• Open Access
    Review

    Autophagy and diabetes

    Milan Obradovic 1
    Sonja Zafirovic 1
    Zoran Gluvic 2
    Jelena Radovanovic 1
    Esma R. Isenovic 1*

    Explor Med. 2023;4:576–588 DOI: https://doi.org/10.37349/emed.2023.00162

    Received: May 04, 2023 Accepted: May 29, 2023 Published: August 31, 2023

    Academic Editor: Jun Ren, Fudan University Zhongshan Hospital, China

    This article belongs to the special issue Genetic and Epigenetic Control of Autophagy in Human Diseases

    Abstract

    The current literature findings on autophagy’s beneficial and detrimental roles in diabetes mellitus (DM) and diabetes-related comorbidities were reviewed. The effects of oral hypoglycaemic medicines and autophagy in DM. Autophagy plays an important function in cellular homeostasis by promoting cell survival or initiating cell death in physiological settings was also assessed. Although autophagy protects insulin-target tissues, organelle failure caused by autophagy malfunction influences DM and other metabolic diseases. Endoplasmic reticulum and oxidative stress enhance autophagy levels, making it easier to regulate stress-induced intracellular changes. Evidence suggests that autophagy-caused cell death can occur when autophagy is overstimulated and constitutively activated, which might prevent or develop DM. Even though the precise role of autophagy in DM complications is uncertain, deregulation of the autophagic machinery is strongly linked to beta cell destruction and the aetiology of DM. Thus, improving autophagy dysfunction is a possible therapeutic objective in treating DM and other metabolic disorders.

    Keywords

    Diabetes, autophagy, anti-hyperglycemic, oxidative stress

    Introduction

    Diabetes mellitus (DM) is an endocrine disease caused by defects in insulin secretion or actions and characterized by hyperglycaemia [1, 2]. The pandemic proportions of DM, with over 425 million people estimated to have DM worldwide, threaten human health [3, 4]. Furthermore, according to the World Health Organization, the prevalence of DM is in a constant upward trend, especially worrying that DM is in the range of the most common causes of death [5]. Apart from genetic predisposition, the high-risk factors for DM development are an unhealthy diet, a sedentary lifestyle, obesity, and other bad habits [6, 7]. Cardiovascular complications arose due to DM-caused cell and tissue damage and dysfunction of various organs [810]. Although all types of DM are treatable and DM pathology research has come a long way, the exact mechanism of pathological processes that trigger the onset of DM and further development of DM-related complications still needs to be clarified. Evidence suggests that autophagy may be a critical regulatory signaling pathway in DM development and prevention [11, 12].

    Autophagy plays a crucial role in cellular homeostasis in physiological conditions by promoting cell survival or initiating cell death [13]. Autophagy is a housekeeping catabolic process that enables the removal of excess or damaged cellular components and organelles and regulates normal cell function, including pancreatic beta cells [14, 15]. In addition, autophagy protects cells in different stress conditions, including ischemia and hypoxia, oxidative stress (OxS), hyperglycemia, hyperlipidemia, and other harmful factors [16, 17]. However, dysregulation of autophagic machinery is tightly linked with beta cell injury and the pathophysiology of DM [12]. In addition, evidence shows that autophagy restoration allows a protective effect, but over-activation of basal autophagy may cause cell death [18]. Thus, the particular role of autophagy in diabetic complications still needs to be elucidated.

    This review discusses recent literature on the role of both the protective and detrimental effects of autophagy in DM and DM-related complications.

    Searching strategy

    From 1978 to April 2023, the MEDLINE and PubMed databases for all English and non-English articles containing an English abstract were searched. All of the publications found, and all of the abstracts from national and international cardiovascular symposia into one thorough evaluation were combined. Search terms were DM, autophagy, autophagy and endoplasmic reticulum stress (ERS), autophagy and OxS, DM and autophagy, autophagy and anti-hyperglycemic.

    DM

    DM is one of the most common endocrinological diseases, with a constantly rising prevalence. Besides, chronic high blood glucose affects fat, protein, and carbohydrate metabolism due to inadequate insulin secretion, insulin effects, or both [19]. DM can have long-term consequences on a person’s health, leading to damage to numerous organs, among which the eyes, kidneys, heart, and blood vessels are most often affected [9, 20]. DM is a consequence of the modern lifestyle and the increase in other factors, among which obesity stands out [21, 22].

    There are several main types of DM: type 1 DM (T1DM), type 2 DM (T2DM), gestational DM (GDM), and specific types of DM caused by different factors [19]. Several pathological processes have been shown to influence the development of DM thus far [23].

    T1DM occurs in fewer people diagnosed with DM, usually 5–10% of all DM cases. T1DM is also known as insulin-dependent DM or juvenile DM. This ailment is caused by T cell-mediated autoimmune disease, which is defined by the destruction of pancreatic cells, the absence of insulin, and hyperglycemia [20, 24]. The destruction of beta cells of the pancreas in the younger population is usually fast and can lead to ketoacidosis, sometimes the initial presentation of the disease. In others, the function of the beta cells of the pancreas can be preserved enough to prevent the occurrence of ketoacidosis [23, 25].

    T2DM is prevalent in most people diagnosed with DM, accounting for 90–95% of all DM cases. T2DM is the most common metabolic disorder. This type of DM is often called insulin-independent DM and affects people who have insulin resistance (IR) or insulin deficiency [19]. With this type of DM, where the peripheral cells resist endogenous insulin, insulin administration is unnecessary to survive [24]. Most patients with T2DM are obese, which affects the development of IR, or have a distinct abdominal type of obesity. Hyperglycemia can develop for years, and the disease remains unnoticed until severe symptoms that indicate the presence of DM develop [26, 27]. Both macro and microvascular complications often occur in patients with T2DM [28]. The risk of T2DM increases with age, obesity, and reduced physical activity [27].

    In addition to these two primary types of DM, it is also important to mention GDM, which occurs in women during pregnancy and is characterized by hyperglycemia. GDM occurs due to insufficient secretion of insulin to compensate for metabolic stress caused by IR. Risk factors for developing GDM are obesity, history of previous GDM, older women, polycystic ovary syndrome, and others. Hyperglycemia often continues even after childbirth, indicating the presence of T2DM [29].

    Specific types of DM include genetic disorders of pancreatic beta cells, genetic disorders of insulin action, diseases of the exocrine pancreas, neonatal DM, and DM caused by various drugs and chemicals, viruses, and others [23].

    It is also worth noting the possible link between Alzheimer’s disease (AD) and DM since AD has been dubbed “type 3 DM” [30]. The exact form of the link is quite complex and needs to be fully known; however, certain essential aspects connect AD with DM, particularly T2DM. DM is linked to an increased incidence of dementia and cognitive decline, both of which are symptoms of AD [31, 32]. DM management and a healthy lifestyle may have potential benefits in lowering the risk of cognitive decline and dementia [33]. Age, obesity, high blood pressure, high cholesterol levels, and a sedentary lifestyle are all risk factors for these diseases. Insulin plays an important role in the brain, and changes in insulin signaling or decreased insulin sensitivity may impact brain function and contribute to the pathology of AD [34]. DM is well-known for its adverse effects on blood vessels, resulting in vascular damage and decreased blood flow to the brain [32, 35]. Vascular issues are also linked to an increased risk of dementia and the development of AD [36, 37].

    Autophagy

    Autophagy represents a mechanism that is involved in cellular homeostasis and defence. It is a catabolic process that degrades and recycles intracellular elements. Hence, autophagy is responsible for pathogen clearance, cellular recycling, and preventing cell toxicity due to the accumulation of damaged organelles and proteins [3840]. Besides, autophagy enables energy production since it disassembles lipids into free fatty acids that could be oxidized via mitochondria [41]. The autophagy process is linked with apoptosis and, therefore, participates in the cell cycle regulation and maintains genomic stability [42, 43]. Autophagy is classified into three categories based on how substances are transported to the lysosome: macroautophagy, microautophagy, and chaperon-mediated autophagy (Figure 1). Microautophagy and macroautophagy can be selective or non-selective (as shown in yeast and advanced organisms), although chaperon-mediated autophagy is exclusively found in mammalian systems [44]. The macroautophagy process implies a double-membrane autophagosome that surrounds the components from the cytosol, which further fuses with the lysosome. All the contents from the autophagosome are revealed to the lysosome (autolysosome) for recycling and deterioration. Microautophagy has different molecular mechanisms, and it is categorized into three forms: type 1 includes lysosomal (vacuolar) membrane protrusion, type 2 involves lysosomal (vacuolar) invagination, as well as type 3 represents the invagination of the endosomal membrane [38, 44, 45]. During chaperon-mediated autophagy, targeted proteins from the cytosol are degraded, not organelles, and there is no configuration of autophagosome and autolysosome. The protein complex is transported into the lysosome via heat shock cognate 70 kDa protein (HSC70) and lysosome-associated membrane protein type 2A (LAMP-2A). The proteins for degradation have pentapeptide KFERQ-like motif, which HSC70 recognizes, and together, they form a compound. Finally, the present complex cooperates with the cytoplasmatic tail of LAMP-2A, and the proteins for degradation are transported into the lysosome [38]. Of these three types of autophagy, macroautophagy is the most examined.

    Presentation of autophagy general molecular mechanisms with three main forms. (A) Macroautophagy; (B) microautophagy; (C) chaperon-mediated autophagy. AMPK: adenosine monophosphate-activated kinase; ATG: autophagy-related genes; Lys: lysosome; mTOR: mammalian target of rapamycin; PI3K: phosphatidylinositol 3-kinase; ULK1: unc-51-like autophagy activating kinase 1. Created via BioRender.com

    According to nutritional status, there is non-selective and selective autophagy. Non-selective autophagy implies stress and nutrient malnourishment, while selective autophagy of organelles, proteins, or pathogens occurs in nutrient-rich circumstances [38, 46, 47]. Selective autophagy preserves cell homeostasis and prevents the incidence of some diseases by preserving the number of organelles—mitochondria (mitophagy), peroxisomes (pexophagy), proteins (aggrephagy), lysosomes (lysophagy), and ribosomes (ribophagy) [46, 48, 49].

    The main cellular signaling pathway responsible for physiological autophagy is PI3K/protein kinase B (AKT)/mTOR [50]. ATG are the most important regulators of autophagy molecular processes and signaling pathways [49, 51]. The macroautophagy process has five stages: initiation, nucleation, elongation with autophagosome production, fusion, and destruction [52, 53]. In the initiation stage, the ULK1 complex and the class III PI3K complex 1 are included. ULK1 is regulated by the mTOR complex 1 (mTORC1) and AMPK, and under starvation circumstances, in the initiation phase, mTORC1 is deactivated, resulting in the repression of ULK1 complex [53, 54]. After the initiation phase, the membrane expands, named a phagophore [55, 56]. Nucleation of the phagophore, i.e. nucleation phase, is followed by ULK1 complex phosphorylation of the PI3K complex, which stimulates phagophore formation [56]. The PI3K complex is regulated by proteins that interrelate Beclin 1 [38]. Elongation of the phagophore is supported by two ubiquitin-like protein complexes-ATG12-ATG5-ATG16L1 and ATG4B-ATG7-ATG3, and they mediate the activation of microtubule-associated protein 1A/1B-light chain 3 (LC3) into LC3I, lipidation with phosphatidylethanolamine (PE) to form LC3-PE conjugate (LC3II) [53, 57, 58]. Phagophore enlarges and eventually creates autophagosome whose dimension differs among organism types [59, 60]. The cytoplasmatic microtubular network is important in autophagosome fusion with the lysosome/vacuole, especially syntaxin 17 (STX17), vacuolar morphogenesis protein 7 and 9, and vesicle-associated membrane protein 8 [53, 61]. In the degradation phase, autolysosome contents are exposed to the acidic lumen, degraded by lysosomal hydrolases, and released back into the cytoplasm through lysosomal permeases for cell reuse [56, 62].

    Autophagy has an important function in cellular physiological and pathological regulation and viability. Therefore, autophagy is associated with cell/tissue injuries due to systematic disorders, hypoxia, starvation, infection, etc. It is speculated that correct autophagy control could improve DM-related systemic problems [63]. DM is disposed to cellular alterations accelerated by autophagy [64]. According to the literature, autophagy has double-edged sword functions in DM pathology since, in the beginning, it reduces ERS and consequently protects the cells [65].

    Nonetheless, systemic activation of autophagy might not always stimulate beneficial properties for non-targeted healthy cells. With the DM pathology progress, impaired autophagy misplaces its protective role and participates in the development of several DM chronic complications (Figure 2), including diabetic retinopathy, cardiomyopathy, vasculopathy, nephropathy, neuropathy, and skeletal alterations [46, 6668]. Besides, the dysregulation of autophagy could be used as a possible therapeutic target in DM [69]. Therefore, autophagy should be monitored at every disease stage of all target organs [70]. Understanding the role of autophagy in cellular maintenance is currently the focus of interest, with the help of various molecular laboratory techniques [71]. Moreover, clarifying the autophagy specifics generated by different disorders in all cells is essential.

    Impact of imbalanced autophagy in the development of diabetic complications in different tissues. ROS: reactive oxygen species; ER: endoplasmic reticulum. Created via BioRender.com

    Autophagy and DM

    Autophagy in the physiology of beta cells

    For cells to survive and function properly, continual intracellular synthesis must occur, and degradation is necessary [72]. DM and other metabolic diseases are influenced by various organelle dysfunctions caused by deranged autophagy [43, 73]. Low autophagy levels regulate survival and function, protecting insulin-targeting tissues [11, 74]. The ERS and OxS increase autophagy levels, controlling stress-induced intracellular changes [14, 43, 7577]. Autophagy-caused cell death can, however, result from excessively stimulated and constitutively activated autophagy [7881].

    Autophagy, DM, and chronic DM complications

    In diabetic animal models, genetic or chemical inhibition of autophagy may cause acceleration of beta cell mass and function loss [11]. Both sexes’ T1DM and T2DM animal models exhibit decreased autophagy [77, 8289]. Defective lysosomes and reduced insulin secretion are associated with abnormal autophagy in T1DM female and male mice [82, 85]. Compared to pancreatic cells from people without DM, those with T2DM show more autophagic vacuoles and autophagosomes and express lower lysosomal genes [90, 91]. Lipotoxicity, OxS, ERS, and inflammation induce autophagy impairment, organelle damage, and faulty lipid and protein accumulation in β cells and insulin-target tissues, promoting IR and T2DM [77, 92]. The resulting glucotoxicity impacts the continued escalation of OxS and ERS and enhances inflammation [11, 77, 93, 94]. Additionally, due to an improper response to the ERS caused by obesity, autophagy deficiency in β cells may trigger the transition from obesity to DM [95]. Although mechanisms are not entirely understood, autophagy may impact the chronic vascular complications of DM [43]. Autophagy is mediated by a variety of mechanisms in the chronic complications of DM, including mTORC1 (an autophagy inhibitor), AMPK (an autophagy activator), OxS, and ERS [9698]. In Figure 3, the three major autophagy signaling pathways—mTORC1, AMPK, and silent information regulator T1 (SIRT1) inhibition for negative autophagy regulation. AMPK activates the ULK1-ATG13-focal adhesion kinase family interacting with protein 200 complex, while mTORC1 inactivates. SIRT1 activates the ATG5-ATG7-LC3 complex and forkhead box O3 (FoxO3) [9698].

    Autophagy intracellular signaling pathways in DM. Created via BioRender.com

    Oral anti-hyperglycemic drugs and autophagy

    One possible preventive and therapeutic target for DM is the derangement of autophagy [69, 82]. Therefore, tapering to sufficient autophagy is a viable therapeutic goal in treating many metabolic abnormalities [77]. Resveratrol and rapamycin are two autophagy activators shown to improve beta cell activity in diabetic animal models [11]. Two current techniques for altering autophagy in DM animal models include using autophagy inhibitors or deleting ATG [43].

    Activating AMPK and SIRT1 signaling by metformin and sodium-glucose cotransporter-2 (SGLT-2) inhibitors may promote autophagy [90, 99, 100]. Metformin’s therapeutic effect on DM-related autophagy regulation is linked to the transcription factor EB (TFEB) [101]. In vitro lipotoxicity results in metformin promoting autophagy in beta cells and inhibiting their apoptosis [98]. It has been observed that the SGLT-2 inhibitor empagliflozin and the dipeptidyl peptidase 4 (DPP-4) inhibitor linagliptin restore glomerular and cardiomyocyte autophagy in db/db male mice and Zucker Diabetic Fatty male rats [102, 103]. Thiazolidinedione member, pioglitazone, promotes autophagy via the AMPK pathway [104]. Alogliptin (DPP-4 inhibitor) induces autophagy through a mechanism that depends on the SIRT1/AMPK/mTORC1 cascade [105]. Liraglutide, a member of glucagon-like peptide 1 receptor agonists, may increase autophagy in male Sprague Dawley rats, a model of chronic renal failure, by modulating the AMPK and mTORC1 pathways [106, 107]. Additionally, glucagon-like peptide 1 receptor agonists promote autophagy by reducing T2DM impairment of autophagosome-lysosome fusion [77].

    Conclusions

    There is now strong evidence that autophagy plays a role in the etiology of DM. The development of drugs that take advantage of autophagy’s putative cytoprotective effect in DM is a potentially intriguing field of research. Future research should clarify contradictory findings, and important questions about the role of autophagy dysfunction in developing IR and T2DM remain unanswered. If the pathogenic role of dysregulated autophagy in the development of DM is proven, a new class of drugs based on a novel concept will be conceivable.

    Because there are multiple perspectives on the relationship between autophagy and DM, it is critical to identify specific molecular targets within the autophagy pathway that could lead to the development of novel therapeutic interventions for DM, such as strategies to enhance autophagy in specific cell types, like as pancreatic beta cells, to improve their function and survival. Furthermore, several dietary interventions activate autophagy and enhance insulin sensitivity; therefore, understanding the appropriate dietary strategies that can modify autophagy to prevent or manage DM is critical.

    Understanding the relationships between autophagy regulation and other therapeutic methods and combining autophagy-targeting interventions with existing DM treatments such as insulin therapy or anti-hyperglycemia medicines, will be critical in designing effective combination therapies.

    Abbreviations

    AD:

    Alzheimer’s disease

    AMPK:

    adenosine monophosphate-activated kinase

    ATG:

    autophagy-related genes

    DM:

    diabetes mellitus

    ERS:

    endoplasmic reticulum stress

    GDM:

    gestational diabetes mellitus

    IR:

    insulin resistance

    LC3:

    microtubule-associated protein 1A/1B-light chain 3

    mTOR:

    mammalian target of rapamycin

    mTORC1:

    mammalian target of rapamycin complex 1

    OxS:

    oxidative stress

    PI3K:

    phosphatidylinositol 3-kinase

    SIRT1:

    silent information regulator T1

    T1DM:

    type 1 diabetes mellitus

    T2DM:

    type 2 diabetes mellitus

    ULK1:

    unc-51-like autophagy activating kinase 1

    Declarations

    Author contributions

    SZ, ZG, and JR: Writing—original draft, Conceptualization. ERI and MO: Investigation, Writing—original draft, Supervision. All authors contributed to manuscript’s revision, and read, and approved the submitted version.

    Conflicts of interest

    The authors declare that they have no conflicts of interest.

    Ethical approval

    Not applicable.

    Consent to participate

    Not applicable.

    Consent to publication

    Not applicable.

    Availability of data and materials

    Not applicable.

    Funding

    This work was funded by the Ministry of Education, Science and Technological Development of the Republic of Serbia [Contract number 451-03-47/2023-01/200017]. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

    Copyright

    © The Author(s) 2023.

    References

    Nair M. Diabetes mellitus, part 1: physiology and complications. Br J Nurs. 2007;16:1848. [DOI] [PubMed]
    Forbes JM, Cooper ME. Mechanisms of diabetic complications. Physiol Rev. 2013;93:13788. [DOI] [PubMed]
    Saeedi P, Petersohn I, Salpea P, Malanda B, Karuranga S, Unwin N, et al. Global and regional diabetes prevalence estimates for 2019 and projections for 2030 and 2045: results from the International Diabetes Federation Diabetes Atlas, 9th edition. Diabetes Res Clin Pract. 2019;157:107843. [DOI] [PubMed]
    Cho NH, Shaw JE, Karuranga S, Huang Y, da Rocha Fernandes JD, Ohlrogge AW, et al. IDF Diabetes Atlas: global estimates of diabetes prevalence for 2017 and projections for 2045. Diabetes Res Clin Pract. 2018;138:27181. [DOI] [PubMed]
    Diabetes [Internet]. Geneva: World Health Organization; c2023 [cited 2023 Apr 4]. Available from: http://www.who.int/news-room/fact-sheets/detail/diabetes
    Roglic G. WHO Global report on diabetes: a summary. Int J Noncommun Dis. 2016;1:38. [DOI]
    Ceriello A, Prattichizzo F. Variability of risk factors and diabetes complications. Cardiovasc Diabetol. 2021;20:101. [DOI] [PubMed] [PMC]
    Obradovic M, Stanimirovic J, Panic A, Bogdanovic N, Sudar-Milovanovic E, Cenic-Milosevic D, et al. Regulation of Na+/K+-ATPase by estradiol and IGF-1 in cardio-metabolic diseases. Curr Pharm Des. 2017;23:155161. [DOI] [PubMed]
    Obradovic M, Sudar E, Zafirovic S, Stanimirovic J, Labudovic-Borovic M, Isenovic ER. Estradiol in vivo induces changes in cardiomyocytes size in obese rats. Angiology. 2015;66:2535. [DOI] [PubMed]
    American Diabetes Association. Diagnosis and classification of diabetes mellitus. Diabetes Care. 2009;32 Suppl 1:S627. [DOI] [PubMed] [PMC]
    Yao D, GangYi Y, QiNan W. Autophagic dysfunction of β cell dysfunction in type 2 diabetes, a double-edged sword. Genes Dis. 2021;8:43847. [DOI] [PubMed] [PMC]
    Sehrawat A, Mishra J, Mastana SS, Navik U, Bhatti GK, Reddy PH, et al. Dysregulated autophagy: a key player in the pathophysiology of type 2 diabetes and its complications. Biochim Biophys Acta Mol Basis Dis. 2023;1869:166666. [DOI] [PubMed]
    Glick D, Barth S, Macleod KF. Autophagy: cellular and molecular mechanisms. J Pathol. 2010;221:312. [DOI] [PubMed] [PMC]
    Marasco MR, Conteh AM, Reissaus CA, Cupit JE 5th, Appleman EM, Mirmira RG, et al. Interleukin-6 reduces β-Cell oxidative stress by linking autophagy with the antioxidant response. Diabetes. 2018;67:157688. [DOI] [PubMed] [PMC]
    Jia G, Sowers JR. Autophagy: a housekeeper in cardiorenal metabolic health and disease. Biochim Biophys Acta. 2015;1852:21924. [DOI] [PubMed] [PMC]
    Chen Y, Hua Y, Li X, Arslan IM, Zhang W, Meng G. Distinct types of cell death and the implication in diabetic cardiomyopathy. Front Pharmacol. 2020;11:42. [DOI] [PubMed] [PMC]
    Abdellatif M, Sedej S, Carmona-Gutierrez D, Madeo F, Kroemer G. Autophagy in cardiovascular aging. Circ Res. 2018;123:80324. [DOI] [PubMed]
    Jung S, Jeong H, Yu SW. Autophagy as a decisive process for cell death. Exp Mol Med. 2020;52:92130. [DOI] [PubMed] [PMC]
    Reed J, Bain S, Kanamarlapudi V. A review of current trends with type 2 diabetes epidemiology, aetiology, pathogenesis, treatments and future perspectives. Diabetes Metab Syndr Obes. 2021;14:3567602. [DOI] [PubMed] [PMC]
    Demir S, Nawroth PP, Herzig S, Ekim Üstünel B. Emerging targets in type 2 diabetes and diabetic complications. Adv Sci (Weinh). 2021;8:e2100275. [DOI] [PubMed] [PMC]
    Chobot A, Górowska-Kowolik K, Sokołowska M, Jarosz-Chobot P. Obesity and diabetes—not only a simple link between two epidemics. Diabetes Metab Res Rev. 2018;34:e3042. [DOI] [PubMed] [PMC]
    Al-Goblan AS, Al-Alfi MA, Khan MZ. Mechanism linking diabetes mellitus and obesity. Diabetes Metab Syndr Obes. 2014;7:58791. [DOI] [PubMed] [PMC]
    Banday MZ, Sameer AS, Nissar S. Pathophysiology of diabetes: an overview. Avicenna J Med. 2020;10:17488. [DOI] [PubMed] [PMC]
    Burrack AL, Martinov T, Fife BT. T cell-mediated beta cell destruction: autoimmunity and alloimmunity in the context of type 1 diabetes. Front Endocrinol (Lausanne). 2017;8:343. [DOI] [PubMed] [PMC]
    Duca LM, Reboussin BA, Pihoker C, Imperatore G, Saydah S, Mayer-Davis E, et al. Diabetic ketoacidosis at diagnosis of type 1 diabetes and glycemic control over time: the SEARCH for diabetes in youth study. Pediatr Diabetes. 2019;20:1729. [DOI] [PubMed] [PMC]
    Wu Y, Ding Y, Tanaka Y, Zhang W. Risk factors contributing to type 2 diabetes and recent advances in the treatment and prevention. Int J Med Sci. 2014;11:1185200. [DOI] [PubMed] [PMC]
    Papaetis GS, Papakyriakou P, Panagiotou TN. Central obesity, type 2 diabetes and insulin: exploring a pathway full of thorns. Arch Med Sci. 2015;11:46382. [DOI] [PubMed] [PMC]
    An J, Nichols GA, Qian L, Munis MA, Harrison TN, Li Z, et al. Prevalence and incidence of microvascular and macrovascular complications over 15 years among patients with incident type 2 diabetes. BMJ Open Diabetes Res Care. 2021;9:e001847. [DOI] [PubMed] [PMC]
    Alfadhli EM. Gestational diabetes mellitus. Saudi Med J. 2015;36:399406. [DOI] [PubMed] [PMC]
    de la Monte SM, Wands JR. Alzheimer’s disease is type 3 diabetes—evidence reviewed. J Diabetes Sci Technol. 2008;2:110113. [DOI] [PubMed] [PMC]
    Chornenkyy Y, Wang WX, Wei A, Nelson PT. Alzheimer’s disease and type 2 diabetes mellitus are distinct diseases with potential overlapping metabolic dysfunction upstream of observed cognitive decline. Brain Pathol. 2019;29:317. [DOI] [PubMed] [PMC]
    Hardigan T, Ward R, Ergul A. Cerebrovascular complications of diabetes: focus on cognitive dysfunction. Clin Sci (Lond). 2016;130:180722. [DOI] [PubMed] [PMC]
    Chowdhary N, Barbui C, Anstey KJ, Kivipelto M, Barbera M, Peters R, et al. Reducing the risk of cognitive decline and dementia: WHO recommendations. Front Neurol. 2021;12:765584. [DOI] [PubMed] [PMC]
    Sędzikowska A, Szablewski L. Insulin and insulin resistance in Alzheimer’s disease. Int J Mol Sci. 2021;22:9987. [DOI] [PubMed] [PMC]
    Liu R, Li L, Shao C, Cai H, Wang Z. The impact of diabetes on vascular disease: progress from the perspective of epidemics and treatments. J Diabetes Res. 2022;2022:1531289. [DOI] [PubMed] [PMC]
    Luchsinger JA, Reitz C, Honig LS, Tang MX, Shea S, Mayeux R. Aggregation of vascular risk factors and risk of incident Alzheimer disease. Neurology. 2005;65:54551. [DOI] [PubMed] [PMC]
    Brain J, Greene L, Tang EYH, Louise J, Salter A, Beach S, et al. Cardiovascular disease, associated risk factors, and risk of dementia: an umbrella review of meta-analyses. Front Epidemiol. 2023;3:1095236. [DOI]
    Bharath LP, Rockhold JD, Conway R. Selective autophagy in hyperglycemia-induced microvascular and macrovascular diseases. Cells. 2021;10:2114. [DOI] [PubMed] [PMC]
    Salemkour Y, Lenoir O. Endothelial autophagy dysregulation in diabetes. Cells. 2023;12:947. [DOI] [PubMed] [PMC]
    White E. The role for autophagy in cancer. J Clin Invest. 2015;125:426. [DOI] [PubMed] [PMC]
    Yang J, Zhou R, Ma Z. Autophagy and energy metabolism. In: Qin ZH, editor. Autophagy: Biology and Diseases. Singapore: Springer; 2019. pp. 329–57. [DOI] [PubMed]
    Yoo SM, Jung YK. A molecular approach to mitophagy and mitochondrial dynamics. Mol Cells. 2018;41:1826. [DOI] [PubMed] [PMC]
    Ge X, Wang L, Fei A, Ye S, Zhang Q. Research progress on the relationship between autophagy and chronic complications of diabetes. Front Physiol. 2022;13:956344. [DOI] [PubMed] [PMC]
    Oku M, Sakai Y. Three distinct types of microautophagy based on membrane dynamics and molecular machineries. Bioessays. 2018;40:e1800008. [DOI] [PubMed]
    Mijaljica D, Prescott M, Devenish RJ. Microautophagy in mammalian cells: revisiting a 40-year-old conundrum. Autophagy. 2011;7:67382. [DOI] [PubMed]
    Zaffagnini G, Martens S. Mechanisms of selective autophagy. J Mol Biol. 2016;428:171424. [DOI] [PubMed] [PMC]
    Gubas A, Dikic I. A guide to the regulation of selective autophagy receptors. FEBS J. 2022;289:7589. [DOI] [PubMed]
    Anding AL, Baehrecke EH. Cleaning house: selective autophagy of organelles. Dev Cell. 2017;41:1022. [DOI] [PubMed] [PMC]
    Levine B, Kroemer G. Biological functions of autophagy genes: a disease perspective. Cell. 2019;176:1142. [DOI] [PubMed] [PMC]
    Jung CH, Ro SH, Cao J, Otto NM, Kim DH. mTOR regulation of autophagy. FEBS Lett. 2010;584:128795. [DOI] [PubMed] [PMC]
    Mizushima N, Levine B. Autophagy in human diseases. N Engl J Med. 2020;383:156476. [DOI] [PubMed]
    Sun K, Deng W, Zhang S, Cai N, Jiao S, Song J, et al. Paradoxical roles of autophagy in different stages of tumorigenesis: protector for normal or cancer cells. Cell Biosci. 2013;3:35. [DOI] [PubMed] [PMC]
    Alvarez-Meythaler JG, Garcia-Mayea Y, Mir C, Kondoh H, LLeonart ME. Autophagy takes center stage as a possible cancer hallmark. Front Oncol. 2020;10:586069. [DOI] [PubMed] [PMC]
    Hurley JH, Young LN. Mechanisms of autophagy initiation. Annu Rev Biochem. 2017;86:22544. [DOI] [PubMed] [PMC]
    He C, Klionsky DJ. Regulation mechanisms and signaling pathways of autophagy. Annu Rev Genet. 2009;43:6793. [DOI] [PubMed] [PMC]
    Parzych KR, Klionsky DJ. An overview of autophagy: morphology, mechanism, and regulation. Antioxid Redox Signal. 2014;20:46073. [DOI] [PubMed] [PMC]
    Geng J, Klionsky DJ. The Atg8 and Atg12 ubiquitin-like conjugation systems in macroautophagy. ‘Protein modifications: beyond the usual suspects’ review series. EMBO Rep. 2008;9:85964. [DOI] [PubMed] [PMC]
    Young ARJ, Chan EYW, Hu XW, Köchl R, Crawshaw SG, High S, et al. Starvation and ULK1-dependent cycling of mammalian Atg9 between the TGN and endosomes. J Cell Sci. 2006;119:3888900. [DOI] [PubMed]
    Mizushima N, Klionsky DJ. Protein turnover via autophagy: implications for metabolism. Annu Rev Nutr. 2007;27:1940. [DOI] [PubMed]
    Pfeifer U. Inhibition by insulin of the formation of autophagic vacuoles in rat liver. A morphometric approach to the kinetics of intracellular degradation by autophagy. J Cell Biol. 1978;78:15267. [DOI] [PubMed] [PMC]
    Fader CM, Sánchez DG, Mestre MB, Colombo MI. TI-VAMP/VAMP7 and VAMP3/cellubrevin: two v-SNARE proteins involved in specific steps of the autophagy/multivesicular body pathways. Biochim Biophys Acta. 2009;1793:190116. [DOI] [PubMed]
    Yorimitsu T, Klionsky DJ. Autophagy: molecular machinery for self-eating. Cell Death Differ. 2005;12 Suppl 2:154252. [DOI] [PubMed] [PMC]
    Yamamoto S, Kazama JJ, Fukagawa M. Autophagy: a two-edged sword in diabetes mellitus. Biochem J. 2013;456:e13. [DOI] [PubMed]
    Sarmah DT, Gujjar S, Mathapati S, Bairagi N, Chatterjee S. Identification of critical autophagy-related proteins in diabetic retinopathy: a multi-dimensional computational study. Gene. 2023;866:147339. [DOI] [PubMed]
    Gong Q, Wang H, Yu P, Qian T, Xu X. Protective or harmful: the dual roles of autophagy in diabetic retinopathy. Front Med (Lausanne). 2021;8:644121. [DOI] [PubMed] [PMC]
    Li R, Du JH, Yao GM, Yao Y, Zhang J. Autophagy: a new mechanism for regulating VEGF and PEDF expression in retinal pigment epithelium cells. Int J Ophthalmol. 2019;12:55762. [DOI] [PubMed] [PMC]
    Lopes de Faria JM, Duarte DA, Montemurro C, Papadimitriou A, Consonni SR, Lopes de Faria JB. Defective autophagy in diabetic retinopathy. Invest Ophthalmol Vis Sci. 2016;57:435666. [DOI] [PubMed]
    Liu P, Zhu W, Wang Y, Ma G, Zhao H, Li P. Chinese herbal medicine and its active compounds in attenuating renal injury via regulating autophagy in diabetic kidney disease. Front Endocrinol (Lausanne). 2023;14:1142805. [DOI] [PubMed] [PMC]
    Dikic I, Elazar Z. Mechanism and medical implications of mammalian autophagy. Nat Rev Mol Cell Biol. 2018;19:34964. [DOI] [PubMed]
    Komatsu M, Waguri S, Koike M, Sou YS, Ueno T, Hara T, et al. Homeostatic levels of p62 control cytoplasmic inclusion body formation in autophagy-deficient mice. Cell. 2007;131:114963. [DOI] [PubMed]
    Nwose EU, Bwititi PT. Autophagy in diabetes pathophysiology: oxidative damage screening as potential for therapeutic management by clinical laboratory methods. Front Cell Dev Biol. 2021;9:651776. [DOI] [PubMed] [PMC]
    Ohsumi Y. Historical landmarks of autophagy research. Cell Res. 2014;24:923. [DOI] [PubMed] [PMC]
    Muralidharan C, Linnemann AK. β-Cell autophagy in the pathogenesis of type 1 diabetes. Am J Physiol Endocrinol Metab. 2021;321:E4106. [DOI] [PubMed] [PMC]
    Barlow AD, Thomas DC. Autophagy in diabetes: β-cell dysfunction, insulin resistance, and complications. DNA Cell Biol. 2015;34:25260. [DOI] [PubMed]
    Yan J, Feng Z, Liu J, Shen W, Wang Y, Wertz K, et al. Enhanced autophagy plays a cardinal role in mitochondrial dysfunction in type 2 diabetic Goto-Kakizaki (GK) rats: ameliorating effects of (-)-epigallocatechin-3-gallate. J Nutr Biochem. 2012;23:71624. [DOI] [PubMed]
    He Q, Sha S, Sun L, Zhang J, Dong M. GLP-1 analogue improves hepatic lipid accumulation by inducing autophagy via AMPK/mTOR pathway. Biochem Biophys Res Commun. 2016;476:196203. [DOI] [PubMed]
    Kitada M, Koya D. Autophagy in metabolic disease and ageing. Nat Rev Endocrinol. 2021;17:64761. [DOI] [PubMed]
    Demirtas L, Guclu A, Erdur FM, Akbas EM, Ozcicek A, Onk D, et al. Apoptosis, autophagy & endoplasmic reticulum stress in diabetes mellitus. Indian J Med Res. 2016;144:51524. [DOI] [PubMed] [PMC]
    Masini M, Lupi R, Bugliani M, Boggi U, Filipponi F, Masiello P, et al. A role for autophagy in β-cell life and death. Islets. 2009;1:1579. [DOI] [PubMed]
    Yang JS, Lu CC, Kuo SC, Hsu YM, Tsai SC, Chen SY, et al. Autophagy and its link to type II diabetes mellitus. Biomedicine (Taipei). 2017;7:8. [PubMed] [PMC]
    Wang S, Sun QQ, Xiang B, Li XJ. Pancreatic islet cell autophagy during aging in rats. Clin Invest Med. 2013;36:E7280. [DOI] [PubMed]
    Muralidharan C, Conteh AM, Marasco MR, Crowder JJ, Kuipers J, de Boer P, et al. Pancreatic beta cell autophagy is impaired in type 1 diabetes. Diabetologia. 2021;64:86577. [DOI] [PubMed] [PMC]
    Chen ZF, Li YB, Han JY, Yin JJ, Wang Y, Zhu LB, et al. Liraglutide prevents high glucose level induced insulinoma cells apoptosis by targeting autophagy. Chin Med J (Engl). 2013;126:93741. [PubMed]
    Ebato C, Uchida T, Arakawa M, Komatsu M, Ueno T, Komiya K, et al. Autophagy is important in islet homeostasis and compensatory increase of beta cell mass in response to high-fat diet. Cell Metab. 2008;8:32532. [DOI] [PubMed]
    Sun-Wada GH, Toyomura T, Murata Y, Yamamoto A, Futai M, Wada Y. The a3 isoform of V-ATPase regulates insulin secretion from pancreatic beta-cells. J Cell Sci. 2006;119:453140. [DOI] [PubMed]
    Barutta F, Bellini S, Kimura S, Hase K, Corbetta B, Corbelli A, et al. Protective effect of the tunneling nanotube-TNFAIP2/M-sec system on podocyte autophagy in diabetic nephropathy. Autophagy. 2023;19:50524. [DOI] [PubMed] [PMC]
    Bugliani M, Mossuto S, Grano F, Suleiman M, Marselli L, Boggi U, 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;10:52. [DOI] [PubMed] [PMC]
    Marasco MR, Linnemann AK. β-cell autophagy in diabetes pathogenesis. Endocrinology. 2018;159:212741. [DOI] [PubMed] [PMC]
    Sheng Q, Xiao X, Prasadan K, Chen C, Ming Y, Fusco J, et al. Autophagy protects pancreatic beta cell mass and function in the setting of a high-fat and high-glucose diet. Sci Rep. 2017;7:16348. [DOI] [PubMed] [PMC]
    Masini M, Bugliani M, Lupi R, del Guerra S, Boggi U, Filipponi F, et al. Autophagy in human type 2 diabetes pancreatic beta cells. Diabetologia. 2009;52:10836. [DOI] [PubMed]
    Fujitani Y, Kawamori R, Watada H. The role of autophagy in pancreatic beta-cell and diabetes. Autophagy. 2009;5:2802. [DOI] [PubMed]
    Kosacka J, Kern M, Klöting N, Paeschke S, Rudich A, Haim Y, et al. Autophagy in adipose tissue of patients with obesity and type 2 diabetes. Mol Cell Endocrinol. 2015;409:2132. [DOI] [PubMed]
    Radovanović J, Banjac K, Obradović MM, Isenović ER. Antioxidant enzymes and vascular diseases. Explor Med. 2021;2:54455. [DOI]
    Panic A, Stanimirovic J, Sudar-Milovanovic E, Isenovic ER. Oxidative stress in obesity and insulin resistance. Explor Med. 2022;3:5870. [DOI]
    Quan W, Lim YM, Lee MS. Role of autophagy in diabetes and endoplasmic reticulum stress of pancreatic β-cells. Exp Mol Med. 2012;44:818. [DOI] [PubMed] [PMC]
    Kim J, Kundu M, Viollet B, Guan KL. AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat Cell Biol. 2011;13:13241. [DOI] [PubMed] [PMC]
    Kim J, Kim YC, Fang C, Russell RC, Kim JH, Fan W, et al. Differential regulation of distinct Vps34 complexes by AMPK in nutrient stress and autophagy. Cell. 2013;152:290303. [DOI] [PubMed] [PMC]
    Jiang Y, Huang W, Wang J, Xu Z, He J, Lin X, et al. Metformin plays a dual role in MIN6 pancreatic β cell function through AMPK-dependent autophagy. Int J Biol Sci. 2014;10:26877. [DOI] [PubMed] [PMC]
    Packer M. Autophagy-dependent and -independent modulation of oxidative and organellar stress in the diabetic heart by glucose-lowering drugs. Cardiovasc Diabetol. 2020;19:62. [DOI] [PubMed] [PMC]
    Xu J, Kitada M, Ogura Y, Liu H, Koya D. Dapagliflozin restores impaired autophagy and suppresses inflammation in high glucose-treated HK-2 cells. Cells. 2021;10:1457. [DOI] [PubMed] [PMC]
    Zhang D, Ma Y, Liu J, Deng Y, Zhou B, Wen Y, et al. Metformin alleviates hepatic steatosis and insulin resistance in a mouse model of high-fat diet-induced nonalcoholic fatty liver disease by promoting transcription factor EB-dependent autophagy. Front Pharmacol. 2021;12:689111. [DOI] [PubMed] [PMC]
    Korbut AI, Taskaeva IS, Bgatova NP, Muraleva NA, Orlov NB, Dashkin MV, et al. SGLT2 inhibitor empagliflozin and DPP4 inhibitor linagliptin reactivate glomerular autophagy in db/db mice, a model of type 2 diabetes. Int J Mol Sci. 2020;21:2987. [DOI] [PubMed] [PMC]
    Aragón-Herrera A, Feijóo-Bandín S, Otero Santiago M, Barral L, Campos-Toimil M, Gil-Longo J, et al. Empagliflozin reduces the levels of CD36 and cardiotoxic lipids while improving autophagy in the hearts of Zucker diabetic fatty rats. Biochem Pharmacol. 2019;170:113677. [DOI] [PubMed]
    Xu L, Xu K, Wu Z, Chen Z, He Y, Ma C, et al. Pioglitazone attenuates advanced glycation end products-induced apoptosis and calcification by modulating autophagy in tendon-derived stem cells. J Cell Mol Med. 2020;24:224051. [DOI] [PubMed] [PMC]
    Zhu B, Li Y, Xiang L, Zhang J, Wang L, Guo B, et al. Alogliptin improves survival and health of mice on a high-fat diet. Aging Cell. 2019;18:e12883. [DOI] [PubMed] [PMC]
    Xue L, Pan Z, Yin Q, Zhang P, Zhang J, Qi W. Liraglutide promotes autophagy by regulating the AMPK/mTOR pathway in a rat remnant kidney model of chronic renal failure. Int Urol Nephrol. 2019;51:230513. [DOI] [PubMed]
    He Y, Ao N, Yang J, Wang X, Jin S, Du J. The preventive effect of liraglutide on the lipotoxic liver injury via increasing autophagy. Ann Hepatol. 2020;19:4452. [DOI] [PubMed]