels. This review synthesizes the current knowledge on microbiome-host coevolution, focusing on three key aspects. First, it examines the evolutionary [...] Read more.
The holobiont paradigm, conceptualizing host–microbiome assemblages as functionally integrated entities, has fundamentally altered interpretations of adaptive responses to environmental pressures spanning multiple organizational levels. This review synthesizes the current knowledge on microbiome-host coevolution, focusing on three key aspects. First, it examines the evolutionary origins of holobionts from primordial microbial consortia. Second, it considers the mechanistic basis of microbiome-mediated stress resilience in plants and animals. Finally, it explores the ecological implications of inter-holobiont interactions. We highlight how early microbial alliances (protomicrobiomes) laid the groundwork for eukaryotic complexity through metabolic cooperation, with modern holobionts retaining this plasticity to confront abiotic and biotic stressors. In plants, compartment-specific microbiomes (e.g., rhizosphere, phyllosphere) enhance drought tolerance or nutrient acquisition, while in animals, the gut microbiome modulates neuroendocrine and immune functions via multi-organ axes (gut–brain, gut–liver, etc.). Critically, we emphasize the role of microbial metabolites (e.g., short-chain fatty acids, VOCs) as universal signaling molecules that coordinate holobiont responses to environmental change. Emerging strategies, like microbiome engineering and probiotics, are discussed as tools to augment stress resilience in agriculture and medicine. By framing adaptation as a collective trait of the holobiont, this work bridges evolutionary biology, microbiology, and ecology to offer a unified perspective on stress biology. Full article
Show Figures

Figure 1

29 pages, 14681 KB  
Article
Single-Nucleus RNA Sequencing and Spatial Transcriptomics Reveal Cellular Heterogeneity and Intercellular Communication Networks in the Hypothalamus–Pituitary–Ovarian Axis of Pregnant Mongolian Cattle
by Yanchun Bao, Fengying Ma, Chenxi Huo, Hongxia Jia, Yunhan Li, Xiaoyi Yang, Jiajing Liu, Pengbo Gu, Caixia Shi, Mingjuan Gu, Lin Zhu, Yu Wang, Bin Liu, Risu Na and Wenguang Zhang
Animals 2025, 15(15), 2277; https://doi.org/10.3390/ani15152277 - 4 Aug 2025
Viewed by 914
Abstract
The hypothalamus–pituitary–ovarian (HPO) axis orchestrates reproductive functions through intricate neuroendocrine crosstalk. Here, we integrated single-nucleus RNA sequencing (snRNA-seq) and spatial transcriptomics (ST) to decode the cellular heterogeneity and intercellular communication networks in the reproductive systems of pregnant Mongolian cattle. We retained a total [...] Read more.
The hypothalamus–pituitary–ovarian (HPO) axis orchestrates reproductive functions through intricate neuroendocrine crosstalk. Here, we integrated single-nucleus RNA sequencing (snRNA-seq) and spatial transcriptomics (ST) to decode the cellular heterogeneity and intercellular communication networks in the reproductive systems of pregnant Mongolian cattle. We retained a total of 6161 high-quality nuclei from the hypothalamus, 14,715 nuclei from the pituitary, and 26,072 nuclei from the ovary, providing a comprehensive cellular atlas across the HPO axis. In the hypothalamus, neurons exhibited synaptic and neuroendocrine specialization, with glutamatergic subtype Glut4 serving as a TGFβ signaling hub to regulate pituitary feedback, while GABAergic GABA1 dominated PRL signaling, likely adapting maternal behavior. Pituitary stem cells dynamically replenished endocrine populations via TGFβ, and lactotrophs formed a PRLPRLR paracrine network with stem cells, synergizing mammary development. Ovarian luteal cells exhibited steroidogenic specialization and microenvironmental synergy: endothelial cells coregulated TGFβ-driven angiogenesis and immune tolerance, while luteal–stromal PRLPRLR interactions amplified progesterone synthesis and nutrient support. Granulosa cells (GCs) displayed spatial-functional stratification, with steroidogenic GCs persisting across pseudotime as luteinization precursors, while atretic GCs underwent apoptosis. Spatial mapping revealed GCs’ annular follicular distribution, mediating oocyte–somatic crosstalk, and luteal–endothelial colocalization supporting vascularization. This study unveils pregnancy-specific HPO axis regulation, emphasizing multi-organ crosstalk through TGFβ/PRL pathways and stem cell-driven plasticity, offering insights into reproductive homeostasis and pathologies. Full article
(This article belongs to the Section Cattle)
Show Figures

Figure 1

28 pages, 1763 KB  
Review
Interaction Between Konjac Glucomannan and Gut Microbiota and Its Impact on Health
by Yufen Yu, Shuo Jin, Yi Yang, Xiaodong Han, Rongfa Guan and Hao Zhong
Biology 2025, 14(8), 923; https://doi.org/10.3390/biology14080923 - 23 Jul 2025
Viewed by 2568
Abstract
Konjac glucomannan (KGM) is a natural polysaccharide polymer. It is degraded by gut microbiota-derived β-mannanase into small-molecule nutrients, which exert diverse physiological regulatory effects. As a prebiotic, KGM modulates gut microbiota composition. It selectively fosters the proliferation of beneficial commensals and suppresses potential [...] Read more.
Konjac glucomannan (KGM) is a natural polysaccharide polymer. It is degraded by gut microbiota-derived β-mannanase into small-molecule nutrients, which exert diverse physiological regulatory effects. As a prebiotic, KGM modulates gut microbiota composition. It selectively fosters the proliferation of beneficial commensals and suppresses potential pathogens, thereby alleviating microbiota-related disorders. Moreover, microbiota fermentation of KGM produces metabolites. Short-chain fatty acids (SCFAs) are particularly notable among these metabolites. They exert multifaceted beneficial effects, including metabolic regulation, intestinal barrier strengthening, and neuroprotective functions. These effects are mediated through inhibition of inflammatory pathways (e.g., NF-κB, MAPK), modulation of lipid metabolism genes (e.g., CD36), and regulation of neurotransmitters (e.g., GABA, 5-HT). This highlights KGM’s therapeutic potential for metabolic, inflammatory, and neurodegenerative diseases. Current clinical use is limited by dose-dependent adverse effects and interindividual response variability, which stem from different microbial communities. This necessitates personalized dosage strategies. Despite these limitations, KGM as a prebiotic polysaccharide exhibits multifaceted bioactivity. Current evidence suggests its potential to synergistically modulate metabolic pathways, gut microbiota composition, immune cell signaling, and neuroendocrine interactions. This highlights its promise for developing novel therapeutic interventions. Full article
(This article belongs to the Special Issue Gut Microbiome in Health and Disease (2nd Edition))
Show Figures

Figure 1

19 pages, 340 KB  
Review
The Role of Selected Proteins in the Pathogenesis of Psoriasis
by Mateusz Matwiejuk, Agnieszka Kulczyńska-Przybik, Hanna Myśliwiec, Adrian Chabowski, Barbara Mroczko and Iwona Flisiak
Int. J. Mol. Sci. 2025, 26(13), 6475; https://doi.org/10.3390/ijms26136475 - 4 Jul 2025
Viewed by 1009
Abstract
Psoriasis is a chronic, immune-mediated inflammatory skin disease with complex genetic, environmental, and immunological determinants. Beyond the skin, it affects multiple systems, including the joints and cardiovascular system. A hallmark of psoriasis is an overactivation of the innate and adaptive immune responses, leading [...] Read more.
Psoriasis is a chronic, immune-mediated inflammatory skin disease with complex genetic, environmental, and immunological determinants. Beyond the skin, it affects multiple systems, including the joints and cardiovascular system. A hallmark of psoriasis is an overactivation of the innate and adaptive immune responses, leading to dysregulated cytokine signaling, altered keratinocyte function, and aberrant expression of structural and regulatory proteins. In recent years, growing attention has been given to the skin as a neuro–immuno–endocrine organ, with evidence showing the role of stress-related neuropeptides, UVB-induced immune modulation, and vitamin D signaling in the disease pathogenesis. This review highlights emerging evidence on key multifunctional proteins—elafin, chemerin, and NAMPT (visfatin)—that exert both pro- and anti-inflammatory actions. Although still underexplored, these molecules appear to contribute significantly to the psoriatic microenvironment by modulating inflammation, immunity, and skin barrier function. Their dual roles suggest complex interactions within the cutaneous immune–neuroendocrine network, positioning them as potential biomarkers or therapeutic targets in psoriasis. By integrating insights into classical and emerging mediators, this review aims to provide a comprehensive perspective on the evolving landscape of psoriasis pathophysiology. Full article
(This article belongs to the Special Issue Molecular Mechanisms and Therapeutic Targets in Skin Diseases)
28 pages, 933 KB  
Review
Therapeutic Horizons: Gut Microbiome, Neuroinflammation, and Epigenetics in Neuropsychiatric Disorders
by Shabnam Nohesara, Hamid Mostafavi Abdolmaleky, Ahmad Pirani and Sam Thiagalingam
Cells 2025, 14(13), 1027; https://doi.org/10.3390/cells14131027 - 4 Jul 2025
Cited by 1 | Viewed by 1394
Abstract
Neuroinflammation is a hallmark of many neuropsychiatric disorders (NPD), which are among the leading causes of disability worldwide. Emerging evidence highlights the significant role of the gut microbiota (GM)–immune system–brain axis in neuroinflammation and the pathogenesis of NPD, primarily through epigenetic mechanisms. Gut [...] Read more.
Neuroinflammation is a hallmark of many neuropsychiatric disorders (NPD), which are among the leading causes of disability worldwide. Emerging evidence highlights the significant role of the gut microbiota (GM)–immune system–brain axis in neuroinflammation and the pathogenesis of NPD, primarily through epigenetic mechanisms. Gut microbes and their metabolites influence immune cell activity and brain function, thereby contributing to neuroinflammation and the development and progression of NPD. The enteric nervous system, the autonomic nervous system, neuroendocrine signaling, and the immune system all participate in bidirectional communication between the gut and the brain. Importantly, the interaction of each of these systems with the GM influences epigenetic pathways. Here, we first explore the intricate relationship among intestinal microbes, microbial metabolites, and immune cell activity, with a focus on epigenetic mechanisms involved in NPD pathogenesis. Next, we provide background information on the association between inflammation and epigenetic aberrations in the context of NPD. Additionally, we review emerging therapeutic strategies—such as prebiotics, probiotics, methyl-rich diets, ketogenic diet, and medications—that may modulate the GM–immune system–brain axis via epigenetic regulation for the prevention or treatment of NPD. Finally, we discuss the challenges and future directions in investigating the critical role of this axis in mental health. Full article
Show Figures

Figure 1

15 pages, 1361 KB  
Review
Gut Microbiome Dysbiosis and Its Impact on Reproductive Health: Mechanisms and Clinical Applications
by Efthalia Moustakli, Sofoklis Stavros, Periklis Katopodis, Anastasios Potiris, Peter Drakakis, Stefanos Dafopoulos, Athanasios Zachariou, Konstantinos Dafopoulos, Konstantinos Zikopoulos and Athanasios Zikopoulos
Metabolites 2025, 15(6), 390; https://doi.org/10.3390/metabo15060390 - 11 Jun 2025
Cited by 3 | Viewed by 2278
Abstract
The human gut microbiome is integral to maintaining systemic physiological balance, with accumulating evidence emphasizing its critical role in reproductive health. This review investigates the bidirectional interactions between the gut microbiota and the female reproductive system, mediated by neuroendocrine, immune, and metabolic pathways, [...] Read more.
The human gut microbiome is integral to maintaining systemic physiological balance, with accumulating evidence emphasizing its critical role in reproductive health. This review investigates the bidirectional interactions between the gut microbiota and the female reproductive system, mediated by neuroendocrine, immune, and metabolic pathways, constituting the gut–reproductive axis. Dysbiosis, characterized by microbial imbalance, has been linked to reproductive disorders such as polycystic ovary syndrome (PCOS), endometriosis, infertility, impaired spermatogenesis, and pregnancy complications. These associations can be explained by immunological dysregulation, systemic inflammation, altered sex hormone metabolism, and hypothalamic–pituitary–gonadal (HPG) axis disturbances. This review aims to clarify the molecular and cellular mechanisms underpinning gut–reproductive interactions and to evaluate the feasibility of microbiome-targeted therapies as clinical interventions for improving reproductive outcomes. Full article
(This article belongs to the Section Endocrinology and Clinical Metabolic Research)
Show Figures

Figure 1

18 pages, 11122 KB  
Article
Serotonin Signaling and Macrophage Subsets in Goldfish Gills: Unraveling the Neuroimmune Network for Gill Homeostasis
by Manal T. Hussein, Giacomo Zaccone, Marco Albano, Alessio Alesci, Sebastian Marino, Rasha Alonaizan and Doaa M. Mokhtar
Life 2025, 15(5), 751; https://doi.org/10.3390/life15050751 - 7 May 2025
Cited by 2 | Viewed by 792
Abstract
Goldfish (Carassius auratus) gills function as both respiratory and immune-regulatory organs, integrating neuroendocrine and immune responses to environmental stimuli. This study explores the spatial organization and interaction of neuroendocrine cells (NECs) and immune cells within goldfish gills using confocal immunohistochemistry and [...] Read more.
Goldfish (Carassius auratus) gills function as both respiratory and immune-regulatory organs, integrating neuroendocrine and immune responses to environmental stimuli. This study explores the spatial organization and interaction of neuroendocrine cells (NECs) and immune cells within goldfish gills using confocal immunohistochemistry and transmission electron microscopy. NECs, identified near blood capillaries and nerve fibers, highlight their role in environmental sensing and physiological regulation. These cells express serotonin (5-HT), a neurotransmitter critical for neuroimmune communication. Two distinct macrophage subsets were observed: iNOS-positive macrophages, concentrated in the basal epithelium, suggest a pro-inflammatory role, whereas 5-HT-positive macrophages, dispersed in the subepithelium, likely contribute to immune modulation. The co-localization of MHC-II and CD68 in macrophages further supports an active antigen-processing system in the gills. Ultrastructural analysis revealed diverse immune cells, including rodlet cells, telocytes, and lymphocytes, within the gill epithelium. Telocytes formed intricate networks with immune cells, highlighting their role in immune coordination and tissue homeostasis. These findings provide new insights into the neuroimmune interactions in fish gills, contributing to a broader understanding of aquatic immune systems and environmental adaptability. Full article
(This article belongs to the Special Issue Feature Papers in Animal Science: 4th Edition)
Show Figures

Figure 1

15 pages, 1143 KB  
Article
Drug Administration Before or After Exposure to Low Temperatures—Does It Matter for the Therapeutic Effect?
by Kadir Bezirci, Boryana Borisova, Konstantinos Papadakis, Dancho Danalev and Hristina Nocheva
Int. J. Mol. Sci. 2025, 26(8), 3883; https://doi.org/10.3390/ijms26083883 - 19 Apr 2025
Viewed by 738
Abstract
The adaptation of the body when exposed to a lower-than-usual temperature is a challenge that involves neuro-endocrine–immune mechanisms and affects the pharmacokinetics and/or pharmacodynamics of drugs taken before or after cold exposure. The experiments presented in this study clearly show differences in the [...] Read more.
The adaptation of the body when exposed to a lower-than-usual temperature is a challenge that involves neuro-endocrine–immune mechanisms and affects the pharmacokinetics and/or pharmacodynamics of drugs taken before or after cold exposure. The experiments presented in this study clearly show differences in the analgesic effect of an exogenously introduced model substance (C-terminal fragment of calcium-binding protein, spermatid-specific 1) before and after cold exposure compared to its effect at an ambient temperature. The model substance used for the experiments is an octapeptide, TDIFELLK, which was synthesized via standard solid-phase peptide synthesis. Preliminary studies proved TDIFELLK’s analgesic activity. The ANOVA analysis performed showed statistically significant differences in the pain thresholds, measured by a paw pressure test, in 109 rats distributed among 14 groups and subjected to cold exposure according to different set-ups. Cold exposure immediately after TDIFELLK administration appears to enhance its analgesic effect, while cold exposure before administration reduces the effect. In some of the set-ups, antagonists of the most significant for analgesia receptors, i.e., opioid, cannabinoid, and serotonergic, were also introduced. The results showed that cold exposure had a modulating influence on the effect of the exogenously administered substances. The modulating effect was manifested differently depending on whether the intake occurred before or after cold exposure. The results also showed that the interaction with individual mediator systems was also subjected to differences depending on intake occurring before and after cold exposure. Full article
Show Figures

Figure 1

41 pages, 2291 KB  
Review
Understanding Merkel Cell Carcinoma: Pathogenic Signaling, Extracellular Matrix Dynamics, and Novel Treatment Approaches
by Maria Konstantaraki, Aikaterini Berdiaki, Monica Neagu, Sabina Zurac, Konstantinos Krasagakis and Dragana Nikitovic
Cancers 2025, 17(7), 1212; https://doi.org/10.3390/cancers17071212 - 2 Apr 2025
Viewed by 3471
Abstract
Merkel cell carcinoma (MCC) is a rare but aggressive neuroendocrine skin cancer, driven by either Merkel cell polyomavirus (MCPyV) integration or ultraviolet (UV)-induced mutations. In MCPyV-positive tumors, viral T antigens inactivate tumor suppressors pRb and p53, while virus-negative MCCs harbor UV-induced mutations that [...] Read more.
Merkel cell carcinoma (MCC) is a rare but aggressive neuroendocrine skin cancer, driven by either Merkel cell polyomavirus (MCPyV) integration or ultraviolet (UV)-induced mutations. In MCPyV-positive tumors, viral T antigens inactivate tumor suppressors pRb and p53, while virus-negative MCCs harbor UV-induced mutations that activate similar oncogenic pathways. Key signaling cascades, including PI3K/AKT/mTOR and MAPK, support tumor proliferation, survival, and resistance to apoptosis. Histologically, MCC consists of small round blue cells with neuroendocrine features, high mitotic rate, and necrosis. The tumor microenvironment (TME) plays a central role in disease progression and immune escape. It comprises a mix of tumor-associated macrophages, regulatory and cytotoxic T cells, and elevated expression of immune checkpoint molecules such as PD-L1, contributing to an immunosuppressive niche. The extracellular matrix (ECM) within the TME is rich in proteoglycans, collagens, and matrix metalloproteinases (MMPs), facilitating tumor cell adhesion, invasion, and interaction with stromal and immune cells. ECM remodeling and integrin-mediated signaling further promote immune evasion and therapy resistance. Although immune checkpoint inhibitors targeting PD-1/PD-L1 have shown promise in treating MCC, resistance remains a major hurdle. Therapeutic strategies that concurrently target the TME—through inhibition of ECM components, MMPs, or integrin signaling—may enhance immune responses and improve clinical outcomes. Full article
(This article belongs to the Special Issue Insights from the Editorial Board Member)
Show Figures

Figure 1

85 pages, 24685 KB  
Review
Adaptogens in Long-Lasting Brain Fatigue: An Insight from Systems Biology and Network Pharmacology
by Alexander Panossian, Terrence Lemerond and Thomas Efferth
Pharmaceuticals 2025, 18(2), 261; https://doi.org/10.3390/ph18020261 - 15 Feb 2025
Cited by 4 | Viewed by 10794
Abstract
Long-lasting brain fatigue is a consequence of stroke or traumatic brain injury associated with emotional, psychological, and physical overload, distress in hypertension, atherosclerosis, viral infection, and aging-related chronic low-grade inflammatory disorders. The pathogenesis of brain fatigue is linked to disrupted neurotransmission, the glutamate-glutamine [...] Read more.
Long-lasting brain fatigue is a consequence of stroke or traumatic brain injury associated with emotional, psychological, and physical overload, distress in hypertension, atherosclerosis, viral infection, and aging-related chronic low-grade inflammatory disorders. The pathogenesis of brain fatigue is linked to disrupted neurotransmission, the glutamate-glutamine cycle imbalance, glucose metabolism, and ATP energy supply, which are associated with multiple molecular targets and signaling pathways in neuroendocrine-immune and blood circulation systems. Regeneration of damaged brain tissue is a long-lasting multistage process, including spontaneously regulating hypothalamus-pituitary (HPA) axis-controlled anabolic–catabolic homeostasis to recover harmonized sympathoadrenal system (SAS)-mediated function, brain energy supply, and deregulated gene expression in rehabilitation. The driving mechanism of spontaneous recovery and regeneration of brain tissue is a cross-talk of mediators of neuronal, microglia, immunocompetent, and endothelial cells collectively involved in neurogenesis and angiogenesis, which plant adaptogens can target. Adaptogens are small molecules of plant origin that increase the adaptability of cells and organisms to stress by interaction with the HPA axis and SAS of the stress system (neuroendocrine-immune and cardiovascular complex), targeting multiple mediators of adaptive GPCR signaling pathways. Two major groups of adaptogens comprise (i) phenolic phenethyl and phenylpropanoid derivatives and (ii) tetracyclic and pentacyclic glycosides, whose chemical structure can be distinguished as related correspondingly to (i) monoamine neurotransmitters of SAS (epinephrine, norepinephrine, and dopamine) and (ii) steroid hormones (cortisol, testosterone, and estradiol). In this narrative review, we discuss (i) the multitarget mechanism of integrated pharmacological activity of botanical adaptogens in stress overload, ischemic stroke, and long-lasting brain fatigue; (ii) the time-dependent dual response of physiological regulatory systems to adaptogens to support homeostasis in chronic stress and overload; and (iii) the dual dose-dependent reversal (hormetic) effect of botanical adaptogens. This narrative review shows that the adaptogenic concept cannot be reduced and rectified to the various effects of adaptogens on selected molecular targets or specific modes of action without estimating their interactions within the networks of mediators of the neuroendocrine-immune complex that, in turn, regulates other pharmacological systems (cardiovascular, gastrointestinal, reproductive systems) due to numerous intra- and extracellular communications and feedback regulations. These interactions result in polyvalent action and the pleiotropic pharmacological activity of adaptogens, which is essential for characterizing adaptogens as distinct types of botanicals. They trigger the defense adaptive stress response that leads to the extension of the limits of resilience to overload, inducing brain fatigue and mental disorders. For the first time, this review justifies the neurogenesis potential of adaptogens, particularly the botanical hybrid preparation (BHP) of Arctic Root and Ashwagandha, providing a rationale for potential use in individuals experiencing long-lasting brain fatigue. The review provided insight into future research on the network pharmacology of adaptogens in preventing and rehabilitating long-lasting brain fatigue following stroke, trauma, and viral infections. Full article
(This article belongs to the Section Natural Products)
Show Figures

Graphical abstract

27 pages, 4623 KB  
Review
Antimicrobial Neuropeptides and Their Receptors: Immunoregulator and Therapeutic Targets for Immune Disorders
by Kaiqi Chen, Xiaojun Wu, Xiaoke Li, Haoxuan Pan, Wenhui Zhang, Jinxi Shang, Yinuo Di, Ruonan Liu, Zhaodi Zheng and Xitan Hou
Molecules 2025, 30(3), 568; https://doi.org/10.3390/molecules30030568 - 27 Jan 2025
Viewed by 2249
Abstract
The interaction between the neuroendocrine system and the immune system plays a key role in the onset and progression of various diseases. Neuropeptides, recognized as common biochemical mediators of communication between these systems, are receiving increasing attention because of their potential therapeutic applications [...] Read more.
The interaction between the neuroendocrine system and the immune system plays a key role in the onset and progression of various diseases. Neuropeptides, recognized as common biochemical mediators of communication between these systems, are receiving increasing attention because of their potential therapeutic applications in immune-related disorders. Additionally, many neuropeptides share significant similarities with antimicrobial peptides (AMPs), and evidence shows that these antimicrobial neuropeptides are directly involved in innate immunity. This review examines 10 antimicrobial neuropeptides, including pituitary adenylate cyclase-activating polypeptide (PACAP), vasoactive intestinal peptide (VIP), α-melanocyte stimulating hormone (α-MSH), ghrelin, adrenomedullin (AM), neuropeptide Y (NPY), urocortin II (UCN II), calcitonin gene-related peptide (CGRP), substance P (SP), and catestatin (CST). Their expression characteristics and the immunomodulatory mechanisms mediated by their specific receptors are summarized, along with potential drugs targeting these receptors. Future studies should focus on further investigating antimicrobial neuropeptides and advancing the development of related drugs in preclinical and/or clinical studies to improve the treatment of immune-related diseases. Full article
(This article belongs to the Section Medicinal Chemistry)
Show Figures

Figure 1

31 pages, 2842 KB  
Review
Endocrine Disrupting Toxicity of Bisphenol A and Its Analogs: Implications in the Neuro-Immune Milieu
by Erica Buoso, Mirco Masi, Roberta Valeria Limosani, Chiara Oliviero, Sabrina Saeed, Martina Iulini, Francesca Carlotta Passoni, Marco Racchi and Emanuela Corsini
J. Xenobiot. 2025, 15(1), 13; https://doi.org/10.3390/jox15010013 - 17 Jan 2025
Cited by 12 | Viewed by 5283
Abstract
Endocrine-disrupting chemicals (EDCs) are natural or synthetic substances that are able to interfere with hormonal systems and alter their physiological signaling. EDCs have been recognized as a public health issue due to their widespread use, environmental persistence and the potential levels of long-term [...] Read more.
Endocrine-disrupting chemicals (EDCs) are natural or synthetic substances that are able to interfere with hormonal systems and alter their physiological signaling. EDCs have been recognized as a public health issue due to their widespread use, environmental persistence and the potential levels of long-term exposure with implications in multiple pathological conditions. Their reported adverse effects pose critical concerns about their use, warranting their strict regulation. This is the case of bisphenol A (BPA), a well-known EDC whose tolerable daily intake (TDI) was re-evaluated in 2023 by the European Food Safety Authority (EFSA), and the immune system has been identified as the most sensitive to BPA exposure. Increasing scientific evidence indicates that EDCs can interfere with several hormone receptors, pathways and interacting proteins, resulting in a complex, cell context-dependent response that may differ among tissues. In this regard, the neuronal and immune systems are important targets of hormonal signaling and are now emerging as critical players in endocrine disruption. Here, we use BPA and its analogs as proof-of-concept EDCs to address their detrimental effects on the immune and nervous systems and to highlight complex interrelationships within the immune–neuroendocrine network (INEN). Finally, we propose that Receptor for Activated C Kinase 1 (RACK1), an important target for EDCs and a valuable screening tool, could serve as a central hub in our toxicology model to explain bisphenol-mediated adverse effects on the INEN. Full article
(This article belongs to the Special Issue The Role of Endocrine-Disrupting Chemicals in the Human Health)
Show Figures

Graphical abstract

31 pages, 3064 KB  
Review
Unraveling the Gut Microbiota: Implications for Precision Nutrition and Personalized Medicine
by Alessio Abeltino, Duaa Hatem, Cassandra Serantoni, Alessia Riente, Michele Maria De Giulio, Marco De Spirito, Flavio De Maio and Giuseppe Maulucci
Nutrients 2024, 16(22), 3806; https://doi.org/10.3390/nu16223806 - 6 Nov 2024
Cited by 30 | Viewed by 10071
Abstract
Recent studies have shown a growing interest in the complex relationship between the human gut microbiota, metabolism, and overall health. This review aims to explore the gut microbiota–host association, focusing on its implications for precision nutrition and personalized medicine. The objective is to [...] Read more.
Recent studies have shown a growing interest in the complex relationship between the human gut microbiota, metabolism, and overall health. This review aims to explore the gut microbiota–host association, focusing on its implications for precision nutrition and personalized medicine. The objective is to highlight how gut microbiota modulate metabolic and immune functions, contributing to disease susceptibility and wellbeing. The review synthesizes recent research findings, analyzing key studies on the influence of gut microbiota on lipid and carbohydrate metabolism, intestinal health, neurobehavioral regulation, and endocrine signaling. Data were drawn from both experimental and clinical trials examining microbiota–host interactions relevant to precision nutrition. Our findings highlight the essential role of gut microbiota-derived metabolites in regulating host metabolism, including lipid and glucose pathways. These metabolites have been found to influence immune responses and gut barrier integrity. Additionally, the microbiota impacts broader physiological processes, including neuroendocrine regulation, which could be crucial for dietary interventions. Therefore, understanding the molecular mechanisms of dietary–microbiota–host interactions is pivotal for advancing personalized nutrition strategies. Tailored dietary recommendations based on individual gut microbiota compositions hold promise for improving health outcomes, potentially revolutionizing future healthcare approaches across diverse populations. Full article
Show Figures

Figure 1

Back to TopTop