Научная статья на тему 'BETA-CELL AUTOPHAGY UNDER THE SCOPE OF HYPOGLYCEMIC DRUGS; POSSIBLE MECHANISM AS A NOVEL THERAPEUTIC TARGET'

BETA-CELL AUTOPHAGY UNDER THE SCOPE OF HYPOGLYCEMIC DRUGS; POSSIBLE MECHANISM AS A NOVEL THERAPEUTIC TARGET Текст научной статьи по специальности «Фундаментальная медицина»

CC BY
67
7
i Надоели баннеры? Вы всегда можете отключить рекламу.
Журнал
Ожирение и метаболизм
Scopus
ВАК
Ключевые слова
HYPOGLYCEMIC DRUGS / β-CELL / PATHOGENESIS / DPP4 & GLP-1 / AUTOPHAGY / AMP-ACTIVATED PROTEIN KINASE PATHWAY & MTOR-1 PATHWAY / SULFONYLUREAS & SGLT2

Аннотация научной статьи по фундаментальной медицине, автор научной работы — Marzoog B.A., Vlasova T.I.

Physiologically, autophagy is a major protective mechanism of β-cells from apoptosis, through can reserve normal β- cell mass and inhibit the progression of β-cells destruction. Beta-cell mass can be affected by differentiation from progenitors and de-differentiation as well as self-renewal and apoptosis. Shred evidence indicated that hypoglycemic drugs can induce β-cell proliferation capacity and neogenesis via autophagy stimulation. However, prolonged use of selective hypoglycemic drugs has induced pancreatitis besides several other factors that contribute to β-cell destruction and apoptosis initiation. Interestingly, some nonhypoglycemic medications possess the same effects on β-cells but depending on the combination of these drugs and the duration of exposure to β-cells. The paper comprehensively illustrates the role of the hypoglycemic drugs on the insulin-producing cells and the pathogeneses of β-cell destruction in type 2 diabetes mellitus, in addition to the regulation mechanisms of β-cells division in norm and pathology. The grasping of the hypoglycemic drug’s role in beta-cell is clinically crucial to evaluate novel therapeutic targets such as new signaling pathways. The present paper addresses a new strategy for diabetes mellitus management via targeting specific autophagy inducer factors (transcription factors, genes, lipid molecules, etc.).

i Надоели баннеры? Вы всегда можете отключить рекламу.
iНе можете найти то, что вам нужно? Попробуйте сервис подбора литературы.
i Надоели баннеры? Вы всегда можете отключить рекламу.

Текст научной работы на тему «BETA-CELL AUTOPHAGY UNDER THE SCOPE OF HYPOGLYCEMIC DRUGS; POSSIBLE MECHANISM AS A NOVEL THERAPEUTIC TARGET»

BETA-CELL AUTOPHAGY UNDER THE SCOPE OF HYPOGLYCEMIC DRUGS; POSSIBLE MECHANISM AS A NOVEL THERAPEUTIC TARGET

© Basheer A. Marzoog*, Tatyana I. Vlasova

Ogarev Mordovia State University, Saransk, Russia

Physiologically, autophagy is a major protective mechanism of p-cells from apoptosis, through can reserve normal p- cell mass and inhibit the progression of p-cells destruction. Beta-cell mass can be affected by differentiation from progenitors and de-differentiation as well as self-renewal and apoptosis. Shred evidence indicated that hypoglycemic drugs can induce p-cell proliferation capacity and neogenesis via autophagy stimulation. However, prolonged use of selective hypoglycemic drugs has induced pancreatitis besides several other factors that contribute to p-cell destruction and apoptosis initiation. Interestingly, some nonhypoglycemic medications possess the same effects on p-cells but depending on the combination of these drugs and the duration of exposure to p-cells. The paper comprehensively illustrates the role of the hypoglycemic drugs on the insulin-producing cells and the pathogeneses of p-cell destruction in type 2 diabetes mellitus, in addition to the regulation mechanisms of p-cells division in norm and pathology. The grasping of the hypoglycemic drug's role in beta-cell is clinically crucial to evaluate novel therapeutic targets such as new signaling pathways. The present paper addresses a new strategy for diabetes mellitus management via targeting specific autophagy inducer factors (transcription factors, genes, lipid molecules, etc.).

KEYWORDS: Hypoglycemic Drugs; ft-Cell; Pathogenesis; DPP4 & GLP-1; Autophagy; AMP-activated protein kinase pathway & mTOR-1 pathway; Sulfonylureas & SGLT2.

INTRODUCTION

Hypoglycemic drugs are extremely effective in relieving and controlling hyperglycemia, but unfortunately, recent data showed serious side effects after their administration [1]. Under normal physiological conditions, the balance between self-renewal and apoptosis maintains the p-cell mass at the familiar level. Whatever perturbance to this balance results in unfavorable outcomes. Diabetes mellitus usually arises on the background of metabolic syndrome that nowadays exponentially increases with the COVID-19 pandemic, especially in the caloric rich intake populations [2, 3]. Shred evidence that dipeptidyl peptidase 4 (DPP4) can be a potential receptor for COVID-19 entrance, but administration of DPP4 inhibitors was not related to decreasing COVID-19 morbidity incidence [4]. Besides, the primary receptor for COVID-19, the angiotensin-converting enzyme receptor, is also expressed on pancreatic beta cells, therefore, COVID-19 has additional potential for beta-cell destruction and ketosis-prone diabetes developing [5]. Three main kinds of autophagy distinguished; macroautophagy, microauophagy, and chaperon mediated autophagy (we use autophagy term to describe macroautophagy in this paper). The paper aimed to analyze data from the present literature datadase (Scopus and Medline) on the effect of problem of the hypoglycemic drug's effect on p-cell and their role in future therapeutic strategies. We searched both databases using keywords: «hypoglycemic drugs», «beta cell», «Metformin, Sulfonylureas», «Thiazoli-dinediones», «Sodium-glucose cotransporter 2 inhibitors», «Autophagy». 54 results in Scopus and 68 results in Medline. Excluding duplicates and not related titles to our paper by title remained 90 papers. Then we read the abstract of these papers, and only 47 papers related to our topic. After that, we read the full text of these 47 papers. The study of the effects of hypoglycemic drugs on p-cells has become an urgent problem in recent decades, as the number of candidates and affected individuals has increased dramatically.

*Автор, ответственный за переписку / Corresponding author.

REGULATION OF B-CELLS DIVISION AND MATURATION

UNDER DIABETES

The p-cells division is strictly controlled by variable factors in norm and pathology. A recent clinical study approved in vitro that p-cells can be differentiated into glucagon-pro-ducing cells through specific regulatory transcription factors [6]. Physiologically, the p-cell has a FOXO1 transcription factor (TF) in addition to the NKX6.1 transcription factor in the nucleus and cytoplasm [7, 8]. The depletion of FOXO1 leads to the inability to retain NKX6.1 in the nucleus and consequently the deprivation of p-cells. At the same time, the p-cells have shown a capacity to transfer into progenitor-like state that have transcription factor Neurogenin3 that can give rise to any type of islets of Langerhans cells of similarity such as a and 5 like cells in human type 2 diabetes [7, 9, 10]. Surprisingly, the study concluded that insulin secretion is inversely correlated with the degree of dedifferentiation, defined as the ratio of Syn-positive/ hormone-negative cells to Syn-positive cells. On the other hand, p-cell differentiation does not depend on the age, weight or duration of diabetes [7]. Scientists believe that p-cells differentiation of -cells in type 2 diabetes is to protect them from apoptosis and immune system attack. Thereafter, when will be favorable metabolic conditions, the differentiated p-cells return into active p-cells. Proinflammatory cytokines such as IL-1/3 and IFN-y were recently been shown to stimulate early phases of autophagy through ER stress-dependent activation of adenosine 5'monophos-phate-activated protein kinase and the formation of reactive oxygen species formation [11]. In addition to inhibiting ^-cell autophagic flux of cells due to impaired lysosomal function, which contributed to ^-cell apoptosis [12]. Contrary, interleukin 22 (IL-22) and IL-6 have a cytoprotective effect on p-cells by stimulating autophagy and protecting p-cells from tumor necrosis factor-a, IL-1/3, and interferon-y induced apoptosis [13, 14].

© Endocrinology Research Centre, 2021_Received: 15.07.21. Accepted: 09.12.21.

REGULATION OF p-CELL FUNCTION

In the few past years, researchers had amplified the role of apoptosis, autophagy, oxidative stress, and nutrient overload in the regulation of insulin-producing cell activity and quantity [15, 16]. Where Autophagy appears to play a significant role in the regulation of insulin homeostasis in addition to its role in ^-cell survival [11, 17]. Moreover, autophagy supports the proper differentiation of p-cells during development and contributing to p-cell function [18, 19]. Studies in vitro have shown that autophagy protects the p-cells from apoptosis induced solely by fatty acids such as palmitate and cholesterol through activation of the Nuclear factor E2-related factor 2 (Nrf2) [20, 21]. The Atg7 gene was found as an important regulator in autophagy deriving while Atg7 depletion results in beta mass decreasing, and glucose tolerance impairment, defective insulin secretion, and increased apoptosis when combined with high-fat and high-glucose diet, indicating Atg7 depletion impairs autophagy stimulation [22]. In contrast, autophagy can be stimulated by specific dietary components, GLP-1, and cytokines [17]. Excess lipid level plays a huge regulatory role in autophagy controlling too [20, 21]. On the other hand, few data suggested that starvation and amino acid deprivation can inhibit autophagy and promote crinophagy via direct fusing of insulin granules with the lysosomes [17, 23]. A single study published in Medicine journal indicated the role of microbiota in the regulation of homeostasis and eventually insulin receptors sensitivity that can be associated with type 2 diabetes mellitus appearance [24]. On the cellular level of regulation, the misbalance between fuel production by the mitochondria and its expenditure can be a positive stimulus to initiate insulin receptor resistance and accordingly type 2 diabetes mellitus [25]. Finally, autophagy plays a key role in driving the pancreatic stem cells (PSC) differentiation, animal models, into insulin-producing cells via an ambiguous mechanism suspected to be through the Wnt/B-catenin signaling pathway [19, 26].

Autophagy maintains beta cell homeostasis through redistribution of energy sources within the cell to preserve the most vital organelles from failure and later degradation [27]. Maintaining the anatomical structure of the beta cell requires autophagy, where the depletion of Atg7 in the beta cells results in impaired transmission activity of LC3-I to LC3-II transmission activity as well as accumulation of p62 and polyubiquitin in the beta cell cytoplasm, which is usually seen in diabetic patients [28]. Moreover, pathoanatomical changes were observed in Atg7 deficient beta cells such as cyst-like formation sized 15-20 mm that were associated with caspase-3-positive apoptotic cells.

PATHOGENESIS OF B-CELLS DYSFUNCTION

Diabetes mellitus can arise under various pathological mechanisms that, in turn, culminate in the diminish of p-cell activity. Therefore, there should be different pathophysiolog-ical pathways for the development of the condition. Many of these mechanisms have been shown to have a genetic predisposition, including; Proopiomelanocortin gene mutation, Melanocortin receptor mutations, brain-derived neurotrophic factor and receptor mutations, glucose kinase mutations, hepatocyte nuclear factor mutations, mitochondrial DNA mutations, Insulin receptor mutations, viral oncogene homolog 2 (AKT2), v-akt murine thymoma and even lipodystrophy

can alter the normal function of p-cell [29-33]. Apoptosis appears to be the leading cause of p-cell destruction in both types of diabetes mellitus through activation of interleukin (IL)-1 p, nuclear factor (NF)-kB, and Fas pathways, through a process, known as insulitis [34]. The p-cells are highly sensitive to destructive pathogenic factors such as environmental regulated by physical activity, diet and intestinal microbiota [32]. High serum free fatty acid (FFA) and hyperglycemia are the primary cause of p-cells dysfunction via ER stress. And this in turn trigger NF-KB-dependent mechanism that culminates in caspase-3 activation for cytokines and an NF-KB-independent mechanism for nutrients "glucose hyper-sensitization" consequently apoptosis of p-cells [8, 34, 35]. In addition to decreasing in glucose-induced insulin secretion by p-cell due to failure of p-cell sensitivity to hyperglycemia and loss of the first phase with a decrease in the second phase of insulin secretion [36]. In most patients who suffer from type 2 diabetes, it is not so crucial the depletion in the mass of p-cells through dyslipidemia and hyperglycemia, via apop-tosis [37, 38]. Several studies stress the neurohormonal role in the pathogenesis of type 2 diabetes mellitus through the dysregulation of leptin hormone that leads to hyperpha-gia and obesity and accordingly insulin resistance [29]. IL-17 showed a piece of shred evidence in the pathophysiology of type 2 diabetes mellitus through enhancement of the inflammatory processes and insulin resistance where administration of IL-17 antagonist decreased the risk of type 2 diabetes mellitus emergence [39]. Recently, a study done by Anne Raimondo and her colleagues demonstrated that Pep-tidylglycine Alpha-amidating Monooxygenase (PAM) contribute to the development of type 2 diabetes mellitus [33].

Beta-cell dysfunction involved autophagy disturbance, where several studies reported that Atg7 knockdown cells suffered from impaired insulin secretion. Impaired autophagy induces the transdifferentiation of beta cells into alpha cells and non functional islet cells as well as impairs proliferation [40].

HYPOGLYCEMIC DRUGS EFFECT ON p-CELL AUTOPHAGY

(EXPERIMENTAL TRIALS)

Many factors contribute to p-cell co-working ability such as hypoglycemic drugs. Therefore, various effects can be revealed on the administration of antihyperglycemic drugs depending on their mechanism of action. The drugs administered most frequently to control hyperglycemia are met-formin and GLP-1 mimetics such as GLP-1 analogs and GLP-1-like molecules, exendin-4/exenatide, and its derivative such as lixisenatide [41]. Although all these drugs have shown a favorable effect on p-cells and reserving their mass and function. The clinical findings have shown that metformin promotes p-cells autophagy and poses an anti-inflammatory effect that enhances even immune response against viral infection when combined with the seasonal influenza vaccine [42-44]. Both groups of GLP-1 mimetics, GLP-1 receptor agonists and DPP-IV inhibitors, have been shown to induce autophagy in diabetic patients and enhance the insulin gene transcription and biosynthesis through the reduction in p-arrestin recruitment and faster agonist dissociation rates [45-47]. Indeed these two groups of medications have various effects on p-cells since they serve differently from each other [41]. The administration of GLP-1 agonists to normal and diabetic rodents has stimulated p-cells proliferation, neogenesis, and

protects against apoptosis and inflammation [48-50]. While GLP-1 agonists long prescribing ended with inducing P-cell mass through the activation of multiple signaling pathways such as PKA, PI3-kinase, and ERK1/2 [51-53]. In particular, the liraglutide and exenatide administration in animal models promoted the first- and second-phase insulin secretion via restoring the beta-cell sensitivity to glucose, besides, the liraglutide and exenatide induced beta cell proliferation/ regeneration and mass of beta cells through the protection from apoptosis [53, 54]. In vivo, few findings have shown that GLP-1 R signaling exerts a vigorous effect on P-cell survival compared with DPP4 inhibition [55, 56]. The prolonged administration of DPP4 inhibitors induced beta cells mass and regeneration capacity in addition to the anti-inflammatory response [57, 58]. The inhibition of the mammalian target of rapamycin complex 1 (mTORC 1) has shown a stimulatory effect on the autophagy of P-cell, an example of such drugs is rapamycin [59, 60]. In many clinical studies, this immunosup-pressant and anti-neoplastic drug has lowered the glucose level and decreased weight gain in addition to insulin resistance [61-65]. However, unfortunately, several studies have shown that Rapamycin has impaired glucose tolerance and elevated insulin resistance even the appearance of frank diabetes in vivo in addition to a decrease in P-cell autophagy and function [62, 66-70].

Sodium-glucose cotransporter 2 inhibitors (SGLT2) have beneficial effects on beta cells by inducing autophagy through the overexpression of adenosine monophosphate-activat-ed protein kinase, sirtuin-1, and/or hypoxia-inducible fac-tors-1a/2a. In vivo, SGLT2 has induced beta-cell proliferation and improved insulin secretion in response to hyperglycemia in addition to ameliorating lipotoxicity [71, 72].

Sulfonylureas (SUs) is a common class of antihyperglycemic drug used in type 2 diabetes mellitus. Glibenclamide, second generation in this group of medications, induces beta cell autophagy by activating the AMPK pathway instead of the mTOR pathway However, the effects of autophagy depend on the state of the beta cells, if it is sensitive to hyperglycemia, thus enhancing autophagy induces insulin secretion. However, in altered beta cell sensitivity to high glucose levels, autophagy induction reduces insulin secretion. Also, it is well known that insulin over secretion induces endoplasmic reticulum stress and oxidative stress. Furthermore, over-induction of autophagy alters insulin storage granules and results in their degradation. studies on Min-6 cells suggested that Glibenclamide induces insulin secretion but its inhibition was accompanied by significant up-regulation of insulin secretion [73].

Thiazolidinediones are another class of antihyperglycemic medication. Rosiglitazone belongs to this class of medications, which is reported to be a well inducer for beta cell autophagy through activation of the AMPK pathway in INS-1 cells. Beta cell autophagy maintains beta cells mass and prevent transdifferentiation (cell linage reprogramming) as well as improves proliferation.

To decrease the speed of P-cell destruction, it's now recommended the early diagnosis of type 2 diabetes mellitus and prefers to not use insulin secretagogues such as sulfonylureas since they impair the P-cell function and leads to their apoptosis [66].

Finally, it seems that prolonged use of incretin-based therapies, particularly sitagliptin, for more than 2.4 years may promote the development of acute pancreatitis, especially when there is a tripling in the level of pancreatic amylase and lipase enzymes level [1, 74]. (Figure 1)

High Glucose

KATP channel

GLUT 2

Nutrients Starvation High fat diet Vitamins Cytokines IL-6 IL-22

MicroRNAs & signaling pathways mR-99a mR-22 miR421 and DRAM1 sirtuin Drugs Intermittent fasting GLP-1 agonists Rapamycin Vitamin D longevinex resveratrol

Increase insulin secretion

Oxidative phosphorylation

Pro-inflammatory cytokines

^Oxidative stress

Endoplasmic Reticulum

Pro-inflammatory cytokines

GLP-1

ß-cell

Figure 1. Autophagy's role in maintaining Beta-cell mass and function through resolving the oxidative stress, endoplasmic reticulum (ER) stress, and

reducing pro-insulin degradation (these functions indicated by asterisks).

Note. GLP-1 group of anti-diabetic medications in addition to other factors can affect autophagy. Oxidative stress and endoplasmic reticulum stress are critical for maintaining autophagy and beta cell mass from apoptosis and cell death, which are necessary to maintain blood glucose levels. Abbreviations: ATP — adenosine triphosphate, IL — interleukin, GLP-1 — glucagon-like peptide-1, ROS — reactive oxygen species, hIAPP — human islet amyloid

polypeptide.

CONCLUSION

Therefore, the observed data have shown the possibility of p-cell mass maintaining in diabetic patients by promoting autophagy promotion via selective hypoglycemic drugs, including DDP-IV inhibitors and GLP-1 mimetics. Several preclinical trials have been performed in the course of stimulating p-cell proliferation by using hypoglycemic drugs. Most were promising and culminated in the possibility of p-cell to maintain self-renewal and protect themselves from transdifferentiation into pathological nonbeta cells. But, this process indirectly occurs, firstly hypoglycemic drugs induce autophagy, then autophagy promoted beta-cell proliferation, therefore there is no direct in vitro or in vivo study shown this, and we in this paper connected the missing parts to complete the chain [7, 17, 19, 42, 43, 73, 75-77].

In preclinical studies, the majority of hypoglycemic drugs possessed therapeutic effects on the beta cell during their administration including; enhance autophagy and insulin biosynthesis, possessing anti-inflammatory effects, protecting against apoptosis and beta cell destruction, as well as enhance p-cell survival. Beta-cell mass can be affected by differentiation from progenitors and de-differentiation as well as self-renewal and apoptosis.

Interestingly, recent investigated data have emphasized the role of transdifferentiation in beta cell regeneration by recruiting endoderm-derived cells by applying specific

niche factors, including transcription factors [78-80]. But, the following question is still ambiguous and needs more research and clinical study; Do neo transdifferentiated beta cells persist or terminate with stop of the administration of the inducing factor administration? While this can be of use in triggering other cells of islets of Langerhans to transfer into p-cells and start producing insulin. This requires more research and clinical studies to prove the clinical importance of the hypoglycemic drugs on p-cells metabolism. We suggest inducing beta cells autophagy in diabetic patients through targeting autophagy signaling pathways ameliorates hyperglycemia. Hypoglycemic drags are well inducers for beta-cells autophagy and can be recruited and modified to be suitable for this purpose.

ADDITIONAL INFORMATION

Funding. No funding.

Conflict of interest. The authors declare no obvious and potential conflicts of interest related to the content of this article.

Contribution of authors. Basheer Abdullah Marzoog — design of the work and the acquisition, analysis, interpretation of data for the work, Drafting the work and revising it critically for important intellectual content. Tatyana Ivanovna Vlasova — revising the work critically for important intellectual content. All of the authors read and approved the final version of the manuscript before publication, agreed to be responsible for all aspects of the work, implying proper examination and resolution of issues relating to the accuracy or integrity of any part of the work.

СПИСОК ЛИТЕРАТУРЫ | REFERENCES

10.

DeVries JH, Rosenstock J. DPP-4 Inhibitor-Related 12.

Pancreatitis: Rare but Real! Diabetes Care. 2017;40:161-163.

doi: https://doi.org/10.2337/dci16-0035

Sada K, Nishikawa T, Kukidome D, et al. Hyperglycemia induces

cellular hypoxia through production of mitochondrial ROS 13.

followed by suppression of aquaporin-1. PLoSOne. 2016;11.

doi: https://doi.org/10.1371/journal.pone.0158619

Marzoog B. Lipid behavior in metabolic syndrome

pathophysiology. Curr Diabetes Rev. 2021;17. 14.

doi: https://doi.org/10.2174/1573399817666210915101321

Lim S, Bae JH, Kwon H-S, Nauck MA. COVID-19 and diabetes mellitus:

from pathophysiology to clinical management. Nat Rev Endocrinol. 15.

2021;17:11-30. doi: https://doi.org/10.1038/s41574-020-00435-4

Rubino F, Amiel SA, Zimmet P, et al. New-Onset

Diabetes in Covid-19. N Engl J Med. 2020;383(8):789-790.

doi: https://doi.org/10.1056/NEJMc2018688 16.

Brereton MF, Iberl M, Shimomura K, et al. Reversible

changes in pancreatic islet structure and function produced

by elevated blood glucose. Nat Commun. 2014;5:4639. 17.

doi: https://doi.org/10.1038/ncomms5639

Cinti F, Bouchi R, Kim-Muller JY, et al. Evidence of ß-Cell

Dedifferentiation in Human Type 2 Diabetes. J Clin Endocrinol Metab. 18.

2016;101:1044-1054. doi: https://doi.org/10.1210/jc.2015-2860

Cheng STW, Li SYT, Leung PS. Fibroblast Growth Factor 21 Stimulates

Pancreatic Islet Autophagy via Inhibition of AMPK-mTOR Signaling. 19.

Int J Mol Sci. 2019;20:2517. doi: https://doi.org/10.3390/ijms20102517.

Talchai C, Xuan S, Lin HV, et al. Pancreatic ß cell dedifferentiation

as a mechanism of diabetic ß cell failure. Cell. 2012;150:1223-1234.

doi: https://doi.org/10.1016/j.cell.2012.07.029. 20.

Bensellam M, Jonas JC, Laybutt DR. Mechanisms

of ß-cell dedifferentiation in diabetes: Recent findings and

future research directions. J Endocrinol. 2018;236:109-143. 21.

doi: https://doi.org/10.1530/JOE-17-0516.

DiNicolantonio JJ, McCarty M. Autophagy-induced degradation

of Notch1, achieved through intermittent fasting, may promote beta 22.

cell neogenesis: implications for reversal of type 2 diabetes. Open Hear.

2019;6(1):e001028. doi: https://doi.org/10.1136/openhrt-2019-001028

Lambelet M, Terra LF, Fukaya M, et al. Dysfunctional autophagy

following exposure to pro-inflammatory cytokines contributes

to pancreatic ß-cell apoptosis. Cell Death Dis. 2018;9:96.

doi: https://doi.org/10.1038/s41419-017-0121-5

Hu M, Yang S, Yang L, et al. Interleukin-22 Alleviated Palmitate-

Induced Endoplasmic Reticulum Stress in INS-1 Cells through

Activation of Autophagy. PLoS One. 2016;11(1):e0146818.

doi: https://doi.org/10.1371/journal.pone.0146818

iНе можете найти то, что вам нужно? Попробуйте сервис подбора литературы.

Linnemann AK, Blumer J, Marasco MR, et al. Interleukin 6 protects

pancreatic b cells from apoptosis by stimulation of autophagy. FASEB

J. 2017;31:4140-4152. doi: https://doi.org/10.1096/fj.201700061RR

Butler PC, Meier JJ, Butler AE, Bhushan A.

The replication of ß cells in normal physiology, in disease and

for therapy. Nat Clin Pract Endocrinol Metab. 2007;3:758-768.

doi: https://doi.org/10.1038/ncpendmet0647

Talchai C, Lin HV, Kitamura T, Accili D. Genetic and biochemical

pathways of ß-cell failure in type 2 diabetes. Diabetes, ObesMetab.

2009;11:38-45. doi: https://doi.org/10.1111/j.1463-1326.2009.01115.x

Marasco MR, Linnemann AK. B-Cell autophagy

in diabetes pathogenesis. Endocrinology. 2018;159:2127-2141.

doi: https://doi.org/10.1210/en.2017-03273

Riahi Y, Wikstrom JD, Bachar-Wikstrom E, et al. Autophagy is

a major regulator of beta cell insulin homeostasis. Diabetologia.

2016;59:1480-1491. doi: https://doi.org/10.1007/s00125-016-3868-9

Ren L, Yang H, Cui Y, et al. Autophagy is essential for

the differentiation of porcine PSCs into insulin-producing

cells. Biochem Biophys Res Commun. 2017;488:471-476.

doi: https://doi.org/10.1016/j.bbrc.2017.05.058

Choi SE, Lee SM, Lee YJ, et al. Protective role of autophagy

in palmitate-induced INS-1 ß-cell death. Endocrinology.

2009;150:126-134. doi: https://doi.org/10.1210/en.2008-0483

Wu J, Kong F, Pan Q, et al. Autophagy protects against cholesterol-

induced apoptosis in pancreatic ß-cells. Biochem Biophys Res Commun.

2017;482:678-685. doi: https://doi.org/10.1016/j.bbrc.2016.11.093

Sheng Q, Xiao X, Prasadan K, et al. Autophagy protects pancreatic beta cell

mass and function in the setting of a high-fat and high-glucose diet. Sci

Rep. 2017;7(1):16348. doi: https://doi.org/10.1038/s41598-017-16485-0

23. Goginashvili A, Zhang Z, Erbs E, et al. Insulin secretory granules control autophagy in Pancreatic ß cells. Science (80-). 2015;347:878-882. doi: https://doi.org/10.1126/science.aaa2628.

24. Li C, Li X, Han H, et al. Effect of probiotics on metabolic profiles

in type 2 diabetes mellitus. Medicine (Baltimore). 2016;95(26):e4088. doi: https://doi.org/10.1097/MD.0000000000004088

25. Patti M-E, Corvera S. The Role of Mitochondria in the Pathogenesis of Type 2 Diabetes. Endocr Rev. 2010;31(3):364-395.

doi: https://doi.org/10.1210/er.2009-0027

26. Xu S, Sun F, Ren L, Yang H, Tian N, Peng S. Resveratrol controlled the fate of porcine pancreatic stem cells through the Wnt/ß-catenin signaling pathway mediated by Sirt1. PLoS One. 2017;12(10):e0187159. doi: https://doi.org/10.1371/journal.pone.0187159

27. Kuma A, Hatano M, Matsui M, et al. The role of autophagy during the early neonatal starvation period. Nature. 2004;432(7020):1032-1036. doi: https://doi.org/10.1038/nature03029

28. Mizukami H, Takahashi K, Inaba W, et al. Involvement of oxidative stress-induced DNA damage, endoplasmic reticulum stress, and autophagy deficits in the decline of ß-cell mass in Japanese type 2 diabetic patients. Diabetes Care. 2014;37:1966-1974.

doi: https://doi.org/10.2337/DC13-2018.

29. Murphy R, Carroll RW, Krebs JD. Pathogenesis of the metabolic syndrome: insights from monogenic disorders. Mediators Inflamm. 2013;2013:920214. doi: https://doi.org/10.1155/2013/920214.

30. Nica AC, Ongen H, Irminger J-C, et al. Cell-type, allelic, and genetic signatures in the human pancreatic beta cell transcriptome. Genome Res. 2013;23:1554-1562. doi: https://doi.org/10.1101/gr.150706.112

31. Brunetti A, Chiefari E, Foti D. Perspectives on the contribution

of genetics to the pathogenesis of type 2 diabetes mellitus. Recenti Prog Med. 2011;102:468-475. doi: https://doi.org/10.1701/998.10858.

32. Kalin MF, Goncalves M, John-Kalarickal J, Fonseca V. Pathogenesis of type 2 diabetes mellitus. Princ. DiabetesMellit. 2017.

doi: https://doi.org/10.1007/978-3-319-18741-9_13

33. Raimondo A, Thomsen SK, Hastoy B, et al. Type 2 Diabetes Risk Alleles Reveal a Role for Peptidylglycine Alpha-amidating Monooxygenase in Beta Cell Function. bioRxiv. 2017.

doi: https://doi.org/10.1101/158642

34. Cnop M, Welsh N, Jonas JC, et al. Mechanisms

of pancreatic ß-cell death in type 1 and type 2 diabetes: Many differences, few similarities. Diabetes. 2005;54:S97-107. doi: https://doi.org/10.2337/diabetes.54.suppl_2.S97.

35. Ozougwu O. The pathogenesis and pathophysiology of type 1 and type 2 diabetes mellitus. J Physiol Pathophysiol. 2013;4:46-57. doi: https://doi.org/10.5897/JPAP2013.0001

36. Ashcroft FM, Rorsman P. Molecular defects in insulin secretion in type-2 diabetes. Rev Endocr Metab Disord. 2004;5:135-142. doi: https://doi.org/10.1023/B:REMD.0000021435.87776.a7.

37. Butler AE, Janson J, Bonner-Weir S, et al. ß-cell deficit and increased ß-cell apoptosis in humans with type 2 diabetes. Diabetes. 2003;52:102-110. doi: https://doi.org/10.2337/diabetes.52.1.102.

38. Kahn SE. The relative contributions of insulin resistance and beta-cell dysfunction to the pathophysiology of Type 2 diabetes. Diabetologia. 2003;46:3-19. doi: https://doi.org/10.1007/s00125-002-1009-0

39. Abdel-Moneim A, Bakery HH, Allam G. The potential pathogenic role of IL-17/Th17 cells in both type 1 and type

2 diabetes mellitus. BiomedPharmacother. 2018;101:287-292. doi: https://doi.org/10.1016/j.biopha.2018.02.103

40. Marzoog BA, Vlasova TI. Transcription Factors in Deriving ß Cell Regeneration; A Potential Novel Therapeutic Target. Curr Mol Med. 2021;21. doi: https://doi.org/10.2174/1566524021666210712144638

41. Dalle S, Burcelin R, Gourdy P. Specific actions of GLP-1 receptor agonists and DPP4 inhibitors for the treatment of pancreatic ß-cell impairments in type 2 diabetes. Cell Signal. 2013;25:570-579. doi: https://doi.org/10.1016/j.cellsig.2012.11.009

42. Jiang Y, Huang W, Wang J, et al. Metformin Plays a Dual Role in MIN6 Pancreatic ß Cell Function through AMPK-dependent Autophagy. Int J Biol Sci. 2014;10:268-277. doi: https://doi.org/10.7150/ijbs.7929

43. Wu J, Wu JJ, Yang LJ, et al. Rosiglitazone protects against palmitate-induced pancreatic beta-cell death by activation of autophagy

via 5'-AMP-activated protein kinase modulation. Endocrine. 2013;44:87-98. doi: https://doi.org/10.1007/s12020-012-9826-5

44. Diaz A, Romero M, Vazquez T, et al. Metformin improves in vivo and in vitro B cell function in individuals with

obesity and Type-2 Diabetes. Vaccine. 2017;35:2694-2700. doi: https://doi.Org/10.1016/j.vaccine.2017.03.078

45. Janzen KM, Steuber TD, Nisly SA. GLP-1 Agonists

in Type 1 Diabetes Mellitus. Ann Pharmacother. 2016;50:656-665. doi: https://doi.org/10.1177/1060028016651279

46. Wajchenberg BL. ß-Cell Failure in Diabetes and Preservation by Clinical Treatment. Endocr Rev. 2007;28(2):187-218.

doi: https://doi.org/10.1210/10.1210/er.2006-0038

47. Jones B, Buenaventura T, Kanda N, et al. Targeting GLP-1 receptor trafficking to improve agonist efficacy. Nat Commun. 2018;9(1):1602. doi: https://doi.org/10.1038/s41467-018-03941-2

48. Donnelly D. The structure and function of the glucagon-like peptide-1 receptor and its ligands. Br J Pharmacol. 2012;166:27-41. doi: https://doi.org/10.1111/j.1476-5381.2011.01687.x

49. Shyangdan DS, Royle P, Clar C, et al. Glucagon-like peptide analogues for type 2 diabetes mellitus. Cochrane Database Syst Rev. 2011.

doi: https://doi.org/10.1002/14651858.CD006423.pub2

50. Piya MK, Tahrani AA, Barnett AH. Emerging treatment options for type 2 diabetes. Br J Clin Pharmacol. 2010;70(5):631-644. doi: https://doi.org/10.1111/j.1365-2125.2010.03711.x

51. Doyle ME, Egan JM. Mechanisms of action of glucagon-like peptide 1 in the pancreas. Pharmacol Ther. 2007;113:546-593. doi: https://doi.org/10.1016/j.pharmthera.2006.11.007

52. Lee Y-S, Jun H-S. Anti-Inflammatory Effects of GLP-1-Based Therapies beyond Glucose Control. Mediators Inflamm. 2016;2016:1-11.

doi: https://doi.org/10.1155/2016/3094642.

53. Lee Y-S, Jun H-S. Anti-diabetic actions of glucagon-like peptide-1 on pancreatic beta-cells. Metabolism. 2014;63(1):9-19. doi: https://doi.org/10.1016/j.metabol.2013.09.010

54. Vilsboll T. The effects of glucagon-like peptide-1

on the beta cell. Diabetes, Obes Metab. 2009;11:11-18. doi: https://doi.org/10.1111/j.1463-1326.2009.01073.x

55. Pratley RE, Nauck M, Bailey T, et al. Liraglutide versus sitagliptin for patients with type 2 diabetes who did not have adequate glycaemic control with metformin: a 26-week, randomised, parallel-group, open-label trial. Lancet. 2010;375:1447-1456. doi: https://doi.org/10.1016/S0140-6736(10)60307-8.

56. Bergenstal RM, Wysham C, MacConell L, et al. Efficacy and safety of exenatide once weekly versus sitagliptin or pioglitazone

as an adjunct to metformin for treatment of type 2 diabetes (DURATION-2): A randomised trial. Lancet. 2010;376:431-439. doi: https://doi.org/10.1016/S0140-6736(10)60590-9.

57. Omar BA, Vikman J, Winzell MS, et al. Enhanced beta cell function and anti-inflammatory effect after chronic treatment with the dipeptidyl peptidase-4 inhibitor vildagliptin in an advanced-aged diet-induced obesity mouse model. Diabetologia. 2013;56(8):1752-1760. doi: https://doi.org/10.1007/s00125-013-2927-8

58. 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(6):473-479. doi: https://doi.org/10.1016/jjcjd.2014.01.008

59. Tanemura M, Ohmura Y, Deguchi T, et al. Rapamycin causes upregulation of autophagy and impairs islets function both in vitro and in vivo. Am J Transplant. 2012;12:102-114. doi: https://doi.org/10.1111/j.1600-6143.2011.03771.x

60. Zhou Z, Wu S, Li X, et al. Rapamycin induces autophagy

and exacerbates metabolism associated complications in a mouse model of type 1 diabetes. Indian J Exp Biol. 2010;48:31-38.

61. Chang G-R, Wu Y-Y, Chiu Y-S, et al. Long-term administration

of rapamycin reduces adiposity, but impairs glucose tolerance in high-fat diet-fed KK/HlJ mice. Basic Clin Pharmacol Toxicol. 2009;105:188-198. doi: https://doi.org/10.1111/j.1742-7843.2009.00427.x

62. Chang G-R, Chiu Y-S, Wu Y-Y, et al. Rapamycin protects against high fat diet-induced obesity in C57BL/6J mice. J Pharmacol Sci. 2009;109:496-503. doi: https://doi.org/10.1254/jphs.08215fp.

63. Gong F-H, Ye Y-N, Li J-M, et al. Rapamycin-ameliorated diabetic symptoms involved in increasing adiponectin expression in diabetic mice on a high-fat diet. Kaohsiung J Med Sci. 2017;33:321-326.

doi: https://doi.org/10.1016/j.kjms.2017.05.008

64. Reifsnyder PC, Flurkey K, Te A, Harrison DE. Rapamycin treatment benefits glucose metabolism in mouse models of type 2 diabetes. Aging (Albany NY). 2016;8:3120-3130. doi: https://doi.org/10.18632/aging.101117

65. Fang Y, Westbrook R, Hill C, et al. Duration of rapamycin treatment has differential effects on metabolism in mice. Cell Metab. 2013;17:456-462. doi: https://doi.org/10.1016/j.cmet.2013.02.008

66.

67.

69.

70.

71.

72.

73.

Lupi R, Del Prato S. Beta-cell apoptosis in type 2 diabetes: quantitative

and functional consequences. Diabetes Metab. 2008;34(S2):56-64.

iНе можете найти то, что вам нужно? Попробуйте сервис подбора литературы.

doi: https://doi.org/10.1016/S1262-3636(08)73396-2

Barlow AD, Nicholson ML, Herbert TP. Evidence for Rapamycin

Toxicity in Pancreatic ß-Cells and a Review of the Underlying

Molecular Mechanisms. Diabetes. 2013;62:2674-2682.

doi: https://doi.org/10.2337/db13-0106

Schindler CE, Partap U, Patchen BK, Swoap SJ. Chronic

rapamycin treatment causes diabetes in male mice.

Am J Physiol Regul Integr Comp Physiol. 2014;307:R434-43.

doi: https://doi.org/10.1152/ajpregu.00123.2014.

Lamming DW, Ye L, Astle CM, et al. Young and old genetically

heterogeneous HET3 mice on a rapamycin diet are glucose

intolerant but insulin sensitive. Aging Cell. 2013;12:712-718.

doi: https://doi.org/10.1111/acel.12097

Lamming DW, Ye L, Katajisto P, et al. Rapamycin-Induced

Insulin Resistance Is Mediated by mTORC2 Loss and

Uncoupled from Longevity. Science (80-). 2012;335:1638-1643.

doi: https://doi.org/10.1126/science.1215135

Okauchi S, Shimoda M, Obata A, et al. Protective effects of SGLT2

inhibitor luseogliflozin on pancreatic ß-cells in obese type 2 diabetic

db/db mice. Biochem Biophys Res Commun. 2016;470:772-782.

doi: https://doi.org/10.1016ZJ.BBRC.2015.10.109

Lalloyer F, Vandewalle B, Percevault F, et al. Peroxisome proliferator-

activated receptor a improves pancreatic adaptation to insulin

resistance in obese mice and reduces lipotoxicity in human islets.

Diabetes. 2006;55:1605-1613. doi: https://doi.org/10.2337/DB06-0016

Zhou J, Kang X, Luo Y, et al. Glibenclamide-Induced

Autophagy Inhibits Its Insulin Secretion-Improving

74.

75.

76.

77.

78.

79.

Function in ß Cells. Int J Endocrinol. 2019;2019:1-8.

doi: https://doi.org/10.1155/2019/1265175.

Ganesan K, Rana MBM, Sultan S. Oral Hypoglycemic Medications.

StatPearls Publishing; 2020.

Bugliani M, Mossuto S, Grano F, et al. Modulation of Autophagy

Influences the Function and Survival of Human Pancreatic

Beta Cells Under Endoplasmic Reticulum Stress Conditions

and in Type 2 Diabetes. Front Endocrinol (Lausanne). 2019;10.

doi: https://doi.org/10.3389/fendo.2019.00052

Chen Z, Li Y-B, Han J, et al. The double-edged effect of autophagy

in pancreatic beta cells and diabetes. Autophagy. 2011;7:12-16.

doi: https://doi.org/10.4161/auto.7.1.13607

Capozzi ME, DiMarchi RD, Tschöp MH, et al. Targeting

the Incretin/Glucagon System With Triagonists to

Treat Diabetes. Endocr Rev. 2018;39(5):719-738.

doi: https://doi.org/10.1210/er.2018-00117

Churchill AJ, Gutiérrez GD, Singer RA, et al. Genetic evidence

that Nkx2.2 acts primarily downstream of Neurog3

in pancreatic endocrine lineage development. Elife. 2017;6.

doi: https://doi.org/10.7554/eLife.20010

Zhu Y, Liu Q, Zhou Z, Ikeda Y. PDX1, Neurogenin-3, and MAFA:

critical transcription regulators for beta cell development

and regeneration. Stem Cell Res Ther. 2017;8(1):240.

doi: https://doi.org/10.1186/s13287-017-0694-z

Donelan W, Li S, Wang H, et al. Pancreatic and duodenal

homeobox gene 1 (Pdx1) down-regulates hepatic transcription

factor 1 alpha (hnf1a) expression during reprogramming

of human hepatic cells into insulin-producing cells. Am J Transl

Res. 2015;7(6):995-1008.

AUTHORS INFO:

*Basheer Abdullah Marzoog, undergraduate student [address: 68 Bolshevitskaya str., 430005 Saransk, Russia]; ORCID: https://orcid.org/0000-0001-5507-2413; Researcher ID: AAD-6284-2021; e-mail: marzug@mail.ru

Tatyana Ivanovna Vlasova, MD, PhD, professor; ORCID: http://orcid.org/0000-0002-2624-6450; eLibrary SPIN: 5314-3771; e-mail: v.t.i@bk.ru

Corresponding author.

TO CITE THIS ARTICLE:

Marzoog BA, Vlasova TI. Beta-cell autophagy under the scope of hypoglycemic drugs; possible mechanism

as a novel therapeutic target // Ожирение и метаболизм. doi: https://doi.org/10.14341/omet12778

2021.

Т. 18.

№4.

C. 465-470.

i Надоели баннеры? Вы всегда можете отключить рекламу.