Next Article in Journal
Sarcopenic Obesity, Pathogenesis, and Treatment with a Focus on Exercise and Protein Intake
Previous Article in Journal
From COVID-19 to Long COVID: The Forms of the Neurological Manifestations
 
 
Journal of Mind and Medical Sciences is published by MDPI from Volume 12 Issue 1 (2025). Previous articles were published by another publisher in Open Access under a CC-BY (or CC-BY-NC-ND) licence, and they are hosted by MDPI on mdpi.com as a courtesy and upon agreement with Valparaiso University (ValpoScholar).
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Diabetes Mellitus: Interdisciplinary Medical, Surgical and Psychological Therapeutic Approach

by
Bogdan Socea
1,2,
Adrian Silaghi
2,†,
Laura Florentina Rebegea
3,4,†,
Daniela Gabriela Balan
5,
Cristian Balalau
1,2,
Tiberiu Ștefăniță Tenea-Cojan
6,
Doina Andrada Mihai
7 and
Ioana Paunica
7,*,†
1
Department of General Surgery, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
2
St. Pantelimon Emergency Clinical Hospital, Department of General Surgery, Bucharest, Romania
3
Faculty of Medicine and Pharmacy, Department of Internal Medicine, Dunarea de Jos University of Galati, Galati, Romania
4
St. Ap. Andrei Emergency Clinical Hospital, Department of Radiology, Galati, Romania
5
Department of Physiology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
6
Department of Surgery, Craiova University of Medicine and Pharmacy, Craiova, Romania
7
N.C. Paulescu National Institute, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work and thus share first authorship.
J. Mind Med. Sci. 2023, 10(2), 217-236; https://doi.org/10.22543/2392-7674.1445
Submission received: 18 June 2023 / Revised: 17 July 2023 / Accepted: 24 July 2023 / Published: 25 October 2023

Abstract

:
Diabetes mellitus is a complex and widespread metabolic disease, having extremely complex implications (biological, psychological, social) for patients. Understanding the pathophysiology of diabetes (majorly influenced by various factors such as genetic predisposition, age, lifestyle choices, etc.) is essential for the prevention of this condition and the establishment of effective treatment strategies. The latest and relevant literature data related to the epidemiology, pathophysiology, and treatment of diabetes are presented, after an exhaustive review of the articles published on this topic and indexed in the WOS, PubMed, Scopus and Google Scholar databases. Preventing or delaying the onset of diabetes can be achieved in some patients with type 2 diabetes. After onset, treatment of diabetes is complex, involving a comprehensive approach (pharmacological interventions, lifestyle changes, surgical interventions in selected cases, as well as psychological support), depending on the stage of the disease and possible associated complications. Finally, diabetes is often asymptomatic in the initial stages, so an early diagnosis remains the essential element for the best subsequent therapeutic control.

Introduction

Diabetes mellitus (DM) is a metabolic disease with a high incidence among the population, being characterized by the presence of high levels of glucose in the blood. Hyperglycemia has both direct consequences on the cells of the body, as well as through the alternative pathways of glucose metabolism [1]. Historically, diabetes has been divided by age of onset into juvenile diabetes (type 1 diabetes) and adult-onset diabetes (type 2 diabetes), the latter being secondary to increased calorie intake from various sources (sugars or lipids). Until the discovery of insulin, diabetes mellitus type 1 was a fatal disease, secondary to the chronic catabolic state and the frequent occurrence of severe episodes of ketoacidosis (newborns in rare cases exceeded 2 years) [2,3].
Most cells in the body suffer when exposed to high blood sugar levels. The complications associated with this disease are multiple, with or without clinical expression, they can appear earlier or later and in different degrees of severity. Microvascular lesions in the persistent hyperglycemic syndrome lead to different forms of manifestation such as retinopathy, nephropathy or diabetic neuropathy [4]. Regarding the large vessels (placed at cerebral, cardiac or peripheral level), the complications of diabetes can lead to myocardial infarction, strokes, and minor or major amputations secondary to the occlusion of peripheral arteries [5].
In addition to all these direct effects of increased blood sugar, there are also a number of psychological changes that occur in these patients. These can be secondary both to the increase in blood sugar and to the treatment of this disease burdened most of the time by high costs, painful and repeated interventions, or various complications that may occur (which often led to the inability to move or take care of own body) [6].
The management of diabetes is complex, reducing the glycemic level being the main goal in controlling the disease. There are also therapies aimed at reducing the preventable mortality rate, as well as increasing the patient's quality of life that can lead to a life as close to normal as possible. Thus, multiple medical, surgical and psychological methods have been developed, sometimes it is necessary to combine them to obtain favorable results [7].
The present study is a review with the objective of identifying the most recent data from the literature regarding epidemiology, etiology, physiopathology and the new treatment methods (medical, surgical and psychological) that have appeared recently.

Discussions

Epidemiology

With the increase in life expectancy, the economic development of countries and the adoption of a different type of diet, there has been an increase in the incidence of diabetes. From a pathophysiological point of view, a consensus has been reached that divide high glycemic level according to age, the main cause and risk factors in diabetes type I, type II, hyperglycemia associated with pregnancy, pancreatic disorders or other endocrinopathies [8]. Thus, in 2014, 422 million patients were registered globally with diabetes, with an increase in prevalence especially in underdeveloped and developing countries (for example, an increase in Africa from 3.1% in 1980 to 7, 1% in 2014) [9].
The diagnosis of diabetes is usually made late, because the appearance of high blood sugar is poorly expressed clinically or even asymptomatic. Thus, 45.8% of all cases of diabetes that occur in adults are undiagnosed from the onset, so they are much more exposed to the development of complications than those who receive treatment. Costs associated with late-diagnosed forms are 3 times higher than in the general population, reaching $673 billion for the underlying disease and associated complications [10,11].
Most patients with diabetes are found in Asia, China together with India (totaling over 180 million), followed by the USA (where 30.2 million people have high blood sugar levels), Brazil (with 12.4 million) followed by Indonesia and Mexico (having 12 and 10.3 million patients respectively). 95% of these cases are considered to be type II diabetes, the remaining 5% are attributed to type I diabetes [9]. In terms of distribution within different populations, both genetic characteristics and living environment have a major contribution. In the US, there is an increase of approximately 1.5 million patients with diabetes per year, with the prevalence of diabetes in non-Hispanic blacks at 17.7%, especially in the male population, followed by Hispanics, Asians, and whites [12].
Type I diabetes appears predominantly around the age of 15, with a high incidence in developed countries such as the USA, Great Britain, China, Brazil or Nigeria, and with global variations between 0.13 and 1.61 respectively per 100,000 inhabitants [13]. The male population is usually the majority, exceeding the percentage of 50% in countries with a Human Development Index of over 0.700, equal values being observed in less economically developed countries. Lower values appear in the non-European population compared to the European one [11].
Diabetes mellitus was the direct cause of death in 6.8% of cases, with approximately 5 million deaths/year associated with acute complications of hyperglycemia, with an increasing number being recorded today. Along with the premature death of the population, diabetes mellitus is associated with an increased risk of disability, ending up being in the first 5 entities that cause its appearance in the population aged between 20 and 70 years (it is responsible for approximately 143 million patients with impairment of activity and quality of life [14].
Gestational diabetes is defined as the presence of glucose intolerance associated with hyperglycemia detected in any trimester of pregnancy, without these features being present before pregnancy [15]. Women and babies who develop during this period are subject to immediate and long-term complications, with the risk of developing type II diabetes after pregnancy being over 60% in the first 16 years after the event [16]. In Europe, the prevalence of diabetes in the pregnant population reaches values of 20% [17,18]. Recent data show a 39% rate of gestational diabetes in obese or overweight women in Western Europe, with prevalence rates ranging from 24% in the UK to 52% in Denmark. Much lower rates, between 2-6% are observed in the northern area of Europe or in the Mediterranean Sea basin [18].
The main complications that can occur in gestational diabetes are: the occurrence of preeclampsia or eclampsia, the presence of a macrosomic fetus, increased rates of caesarean section or low birth weight with the admission of the newborn to the neonatal intensive care unit. All these parameters were influenced by the presence of elevated glycemic levels, thus the rate of hypertension associated with pregnancy reached 17%, along with high birth weight that occurred in 18.5% of cases and premature birth resulted in a rate of caesarean section in this population of 40.2% of cases. Females born to these mothers with high glycemic levels required admission to the intensive care unit in 6.65% of cases [19].
Type II diabetes is secondary to specific causes and can occur in a series of endocrinopathies such as excess hyperglycemic hormones (growth hormone, cortisol, catecholamines), in the context of treatments with protease inhibitors for HIV, antipsychotics administered in psychotic/schizoaffective disorders or from an immune cause (immune checkpoint inhibitors for some forms of cancer). Treatment of the underlying disease may result in resolution of elevated glycemic levels, and when this is determined by treatment, a risk versus benefit assessment must be considered [19].

Pathophysiology

Type I diabetes is characterized by low insulin secretion secondary to accelerated destruction of pancreatic β-cells by autoantibodies, which will eventually lead to hyperglycemia and acidosis as a result of low blood insulin levels. The destruction of pancreatic β-cells begins with the loss of tolerance of the immune system, being considered foreign and attacked by immune cells, a process controlled by T cells. Thus, the identification of the steps leading to the attack of β-cells by specific antibodies and immune cells may be sooner or later identified and modified in the sense of stopping aggression and thus preventing the onset of diabetes [20].
The link between immune cells and type I diabetes was highlighted in some patients who had certain types of antigens of the Human Leukocyte Antigen (HLA) system and elevated blood sugar levels. Later through research on the human genome, HLA class II was identified as a genetic risk factor, being responsible for 50% of the cases of type I diabetes, which suggests that the presentation of a certain peptide to the immune system is involved in the pathogenesis of this diseases [21]. Genetic factors that predispose to type I diabetes include genes that control immunity, tyrosine phosphatase, non-receptor type CTLA4, and lns [22]. Thus, in most cases type I diabetes can be a polygenic syndrome, but they are also found in isolated cases where only a single gene can be involved, such as the Autoimmune Regulator gene associated with autoimmune polyglandular syndrome type 1 [23].
During the course of the disease, a series of antibodies against insulin, glutamate decarboxylase, insulinoma antigen 2 or zinc transporter 8 may appear, which may represent the sign of autoimmunity, their level can be determined in the patient's plasma and correlated with the severity of the disease [24]. The synthesis of these autoantibodies could be influenced by CD4+ and CD8+ T cells, which are found in the peripheral blood, in the pancreatic tissue and even in the peripancreatic lymph nodes, with the difference that in the case of patients with type I diabetes the cells have activity against β cells [25,26]. The presence of these autoreactive species in the peripheral blood is quite small and is similar to the concentration found in a healthy individual, the only difference being an abnormal activity in the periphery of CD 8+ cells, secondary to a series of changes that occur during their activation [27]. These CD8+ cells have functional characteristics different from normal ones, that is they show a low epitope affinity for HLA complexes and low interaction with antigen-presenting cells. Some localized changes in T receptors do not elicit an extensive immune response when interaction with the HLA-epitope complex is present. All this is the consequence of their incomplete maturation during development at the thymic level [28,29,30].
Various extrinsic factors have been proposed for autoactivation of the immune system and pancreatic β-cell destruction. Metabolic stress, viral infections or the presence of pro-inflammatory agents that reach the pancreatic tissue from the digestive tract through different pathways are usually involved in the autoimmune response [31]. Thus, during a viral infection, pancreatic β cells express adhesion factors and receptors, including coxsackievirus and adenovirus receptor (CAR). They have been identified in insulin-containing granules, leading to a vulnerability of pancreatic tissue to infection with coxsackievirus B4 [32] and could trigger type I diabetes in patients who had enterovirosis with such a virus. For other viral particles, such as rotavirus and cytomegalovirus, the pancreatic cell response may be similar to coxsackievirus B4 infection [33,34].
Other factors involved in the onset and worsening of type I diabetes are diet and gut microbiota. One of the roles of the bacterial population of the digestive tract is represented by the modulation and differentiation of local immune cells, in order to maintain homeostasis which is achieved through cellular immunity but also through the short-chain fatty acids synthesized by it [35]. The roles of fatty acids are varied, from direct inhibition of neutrophils and dendritic cells (through the synthesis of antimicrobial peptides by lymphoid cells or pancreatic B cells), to the role of moderating the immune response generated by T cells (through inhibition of histone deacetylase), as well as activation of pathways mammalian target of rapamycin (mTOR) [36]. Experimental studies on mouse populations demonstrated the protective role of these antibacterial cytokines against the onset of type I diabetes and the ability of short-chain fatty acids to decrease the synthesis of cellular messengers that could cause pancreatic β-cell destruction [37].
The patient's diet can influence the onset of type I diabetes both directly and through the microbiota. Thus, a low-gluten diet during pregnancy may lead to low rates of autoimmune diabetes in newborns [38]. Administration of vitamin D, omega-3 fatty acids did not have clear results in terms of decreasing the risk of developing type I diabetes [39].
In type II diabetes there is a dysregulation of the control mechanisms between the glycemic level and insulin secretion, thus leading to hyperglycemia [40]. One of the main causes of this phenomenon is β-cell dysfunction, which leads to inadequate secretion compared to the need to maintain an adequate glycemic level. At the same time, there is an increase in glucose synthesis in the liver and a decrease in its use in muscle and adipose tissue, secondary to the inability of specific receptors to respond to these stimuli, a phenomenon called insulin resistance [41].
The synthesis of insulin at the level of pancreatic β cells is done in stages by conformational changes of proinsulin and keeping the active form in vesicles that are secreted when needed. The main stimulus for its release from blood cells is glucose via the glucose transporter, a cation transport protein that causes the change in the ATP/ADP ratio that closes ATP-dependent potassium channels. This subsequently leads to an increase in the concentration of ATP, but also the accumulation of intracellular calcium that will mobilize the granules in which insulin is stored and thus they will migrate to the cell membrane where they will be eliminated by exocytosis [42]. Other stimuli that cause the release of insulin from cells are represented by large amounts of amino acids, fatty acids, hormones [43] and also extracellular cAMP or ATP by influencing intracellular calcium levels [44,45].
If there are elevated levels of blood glucose or serum lipids secondary to persistent food excess, a state of chronic inflammation is observed and with it a series of consequences such as: oxidative, amyloid and metabolic stress, which will lead to the loss of the integrity of the pancreatic islet tissue [46]. Lipotoxicity, glucotoxicity, and glycoprotein toxicity occur in the context of obesity and metabolic stress secondary to activation of the cellular apoptotic unfolded protein response (UPR) at the β-cell level [47]. The increased level of fatty acids can cause inhibition of the sarcoplasmic reticulum with decreased intracellular calcium mobilization. Persistent glycemic status may increase β-cell synthesis of proinsulin and islet amyloid polypeptides (IAAPs), which will lead to a three-dimensional misfolding of proinsulin, with its accumulation in the cytoplasm increasing oxidative stress that will ultimately lead to a cascade of proapoptotic elements with tissue destruction and chronic inflammation [48].
Another consequence of hyperglycemia and glucotoxicity is the increased synthesis of reactive oxygen species (ROS) that will generate an inflammatory syndrome and increased oxidative stress. Excess ROS led to an inactivation of mitochondria that will not be able to metabolize superoxide ions. They are directly involved in the pathogenesis of diabetes, so they are responsible for the formation of advanced glycation end products (AGEs) by stimulating the polyol pathway. Thus, a series of changes in normal cell physiology occur, including for example increased expression of AGE receptors and activation of protein kinase C and its isoforms [49,50]. Secondary to these metabolic products, a state of hypoxia and ischemia occurs, which will lead to the release of inflammatory markers, and their activity can lead to neo-formation of vessels and irreversible changes in the normal structure of tissues, even if the glycemic level will normalize [51].
Reduced physical activity predisposes to obesity and type II diabetes. A variable degree of systemic inflammation can occur which will lead to the release of cytokines such as IL-6, TNFα and IL-1 into the bloodstream, known as metabolic inflammation [52]. Elevated levels of IL-1 directly affect pancreatic β-cells by releasing a large amount of nuclear factor kappa-enhancer of activated B-cell light chain (NF-κB), which will lead to a decrease in their ability to release insulin in blood flow and further induce apoptosis. If the patient decides to lose weight, there is a sensitization of the tissues to insulin, thus improving the glycemic level and reducing the overall cardiovascular risk. The underlying mechanism for this is the release of anti-inflammatory cytokines such as IL-1 and TNF-α antagonists and the release of antioxidant factors such as glutathione, which will neutralize free radicals released as a result of aberrant metabolic pathways [53,54,55].
Similar to type I diabetes, the microbiota has a determining role in the occurrence of type II diabetes [56]. A diet rich in fatty acids causes an increase in the level of lipopolysaccharide synthesized by intestinal bacteria, which will lead to local inflammation and insulin resistance along with a change in the ratio of bacteria present at this level [57]. The resulting dysbiosis can cause a decrease in the production of amino acids and trimethylamine that will cause the alteration of blood sugar regulation mechanisms, thus favoring the onset of type II diabetes [58,59].
During pregnancy, a series of changes occur that favor the development of the fetus. In the first period of pregnancy, there is a general sensitization of the tissues to insulin, which will determine the capture of glucose in the tissues secondary to the increased need [60]. Following the increased levels of estrogens, cortisol, leptin, progesterone and placental growth hormones, insulin resistance will appear [61], which will determine a higher glycemic level necessary for fetal growth. For a good glycemic control, it is necessary to compensate the function of the pancreatic β-cells. In normal pregnancy, pancreatic islet hypertrophy and hyperplasia may occur along with increased insulin secretion [62].
In the case of gestational diabetes, the damage can be multifactorial, affecting all phases of insulin synthesis and release. Most often, there are genetic lesions of the potassium channel KQR-like 1 and glucokinase. In the case of minor deficits, any stressful factor can cause diabetes, especially pregnancy. In addition to the genetic damage of these patients, the above-normal glycemic levels cause the appearance of glucotoxicity, which negatively influences β-cell function with decreased insulin secretion and stimulation of apoptosis, thus forming a vicious circle in which gestational diabetes occurs [63].
The placenta, through the synthesis of the hormones and cytokines it secretes, causes hyperglycemia and later, in selected cases, gestational diabetes. Transplacental glucose transfer occurs via GLUT1 without the need for insulin [64]. The changes observed in the placenta of babies born to mothers who had gestational diabetes were mainly in the area of lipid metabolism compared to that of glucose (67% vs 9%) [65], a fact confirmed by the relationship between maternal obesity, glucose and excess growth of fat sizes [66].
A number of products such as Bisphenol A have been associated with the presence of elevated glucose levels in pregnancy by altering methylation at the cellular level, thus altering the signals received by the placenta [67].
Elevated glycemic levels in patients' plasma cause a series of changes in the structure and physiology of cells through the aberrant activation of various metabolic pathways. Aberrant activation of protein kinases C has been observed in association with increased formation of polyols, hexosamines, glucose autoxidation, and elevated levels of advanced glycation end products, all of these metabolic adaptations serving to reduce elevated glycemic levels [68].
Elevated glycemic levels in patients' plasma cause a series of changes in the structure and physiology of cells through the aberrant activation of various metabolic pathways. Aberrant activation of protein kinases C has been observed in association with increased formation of polyols, hexosamines, glucose autoxidation, and elevated levels of advanced glycation end products, all of these metabolic adaptations serving to reduce elevated glycemic levels [68].
Protein kinases are a family of proteins involved in the growth, differentiation and modulation of vascular permeability being an important factor for the neoformation of blood vessels [69]. In the event of an increase in the glycemic level, there is an increase in the activity of these molecules in various tissues such as the myocardial, retinal, or renal, etc., which will lead to complications that influence the formation of polyols, thus amplifying the changes in the cell structure [70,71]. The cellular mechanisms by which these changes occur are the activation of different growth factors such as transforming growth factor beta (TGF-β), platelet-derived growth factor (PDGF) and epidermal growth factor (EGF). All these factors will lead to the exacerbation of the extracellular matrix, which may play an important role in the progression of chronic diseases such as diabetic nephropathy [68].
Polyols are products that result in an alternative way of glucose metabolism mediated by 2 enzymes: aldose reductase and sorbitol dehydrogenase using Nicotinamide adenine dinucleotide phosphate (NADPH) as catalysts [72]. Physiologically, these 2 enzymes are involved in the formation of fructose used in different organs such as: kidneys, intestinal cells and spermatocytes [73]. Sorbitol, one of the polyols, can cause changes in ATP function and thus (due to the strong electronegative charge) changes in cellular osmotic pressure [74]. Being a polar molecule, sorbitol cannot easily cross the cell membrane accumulating at this level, mainly in the retina, kidneys and Schwann cells, which causes the increase of osmotic pressure and subsequently the attraction of water increasing their volume and destroying the cells [75]. Thus, cataracts appear in diabetic patients, with much higher rates and in a shorter time than the healthy population, enzymes and renal tubular cell dysfunction, but also poor myelination of axons due to the poor differentiation of Schwann cells through the low expression of Insulin-like growth factor 1 (IGF-1) receptors [76]. Through the excess use of NADPH and the inability to regenerate it, there is a surplus of NADH that causes changes in lipid metabolism, the formation of growth factors and the activation of protein kinase C, causing increased oxidative stress.
Advanced glycation end products (AGEs) represent non-enzymatic reaction products between free amino groups of various compounds and glucose or aldehydes [77]. These products are responsible for some of the micro- and macrovascular complications of diabetes. A specific phenomenon is observed in structural proteins such as collagen, where the reversible reaction between amino groups and sugars forms a covalent bond that cannot be easily broken, thus causing changes in the spatial structure of these structures. The clinical symptom that occurs is atheroma plaque formation along with basement membrane thickening, reduced elasticity and subsequent tissue dysfunction [78]. At the cellular level, a number of cellular signaling mechanisms are affected. Nitric oxide-induced vasodilatation through its secondary inactivation to the pro-oxidant environment, activation of nuclear factor kB, cellular expression of adhesion molecules and activation of transmembrane transport proteins such as Rac type can cause systemic inflammation, endothelial dysfunction, atherosclerosis and thrombosis [79].
In large quantities, glucose can undergo autoxidation phenomena that will lead to the formation of reactive oxygen species such as H2O2, malondialdehyde or methylglyoxal, glyoxal [80]. The effects of these highly reactive products are the intensification of oxidative stress, and the resulting substances can change the spatial structure of proteins, because the reactive carbonyl species contained in it can bind to arginine and lysine, which will cause the same result as the large amount of AGEs [81].

Treatment

Secondary to the high level of blood sugar, micro and macrovascular complications appear, thus increasing the general cardiovascular risk, premature aging of the tissues secondary to oxidative stress and the decrease in the quality of life due to the presence of various conditions that can cause a marked decrease in visual and locomotor activities. Acute complications such as hypoglycemic or hyperglycemic coma can be life-threatening for patients, direct consequences of inadequate insulin synthesis and release. Thus, DM treatment requires a multidisciplinary approach in controlling metabolic and cellular abnormalities and their consequences [82].
The main goal of DM treatment is to normalize the glycemic level, so since the development of insulin, numerous antihyperglycemic substances have been introduced, which function both as insulin agonists and as secretion stimulators or as sensitizers of insulin receptors in various organs. Most of the time, combinations of these are used for adequate blood sugar control. Rapid-acting insulin and its analogs act during meal periods by reducing postprandial blood glucose, and prolonged-release insulin has a similar action to continuous insulin release to maintain an adequate blood glucose level [83]. In addition to these direct methods of insulin administration, a series of adjunctive measures must be taken, such as lifestyle modification, administration of compounds that reduce oxidative and general cardiovascular stress and thus reduce the need for medication and improve quality of life [84]. Thus, the administration of insulin solves only part of the problem, the disease will continue its evolution and large doses of drugs will be needed for the same result, therefore a series of adjuvant therapies have been developed that have the role of either sensitizing the tissues, to respond better to insulin stimulation or to increase the release of insulin from stores [85].

Medical treatment Insulins

Basal insulin analogs have the ability to maintain a glycemic level as constant as possible, with fewer episodes of nocturnal hypoglycemia [86], with a level closer to the limit value in the case of glycosylated hemoglobin. Regarding the different preparations available, glargine is an insulin analogue that precipitates at the level of the subcutaneous tissue secondary to the constant pH of this level, causing the formation of dimers and monomers, which will lead to a constant release [87]. Compared to Neutral Protamine Hagedorn (NPH), insulin glargine has much lower peak release levels and minimal individual variability [88]. The rate of administration of the 2 substances is different, so a single dose of Glargine is similar to taking 2 doses of NPH insulin in terms of long-term blood sugar control. Hypoglycemia, which can occur with inadequate dosing of insulin preparations, showed fewer hypoglycemic events, particularly at night, for NPH-insulin. However, the satisfaction level of patients using Gla-100 is much better than for NPH-insulin [89].
Detemir is a product that contains a fatty acid chain that favors the formation of dihexamers that can attach more easily to albumin. Thus, there is a slower release of it from the injection site, with less individual variability in the pharmacodynamics of the prepared insulin and with a longer duration of action [86]. Compared to NPH insulin, Detemir has a better safety profile, with a lower risk of hypoglycemia, including at night, and with much smaller daily variations. These characteristics are similar to those of Gla-100, but with a requirement of 2 doses per day [90], with the effect diminishing after 12 hours [91]. In the long term, the glycemic level monitored by glycosylated hemoglobin was similar (8.8 Glargine vs 8.9 Detemir), which shows no major differences between the efficiency and effectiveness of Detemir vs Gla-100 [86].
Degludec is a newer generation insulin product in the form of a hexamer that dissociates locally into monomers and is subsequently absorbed and circulates in the plasma via albumin. Comparative results with GLA-100 are better in terms of nocturnal hypoglycemia and glycemic level [92], but with similar results in terms of maintaining basal glycemic level [93].
The method of basal insulin administration is important in disease control. Continuous subcutaneous administration mimics the continuous basal secretion of pancreatic cells, which reduces the risk of hypoglycemia compared with multiple-dose administration based on circadian cycle variations [94]. In addition, a better quality of life and a better degree of satisfaction were observed in the first case [95].
Fast insulin or during the meal is used most of the time in the case of a sudden increase in the glycemic level, such as in the case of a rich meal or in case of complications. It is often administered as a bolus in combinations of short-acting insulin and rapid analogues, depending on the amount of carbohydrates a patient consumes with a meal, the glycemic level before the meal and the possible physical activity he will perform [86].
The use of rapid-acting human insulin compared with rapid-acting insulin analogues such as lispro and aspart showed better glycemic control and fewer complications than insulin analogues [96], the main factor monitored being the risk of nocturnal hypoglycemia [86]. Concomitant administration of human insulin and glargine provided long-term inadequate glycemic control compared to administration of the analog + glargine, with a significant decrease in glycosylated hemoglobin when administered 30-45 minutes before a meal. The risk of hypoglycemia for these 2 treatment strategies is similar. In addition to the classic method of bolus administration, the continuous infusion administration of prandial insulin showed similar glycosylated hemoglobin values for analogs, namely human insulin [97].

New formulas of insulin preparations - smart insulins

The concept of smart insulins is based on the ability of some systems to release insulin endogenously under conditions of hyperglycemia and to decrease the insulin release concentration if the blood glucose concentration decreases [98]. The system that controls the release of insulin relies on a sensor, a chemical that can change its chemical or physical structure that can trigger the release of insulin or other molecules [99]. Insulin delivery systems are largely based on polymers that form an impenetrable matrix under normoglycemic conditions, but once the glycemic level rises above a certain level, structural changes occur, such as swelling or shrinking, which alters the permeability to insulin, allowing it to release the nanostructure [100]. The factor that changes the spatial structure of the matrix is in many cases the pH for polymers such as poly(2-dimethylamino), poly(acrylic acid), hyaluronic acid, but also hypoxia or oxygenated water resulting from the oxidation of glucose with a better control in regarding the release of incorporated substances [101].
An advantage of incorporating insulin into polymer macrostructures is the ability to add multiple types of insulin, within multiple polymer types, which will result in on-demand release, thus providing better stability and response when a stimulus [102].

Insulin therapy devices

Since the discovery of insulin, the predominant method of administration has been through subcutaneous injections, and with the development of technology there have been a number of devices such as smart pens, pumps and needle-free devices, as well as alternative ways of administering insulin that are less invasive. Smart insulin pens are devices that can be connected via Bluetooth to an application on any smartphone device through which the dose of insulin can be better adjusted according to physical activity and carbohydrate intake [103]. Recorded data can be sent to a server where the attending physician and patient can better dose the amount of insulin based on the patient's history. One of the disadvantages of subcutaneous administration is the repeated puncturing of the integuments. Insulin jet injectors represent a new method of administration by which a high-pressure jet pierces the skin, and the insulin reaches the level of the subcutaneous tissue where it is absorbed. The advantages of this system are represented by a better control of postprandial blood glucose and a shorter duration until the action takes place with the risk of ecchymosis in the case of too high pressure [104].
Among the insulin pumps developed on the market, the insulin patch-pump system is the newest model, which presents several features that make it superior to other models, such as simplicity of use or the absence of skin penetration with a sharp object (delivering insulin through a packet) [105]. Another way of infusing insulin through the pump is the peritoneal one, recommended for patients with skin and subcutaneous tissue lesions and whose insulin administration may have variations, with very good results in terms of glycemic control but with the risk of infection at the level catheter [106].
The advantages of using insulin pumps are the low incidence of hypoglycemic episodes secondary to a better dose of insulin, a better quality of life secondary to the more flexible lifestyle it brings, and the small number of injections required to administer the treatment. The main disadvantage of these devices is the high maintenance and purchase price together with possible infections and irritation at the insertion site [107].
The artificial pancreas is a pump that administers insulin based on the blood glucose level at a given time. Thus, a sensor is used that measures blood sugar, the data being sent to a computer that analyzes the result received and, according to an algorithm, determines the release of the substance [102]. The efficiency and safety offered by these systems depend on the algorithm underlying insulin delivery, so the addition of a hypoglycemia control system leads to lower rates of nocturnal hypoglycemia compared to pumps that deliver insulin only according to the electrical signal received from the sensor (the algorithm can anticipate a possible decrease in the glycemic level) [108].
From this idea was developed the bionic pancreas, which is a dual infusion pump that releases both glucagon and insulin into the bloodstream, controlled by a smartphone app. By measuring blood glucose over a short period of time, along with other factors such as previous blood sugar levels, the amount of insulin administered, the level of activity and carbohydrate intake, the artificial intelligence determines the amount of insulin needed and subsequently releases glucagon if there is excess. By using these smart devices, patients' quality of life has increased significantly, with better glycemic control (fewer episodes of nocturnal hypoglycemia with constant glycosylated hemoglobin), but with increased acquisition and maintenance costs [109].

Other hypoglycemic treatments

In addition to the classic treatment with insulin, which has the role of completely replacing pancreatic function, a number of other substances have been developed with the aim of lowering blood sugar levels, reducing body weight in obesity and general cardiovascular risk [110]. All of these have been classified as non-insulin treatment, acting either by stimulating the release of insulin from the pancreas, by improving the affinity of cells for the hypoglycemic hormone, by decreasing the formation of glucose in the liver, or by inhibiting glucose transport [111].
Biguanides and thiazolidinediones are substances that have the ability to reduce insulin resistance and at the same time positively influence other abnormal metabolic processes such as aberrant lipid metabolism and atherosclerosis [112]. Metformin, a biguanide, has the ability to decrease hepatic glucose synthesis by activating AMP-activated protein kinase (AMPK) at the mitochondrial level, with an increase in tissue sensitivity to insulin due to the influence of tyrosine kinase pathways and acceleration of insulin secretion at the β-cell level [113]. All these effects are associated with a better proliferation of type I collagen at the bone level with the ability to maintain normal bone density but also with a greater risk of developing metabolic acidosis or diarrhea [114,115].
Thiazolidinediones are proliferator-activated receptor-γ (PPAR-γ) agonists that have effects similar to biguanides, causing an increase in insulin secretion through stimulation of pancreatic β-cells and a sensitization of peripheral tissues, especially liver and adipose, to hypoglycemic hormone. The hypoglycemic effects are associated with a decrease in circulating fatty acid levels and an anti-inflammatory effect, which will lead to a decrease in overall cardiovascular risk, but with a higher risk of weight gain secondary to the stimulation of adipocyte formation from mesenchymal stem cells and fractures by primary differentiation of osteoblasts into osteoclasts, secondary to PPAR-γ receptor activation [83].
The category of insulin secretagogues includes sulfonylureas and meglitinides, which have the ability to stimulate pancreatic β-cells at various stages of insulin formation. Sulfonylureas cause an increase in insulin release by binding to the transmembrane receptor SUR-1 that controls the ATP-dependent K channel that closes and releases preformed insulin regardless of the glycemic level. Associated adverse reactions are mainly represented by marked hypoglycemia, especially in the elderly who have multiple drug combinations, impaired renal or hepatic function, marked weight gain, skin rashes or porphyria [83].
Meglitinides are synthetic antidiabetics that also bind to pancreatic β-cell K channels, but with a much weaker effect than sulphonylureas [116]. The main use of meglitinide is to control postprandial blood glucose by increasing insulin secretion [117], but with fewer side effects (such as hypoglycemia or cardiovascular events) due to its shorter half-life [118].
Glycosidase inhibitors are nitrogen-rich substances that have the ability to inhibit the breakdown of disaccharides and oligosaccharides in glucose, and thus its absorption at the intestinal level is lower with a decrease in postprandial blood glucose [119]. An improvement in pancreatic β-cell function was observed after initiation of treatment. The overall effect will lead to weight loss, improvement in hypertriglyceridemia and systemic blood pressure, reducing overall cardiovascular risk, but with digestive symptoms such as diarrhea, flatulence, and vomiting secondary to the processing of sugars by the microbial flora in the colon [120].
Incretin treatment was based on the ability of glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic peptide (GIP) to stimulate pancreatic β-cells by coupling to G protein-coupled receptors that will close or open the K-ATP channel, along with stimulation of β-cell proliferation, inhibition of apoptosis and glucagon secretion. The release of these molecules is controlled by nutrients in the form of glucose and carbohydrates, during fasting the level is low but after ingestion of food they can reach high values [121].
Thus, by administering GLP-1 receptor agonists, such as dulaglutide, albiglutide and semaglutide [122], there is a decrease in glucagon synthesis and secondary hepatic glucose production, with a significant improvement in pancreatic β and α cell function, along with weight loss through decreased appetite due to delayed gastric emptying [123]. Due to the effects of GLP-1 agonists, they are recommended as the first line of treatment for diabetic patients with cardiovascular risk factors, preventing heart attacks and strokes by maintaining good glycemic levels. After the introduction of such a product, the level of glycosylated hemoglobin can decrease by up to 52.7% [124].
Among the adverse reactions of GLP-1 agonists are described local reactions at the site of administration, such as local erythema, pain and inflammation along with pancreatitis or worsening of renal failure in patients with chronic kidney disease [125].
Dipeptidyl Peptidase-4 is an enzyme that determines the metabolism of GLP-1, decreasing their effectiveness and increasing the glycemic level. Antagonists of this enzyme (such as saxagliptin, alogliptin) mimic the effect of GLP-1 agonist administration by maintaining pancreatic β-cell numbers along with anti-inflammatory and protective effects on overall cardiovascular risk [126,127]. Unlike GLP-1 receptor agonists, adverse reactions are rarer, the rate of hypoglycemia being lower in case of long-term administration, without an increase in body mass index. All of these are burdened by the risk of myocardial remodeling and fibrosis which are accentuated in patients with heart failure [128].
Sodium-glucose cotransporter 2 (SGLT2) is a protein that has the role of reabsorbing glucose that is filtered along with primary urine. Under conditions of hyperglycemia, SGLT2 is overexpressed in the nephron and hyper-uptake can occur. By blocking this transporter, there is a decrease in the resorption of glucose in the urine and a decrease in blood sugar [129]. In addition to the hypoglycemic effect that canagliflozin, dapagliflozin, empagliflozin have, other effects have been observed, such as the ability to improve the functionality of the cardiovascular system, reduce the rate of chronic kidney disease and normalize the lipid profile of patients [130].

Immunomodulatory treatment in type I DM

In type I diabetes, there is a particular situation, namely autoimmunity, by addressing it the destruction of pancreatic β cells would be reduced, the disease would not progress, and if the onset could be observed it could be prevented [20]. The first immunomodulatory treatment initiated in the case of DM type I was that with cyclosporine, which has a direct inhibitory effect on T cells, which caused a decrease in glycosylated hemoglobin below 7.5%, thus causing remission of the disease after 2 months of continuous treatment [131]. After stopping the treatment, a relapse of the disease occurred, with increased destruction of the remaining β-cells. Although it has the ability to induce disease remission, ciclosporin has significant adverse effects such as nephrotoxicity, the risk of various neoplasms or various allergic reactions. This demonstrated once again the autoimmune component of this pathology and the approach of a new strategy, instead of an intense depression of the immune system, with the aim that the β-cells become accepted and recognized as self, thus having an increased tolerance towards insulin-synthesizing cells [20].
A number of substances have been tried to restore self-tolerance of the immune system in type I diabetes. They have targeted both B cells and T cells, with mediocre results in most cases. Alefacept, a CD+2-targeting monoclonal antibody, was administered for 12 weeks to patients with newly diagnosed type 1 DM compared to placebo, with less C-peptide release but no reduction in the need of insulin, and no decrease in glycosylated hemoglobin [132].
Abatacept is a compound that inhibits T cell activation by interfering with CD80 and CD86 receptors on antigen-presenting cells that can no longer interact with defective T cells and are no longer activated, thus having an early effect on the development of autoimmune cells. Administered for 24 months in patients who had been diagnosed with type I diabetes for less than 1 year, the rate of decrease in pancreatic β-cell counts was similar to that of the control group. Orban et al. concluded that this cause of similar decrease is due to low doses that cannot block antigen-presenting cells or because of alternative pathways of stimulation in the etiology of autoimmune diabetes [133].
Rituximab, an anti-CD20 monoclonal antibody, delayed C-peptide decline by 8.2 months in a cohort of patients with recent-onset type 1 diabetes, but long-term pancreatic β-cell destruction follows its natural course. Thus, 2 years after the onset of C-peptide levels in the group of control patients and those who received monoclonal antibody the results were similar [134].
Another possible solution in the case of type I diabetes is the blocking of some stages of the inflammatory cascade that occurs as a result of the loss of tolerance to pancreatic cells. Administration of canakinumab (an IL-1 blocker) showed no response in reducing C-peptide as a marker of insulin synthesis [135]. Other metabolic pathways that could be altered are those of IL-6 and IL-21. Imatinib is a tyrosine kinase inhibitor used in the treatment of myeloid leukemia. Administered to mice, NOD causes an increase in self-tolerance, with the normalization of blood sugar after 2 weeks of treatment in 80% of cases. Its effects are secondary to inhibition of inflammation through antagonism of platelet-derived growth factor receptors (PDGF-R). In the long term it can lead to prolongation of β-cell life by activating NF-κB and increasing nitric oxide production [136].

Surgical treatment

Metabolic surgery is a new surgical concept in which various organs or systems are manipulated and change their function to achieve a better biological outcome [137]. To this end, a number of interventions have been developed such as duodenal and vague nerve stimulation for type II diabetes, pancreas transplantation or various bariatric surgery procedures with the aim of reducing body weight and the associated metabolic syndrome [138].
Surgical interventions that have demonstrated efficacy in type II diabetes are sleeve gastrectomy, roux-en-y gastric bypass, laparoscopic adjustable gastric bands, and biliopancreatic bypass/duodenal switch, all associated with good results in increasing glycemic control and decreasing substantial weight loss [139].
Standard sleeve gastrectomy is the surgical intervention developed for bariatric surgery in the 2000s, currently being the most well-known and well-researched of all, with good long- and short-term results [140]. The indications for this intervention are represented by morbid obesity with metabolic syndrome, the presence of a liver or kidney transplant and in patients with a body mass index between 30-35 kg/m2, who have associated comorbidities [141]. The results at 5 years showed that in 58.4% of cases there was a marked decrease in weight [142], and in patients with diabetes associated with obesity, the level of glycosylated hemoglobin decreased from an average from 8.3 to 6.7%, and fasting blood glucose decreased from a mean value of 170.3 to 112.0 mg/dl [143].
Safety studies of sleeve gastrectomy have shown a mortality rate of 0% but a morbidity of 2.2% [144]. The most frequently observed long-term occurrence of stenoses or strictures but also anastomotic fistula associated with high morbidity, or gastroesophageal reflux disease with an increased risk of Barrett's esophagus (a fact for which the presence of erosive esophagitis before bariatric surgery is a relative contraindication) [145].
Roux-en-Y gastric bypass was first described in 1966 and performed laparoscopically in 1993 [146], being one of the best metabolic interventions to improve hypertension, dyslipidemia, sleep apnea, heart failure and depression [147]. The main indication for this intervention is morbid obesity. In the first months there is a decrease of > 50% which can be maintained in the long term, and in patients with diabetes there is a remission of the disease between 50-80% depending on the duration of the disease and its severity [148]. At an interval of more than 7 years, it was observed that in patients who underwent gastric bypass, the mortality rate from cardiac causes (associated with complications) decreased by 40% in diabetes and cancer [149]. Regarding the rate of perioperative complications, it has been observed that they can be present in 3.5% of cases, along with a mortality between 0.14 and 0.25% [144].
Laparoscopic adjustable gastric banding (LAGB) is the third most common procedure for the relief of morbid obesity and metabolic syndrome [139]. Studies show that it is a simple, effective, reversible and durable procedure if patients are well selected and counseled in the peri-interventional period [150,151]. In studies of 89 patients with morbid obesity and metabolic syndrome, 60 showed improvements at 12 months and 2 years after surgery, with 80 of them at near-normal weight [152]. In patients with diabetes and a high body mass index, a decrease in glycosylated hemoglobin between 1.4 and 2% was observed from the value of 7.9, and the discontinuation of hypoglycemic treatment was reduced from 36.2% before surgery in 12.3% [153].
LAGB is associated with very low mortality and complication rates compared to other bariatric procedures. Perioperative mortality and morbidity rates reached 0.04 for death and 0.9% for morbidity [144]. The possible complications and shortcomings that may appear within this therapeutic approach are represented by the potential of the band to erode the gastric wall, along with its migration or sliding on the gastric wall, esophageal dilatation, all burdened by insufficient weakening [151]. After 15 years after the intervention, it was observed that 29% of the patients had the device intact and its position was the original, while the body mass index was around 35+/-7kg/m2, with an excellent BAROS score [154].
Biliopancreatic diversion with duodenal switch was first performed laparoscopically in 1999 [155] and is currently the most effective weight loss method [156]. This method has the highest resolution rate of comorbidities associated with excess weight, especially type II diabetes, with cure rates reaching 95% [157]. Indications for the procedure are related to morbid obesity, especially in patients with comorbidities, having the ability to maintain weight within normal limits even after several years [158]. Among the complications that can occur perioperatively, anastomotic fistula, along with pulmonary embolism and respiratory failure, are associated with super-obesity, with death rates between 0-2.7% depending on the technique used, higher rates being for laparoscopic techniques [157], while in 7% of cases reintervention is necessary in the first days [159]. In the long term, the results are satisfactory, only 5% of patients need re-intervention to treat possible metabolic complications. Although it has very good cure rates for type II diabetes, with weight loss and weight maintenance, it is burdened by numerous complications, being reserved only for experienced bariatric surgeons who must monitor the patient throughout life to detect possible subsequent complications [141].
Another alternative to surgical treatment is pancreas transplantation, one of the few treatments that can cure insulin-dependent diabetes, preventing its worsening and associated complications. Simultaneous kidney and pancreas transplantation is the most common form of transplantation in the US, and is most often indicated for those with associated chronic disease. Implantation of a pancreas without kidneys can be performed in patients with acute complications of hyperglycemia, such as ketoacidosis, poor glycemic control with insulin therapy, or in cases where patients cannot achieve their hypoglycemic state at certain times of the day. Renal function must be preserved, as immunosuppressive treatment with calcineurin has nephrotoxic potential [160]. An alternative to total pancreatic transplantation is pancreatic graft transplantation with or without associated renal transplantation, but associated with a high rate of complications such as pancreatic pseudocyst formation and anastomotic or pancreatic fistula [161].
Pancreatic B-cell transplantation is an alternative treatment method that does not require invasive surgical treatment, with moderate efficacy associated with acceptable post-transplant immunosurveillance. Most of the time the cells are infused into the portal vein or under the renal capsule according to the Edmonton protocol [162]. Regarding the results of the pancreas transplant operation, the mortality rate 1 year after the intervention reaches 4%, exceeding 9% in 5 years, in most cases the cause being an acute cardio-vascular event [160].
Comparatively, simultaneous pancreas and kidney transplantation has better survival outcomes than renal transplantation with associated hypoglycemic treatment. Similar results have been observed if only pancreatic transplantation is performed, provided that renal function is good [163]. Pancreatic graft rejection rate in pancreas transplantation is assessed by calculating insulin requirement and hemoglobin A1c level, so in patients who underwent pancreas transplantation after kidney transplantation, the risk of rejection at 1 year was only 14%, and 44% at 10 years (depending on recipient age, donor age, body mass index and severity of cardiovascular comorbidities) [164].
Following pancreatic transplantation, there was a normalization of carbohydrate metabolism, a decrease in serum lipids and the resumption of normal endothelial function with improvement of neuro and nephropathy, reduction of retinal eye complications but with an increase in the frequency of cataract secondary to treatment with calcineurin inhibitors [160].
Last but not least, diabetic patients with severe complicated peripheral arteriopathy (such as wet gangrene, etc.) may even end up with limb amputations [165].

Psychological treatment

In addition to medical, surgical, dietary treatment, the psychological aspect should not be neglected in the case of patients with diabetes, being able to improve the therapeutic effect of standard treatment methods [166]. By applying cognitive-behavioral therapy, motivational interviewing and patient-centered therapy, better levels of glycosylated hemoglobin were observed in adults with better emotional status. Thus, a concept was developed according to which psychological intervention through specific methods can additionally improve the quality of life of a chronic patient, as well as the level of blood sugar [167].

Cognitive behavioral therapy

Cognitive-behavioral treatment helps patients reorganize their thoughts, dysfunctional beliefs, and change their behavior so that their mental health improves. Since its first use in the treatment of depression, cognitive behavioral therapy has also been developed for various chronic diseases, including diabetes or heart failure [168]. Thus, a manual was developed to change the behavior of the patient with diabetes, giving him tasks that have the ability to change thoughts, negative habits that could aggravate a hyperglycemia, and at the same time help to develop healthy daily routines such as exercise at certain times and a low-carbohydrate diet [169].
Through sustained cognitive behavioral treatment, the effects of persistent hyperglycemia can be modulated, with a decrease in glycosylated hemoglobin levels and a reduction in the risk of general cardiovascular complications in these patients (the effect being felt in both type II and type I patients). Another parameter that was improved by cognitive-behavioral therapy is adherence to the treatment that directly influences the glycemic level, so patients who followed such therapy had better glycemic control and lower rates of giving up the basic treatment (or neglect regimen), compared to those who were not psychologically counseled. The best results were obtained if the treatment period was short, less than 6 weeks, while after this period patients become non-compliant, the mental fatigue and the cost of treatment being the main factors responsible for this response [170].
The interaction between patient and therapist can vary from remote (via a mobile phone or the Internet) to classic face-to-face or group therapy. Among patients with diabetes, good results were seen in patients who were counseled face to face because it was done in a group and the stimulating effect of this method made it more effective [171].
Comparatively, patients with type I diabetes respond better to cognitive behavioral therapy than those with type II diabetes. The first category of consumers of health services are young people. They have a life dependent on insulin therapy and sometimes feel burdened by life-threatening complications, being thus more prone to developing stress related to the disease compared to those with diabetes type II. Thus, psychological treatment in their case is based on acceptance, understanding the disease and developing a lifestyle that can better control the glycemic level [170].
With the onset of diabetes, a series of complications may occur such as chronic pain or bed rest secondary to vascular accidents or amputations, which will lead to high depression and anxiety [172]. Thus, it is necessary that once cognitive-behavioral therapy is initiated, it must also manage or prevent pre-existing psychiatric symptoms. A long-term therapy of more than 6 weeks that also includes a treatment focused on mood disorders improves both glycemic levels and improves or even eliminates anxiety and depression [173].
Through cognitive behavioral treatment, an improvement in patients' weight, cholesterol and HDL-cholesterol was attempted, but no statistical differences were identified between the patient with and without therapy. Only an improvement in the level of serum lipids secondary to weight loss and better glycemic control was noted [174].

Motivational interviewing

One of the best ways in which patients can change some negative aspects of their lives is motivational interviewing [175]. This technique relies on patients' ability to motivate themselves and engage in changing different behaviors by exploring different situations and eliminating uncertainties and hesitations [176]. Thus, in the treatment of chronic diseases such as diabetes, hypertension and obesity, the association of pharmacotherapy with lifestyle changes determined by motivational interviewing proves to be effective. It increases the degree of acceptance of the disease, and understanding it helps to remove its negative effects through a better ability to reduce the psychological and physical stress associated with the condition [177]. Within this technique, most of the time the psychotherapist has the role of working together with the patient as a partnership, they go together on the path of the disease and when they reach a certain point, the specialist informs the client about the possible options (what else could be done, which are the advantages and disadvantages of the possible methods), the patient being thus helped to solve his problem, but with his own contribution [178].
By applying this method in diabetes, it is possible to achieve decreases in glycosylated hemoglobin along with a reduction in cardiovascular complications of up to 25-35%. It could be a direct consequence, but also secondary to an improved lifestyle with physical activity, a diet rich in vegetables, fruits, lean products with a low intake of processed foods, rich in salt or sugar [179], as well as a better knowledge of the disease through the materials and information provided by the specialist. Similar results were obtained for serum lipid levels [170].
The impact of motivational interviewing was substantial in terms of self-management of the patient with diabetes. Dietary control, exercise and glucose monitoring were better accepted by patients following such therapy than those who were not motivated [180]. The reasons why patients do not comply with the associated diet were represented by not understanding the total calculation of calories consumed, maintaining a fixed meal schedule, what is the relationship between glycemic control and carbohydrate intake, how to get rid of unhealthy habits in the family or how to accept the feeling of hunger. Many of the patients also had problems accepting the need for exercise, integrating it into their lifestyle and work, or not understanding this need. Furthermore, without support from relatives, the exercise program may be initially accepted and later abandoned [181].

Conclusions

Diabetes mellitus is a complex and highly prevalent metabolic disease with significant implications for individuals and health systems worldwide. Its epidemiology is influenced by various factors, including age, genetic predisposition, lifestyle choices, etc. Understanding the pathophysiology of diabetes is crucial for the development of effective treatment strategies. Diabetes management involves a comprehensive approach that includes lifestyle changes, pharmacological interventions, surgical interventions in selected cases as well as psychological support. Ongoing research and advances in medical science aim to improve the prevention, early diagnosis and treatment of diabetes, to significantly improve the quality of life for people affected by the condition.

Compliance with ethical standards

Any aspect of the work covered in this manuscript has been conducted with the ethical approval of all relevant bodies and that such approvals are acknowledged within the manuscript. Informed consent was obtained from all subjects involved in the study.

Conflict of interest disclosure

There are no known conflicts of interest in the publication of this article. The manuscript was read and approved by all authors.

References

  1. Harreiter, J.; Roden, M. Diabetes mellitus-Definition, classification, diagnosis, screening and prevention (Update 2019). Wien Klin Wochenschr. 2019, 131, 6–15. [Google Scholar] [CrossRef] [PubMed]
  2. Nathan, D.M. Long-term complications of diabetes mellitus. N Engl J Med. 1993, 328, 1676–1685. [Google Scholar] [CrossRef] [PubMed]
  3. Cole, J.B.; Florez, J.C. Genetics of diabetes mellitus and diabetes complications. Nat Rev Nephrol. 2020, 16, 377–390. [Google Scholar] [CrossRef] [PubMed]
  4. Deckert, T.; Poulsen, J.E.; Larsen, M. Prognosis of diabetics with diabetes onset before the age of thirty-one. I. Survival, causes of death, and complications. Diabetologia. 1978, 14, 363–370. [Google Scholar] [CrossRef]
  5. Mauricio, D.; Alonso, N.; Gratacòs, M. Chronic Diabetes Complications: The Need to Move beyond Classical Concepts. Trends Endocrinol Metab. 2020, 31, 287–295. [Google Scholar] [CrossRef]
  6. Goldney, R.D.; Phillips, P.J.; Fisher, L.J.; Wilson, D.H. Diabetes, depression, and quality of life: a population study. Diabetes Care. 2004, 27, 1066–1070. [Google Scholar] [CrossRef]
  7. Kalyani, R.R.; Golden, S.H.; Cefalu, W.T. Diabetes and Aging: Unique Considerations and Goals of Care. Diabetes Care. 2017, 40, 440–443. [Google Scholar] [CrossRef]
  8. Forouhi, N.G.; Wareham, N.J. Epidemiology of diabetes. Medicine 2019, 47, 22–27. [Google Scholar] [CrossRef]
  9. Sun, H.; Saeedi, P.; Karuranga, S.; et al. IDF Diabetes Atlas: Global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Res Clin Pract. 2022, 183, 109119. [Google Scholar] [CrossRef]
  10. Rubin, R.J.; Altman, W.M.; Mendelson, D.N. Health care expenditures for people with diabetes mellitus, 1992. J Clin Endocrinol Metab. 1994, 78, 809A–809F. [Google Scholar] [CrossRef]
  11. Zheng, Y.; Ley, S.H.; Hu, F.B. Global aetiology and epidemiology of type 2 diabetes mellitus and its complications. Nat Rev Endocrinol. 2018, 14, 88–98. [Google Scholar] [CrossRef] [PubMed]
  12. Mekala, K.C.; Bertoni, A.G. Epidemiology of diabetes mellitus. Transplantation, Bioengineering, and Regeneration of the Endocrine Pancreas. 2020, 1, 49–58. [Google Scholar] [CrossRef]
  13. DIAMOND Project Group. Incidence and trends of childhood Type 1 diabetes worldwide 1990-1999. Diabet Med. 2006, 23, 857–866. [Google Scholar] [CrossRef]
  14. GBD 2015 Risk Factors Collaborators. Global, regional, and national comparative risk assessment of 79 behavioural, environmental and occupational, and metabolic risks or clusters of risks, 1990-2015: a systematic analysis for the Global Burden of Disease Study 2015. Lancet. 2016, 388, 1659–1724. [Google Scholar] [CrossRef]
  15. Diagnostic criteria and classification of hyperglycaemia first detected in pregnancy: a World Health Organization Guideline. Diabetes Res Clin Pract. 2014, 103, 341–363. [CrossRef]
  16. O'Dea, A.; Tierney, M.; McGuire, B.E.; et al. Can the Onset of Type 2 Diabetes Be Delayed by a Group-Based Lifestyle Intervention in Women with Prediabetes following Gestational Diabetes Mellitus (GDM)? Findings from a Randomized Control Mixed Methods Trial. J Diabetes Res. 2015, 2015, 798460. [Google Scholar] [CrossRef]
  17. Jelsma, J.G.; van Poppel, M.N.; Galjaard, S.; et al. DALI: Vitamin D and lifestyle intervention for gestational diabetes mellitus (GDM) prevention: an European multicentre, randomised trial - study protocol. BMC Pregnancy Childbirth. 2013, 13, 142, Published 2013 Jul 5. [Google Scholar] [CrossRef]
  18. Buckley, B.S.; Harreiter, J.; Damm, P.; et al. Gestational diabetes mellitus in Europe: prevalence, current screening practice and barriers to screening. A review. Diabet Med. 2012, 29, 844–854. [Google Scholar] [CrossRef]
  19. Egan, A.M.; Vellinga, A.; Harreiter, J.; et al. Epidemiology of gestational diabetes mellitus according to IADPSG/WHO 2013 criteria among obese pregnant women in Europe. Diabetologia. 2017, 60, 1913–1921. [Google Scholar] [CrossRef]
  20. Warshauer, J.T.; Bluestone, J.A.; Anderson, M.S. New Frontiers in the Treatment of Type 1 Diabetes. Cell Metab. 2020, 31, 46–61. [Google Scholar] [CrossRef]
  21. Barrett, J.C.; Clayton, D.G.; Concannon, P.; et al. Genome-wide association study and meta-analysis find that over 40 loci affect risk of type 1 diabetes. Nat Genet. 2009, 41, 703–707. [Google Scholar] [CrossRef] [PubMed]
  22. Concannon, P.; Rich, S.S.; Nepom, G.T. Genetics of type 1A diabetes. N Engl J Med. 2009, 360, 1646–1654. [Google Scholar] [CrossRef]
  23. Hansen, M.P.; Matheis, N.; Kahaly, G.J. Type 1 diabetes and polyglandular autoimmune syndrome: A review. World J Diabetes. 2015, 6, 67–79. [Google Scholar] [CrossRef]
  24. Bloem, S.J.; Roep, B.O. The elusive role of B lymphocytes and islet autoantibodies in (human) type 1 diabetes. Diabetologia. 2017, 60, 1185–1189. [Google Scholar] [CrossRef]
  25. Velthuis, J.H.; Unger, W.W.; van der Slik, A.R.; et al. Accumulation of autoreactive effector T cells and allo-specific regulatory T cells in the pancreas allograft of a type 1 diabetic recipient. Diabetologia. 2009, 52, 494–503. [Google Scholar] [CrossRef]
  26. Michels, A.W.; Landry, L.G.; McDaniel, K.A.; et al. Islet-Derived CD4 T Cells Targeting Proinsulin in Human Autoimmune Diabetes. Diabetes. 2017, 66, 722–734. [Google Scholar] [CrossRef]
  27. Kuric, E.; Seiron, P.; Krogvold, L.; et al. Demonstration of Tissue Resident Memory CD8 T Cells in Insulitic Lesions in Adult Patients with Recent-Onset Type 1 Diabetes. Am J Pathol. 2017, 187, 581–588. [Google Scholar] [CrossRef]
  28. Abreu, J.R.; Martina, S.; Verrijn Stuart, A.A.; et al. CD8 T cell autoreactivity to preproinsulin epitopes with very low human leucocyte antigen class I binding affinity. Clin Exp Immunol. 2012, 170, 57–65. [Google Scholar] [CrossRef]
  29. Unger, W.W.; Velthuis, J.; Abreu, J.R.; et al. Discovery of low-affinity preproinsulin epitopes and detection of autoreactive CD8 T-cells using combinatorial MHC multimers. J Autoimmun. 2011, 37, 151–159. [Google Scholar] [CrossRef]
  30. Beringer, D.X.; Kleijwegt, F.S.; Wiede, F.; et al. T cell receptor reversed polarity recognition of a self-antigen major histocompatibility complex. Nat Immunol. 2015, 16, 1153–1161. [Google Scholar] [CrossRef]
  31. Krogvold, L.; Edwin, B.; Buanes, T.; et al. Detection of a low-grade enteroviral infection in the islets of langerhans of living patients newly diagnosed with type 1 diabetes. Diabetes. 2015, 64, 1682–1687. [Google Scholar] [CrossRef] [PubMed]
  32. Roep, B.O. A viral link for type 1 diabetes. Nat Med. 2019, 25, 1816–1818. [Google Scholar] [CrossRef] [PubMed]
  33. Perrett, K.P.; Jachno, K.; Nolan, T.M.; Harrison, L.C. Association of Rotavirus Vaccination With the Incidence of Type 1 Diabetes in Children. JAMA Pediatr. 2019, 173, 280–282. [Google Scholar] [CrossRef] [PubMed]
  34. Hiemstra, H.S.; Schloot, N.C.; van Veelen, P.A.; et al. Cytomegalovirus in autoimmunity: T cell crossreactivity to viral antigen and autoantigen glutamic acid decarboxylase. Proc Natl Acad Sci U S A. 2001, 98, 3988–3991. [Google Scholar] [CrossRef]
  35. Morrison, D.J.; Preston, T. Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes. 2016, 7, 189–200. [Google Scholar] [CrossRef]
  36. Miani, M.; Le Naour, J.; Waeckel-Enée, E.; et al. Gut Microbiota-Stimulated Innate Lymphoid Cells Support β-Defensin 14 Expression in Pancreatic Endocrine Cells, Preventing Autoimmune Diabetes. Cell Metab. 2018, 28, 557–572.e6. [Google Scholar] [CrossRef]
  37. Pingitore, A.; Gonzalez-Abuin, N.; Ruz-Maldonado, I.; Huang, G.C.; Frost, G.; Persaud, S.J. Short chain fatty acids stimulate insulin secretion and reduce apoptosis in mouse and human islets in vitro: Role of free fatty acid receptor 2. Diabetes Obes Metab. 2019, 21, 330–339. [Google Scholar] [CrossRef]
  38. Antvorskov, J.C.; Halldorsson, T.I.; Josefsen, K.; et al. Association between maternal gluten intake and type 1 diabetes in offspring: national prospective cohort study in Denmark. BMJ. 2018, 362, k3547, Published 2018 Sep 19. [Google Scholar] [CrossRef]
  39. TRIGR Study Group; Akerblom, H.K.; Krischer, J.; et al. The Trial to Reduce IDDM in the Genetically at Risk (TRIGR) study: recruitment, intervention and follow-up. Diabetologia. 2011, 54, 627–633. [Google Scholar] [CrossRef]
  40. Stumvoll, M.; Goldstein, B.J.; van Haeften, T.W. Type 2 diabetes: principles of pathogenesis and therapy. Lancet. 2005, 365, 1333–1346. [Google Scholar] [CrossRef]
  41. Cerf, M.E. Beta cell dysfunction and insulin resistance. Front Endocrinol (Lausanne). 2013, 4, 37, Published 2013 Mar 27. [Google Scholar] [CrossRef] [PubMed]
  42. Rorsman, P.; Ashcroft, F.M. Pancreatic β-Cell Electrical Activity and Insulin Secretion: Of Mice and Men. Physiol Rev. 2018, 98, 117–214. [Google Scholar] [CrossRef] [PubMed]
  43. Boland, B.B.; Rhodes, C.J.; Grimsby, J.S. The dynamic plasticity of insulin production in β-cells. Mol Metab. 2017, 6, 958–973. [Google Scholar] [CrossRef]
  44. Cuíñas, A.; García-Morales, V.; Viña, D.; Gil-Longo, J.; Campos-Toimil, M. Activation of PKA and Epac proteins by cyclic AMP depletes intracellular calcium stores and reduces calcium availability for vasoconstriction. Life Sci. 2016, 155, 102–109. [Google Scholar] [CrossRef]
  45. Motofei, I.G.; Rowland, D.L.; Tampa, M.; et al. Finasteride and androgenic alopecia; from therapeutic options to medical implications. J Dermatolog Treat. 2020, 31, 415–421. [Google Scholar] [CrossRef]
  46. Christensen, A.A.; Gannon, M. The Beta Cell in Type 2 Diabetes. Curr Diab Rep. 2019, 19, 81, Published 2019 Aug 9. [Google Scholar] [CrossRef]
  47. Yamamoto, W.R.; Bone, R.N.; Sohn, P.; et al. Endoplasmic reticulum stress alters ryanodine receptor function in the murine pancreatic β cell. J Biol Chem. 2019, 294, 168–181. [Google Scholar] [CrossRef]
  48. Halban, P.A.; Polonsky, K.S.; Bowden, D.W.; et al. β-cell failure in type 2 diabetes: postulated mechanisms and prospects for prevention and treatment. Diabetes Care. 2014, 37, 1751–1758. [Google Scholar] [CrossRef]
  49. Dali-Youcef, N.; Mecili, M.; Ricci, R.; Andrès, E. Metabolic inflammation: connecting obesity and insulin resistance. Ann Med. 2013, 45, 242–253. [Google Scholar] [CrossRef]
  50. Roca-Rivada, A.; Castelao, C.; Senin, L.L.; et al. FNDC5/irisin is not only a myokine but also an adipokine. PLoS One. 2013, 8, e60563, Published 2013 Apr 11. [Google Scholar] [CrossRef]
  51. Giacco, F.; Brownlee, M. Oxidative stress and diabetic complications. Circ Res. 2010, 107, 1058–1070. [Google Scholar] [CrossRef] [PubMed]
  52. Bunney, P.E.; Zink, A.N.; Holm, A.A.; Billington, C.J.; Kotz, C.M. Orexin activation counteracts decreases in nonexercise activity thermogenesis (NEAT) caused by high-fat diet. Physiol Behav. 2017, 176, 139–148. [Google Scholar] [CrossRef]
  53. American Diabetes Association. 3. Prevention or Delay of Type 2 Diabetes: Standards of Medical Care in Diabetes-2019. Diabetes Care. 2019, 42, S29–S33. [Google Scholar] [CrossRef]
  54. Shamsuzzaman, A.S.; Winnicki, M.; Wolk, R.; et al. Independent association between plasma leptin and C-reactive protein in healthy humans. Circulation. 2004, 109, 2181–2185. [Google Scholar] [CrossRef]
  55. Leeuwenburgh, C.; Fiebig, R.; Chandwaney, R.; Ji, L.L. Aging and exercise training in skeletal muscle: responses of glutathione and antioxidant enzyme systems. Am J Physiol. 1994, 267, R439–R445. [Google Scholar] [CrossRef]
  56. Lynch, S.V.; Pedersen, O. The Human Intestinal Microbiome in Health and Disease. N Engl J Med. 2016, 375, 2369–2379. [Google Scholar] [CrossRef]
  57. Li, X.; Watanabe, K.; Kimura, I. Gut Microbiota Dysbiosis Drives and Implies Novel Therapeutic Strategies for Diabetes Mellitus and Related Metabolic Diseases. Front Immunol. 2017, 8, 1882. [Google Scholar] [CrossRef]
  58. Shan, Z.; Sun, T.; Huang, H.; et al. Association between microbiota-dependent metabolite trimethylamine-N-oxide and type 2 diabetes. Am J Clin Nutr. 2017, 106, 888–894. [Google Scholar] [CrossRef]
  59. Neis, E.P.; Dejong, C.H.; Rensen, S.S. The role of microbial amino acid metabolism in host metabolism. Nutrients. 2015, 7, 2930–2946, Published 2015 Apr 16. [Google Scholar] [CrossRef]
  60. Di Cianni, G.; Miccoli, R.; Volpe, L.; Lencioni, C.; Del Prato, S. Intermediate metabolism in normal pregnancy and in gestational diabetes. Diabetes Metab Res Rev. 2003, 19, 259–270. [Google Scholar] [CrossRef]
  61. Catalano, P.M.; Tyzbir, E.D.; Roman, N.M.; Amini, S.B.; Sims, E.A. Longitudinal changes in insulin release and insulin resistance in nonobese pregnant women. Am J Obstet Gynecol. 1991, 165, 1667–1672. [Google Scholar] [CrossRef] [PubMed]
  62. Parsons, J.A.; Brelje, T.C.; Sorenson, R.L. Adaptation of islets of Langerhans to pregnancy: increased islet cell proliferation and insulin secretion correlates with the onset of placental lactogen secretion. Endocrinology. 1992, 130, 1459–1466. [Google Scholar] [CrossRef] [PubMed]
  63. Prentki, M.; Nolan, C.J. Islet beta cell failure in type 2 diabetes. J Clin Invest. 2006, 116, 1802–1812. [Google Scholar] [CrossRef]
  64. Loghin, M.G.; Gorescki, P.G.; Sima, R.M.; Pleș, L.; Balan, D.G.; et al. The obstetrical management of HIV-positive pregnancy. J Mind Med Sci. 2022, 9, 111–117. [Google Scholar] [CrossRef]
  65. Radaelli, T.; Lepercq, J.; Varastehpour, A.; Basu, S.; Catalano, P.M.; Hauguel-De Mouzon, S. Differential regulation of genes for fetoplacental lipid pathways in pregnancy with gestational and type 1 diabetes mellitus. Am J Obstet Gynecol. 2009, 201, 209.e1–209.e10. [Google Scholar] [CrossRef]
  66. Catalano, P.M.; McIntyre, H.D.; Cruickshank, J.K.; et al. The hyperglycemia and adverse pregnancy outcome study: associations of GDM and obesity with pregnancy outcomes. Diabetes Care. 2012, 35, 780–786. [Google Scholar] [CrossRef]
  67. Dolinoy, D.C. The agouti mouse model: an epigenetic biosensor for nutritional and environmental alterations on the fetal epigenome. Nutr Rev. 2008, 66, S7–S11. [Google Scholar] [CrossRef]
  68. Park, S.; Kang, H.J.; Jeon, J.H.; Kim, M.J.; Lee, I.K. Recent advances in the pathogenesis of microvascular complications in diabetes. Arch Pharm Res. 2019, 42, 252–262. [Google Scholar] [CrossRef]
  69. Forbes, J.M.; Cooper, M.E. Mechanisms of diabetic complications. Physiol Rev. 2013, 93, 137–188. [Google Scholar] [CrossRef]
  70. Pathak, D.; Gupta, A.; Kamble, B.; Kuppusamy, G.; Suresh, B. Oral targeting of protein kinase C receptor: promising route for diabetic retinopathy? Curr Drug Deliv. 2012, 9, 405–413. [Google Scholar] [CrossRef]
  71. Shi, G.J.; Shi, G.R.; Zhou, J.Y.; et al. Involvement of growth factors in diabetes mellitus and its complications: A general review. Biomed Pharmacother. 2018, 101, 510–527. [Google Scholar] [CrossRef] [PubMed]
  72. Soltesova Prnova, M.; Ballekova, J.; Gajdosikova, A.; Gajdosik, A.; Stefek, M. A novel carboxymethylated mercaptotriazinoindole inhibitor of aldose reductase interferes with the polyol pathway in streptozotocin-induced diabetic rats. Physiol Res. 2015, 64, 587–591. [Google Scholar] [CrossRef] [PubMed]
  73. Gugliucci, A. Formation of Fructose-Mediated Advanced Glycation End Products and Their Roles in Metabolic and Inflammatory Diseases. Adv Nutr. 2017, 8, 54–62. [Google Scholar] [CrossRef] [PubMed]
  74. Brownlee, M. Biochemistry and molecular cell biology of diabetic complications. Nature. 2001, 414, 813–820. [Google Scholar] [CrossRef]
  75. Spector, A. Some aspects of Dr Kinoshita's contributions to lens protein chemistry. Exp Eye Res. 1990, 50, 689–694. [Google Scholar] [CrossRef]
  76. Srikanth, K.K.; Orrick, J.A. Biochemistry, Polyol Or Sorbitol Pathways; StatPearls Publishing: Treasure Island, FL, USA, 14 November 2022. [Google Scholar]
  77. Brahma, M.K.; Pepin, M.E.; Wende, A.R. My Sweetheart Is Broken: Role of Glucose in Diabetic Cardiomyopathy. Diabetes Metab J. 2017, 41, 1–9. [Google Scholar] [CrossRef]
  78. Ulrich, P.; Cerami, A. Protein glycation, diabetes, and aging. Recent Prog Horm Res. 2001, 56, 1–21. [Google Scholar] [CrossRef]
  79. Rhee, S.Y.; Kim, Y.S. The Role of Advanced Glycation End Products in Diabetic Vascular Complications. Diabetes Metab J. 2018, 42, 188–195. [Google Scholar] [CrossRef]
  80. Thornalley, P.J.; Langborg, A.; Minhas, H.S. Formation of glyoxal, methylglyoxal and 3-deoxyglucosone in the glycation of proteins by glucose. Biochem J. 1999, 344, 109–116. [Google Scholar] [CrossRef]
  81. Chetyrkin, S.; Mathis, M.; Pedchenko, V.; et al. Glucose autoxidation induces functional damage to proteins via modification of critical arginine residues. Biochemistry. 2011, 50, 6102–6112. [Google Scholar] [CrossRef]
  82. Dahlén, A.D.; Dashi, G.; Maslov, I.; et al. Trends in Antidiabetic Drug Discovery: FDA Approved Drugs, New Drugs in Clinical Trials and Global Sales. Front Pharmacol. 2022, 12, 807548, Published 2022 Jan 19. [Google Scholar] [CrossRef] [PubMed]
  83. Blahova, J.; Martiniakova, M.; Babikova, M.; Kovacova, V.; Mondockova, V.; Omelka, R. Pharmaceutical Drugs and Natural Therapeutic Products for the Treatment of Type 2 Diabetes Mellitus. Pharmaceuticals (Basel). 2021, 14, 806. [Google Scholar] [CrossRef] [PubMed]
  84. Marín-Peñalver, J.J.; Martín-Timón, I.; Sevillano-Collantes, C.; Del Cañizo-Gómez, F.J. Update on the treatment of type 2 diabetes mellitus. World J Diabetes. 2016, 7, 354–395. [Google Scholar] [CrossRef]
  85. Lazzaroni, E.; Ben Nasr, M.; Loretelli, C.; et al. Anti-diabetic drugs and weight loss in patients with type 2 diabetes. Pharmacol Res. 2021, 171, 105782. [Google Scholar] [CrossRef]
  86. Janež, A.; Guja, C.; Mitrakou, A.; et al. Insulin Therapy in Adults with Type 1 Diabetes Mellitus: a Narrative Review. Diabetes Ther. 2020, 11, 387–409. [Google Scholar] [CrossRef]
  87. Heise, T.; Mathieu, C. Impact of the mode of protraction of basal insulin therapies on their pharmacokinetic and pharmacodynamic properties and resulting clinical outcomes. Diabetes Obes Metab. 2017, 19, 3–12. [Google Scholar] [CrossRef]
  88. Lepore, M.; Pampanelli, S.; Fanelli, C.; et al. Pharmacokinetics and pharmacodynamics of subcutaneous injection of long-acting human insulin analog glargine, NPH insulin, and ultralente human insulin and continuous subcutaneous infusion of insulin lispro. Diabetes. 2000, 49, 2142–2148. [Google Scholar] [CrossRef]
  89. Fulcher, G.R.; Gilbert, R.E.; Yue, D.K. Glargine is superior to neutral protamine Hagedorn for improving glycated haemoglobin and fasting blood glucose levels during intensive insulin therapy. Intern Med J. 2005, 35, 536–542. [Google Scholar] [CrossRef]
  90. Hopkinson, H.E.; Jacques, R.M.; Gardner, K.J.; Amiel, S.A.; Mansell, P. Twice-rather than once-daily basal insulin is associated with better glycaemic control in Type 1 diabetes mellitus 12 months after skills-based structured education in insulin self-management. Diabet Med. 2015, 32, 1071–1076. [Google Scholar] [CrossRef]
  91. Renard, E.; Dubois-Laforgue, D.; Guerci, B.; Variability Study Group. Non-inferiority of insulin glargine versus insulin detemir on blood glucose variability in type 1 diabetes patients: a multicenter, randomized, crossover study. Diabetes Technol Ther. 2011, 13, 1213–1218. [Google Scholar] [CrossRef]
  92. Laranjeira, F.O.; de Andrade, K.R.C.; Figueiredo, A.C.M.G.; Silva, E.N.; Pereira, M.G. Long-acting insulin analogues for type 1 diabetes: An overview of systematic reviews and meta-analysis of randomized controlled trials. PLoS One. 2018, 13, e0194801, Published 2018 Apr 12. [Google Scholar] [CrossRef] [PubMed]
  93. Heller, S.; Buse, J.; Fisher, M.; et al. Insulin degludec, an ultra-longacting basal insulin, versus insulin glargine in basal-bolus treatment with mealtime insulin aspart in type 1 diabetes (BEGIN Basal-Bolus Type 1): a phase 3, randomised, open-label, treat-to-target non-inferiority trial. Lancet. 2012, 379, 1489–1497. [Google Scholar] [CrossRef] [PubMed]
  94. International Hypoglycaemia Study Group. Minimizing Hypoglycemia in Diabetes. Diabetes Care. 2015, 38, 1583–1591. [Google Scholar] [CrossRef] [PubMed]
  95. EQuality1 Study Group--Evaluation of QUALITY of Life and Costs in Diabetes Type 1; Nicolucci, A.; Maione, A.; et al. Quality of life and treatment satisfaction in adults with Type 1 diabetes: a comparison between continuous subcutaneous insulin infusion and multiple daily injections. Diabet Med. 2008, 25, 213–220. [Google Scholar] [CrossRef]
  96. Singh, S.R.; Ahmad, F.; Lal, A.; Yu, C.; Bai, Z.; Bennett, H. Efficacy and safety of insulin analogues for the management of diabetes mellitus: a meta-analysis. CMAJ. 2009, 180, 385–397. [Google Scholar] [CrossRef]
  97. Renner, R.; Pfützner, A.; Trautmann, M.; Harzer, O.; Sauter, K.; Landgraf, R. Use of insulin lispro in continuous subcutaneous insulin infusion treatment. Results of a multicenter trial. German Humalog-CSII Study Group. Diabetes Care. 1999, 22, 784–788. [Google Scholar] [CrossRef]
  98. Gao, L.; Wang, T.; Jia, K.; et al. Glucose-responsive supramolecular vesicles based on water-soluble pillar[5]arene and pyridylboronic acid derivatives for controlled insulin delivery. Chemistry. 2017, 23, 6605–6614. [Google Scholar] [CrossRef]
  99. Kalra, S.; Joshi, A.; Parmar, G. Insulin therapy: going the "smarter" way. Recent Pat Endocr Metab Immune Drug Discov. 2014, 8, 79–84. [Google Scholar] [CrossRef]
  100. Rege, N.K.; Phillips, N.F.B.; Weiss, M.A. Development of glucose-responsive 'smart' insulin systems. Curr Opin Endocrinol Diabetes Obes. 2017, 24, 267–278. [Google Scholar] [CrossRef]
  101. Yu, J.; Qian, C.; Zhang, Y.; et al. Hypoxia and H2O2 Dual-Sensitive Vesicles for Enhanced Glucose-Responsive Insulin Delivery. Nano Lett. 2017, 17, 733–739. [Google Scholar] [CrossRef]
  102. Cernea, S.; Raz, I. Insulin Therapy: Future Perspectives. Am J Ther. 2020, 27, e121–e132. [Google Scholar] [CrossRef] [PubMed]
  103. Bailey, T.S.; Stone, J.Y. A novel pen-based Bluetooth-enabled insulin delivery system with insulin dose tracking and advice. Expert Opin Drug Deliv. 2017, 14, 697–703. [Google Scholar] [CrossRef] [PubMed]
  104. Guo, L.; Xiao, X.; Sun, X.; Qi, C. Comparison of jet injector and insulin pen in controlling plasma glucose and insulin concentrations in type 2 diabetic patients. Medicine (Baltimore). 2017, 96, e5482. [Google Scholar] [CrossRef] [PubMed]
  105. Thompson, B.; Cook, C.B. Insulin Pumping Patches: Emerging Insulin Delivery Systems. J Diabetes Sci Technol. 2019, 13, 8–10. [Google Scholar] [CrossRef]
  106. Giménez, M.; Purkayajtha, S.; Moscardó, V.; Conget, I.; Oliver, N. Intraperitoneal insulin therapy in patients with type 1 diabetes. Does it fit into the current therapeutic arsenal? Endocrinol Diabetes Nutr (Engl Ed). 2018, 65, 182–184. [Google Scholar] [CrossRef]
  107. Heinemann, L.; Krinelke, L. Insulin infusion set: the Achilles heel of continuous subcutaneous insulin infusion. J Diabetes Sci Technol. 2012, 6, 954–964. [Google Scholar] [CrossRef]
  108. Gómez, A.M.; Henao, D.C.; Taboada, L.B.; et al. Impact of sensor-augmented pump therapy with predictive low-glucose management on hypoglycemia and glycemic control in patients with type 1 diabetes mellitus: 1-year follow-up. Diabetes Metab Syndr. 2019, 13, 2625–2631. [Google Scholar] [CrossRef]
  109. Bux Rodeman, K.; Hatipoglu, B. Beta-cell therapies for type 1 diabetes: Transplants and bionics. Cleve Clin J Med. 2018, 85, 931–937. [Google Scholar] [CrossRef]
  110. Rendell, M. The role of sulphonylureas in the management of type 2 diabetes mellitus. Drugs. 2004, 64, 1339–1358. [Google Scholar] [CrossRef]
  111. van Baar, M.J.B.; van Ruiten, C.C.; Muskiet, M.H.A.; van Bloemendaal, L.; IJzerman, R.G.; van Raalte, D.H. SGLT2 Inhibitors in Combination Therapy: From Mechanisms to Clinical Considerations in Type 2 Diabetes Management. Diabetes Care. 2018, 41, 1543–1556. [Google Scholar] [CrossRef]
  112. Zangeneh, F.; Kudva, Y.C.; Basu, A. Insulin sensitizers. Mayo Clin Proc. 2003, 78, 471–479. [Google Scholar] [CrossRef] [PubMed]
  113. Rena, G.; Hardie, D.G.; Pearson, E.R. The mechanisms of action of metformin. Diabetologia. 2017, 60, 1577–1585. [Google Scholar] [CrossRef] [PubMed]
  114. Simpson, C.M.; Calori, G.M.; Giannoudis, P.V. Diabetes and fracture healing: the skeletal effects of diabetic drugs. Expert Opin Drug Saf. 2012, 11, 215–220. [Google Scholar] [CrossRef] [PubMed]
  115. Rena, G.; Lang, C.C. Repurposing Metformin for Cardiovascular Disease. Circulation. 2018, 137, 422–424. [Google Scholar] [CrossRef]
  116. Bailey, T. Options for combination therapy in type 2 diabetes: comparison of the ADA/EASD position statement and AACE/ACE algorithm. Am J Med. 2013, 126, S10–S20. [Google Scholar] [CrossRef]
  117. Fuhlendorff, J.; Rorsman, P.; Kofod, H.; et al. Stimulation of insulin release by repaglinide and glibenclamide involves both common and distinct processes. Diabetes. 1998, 47, 345–351. [Google Scholar] [CrossRef]
  118. Guardado-Mendoza, R.; Prioletta, A.; Jiménez-Ceja, L.M.; Sosale, A.; Folli, F. The role of nateglinide and repaglinide, derivatives of meglitinide, in the treatment of type 2 diabetes mellitus. Arch Med Sci. 2013, 9, 936–943. [Google Scholar] [CrossRef]
  119. Simone, M.I.; Wood, A.; Campkin, D.; Kiefel, M.J.; Houston, T.A. Recent results from non-basic glycosidase inhibitors: How structural diversity can inform general strategies for improving inhibition potency. Eur J Med Chem. 2022, 235, 114282. [Google Scholar] [CrossRef]
  120. Nguyen, V.B.; Nguyen, A.D.; Kuo, Y.H.; Wang, S.L. Biosynthesis of α-Glucosidase Inhibitors by a Newly Isolated Bacterium, Paenibacillus sp. TKU042 and Its Effect on Reducing Plasma Glucose in a Mouse Model. Int J Mol Sci. 2017, 18, 700, Published 2017 Mar 25. [Google Scholar] [CrossRef]
  121. Tan, Q.; Akindehin, S.E.; Orsso, C.E.; et al. Recent Advances in Incretin-Based Pharmacotherapies for the Treatment of Obesity and Diabetes. Front Endocrinol (Lausanne). 2022, 13, 838410, Published 2022 Mar 1. [Google Scholar] [CrossRef]
  122. Nauck, M.A.; Meier, J.J. MANAGEMENT OF ENDOCRINE DISEASE: Are all GLP-1 agonists equal in the treatment of type 2 diabetes? Eur J Endocrinol. 2019, 181, R211–R234. [Google Scholar] [CrossRef] [PubMed]
  123. Scheen, A.J. Dual GIP/GLP-1 receptor agonists: New advances for treating type-2 diabetes. Ann Endocrinol (Paris). 2023, 84, 316–321. [Google Scholar] [CrossRef] [PubMed]
  124. Berra, C.C.; Resi, V.; Mirani, M.; et al. Clinical efficacy and predictors of response to dulaglutide in type-2 diabetes. Pharmacol Res. 2020, 159, 104996. [Google Scholar] [CrossRef] [PubMed]
  125. Kendall, D.M.; Cuddihy, R.M.; Bergenstal, R.M. Clinical application of incretin-based therapy: therapeutic potential, patient selection and clinical use. Am J Med. 2009, 122, S37–S50. [Google Scholar] [CrossRef]
  126. Kazafeos, K. Incretin effect: GLP-1, GIP, DPP4. Diabetes Res Clin Pract. 2011, 93, S32–S36. [Google Scholar] [CrossRef]
  127. Yang, L.; Yuan, J.; Zhou, Z. Emerging roles of dipeptidyl peptidase 4 inhibitors: anti-inflammatory and immunomodulatory effect and its application in diabetes mellitus. Can J Diabetes. 2014, 38, 473–479. [Google Scholar] [CrossRef]
  128. Packer, M. Is the Popularity of Dipeptidyl-Peptidase-4 Inhibitors Justified? Insights From Mechanistic Studies and Clinical Trials. Am J Med. 2018, 131, e287–e289. [Google Scholar] [CrossRef]
  129. Moradi-Marjaneh, R.; Paseban, M.; Sahebkar, A. Natural products with SGLT2 inhibitory activity: Possibilities of application for the treatment of diabetes. Phytother Res. 2019, 33, 2518–2530. [Google Scholar] [CrossRef]
  130. Tentolouris, A.; Vlachakis, P.; Tzeravini, E.; Eleftheriadou, I.; Tentolouris, N. SGLT2 Inhibitors: A Review of Their Antidiabetic and Cardioprotective Effects. Int J Environ Res Public Health. 2019, 16, 2965. [Google Scholar] [CrossRef]
  131. Feutren, G.; Papoz, L.; Assan, R.; et al. Cyclosporin increases the rate and length of remissions in insulin-dependent diabetes of recent onset. Results of a multicentre double-blind trial. Lancet. 1986, 2, 119–124. [Google Scholar] [CrossRef]
  132. Rigby, M.R.; Harris, K.M.; Pinckney, A.; et al. Alefacept provides sustained clinical and immunological effects in new-onset type 1 diabetes patients. J Clin Invest. 2015, 125, 3285–3296. [Google Scholar] [CrossRef] [PubMed]
  133. Orban, T.; Bundy, B.; Becker, D.J.; et al. Co-stimulation modulation with abatacept in patients with recent-onset type 1 diabetes: a randomised, double-blind, placebo-controlled trial. Lancet. 2011, 378, 412–419. [Google Scholar] [CrossRef] [PubMed]
  134. Pescovitz, M.D.; Greenbaum, C.J.; Bundy, B.; et al. B-lymphocyte depletion with rituximab and β-cell function: two-year results. Diabetes Care. 2014, 37, 453–459. [Google Scholar] [CrossRef] [PubMed]
  135. Dinarello, C.A.; van der Meer, J.W. Treating inflammation by blocking interleukin-1 in humans. Semin Immunol. 2013, 25, 469–484. [Google Scholar] [CrossRef]
  136. Hägerkvist, R.; Sandler, S.; Mokhtari, D.; Welsh, N. Amelioration of diabetes by imatinib mesylate (Gleevec): role of beta-cell NF-kappaB activation and anti-apoptotic preconditioning. FASEB J. 2007, 21, 618–628. [Google Scholar] [CrossRef]
  137. Pareek, M.; Schauer, P.R.; Kaplan, L.M.; Leiter, L.A.; Rubino, F.; Bhatt, D.L. Metabolic Surgery: Weight Loss, Diabetes, and Beyond. J Am Coll Cardiol. 2018, 71, 670–687. [Google Scholar] [CrossRef]
  138. Buchwald, H. The evolution of metabolic/bariatric surgery. Obes Surg. 2014, 24, 1126–1135. [Google Scholar] [CrossRef]
  139. Angrisani, L.; Santonicola, A.; Iovino, P.; et al. Bariatric Surgery and Endoluminal Procedures: IFSO Worldwide Survey 2014. Obes Surg. 2017, 27, 2279–2289. [Google Scholar] [CrossRef]
  140. Salminen, P.; Helmiö, M.; Ovaska, J.; et al. Effect of Laparoscopic Sleeve Gastrectomy vs Laparoscopic Roux-en-Y Gastric Bypass on Weight Loss at 5 Years Among Patients With Morbid Obesity: The SLEEVEPASS Randomized Clinical Trial. JAMA. 2018, 319, 241–254. [Google Scholar] [CrossRef]
  141. Brethauer, S.A.; Hammel, J.P.; Schauer, P.R. Systematic review of sleeve gastrectomy as staging and primary bariatric procedure. Surg Obes Relat Dis. 2009, 5, 469–475. [Google Scholar] [CrossRef]
  142. Juodeikis, Ž.; Brimas, G. Long-term results after sleeve gastrectomy: A systematic review. Surg Obes Relat Dis. 2017, 13, 693–699. [Google Scholar] [CrossRef] [PubMed]
  143. Switzer, N.J.; Prasad, S.; Debru, E.; Church, N.; Mitchell, P.; Gill, R.S. Sleeve Gastrectomy and Type 2 Diabetes Mellitus: a Systematic Review of Long-Term Outcomes. Obes Surg. 2016, 26, 1616–1621. [Google Scholar] [CrossRef] [PubMed]
  144. Birkmeyer, N.J.; Dimick, J.B.; Share, D.; et al. Hospital complication rates with bariatric surgery in Michigan. JAMA. 2010, 304, 435–442. [Google Scholar] [CrossRef] [PubMed]
  145. Alvarenga, E.S.; Lo Menzo, E.; Szomstein, S.; Rosenthal, R.J. Safety and efficacy of 1020 consecutive laparoscopic sleeve gastrectomies performed as a primary treatment modality for morbid obesity. A single-center experience from the metabolic and bariatric surgical accreditation quality and improvement program. Surg Endosc. 2016, 30, 2673–2678. [Google Scholar] [CrossRef]
  146. Wittgrove, A.C.; Clark, G.W.; Tremblay, L.J. Laparoscopic Gastric Bypass, Roux-en-Y: Preliminary Report of Five Cases. Obes Surg. 1994, 4, 353–357. [Google Scholar] [CrossRef]
  147. Rubino, F.; Nathan, D.M.; Eckel, R.H.; et al. Metabolic Surgery in the Treatment Algorithm for Type 2 Diabetes: a Joint Statement by International Diabetes Organizations. Obes Surg. 2017, 27, 2–21. [Google Scholar] [CrossRef]
  148. Bhandari, M.; Fobi, M.A.L.; Buchwald, J.N.; Bariatric Metabolic Surgery Standardization (BMSS) Working Group:. Standardization of Bariatric Metabolic Procedures: World Consensus Meeting Statement. Obes Surg. 2019, 29, 309–345. [Google Scholar] [CrossRef]
  149. Silaghi, A.; Gaspar, B.S.; Epistatu, D.; et al. Upper gastrointestinal bleeding in the COVID-19 pandemic; particularities of diagnosis and therapy. J Mind Med Sci. 2022, 9, 276–284. [Google Scholar] [CrossRef]
  150. Brown, W.A.; O'Brien, P.E. The Band Must Not Be Abandoned. Obes Surg. 2017, 27, 1911–1913. [Google Scholar] [CrossRef]
  151. Giet, L.; Baker, J.; Favretti, F.; et al. Medium and long-term results of gastric banding: outcomes from a large private clinic in UK. BMC Obes. 2018, 5, 12, Published 2018 Apr 12. [Google Scholar] [CrossRef]
  152. Ooi, G.J.; Doyle, L.; Tie, T.; et al. Weight loss after laparoscopic adjustable gastric band and resolution of the metabolic syndrome and its components. Int J Obes (Lond). 2017, 41, 902–908. [Google Scholar] [CrossRef] [PubMed]
  153. Mistry, P.; Currie, V.; Super, P.; le Roux, C.W.; Tahrani, A.A.; Singhal, R. Changes in glycaemic control, blood pressure and lipids 5 years following laparoscopic adjustable gastric banding combined with medical care in patients with type 2 diabetes: a longitudinal analysis. Clin Obes. 2018, 8, 151–158. [Google Scholar] [CrossRef] [PubMed]
  154. Vinzens, F.; Kilchenmann, A.; Zumstein, V.; Slawik, M.; Gebhart, M.; Peterli, R. Long-term outcome of laparoscopic adjustable gastric banding (LAGB): results of a Swiss single-center study of 405 patients with up to 18 years' follow-up. Surg Obes Relat Dis. 2017, 13, 1313–1319. [Google Scholar] [CrossRef] [PubMed]
  155. Ren, C.J.; Patterson, E.; Gagner, M. Early results of laparoscopic biliopancreatic diversion with duodenal switch: a case series of 40 consecutive patients. Obes Surg. 2000, 10, 514–524. [Google Scholar] [CrossRef]
  156. Buchwald, H.; Avidor, Y.; Braunwald, E.; et al. Bariatric surgery: a systematic review and meta-analysis. JAMA. 2004, 292, 1724–1737. [Google Scholar] [CrossRef]
  157. Buchwald, H.; Estok, R.; Fahrbach, K.; et al. Weight and type 2 diabetes after bariatric surgery: systematic review and meta-analysis. Am J Med. 2009, 122, 248–256.e5. [Google Scholar] [CrossRef]
  158. Ballesteros-Pomar, M.D.; González de Francisco, T.; Urioste-Fondo, A.; et al. Biliopancreatic Diversion for Severe Obesity: Long-Term Effectiveness and Nutritional Complications. Obes Surg. 2016, 26, 38–44. [Google Scholar] [CrossRef]
  159. Biertho, L.; Lebel, S.; Marceau, S.; et al. Perioperative complications in a consecutive series of 1000 duodenal switches. Surg Obes Relat Dis. 2013, 9, 63–68. [Google Scholar] [CrossRef]
  160. Samoylova, M.L.; Borle, D.; Ravindra, K.V. Pancreas Transplantation: Indications, Techniques, and Outcomes. Surg Clin North Am. 2019, 99, 87–101. [Google Scholar] [CrossRef]
  161. Sutherland, D.E.; Gruessner, R.; Dunn, D.; Moudry-Munns, K.; Gruessner, A.; Najarian, J.S. Pancreas transplants from living-related donors. Transplant Proc. 1994, 26, 443–445. [Google Scholar]
  162. Shapiro, A.M.; Ricordi, C.; Hering, B.J.; et al. International trial of the Edmonton protocol for islet transplantation. N Engl J Med. 2006, 355, 1318–1330. [Google Scholar] [CrossRef] [PubMed]
  163. Kleinclauss, F.; Fauda, M.; Sutherland, D.E.; et al. Pancreas after living donor kidney transplants in diabetic patients: impact on long-term kidney graft function. Clin Transplant. 2009, 23, 437–446. [Google Scholar] [CrossRef] [PubMed]
  164. Gruessner, AC. 2011 update on pancreas transplantation: comprehensive trend analysis of 25,000 cases followed up over the course of twenty-four years at the International Pancreas Transplant Registry (IPTR). Rev Diabet Stud. 2011, 8, 6–16. [Google Scholar] [CrossRef]
  165. Constantin, V.D.; Socea, B.; Gaspar, B.S.; Epistatu, D.; Paunica, I.; Dumitriu, A.S.; Paunica, S.; Silaghi, A. Limb amputations; etiopathogenesis, diagnosis and the multidisciplinary therapeutic approach. J Mind Med Sci. 2022, 9, 209–223. [Google Scholar] [CrossRef]
  166. American Diabetes Association. Standards of medical care in diabetes--2013. Diabetes Care. 2013, 36, S11–S66. [Google Scholar] [CrossRef]
  167. Chapman, A.; Liu, S.; Merkouris, S.; et al. Psychological Interventions for the Management of Glycemic and Psychological Outcomes of Type 2 Diabetes Mellitus in China: A Systematic Review and Meta-Analyses of Randomized Controlled Trials. Front Public Health. 2015, 3, 252. [Google Scholar] [CrossRef]
  168. Motofei, I.G. A dual physiological character for cerebral mechanisms of sexuality and cognition: common somatic peripheral afferents. BJU Int. 2011, 108, 1634–1639. [Google Scholar] [CrossRef]
  169. Pinhas-Hamiel, O.; Hamiel, D. Cognitive Behavioral Therapy and Mindfulness-Based Cognitive Therapy in Children and Adolescents with Type 2 Diabetes. Curr Diab Rep. 2020, 20, 55, Published 2020 Sep 22. [Google Scholar] [CrossRef]
  170. Yang, X.; Li, Z.; Sun, J. Effects of Cognitive Behavioral Therapy-Based Intervention on Improving Glycaemic, Psychological, and Physiological Outcomes in Adult Patients With Diabetes Mellitus: A Meta-Analysis of Randomized Controlled Trials. Front Psychiatry. 2020, 11, 711. [Google Scholar] [CrossRef]
  171. Kumar, V.; Sattar, Y.; Bseiso, A.; Khan, S.; Rutkofsky, I.H. The Effectiveness of Internet-Based Cognitive Behavioral Therapy in Treatment of Psychiatric Disorders. Cureus. 2017, 9, e1626, Published 2017 Aug 29. [Google Scholar] [CrossRef]
  172. Wang, S.B.; Wang, Y.Y.; Zhang, Q.E.; et al. Cognitive behavioral therapy for post-stroke depression: A meta-analysis. J Affect Disord. 2018, 235, 589–596. [Google Scholar] [CrossRef] [PubMed]
  173. Uchendu, C.; Blake, H. Effectiveness of cognitive-behavioural therapy on glycaemic control and psychological outcomes in adults with diabetes mellitus: a systematic review and meta-analysis of randomized controlled trials. Diabet Med. 2017, 34, 328–339. [Google Scholar] [CrossRef] [PubMed]
  174. Serlachius, A.S.; Scratch, S.E.; Northam, E.A.; Frydenberg, E.; Lee, K.J.; Cameron, F.J. A randomized controlled trial of cognitive behaviour therapy to improve glycaemic control and psychosocial wellbeing in adolescents with type 1 diabetes. J Health Psychol. 2016, 21, 1157–1169. [Google Scholar] [CrossRef] [PubMed]
  175. Alvarado-Martel, D.; Boronat, M.; Alberiche-Ruano, M.D.P.; Algara-González, M.A.; Ramallo-Fariña, Y.; Wägner, A.M. Motivational Interviewing and Self-Care in Type 1 Diabetes: A Randomized Controlled Clinical Trial Study Protocol. Front Endocrinol (Lausanne). 2020, 11, 574312, Published 2020 Dec 10. [Google Scholar] [CrossRef]
  176. Soderlund, P.D. Effectiveness of motivational interviewing for improving physical activity self-management for adults with type 2 diabetes: A review. Chronic Illn. 2018, 14, 54–68. [Google Scholar] [CrossRef]
  177. Ekong, G.; Kavookjian, J. Motivational interviewing and outcomes in adults with type 2 diabetes: A systematic review. Patient Educ Couns. 2016, 99, 944–952. [Google Scholar] [CrossRef]
  178. Powell, P.W.; Hilliard, M.E.; Anderson, B.J. Motivational interviewing to promote adherence behaviors in pediatric type 1 diabetes. Curr Diab Rep. 2014, 14, 531. [Google Scholar] [CrossRef]
  179. Swoboda, C.M.; Miller, C.K.; Wills, C.E. Setting Single or Multiple Goals for Diet and Physical Activity Behaviors Improves Cardiovascular Disease Risk Factors in Adults With Type 2 Diabetes: A Pragmatic Pilot Randomized Trial. Diabetes Educ. 2016, 42, 429–443. [Google Scholar] [CrossRef]
  180. Young, H.M.; Miyamoto, S.; Dharmar, M.; Tang-Feldman, Y. Nurse Coaching and Mobile Health Compared With Usual Care to Improve Diabetes Self-Efficacy for Persons With Type 2 Diabetes: Randomized Controlled Trial. JMIR Mhealth Uhealth. 2020, 8, e16665. [Google Scholar] [CrossRef]
  181. Bilgin, A.; Muz, G.; Yuce, G.E. The effect of motivational interviewing on metabolic control and psychosocial variables in individuals diagnosed with diabetes: Systematic review and meta-analysis. Patient Educ Couns. 2022, 105, 2806–2823. [Google Scholar] [CrossRef]

Share and Cite

MDPI and ACS Style

Socea, B.; Silaghi, A.; Rebegea, L.F.; Balan, D.G.; Balalau, C.; Tenea-Cojan, T.Ș.; Mihai, D.A.; Paunica, I. Diabetes Mellitus: Interdisciplinary Medical, Surgical and Psychological Therapeutic Approach. J. Mind Med. Sci. 2023, 10, 217-236. https://doi.org/10.22543/2392-7674.1445

AMA Style

Socea B, Silaghi A, Rebegea LF, Balan DG, Balalau C, Tenea-Cojan TȘ, Mihai DA, Paunica I. Diabetes Mellitus: Interdisciplinary Medical, Surgical and Psychological Therapeutic Approach. Journal of Mind and Medical Sciences. 2023; 10(2):217-236. https://doi.org/10.22543/2392-7674.1445

Chicago/Turabian Style

Socea, Bogdan, Adrian Silaghi, Laura Florentina Rebegea, Daniela Gabriela Balan, Cristian Balalau, Tiberiu Ștefăniță Tenea-Cojan, Doina Andrada Mihai, and Ioana Paunica. 2023. "Diabetes Mellitus: Interdisciplinary Medical, Surgical and Psychological Therapeutic Approach" Journal of Mind and Medical Sciences 10, no. 2: 217-236. https://doi.org/10.22543/2392-7674.1445

APA Style

Socea, B., Silaghi, A., Rebegea, L. F., Balan, D. G., Balalau, C., Tenea-Cojan, T. Ș., Mihai, D. A., & Paunica, I. (2023). Diabetes Mellitus: Interdisciplinary Medical, Surgical and Psychological Therapeutic Approach. Journal of Mind and Medical Sciences, 10(2), 217-236. https://doi.org/10.22543/2392-7674.1445

Article Metrics

Back to TopTop