Advertisement
Review Article| Volume 197, 110568, March 2023

Clinical expert consensus on the assessment and protection of pancreatic islet β-cell function in type 2 diabetes mellitus

Open AccessPublished:February 02, 2023DOI:https://doi.org/10.1016/j.diabres.2023.110568

      Abstract

      Islet β-cell dysfunction is a basic pathophysiological characteristic of type 2 diabetes mellitus (T2DM). Appropriate assessment of islet β-cell function is beneficial to better management of T2DM. Protecting islet β-cell function is vital to delay the progress of type 2 diabetes mellitus. Therefore, the Pancreatic Islet β-cell Expert Panel of the Chinese Diabetes Society and Endocrinology Society of Jiangsu Medical Association organized experts to draft the “Clinical expert consensus on the assessment and protection of pancreatic islet β-cell function in type 2 diabetes mellitus.” This consensus suggests that β-cell function can be clinically assessed using blood glucose-based methods or methods that combine blood glucose and endogenous insulin or C-peptide levels. Some measures, including weight loss and early and sustained euglycemia control, could effectively protect islet β-cell function, and some newly developed drugs, such as Sodium-glucose cotransporter-2 inhibitor and Glucagon-like peptide-1 receptor agonists, could improve islet β-cell function, independent of glycemic control.

      Keywords

      The prevalence of adult diabetes mellitus in China has increased to 11.2%, and more than 90% of the cases is type 2 diabetes mellitus (T2DM) [
      • Chinese Diabetes Society
      Guideline for the prevention and treatment of type 2 diabetes mellitus in China (2020 edition).
      ]. Although the precise mechanism of T2DM development has not been fully clarified, pancreatic islet β-cell dysfunction and insulin resistance are considered two major factors in the pathogenesis of T2DM. Islet β-cell function in T2DM patients decreases at an average rate of 2% yearly and considerably declines in patients with a disease duration of more than 10 years [
      • Gao Z.
      • Yan W.
      • Fang Z.
      • Zhang Z.
      • Yuan L.
      • Wang X.
      • et al.
      Annual decline in β-cell function in patients with type 2 diabetes in China.
      ]. An extensive amount of time is elapsed from the full compensation of islet β-cells for glycemia control to complete decompensation, and this period may allow physicians to adopt useful measures to protect islet β-cell function. Therefore, appropriately assessing islet β-cell function and developing an optimal treatment regimen early should contribute to better delaying the progression of T2DM.
      At present, there is a lack of guidance specifically documented for the assessment and protection of islet β-cell function in T2DM patients in China. To address this, the Pancreatic Islet β-cell Expert Panel of the Chinese Diabetes Society and the Endocrinology Society of Jiangsu Medical Association organized some experts to jointly draft this consensus, focusing on the mechanism of islet β-cell dysfunction, assessment methods, and treatment strategies for the protection of islet β-cell function. They searched and discussed relevant articles from PubMed, Embase, Chinese BioMedical Literature, WanFang Medicine, and China National Knowledge Infrastructure databases, ultimately reaching this consensus. The aim of the consensus is to help clinicians understand and apply assessment methods of islet β-cell function appropriately and implement effective measures to protect islet β-cell function, thus improving the clinical outcomes of T2DM patients.

      1. Concept of islet β-cell function and its dysfunction mechanism

      1.1 Key points

      • 1.
        Islet β-cell function can be defined as the ability of β-cells to synthesize, store, and secrete insulin in appropriate amounts to maintain euglycemia.
      • 2.
        Chronic nonspecific inflammatory reactions, oxidative stress, endoplasmic reticulum stress, and mitochondrial dysfunction caused by glycolipid toxicity, glycemic variability, and environmental endocrine-disrupting compounds are the key factors that lead to a decline in insulin secretion ability and aging of islet β-cells.
      • 3.
        β-cell apoptosis, dedifferentiation of pancreatic endocrine progenitor cells, and trans-differentiation to α-cell-like cells are the main reasons for dysregulation of functional islet β-cell mass.
      Narrowly speaking, islet β-cell function can be defined as the ability of β-cells to synthesize, store, and secrete insulin in appropriate amounts to maintain euglycemia [
      • Xiang K.
      Special types of diabetes mellitus. [M].
      ,
      • Hannon T.S.
      • Kahn S.E.
      • Utzschneider K.M.
      • Buchanan T.A.
      • Nadeau K.J.
      • Zeitler P.S.
      • et al.
      Review of methods for measuring β-cell function: design considerations from the restoring insulin secretion (RISE) consortium.
      ]. Pulsatile insulin secretion is an indicator of normal islet β-cell function. After glucose stimulation, islet β-cells secrete insulin in a biphasic mode, usually the first and second phases. One to two minutes after intravenous glucose stimulation, in normal individuals, insulin level in the portal vein increases, and that in the peripheral vein peaks at 3-5 min, lasting for approximately 10 min (the first phase). However, when glucose is administered orally, peak insulin level is usually attained after 20-30 min, called the early secretory phase. The amount of insulin secreted in the first or early secretory phase accounts for 2-3% of the total insulin in β-cells. After the first phase or early secretory phase (10-30 min later), insulin secretion slowly increases, reaching a plateau at 2-3 h and lasting for several hours. This is the second phase, accounting for approximately 20% of total insulin secretion in islet β-cells [
      • Xiang K.
      Special types of diabetes mellitus. [M].
      . In term of subject with normal glucose tolerance (NGT), after glucose stimulation, the peak value of insulin can reach 5-10 times the fasting value, while the peak value of C-peptide can reach 5-8 times the fasting value and gradually returns to the fasting level within 3-4 h. For patient with type 1 diabetes mellitus (T1DM), the fasting insulin level is lower than the normal value, and blood glucose levels rise dramatically after glucose stimulation; however, insulin and C-peptide levels cannot increase simultaneously, often showing no peak and a low flat curve [
      • Eisenbarth G.S.
      Type I diabetes mellitus. A chronic autoimmune disease.
      ]. For T2DM patient, during the early stage of disease, fasting insulin level was often in the normal range, but insulin secretion rose slowly after glucose stimulation, and the early secretory phase subsided, reaching its peak 1-2 h after stimulation. This may be higher or lower than that of NGT subjects, and cannot fall back to the fasting level 3-4 h after stimulation in most non-overweight/obese patients. The insulin secretion curve of overweight/obesity was similar to that of non-overweight/obesity, but insulin secretion level increased significantly during fasting and after glucose stimulation. As the disease progresses, islet β-cell function gradually declines, similar to T1DM [
      • DeFronzo R.A.
      Pathogenesis of type 2 diabetes mellitus.
      ].
      In addition to insulin, islet β-cells also secrete a variety of other hormones and peptides. Generally, islet β-cell dysfunction is believed to be first exhibited in the loss of sensitivity to glucose stimulation, especially the disappearance of the first or early secretory phase, while the non-glucose-stimulated insulin secretion (NGSIS) response may still exist. However, with prolongation of the disease course, NGSIS function also clearly decreases.
      The mechanism underlying islet β-cell dysfunction is complex. Together, environmental and genetic factors lead to islet β-cell failure. Chronic nonspecific inflammatory reactions, oxidative stress, endoplasmic reticulum stress, and mitochondrial dysfunction caused by glycolipid toxicity, glycemic variability, and environmental endocrine-disrupting compounds are the key factors that lead to a decline in insulin secretion ability and aging of islet β-cells [
      • Yang Y.
      • Kim J.W.
      • Park H.S.
      • Lee E.Y.
      • Yoon K.H.
      Pancreatic stellate cells in the islets as a novel target to preserve the pancreatic β-cell mass and function.
      ,
      • Ying W.
      • Fu W.
      • Lee Y.S.
      • Olefsky J.M.
      The role of macrophages in obesity-associated islet inflammation and β-cell abnormalities.
      ,
      • Weir G.C.
      • Gaglia J.
      • Bonner-Weir S.
      Inadequate β-cell mass is essential for the pathogenesis of type 2 diabetes.
      ]. β-cell apoptosis, dedifferentiation to pancreatic endocrine progenitor cells, and trans-differentiation to α-cell-like cells are the main reasons for the dysregulation of the islet β-cell functional mass [
      • Talchai C.
      • Xuan S.
      • Lin H.V.
      • Sussel L.
      • Accili D.
      Pancreatic β cell dedifferentiation as a mechanism of diabetic β cell failure.
      ]. In addition, variation in multiple susceptibility genes is closely related to islet β-cell dysfunction [
      • Tian Q.
      • Hong T.
      The pathophysiology of insulin secretory dysfunction and its therapeutic strategy.
      ].

      2. Clinical significance of assessing islet β-cell function

      2.1 Key points

      Appropriate assessment of islet β-cell function is beneficial to better selecting an individualized treatment regimen, and predicting the prognosis of T2DM patients.

      2.2 Selecting a reasonable treatment regimen

      In T2DM patients, it is necessary to analyze the severity of insulin resistance and select metformin and other insulin sensitizers to improve insulin sensitivity. Patients with insufficient insulin secretion require further analysis to determine whether islet β-cell function is temporarily inhibited by glucolipid toxicity or permanently damaged by disease progression. These two possibilities can be briefly distinguished by combining the disease duration and NGSIS (such as arginine) response. Regarding the former, especially those with newly diagnosed T2DM and certain NGSIS, glucose toxicity should be relieved rapidly by intensive insulin therapy to restore β-cell function, and the following treatment regimen [
      • Weng J.
      • Li Y.
      • Xu W.
      • Shi L.
      • Zhang Q.
      • Zhu D.
      • et al.
      Effect of intensive insulin therapy on beta-cell function and glycaemic control in patients with newly diagnosed type 2 diabetes: a multicentre randomised parallel-group trial.
      ] can be optimized according to the reassessment results of islet β-cell function. Regarding the latter, especially for patients with a longer disease duration and poor NGSIS response, insulin supplementation or replacement therapy [
      • Zhu X.
      • Yan H.
      • Chang X.
      • Xia M.
      • Wang L.
      • Bian H.
      • et al.
      The value of arginine stimulation test in evaluating the first-phase insulin secretion and its guiding role for the treatment and its guiding role for the treatment of type 2diabetes mellitus.
      ] should be reasonably performed.

      2.3 Predicting the prognosis

      Islet β-cell function can be used as an important parameter to predict the prognosis of patients with diabetes. There is a negative correlation between islet β-cell function and diabetic microvascular complications [
      • Bo S.
      • Cavallo-Perin P.
      • Gentile L.
      • Repetti E.
      • Pagano G.
      Relationship of residual beta-cell function, metabolic control and chronic complications in type 2 diabetes mellitus.
      ,
      • Tung T.H.
      • Shih H.C.
      • Tsai S.T.
      • Chou P.
      • Chen S.J.
      • Lee F.L.
      • et al.
      A community-based study of the relationship between insulin resistance/beta-cell dysfunction and diabetic retinopathy among type II diabetics in Kinmen.
      ].

      3. Assessment methods of islet β-cell function

      3.1 Key points

      Each method for assessing islet β-cell function has its own merits and drawbacks. In clinical applications, it is necessary to comprehensively consider the purpose of assessment and sensitivity and specificity of different methods during the natural course of diabetes mellitus to maximize application potential. The hyperglycemia clamp test, IVGTT, and other glucose stimulation tests are preferred among diabetes mellitus high-risk subjects with normal glucose tolerance and are helpful in evaluating diabetes risk. IVGTT, OGTT, and insulin release tests are suitable for assessing islet β-cell function in prediabetes patients. Residual β-cell function can be measured using OGTT, arginine, or glucagon stimulation in patients diagnosed with diabetes.
      • I.
        Methods for islet β-cell function assessment
      • (1)
        Blood glucose-based assessment methods
      Blood glucose levels, including those of fasting blood glucose, postprandial blood glucose, and glycosylated hemoglobin A1c (HBA1c), can reflect islet β-cell function in T2DM patients. Higher blood glucose levels indicate more serious β-cell dysfunction. In addition, the mean amplitude of glycemic excursion in continuous glucose monitoring is negatively correlated with islet β-cell function; larger glycemic variability corresponds to worse islet β-cell function in T2DM patients [
      • Chen T.
      • Xu F.
      • Su J.B.
      • Wang X.Q.
      • Chen J.F.
      • Wu G.
      • et al.
      Glycemic variability in relation to oral disposition index in the subjects with different stages of glucose tolerance.
      ,
      • Fang F.S.
      • Cheng X.L.
      • Gong Y.P.
      • Wang L.C.
      • Li L.
      • Li J.
      • et al.
      Association between glycemic indices and beta cell function in patients with newly diagnosed type 2 diabetes.
      ]. The parameters of glycemic variability may serve as indirect indices for assessing islet β-cell function.
      • (2)
        Assessment methods combining blood glucose and endogenous insulin/C-peptide (Table 1)
        Table 1Methods of islet β-cell function assessment based on insulin or C-peptide [
        • Hannon T.S.
        • Kahn S.E.
        • Utzschneider K.M.
        • Buchanan T.A.
        • Nadeau K.J.
        • Zeitler P.S.
        • et al.
        Review of methods for measuring β-cell function: design considerations from the restoring insulin secretion (RISE) consortium.
        ,
        • Matthews D.
        • Hosker J.
        • Rudenski A.
        • Naylor B.
        • Treacher D.
        • Turner R.
        Homeostasis model assessment: insulin resistance and beta-cell function from fasting plasma glucose and insulin concentrations in man.
        ,
        • Zhou Z.G.
        • Qi H.Y.
        • Chen X.Y.
        • Huang G.
        Replacement of insulin by fasting C-peptide in modified homeostasis model assessment to evaluate insulin resistance and islet βcell function.
        ,
        • Li G.W.
        • Yang W.Y.
        • Jiang Y.Y.
        • Hu Y.H.
        The possibility of (FINS ×FPG) / (PG2h + PG1h - 2FPG) being taken as an index of pancreaticβ cell insulin secretion in a population-based study.
        ,
        • Pacini G.
        • Mari A.
        Methods for clinical assessment of insulin sensitivity and beta-cell function.
        ,
        • Rayman G.
        • Clark P.
        • Schneider A.
        • Hales C.
        The first phase insulin response to intravenous glucose is highly reproducible.
        ,
        • DeFronzo R.
        • Tobin J.
        • Andres R.
        Glucose clamp technique: a method for quantifying insulin secretion and resistance.
        ,
        • Sjostrand M.
        • Carlson K.
        • Arnqvist H.
        • Gudbjörnsdottir S.
        • Landin-Olsson M.
        • Lindmark S.
        • et al.
        Assessment of beta-cell function in young patients with type 2 diabetes: arginine-stimulated insulin secretion may reflect beta-cell reserve.
        ,
        • Scheen A.
        • Castillo M.
        • Lefèbvre P.
        Assessment of residual insulin secretion in diabetic patients using the intravenous glucagon stimulatory test: methodological aspects and clinical applications.
        ].
        Appraisal procedureMain parameters, drug required, and calculation formulaClinical significanceAdvantagesLimitations
        Fasting serum insulinNon-insulin-treated diabetic patients can be used to assess baseline islet function.Simple and mature methodTesting methods and units are not uniform.
        Fasting serum C-peptideDiabetic patients treated with insulin or producing insulin antibodies can be used to assess islet β-cell function at baseline.Simple method; determination is not interfered by insulin.C-peptide level is affected by age, sex, body shape, and glucose tolerance level and has a long biological half- life.
        Ratio of proinsulin to fasting serum insulin (PI/FINS)The normal value was 7–9%, and the ratio progressively increased from NGT to IGT to diabetesSimple methodTesting methods and units are not uniform, and proinsulin testing has not been commercialized and standardized.
        Homeostasis model assessment-β (HOMA-β) [
        • Matthews D.
        • Hosker J.
        • Rudenski A.
        • Naylor B.
        • Treacher D.
        • Turner R.
        Homeostasis model assessment: insulin resistance and beta-cell function from fasting plasma glucose and insulin concentrations in man.
        ,
        • Zhou Z.G.
        • Qi H.Y.
        • Chen X.Y.
        • Huang G.
        Replacement of insulin by fasting C-peptide in modified homeostasis model assessment to evaluate insulin resistance and islet βcell function.
        ]
        HOMA-β = 20 × FINS/ (FPG − 3.5)

        HOMA-β (normal population)

        =0.27 × FCP / (FPG − 3.5) + 50;

        HOMA-β (diabetic population) = 0.27 × FCP / (FPG − 3.5)
        It is a correction of the FINS/FPG simplification index,Suitable for epidemiological research and assessment after clinical treatment (except for treatment with secretagogues such as insulin and sulfonylureas); if the influence of exogenous insulin is considered, C-peptide can be used instead of insulin for assessment.The interference of insulin resistance overestimates the function of β-cells.
        Modified β-cell function index (MBCI) [
        • Li G.W.
        • Yang W.Y.
        • Jiang Y.Y.
        • Hu Y.H.
        The possibility of (FINS ×FPG) / (PG2h + PG1h - 2FPG) being taken as an index of pancreaticβ cell insulin secretion in a population-based study.
        ]
        MBCI = (FINS × FPG)/ (PG120 + PG60–2 × FPG)

        75 g oral glucose tolerance test
        Represents insulin secretion stimulated by glucoseConsiders the influence of blood sugar level on insulin secretionOnly represents the response of islet β-cells to glucose, which is influenced by insulin sensitivity
        Ratio of peak insulin secretion to basal insulin level after stimulation (Ip/I0)

        75 g oral glucose tolerance test
        After sugar loading, the peak insulin level of healthy individuals can be increased by 5–10 times compared with the basic value.Reflects the ability of insulin secretion after stimulationThe IGT population may be compensated
        Ratio of insulin increment to blood sugar increment after sugar loading [
        • Pacini G.
        • Mari A.
        Methods for clinical assessment of insulin sensitivity and beta-cell function.
        ]
        (I30 − I0)/ (G30 − G0)

        75 g oral glucose tolerance test
        Represents early insulin secretionSimple calculationIslet β-cell function of individuals with flat insulin secretion curve cannot be compared owing to interference by insulin resistance
        Area under the insulin curve after glucose loading (AUCI) [
        • Pacini G.
        • Mari A.
        Methods for clinical assessment of insulin sensitivity and beta-cell function.
        ]
        AUCI = (I0 + 2 × I30 + 3 × I60 + 4 × I120 + 2 × I180)/4

        75 g oral glucose tolerance test
        Roughly determines β-cell insulin secretion functionComprehensively reflects the amount of insulin secretionCannot reflect the peak time of IGT; will misjudge β-cell hyperfunction in IGT population owing to insulin resistance
        Micro-model method: insulin secretion in the first phase [
        • Rayman G.
        • Clark P.
        • Schneider A.
        • Hales C.
        The first phase insulin response to intravenous glucose is highly reproducible.
        ]
        AIR (I0 + I3 + I4 + I5 + I8 + I10)

        The loading amount of glucose is 0.3 g/kg standard body weight, which is prepared into a 50% solution, and the intravenous injection is completed within 2–4 min.
        Represents the acute reaction of islet β-cells stimulated by intravenous high glucose, which is less affected by insulin resistanceEliminates the effects of gastrointestinal hormones and gastric emptying and is suitable for precise research with a small sample sizeWas influenced by insulin clearance variability and blood sugar level. Does not consider the effects of nutrients and glucose-regulated hormones
        Hyperglycemic clamp test [
        • DeFronzo R.
        • Tobin J.
        • Andres R.
        Glucose clamp technique: a method for quantifying insulin secretion and resistance.
        ]
        The first and second phases of

        insulin secretion, maximum insulin secretion
        Represents the true insulin secretion function of islet β-cellAccurately reflects insulin secretion in each phase, including the first phase, second phase, and maximum insulin secretion, which is the golden index for evaluating islet β-cell functionThe operation is complicated and time-consuming and requires professional technicians.
        Arginine stimulation test [
        • Sjostrand M.
        • Carlson K.
        • Arnqvist H.
        • Gudbjörnsdottir S.
        • Landin-Olsson M.
        • Lindmark S.
        • et al.
        Assessment of beta-cell function in young patients with type 2 diabetes: arginine-stimulated insulin secretion may reflect beta-cell reserve.
        ]
        (I2 + I4 + I6)/3 − I0; (C2 + C4 + C6)/3 − C0

        Inject 50 ml of 10% arginine hydrochloride intravenously and complete the injection within 30–60 s. Start timing after the injection.
        Represents the insulin secretion function of islet β-cells under non-glucose stimulation; often used to judge the reserve function of islet β-cells and whether secretagogue use is effective [
        • DeFronzo R.
        • Tobin J.
        • Andres R.
        Glucose clamp technique: a method for quantifying insulin secretion and resistance.
        ]
        Simple, easy to operate, economical, safe, easy to standardize, and repeatableFor T2DM patients with blood glucose >11 mmoL/L, the results of this experiment were biased.
        Glucagon stimulation test [
        • Scheen A.
        • Castillo M.
        • Lefèbvre P.
        Assessment of residual insulin secretion in diabetic patients using the intravenous glucagon stimulatory test: methodological aspects and clinical applications.
        ]
        C6-C0

        Quickly inject 1 mg glucagon intravenously and start timing after injection.
        Represents residual

        islet β-cell function.
        Islet β-cell function assessment for T1DMGlucagon is difficult to obtain.
        Abbreviations: FINS (mU/L)is fasting insulin; Ip (mU/L) is insulin peak; I0 (mU/L) is the basic insulin value; FPG (mmoL/L) is fasting plasma glucose; FCP (pmoL/L) is fasting C-peptide; PG(n) (mmoL/L) is postprandial blood glucose at a specific time, n represents the blood drawing time (minutes); I(n) (mU/L) is insulin at a specific time, n represents the blood drawing time (minutes); G(n) (mmoL/L) is blood glucose at a specific time, n represents the blood drawing time (minutes); AIR is an acute insulin reaction; C(n) (nmoL/L) is the C-peptide at a specific time, n represents the blood drawing time (minutes); NGT is normal glucose tolerance; IGT is impaired glucose tolerance; T1DM is type 1 diabetes; T2DM is type 2 diabetes; -This item is not available.
      Owing to their simplicity, fasting insulin or C-peptide level, ratio of proinsulin to fasting serum insulin and homeostasis model to assess β-cell function (HOMA-β) are generally used to represent islet β-cell function in epidemiological studies. Stimulation tests which can assess islet β-cell function both dynamically and comprehensively are commonly used in clinics. There are two types of insulin or C-peptide release test according to the stimuli: a glucose-stimulated test and a non-glucose-stimulated test. Glucose-stimulated insulin release tests mainly comprise high glucose clamp tests, intravenous glucose tolerance tests (IVGTT), and oral glucose tolerance tests (OGTT). Non-glucose-stimulated insulin release tests mainly include arginine and glucagon tests. Finally, the corresponding indices reflecting β-cell function are calculated using mathematical models and formulas.
      • II.
        Methods for measuring insulin or C-peptide
      There are many methods to measure insulin in humans, including radioimmunoassay (RIA), enzyme-linked immunoassay, chemiluminescence immunoassay, and electrochemiluminescence immunoassay. RIA is a classical manual measurement method, whereas the others are automatic measurement methods. RIA recorded the lowest detection precision, while the other methods had higher precision. Manual operation may be the main factor leading to a large deviation in RIA measurement. The results of insulin measurements in non-RIA are 20-40% lower than those in RIA owing to their high antibody specificity. Although the different methods presented significantly different results, they were still highly correlated. Insulin analogs, anti-insulin antibodies, and heterophile antibodies may cross-react with insulin detection antibodies; however, if the same method is used to evaluate insulin level, this will not result in misjudgment of islet β-cell function, especially for dynamic analysis [
      • Luo X.
      • Lu H.
      • Gao Y.
      • Lu J.
      • Gu C.
      • Jia W.
      Comparative study of serum insulin immunoassays and its clinical significance.
      ].
      The methods for measuring C-peptides in the laboratory include RIA, enzyme-linked immunoassay, chemiluminescence immunoassay, and electrochemiluminescence immunoassay. The ratio of serum C-peptide peak value to the basic value and the time from baseline to peak values are consistent among different methods [
      • Luo X.
      • Lu H.
      • Gao Y.
      • Lu J.
      • Gu C.
      • Jia W.
      Evaluation and clinical significance of four different immunoassays in detection of serum C-peptide.
      ] and thus will not lead to misjudgments in assessing islet β-cell function in T2DM patients.
      • I.
        Assessment method of islet β-cell function based on insulin or C-peptide
      Assessment method of islet β-cell function is shown in Table 1.
      • II.
        Recommendation for clinical methods for detecting islet β-cell function
      Appropriate method selection is the premise for assessment of islet β-cell function. It is necessary to comprehensively consider the purpose of assessment and the sensitivity and specificity of different methods for detecting islet β-cell function during the natural course of T2DM. Hyperglycemic clamp test, IVGTT, and other OGTT can be used to determine the potential damage to β-cell function among high-risk groups whose glucose regulation is still in the normal stage, which is helpful in assessing the risk of diabetes mellitus. Prediabetic patients prefer IVGTT and OGTT combined with insulin release tests to calculate different parameters for assessment. Residual islet β-cell function can be assessed using OGTT parameters, arginine, or glucagon stimulation in patients diagnosed with diabetes mellitus.
      At present, although many indices derived from different stimulation tests are used to represent the changes in the amount and phase of insulin secretion, islet β-cell function can only be “roughly” quantified. It is easy to misunderstand if we want to define a tangent point or normal range for different indices, and the dynamic assessment of islet β-cell function may be more helpful for comparative judgment. All types of assessment methods have advantages and limitations and should therefore be used reasonably. Thus, the appropriate research object and detection indicators should be selected carefully, and statistical analysis should be carefully performed to eliminate the interference of confounding factors. When one method is insufficient to reflect the profile of β-cell function, several tests can be combined according to the research purpose. Accurate, scientific, and comprehensive assessment of islet β-cell function provides a solid foundation for developing individualized treatment plans.

      4. Therapeutic strategies for protecting islet β-cell function

      4.1 Key points

      Measures to protect islet β-cell function include the following:
      • 1.
        Reducing body weight by strengthening lifestyle interventions, weight loss drugs, metabolic surgery.
      • 2.
        Sustainedly near-normoglycemic control and minimization of glycemic variability via treatment with antidiabetic drugs to eliminate glucolipid toxicity over time using short-term intensive insulin therapy.
      • 3.
        Paying attention to the side effects of long-term use of statins, glucocorticoids, immune checkpoint inhibitors, and other drugs on islet β-cell function.
      These measures aim to eliminate glucolipid toxicity, minimize glycemic variability, control chronic low-degree inflammation, improve microcirculation of islet β-cells, and protect islet β-cell function effectively.
      • I.
        Reducing body weight
      Previous studies have evaluated the metabolic effects of different degrees of weight loss (5%, 10%, and 15%) in participants with obesity. The results showed that 5% weight loss was sufficient to improve islet β-cell function and various cardiovascular risk factors, and greater weight loss corresponded to greater improvement in islet β-cell function [
      • Magkos F.
      • Fraterrigo G.
      • Yoshino J.
      • Luecking C.
      • Kirbach K.
      • Kelly S.C.
      • et al.
      Effects of moderate and subsequent progressive weight loss on metabolic function and adipose tissue biology in humans with obesity.
      ]. Measures, including intensive lifestyle interventions, weight-loss drugs, and metabolic surgery, aim to reduce body weight, resulting in reduced ectopic fat deposition in the liver, muscle, and pancreas [
      • Taylor R.
      • Al-Mrabeh A.
      • Zhyzhneuskaya S.
      • Peters C.
      • Barnes A.C.
      • Aribisala B.S.
      • et al.
      Remission of human type 2 diabetes requires decrease in liver and pancreas fat content but is dependent upon capacity for β cell recovery.
      ], optimized glycemic control [
      • Li D.
      • Zou H.
      • Yin P.
      • Li W.
      • He J.
      • Wang S.
      • et al.
      Durability of glycaemic control in type 2 diabetes: a systematic review and meta-analysis for its association with body weight changes.
      ], relieved insulin resistance, and improved islet β-cell function.
      • (1)
        Intensive lifestyle intervention.
      In obese individuals with T2DM, intensive lifestyle intervention results in reduced body weight, relieving glycolipid toxicity [
      • Lim E.L.
      • Hollingsworth K.G.
      • Aribisala B.S.
      • Chen M.J.
      • Mathers J.C.
      • Taylor R.
      Reversal of type 2 diabetes: normalisation of beta cell function in association with decreased pancreas and liver triacylglycerol.
      ]and alleviating chronic low-degree inflammation [
      • Speakman J.R.
      • Mitchell S.E.
      Caloric restriction.
      ,
      • de Lemos E.T.
      • Oliveira J.
      • Pinheiro J.P.
      • Reis F.
      Regular physical exercise as a strategy to improve antioxidant and anti-inflammatory status: benefits in type 2 diabetes mellitus.
      ]. Basic research has shown that long-term caloric restriction reduces apoptosis, trans-differentiation, and dedifferentiation of islet β-cells and induces their redifferentiation [
      • Sheng C.
      • Li F.
      • Lin Z.
      • Zhang M.
      • Yang P.
      • Bu L.
      • et al.
      Reversibility of β-cell-specific transcript factors expression by long-term caloric restriction in db/db mouse.
      ]. The Diabetes Remission Clinical Trial demonstrated that 1 and 2 years after initiation of an intensive lifestyle intervention program for 3–5 months, 46% and 36% of overweight and obese T2DM patients, respectively, achieved clinical remission, compared to 4% and 3% of control participants, respectively. Further analysis showed that the complete remission rate of diabetes reached 86% in patients with weight loss >15 kg. The high glucose clamp test and arginine stimulation test demonstrated that the first phase and maximum insulin secretion rate of participants in the clinical remission group were restored to levels close to those of non-diabetic individuals [
      • Zhyzhneuskaya S.V.
      • Al-Mrabeh A.
      • Peters C.
      • Barnes A.
      • Aribisala B.
      • Hollingsworth K.G.
      • et al.
      Time course of normalization of functional β-cell capacity in the diabetes remission clinical trial after weight loss in type 2 diabetes.
      ]. A very low-calorie diet also effectively improved islet β-cell function in overweight or obese T2DM patients [
      • Al-Mrabeh A.
      • Hollingsworth K.G.
      • Steven S.
      • Taylor R.
      Morphology of the pancreas in type 2 diabetes: effect of weight loss with or without normalisation of insulin secretory capacity.
      ]).
      Exercise improves glucose and lipid metabolism and repairs islet β-cell dysfunction [
      • Cao W.
      • Fang Y.
      • Sun H.
      • Xiang P.
      • Liu C.
      • Wang K.
      Effects of exercise on islet β cell function.
      ]. Newly diagnosed obese T2DM patients demonstrated a 22% increase in HOMA-β levels compared to the baseline value by increasing exercise [
      • Kong X.
      • Cai Y.
      • Xie F.
      Effect of the increased excise intersity on newly diagnosed obese T2DM paitients.
      ]. Moderate-intensity aerobic exercise reduces body weight and visceral fat, thus significantly improving the insulin secretion ability of patients [
      • Dela F.
      • von Linstow M.E.
      • Mikines K.J.
      • Galbo H.
      Physical training may enhance beta-cell function in type 2 diabetes.
      ].
      • (2)
        Weight loss drugs
      After 52 weeks of orlistat add-on, T2DM patients who received lifestyle intervention and metformin treatment lost −5.0% body weight compared to −1.8% in the placebo group, and HOMA-β increased from 50.4% ± 38.5% at baseline to 58.2% ± 37.5% compared to a decrease from 63.5 ± 15.7% at baseline to 53.5 ± 52.3% in the placebo group [
      • Berne C.
      A randomized study of orlistat in combination with a weight management programme in obese patients with type 2 diabetes treated with metformin.
      ]. A small-scale study in China also found that in obese T2DM patients, short-term (12 weeks) addition of orlistat to lifestyle intervention and metformin further reduced body weight, and the amplitude of HOMA-β improvement was 1.8 times that of the control group [
      • Jia X.
      • Xu J.
      • Liu Y.
      • Shen J.
      • Chen Y.
      Clinical trial of orlistat capsules combined with metformin tablets in the treatment of obese type 2 diabetes mellitus.
      ].
      Glucagon-like peptide-1 receptor agonist (GLP-1RA) reduces body weight by inhibiting the feeding center, delaying gastric emptying, and promoting white adipose tissue and adipocyte browning. GLP-1RAs, including liraglutide, dulaglutide, exenatide, and meaglutide, reduced the body weight of obese patients in a dose-dependent manner. With an increase in dosage, both the proportion of weight loss >5% and the amplitude of islet β-cell function improvement increased significantly [
      • Ottney A.
      Glucagon-like peptide-1 receptor agonists for weight loss in adult patients without diabetes.
      ,
      • Ard J.
      • Fitch A.
      • Fruh S.
      • Herman L.
      Weight loss and maintenance related to the mechanism of action of glucagon-like peptide 1 receptor agonists.
      ,
      • O’Neil P.M.
      • Birkenfeld A.L.
      • McGowan B.
      • Mosenzon O.
      • Pedersen S.D.
      • Wharton S.
      • et al.
      Efficacy and safety of semaglutide compared with liraglutide and placebo for weight loss in patients with obesity: a randomised, double-blind, placebo and active controlled, dose-ranging, phase 2 trial.
      ,
      • Anholm C.
      • Kumarathurai P.
      • Pedersen L.R.
      • Nielsen O.W.
      • Kristiansen O.P.
      • Fenger M.
      • et al.
      Liraglutide effects on beta-cell, insulin sensitivity and glucose effectiveness in patients with stable coronary artery disease and newly diagnosed type 2 diabetes.
      ,
      • Mari A.
      • Del Prato S.
      • Ludvik B.
      • Milicevic Z.
      • de la Peña A.
      • Shurzinske L.
      • et al.
      Differential effects of once-weekly glucagon-like peptide-1 receptor agonist dulaglutide and metformin on pancreatic β-cell and insulin sensitivity during a standardized test meal in patients with type 2 diabetes.
      ,
      • Kapitza C.
      • Dahl K.
      • Jacobsen J.B.
      • Axelsen M.B.
      • Flint A.
      Effects of semaglutide on beta cell function and glycaemic control in participants with type 2 diabetes: a randomised, double-blind, placebo-controlled trial.
      ]).
      • (3)
        Metabolic surgery
      According to the guidelines for the surgical treatment of type 2 diabetes in China [
      • Thyroid and Metabolic Surgery Group, Chinese Society of Surgery, Chinese Medical Association, Obesity and Diabetes Department of Chinese College of Surgeons, Chinese Medical Doctor Association
      Guidelines for surgical treatment of obesity and type 2 diabetes in China (2019 full version).
      ], metabolic surgery is recommended as an option to treat T2DM if the patient is aged 16–65 years, with partly residual insulin secretion function and (1) BMI ≥ 32.5 kg/m2 (highly recommended), (2) 27.5 kg/m2 ≤ BMI < 32.5 kg/m2 (recommended), or (3) 25.0 kg/m2 ≤ BMI < 27.5 kg/m2, but difficult to control hyperglycemia after lifestyle intervention and antidiabetic drug treatment, and accompanied by at least two metabolic syndrome components or complications (considered carefully). Weight loss is a major contributor to the benefits of metabolic surgery [
      • Wilding J.
      Weight loss is the major player in bariatric surgery benefits.
      ,
      • Yoshino M.
      • Kayser B.D.
      • Yoshino J.
      • Stein R.I.
      • Reeds D.
      • Eagon J.C.
      • et al.
      Effects of diet versus gastric bypass on metabolic function in diabetes.
      ].
      Metabolic surgery improves islet β-cell function in obese patients with T2DM, which is conducive to clinical remission of diabetes [
      • Zhang Y.
      • Zhu C.
      • Wen X.
      • Wang X.
      • Li L.
      • Rampersad S.
      • et al.
      Laparoscopic sleeve gastrectomy improves body composition and alleviates insulin resistance in obesity related acanthosis nigricans.
      ]). After gastric bypass surgery, fasting and postprandial blood glucose levels were dramatically reduced, compensatory hyperinsulinemia was relieved significantly, and the time to insulin peak was greatly shortened in obese patients with T2DM. Notably, at 3 months and 1 year after gastric bypass surgery, the glucose disposal index of patients increased 5- and 8-fold compared to the preoperational value, respectively, and the peak value of GLP-1 secretion after meals reached 10 times the baseline value [
      • Jørgensen N.B.
      • Jacobsen S.H.
      • Dirksen C.
      • Bojsen-Møller K.N.
      • Naver L.
      • Hvolris L.
      • et al.
      Acute and long-term effects of roux-en-Y gastric bypass on glucose metabolism in subjects with type 2 diabetes and normal glucose tolerance.
      ]. The acute insulin reaction in the IVGTT increased by 770% and 93.5% [
      • Polyzogopoulou E.V.
      • Kalfarentzos F.
      • Vagenakis A.G.
      • Alexandrides T.K.
      Restoration of euglycemia and normal acute insulin response to glucose in obese subjects with type 2 diabetes following bariatric surgery.
      ] at 3 months and 1 year after biliopancreatic diversion with gastric bypass, respectively, in obese patients with T2DM.
      • II.
        Early and sustainedly near-normoglycemic control
      Glucotoxicity and glycemic variability are important causes of islet β-cell dysfunction. Early and sustainedly near-normoglycemic control is beneficial for the long-term protection of islet β-cell function.
      • (1)
        Short-term intensive insulin therapy
      Short-term intensive insulin therapy (IIT) significantly improves islet β-cell function in newly diagnosed T2DM patients, and approximately half of the participants achieved at least 1 year of clinical remission after short-term IIT [
      • Li Y.
      • Xu W.
      • Liao Z.
      • Yao B.
      • Chen X.
      • Huang Z.
      • et al.
      Induction of long-term glycemic control in newly diagnosed type 2 diabetic patients is associated with improvement of beta-cell function.
      ,
      • Wang H.
      • Kuang J.
      • Xu M.
      • Gao Z.
      • Li Q.
      • Liu S.
      • et al.
      Predictors of long-term glycemic remission after 2-week intensive insulin treatment in newly diagnosed type 2 diabetes.
      ,
      • Li Y.
      • Zeng L.
      • Shi L.
      • Zhu D.
      • Zhou Z.
      • Yan L.
      • et al.
      The effects of early intensive therapy on islet beta cell function and long-term glycemia control in newly diagnosed type 2 diabetic patients with different fasting plasma glucose levels.
      ,
      • Liu J.
      • Tuladhar J.
      • Ke W.
      • Deng W.
      • Liu J.
      • Cao X.
      • et al.
      Effect on prognosis in type 2 diabetic patients of the changes of islet β-cell function during short-term continuous subcutaneous insulin infusion.
      ,
      • Liu L.
      • Yang S.
      • Liu J.
      • Li H.
      • Liu J.
      • Cao X.
      • et al.
      Fasting plasma glucose indicates reversibility of the acute insulin response after short-term intensive insulin therapy in patients with various duration of type 2 diabetes.
      ]). Eliminating glucotoxicity maximally and rapidly is the main contributor to IIT improvement of islet β-cell function. Reduction in β-cell metabolic stress and endogenous insulin secretion requirements is closely related to the therapeutic effect of IIT, better glycemic control, lower average blood glucose level in the normal range, and higher inhibition of the increased amplitude of C-peptide (peak C-peptide minus fasting C-peptide) after arginine stimulation during ITT, thereby resulting in better recovery of β-cell function and a higher rate of clinical remission [
      • Liu J.
      • Tuladhar J.
      • Ke W.
      • Deng W.
      • Liu J.
      • Cao X.
      • et al.
      Effect on prognosis in type 2 diabetic patients of the changes of islet β-cell function during short-term continuous subcutaneous insulin infusion.
      ,
      • Liu L.
      • Liu J.
      • Xu L.
      • Ke W.
      • Wan X.
      • Li H.
      • et al.
      Lower mean blood glucose during short-term intensive insulin therapy is associated with long-term glycemic remission in patients with newly diagnosed type 2 diabetes: evidence-based recommendations for standardization.
      ]).
      • (2)
        Non-insulin antidiabetic drugs
      Based on the patient's age, disease course, islet β-cell function, BMI, and other characteristics, combined use of antidiabetic drugs with different mechanisms of action is key to long-term euglycemia control and an important measure to prevent islet β-cell dysfunction. Different non-insulin antidiabetic drugs, including metformin, sulfonylureas, glinides, α-glucosidase inhibitors, thiazolidinediones, and newly developed drugs, indirectly protect islet β-cell function by relieving glucotoxicity. Basic research has shown that mechanisms other than glycemia control contribute to the protection of islet β-cell function by GLP-1RA and sodium-glucose cotransporter-2 inhibitor (SGLT2i).
      • 1.
        GLP-1RA: Animal experiments demonstrated that GLP-1RAs, including liraglutide, dulaglutide, and supaglutide, enhanced insulin secretion in a glucose concentration-dependent manner, promoted the conversion of proinsulin to insulin, induced β-cell proliferation and differentiation, and protected β-cells against apoptotic injury [
        • Ding M.
        • Fang Q.H.
        • Cui Y.T.
        • Shen Q.L.
        • Liu Q.
        • Wang P.H.
        • et al.
        Liraglutide prevents β-cell apoptosis via inactivation of NOX2 and its related signaling pathway.
        ,
        • Kimura T.
        • Obata A.
        • Shimoda M.
        • Hirukawa H.
        • Kanda-Kimura Y.
        • Nogami Y.
        • et al.
        Durability of protective effect of dulaglutide on pancreatic β-cells in diabetic mice: GLP-1 receptor expression is not reduced despite long-term dulaglutide exposure.
        ,
        • Cui Q.
        • Liao Y.
        • Jiang Y.
        • Huang X.
        • Tao W.
        • Zhou Q.
        • et al.
        Novel GLP-1 analog supaglutide improves glucose homeostasis in diabetic monkeys.
        ]. In vitro studies based on human islet cells found that GLP-1RA could restore the insulin secretion ability of β-cells impaired by harmful stimuli, such as palmitic acid [
        • Chowdhury A.I.
        • Bergsten P.
        GLP-1 analogue recovers impaired insulin secretion from human islets treated with palmitate via down-regulation of SOCS2.
        ], high glucose [
        • Park Y.J.
        • Ao Z.
        • Kieffer T.J.
        • Chen H.
        • Safikhan N.
        • Thompson D.M.
        • et al.
        The glucagon-like peptide-1 receptor agonist exenatide restores impaired pro-islet amyloid polypeptide processing in cultured human islets: implications in type 2 diabetes and islet transplantation.
        ] and inflammatory cytokines [
        • Son D.O.
        • Liu W.
        • Li X.
        • Prud’homme G.J.
        • Wang Q.
        Combined effect of GABA and glucagon-like peptide-1 receptor agonist on cytokine-induced apoptosis in pancreatic β-cell line and isolated human islets.
        ], and mildly increase the proliferation rate and number of β-cells in unsorted human islet cells [ [
        • Toso C.
        • McCall M.
        • Emamaullee J.
        • Merani S.
        • Davis J.
        • Edgar R.
        • et al.
        Liraglutide, a long-acting human glucagon-like peptide 1 analogue, improves human islet survival in culture.
        ,
        • Rutti S.
        • Sauter N.S.
        • Bouzakri K.
        • Prazak R.
        • Halban P.A.
        • Donath M.Y.
        In vitro proliferation of adult human beta-cells.
        ]. It should be noted that the human ß-cell proliferation in vivo is rarely observed, in addition, the ß-cell proliferation in response to GLP-1 or GLP-1RAs has only been shown in young animals and probably is irrelevant for human type 2 diabetes occurring at older age. Nevertheless, clinical researches showed that GLP-1RAs improved first- and second-phase insulin secretion function in T2DM patients [
        • Anholm C.
        • Kumarathurai P.
        • Pedersen L.R.
        • Nielsen O.W.
        • Kristiansen O.P.
        • Fenger M.
        • et al.
        Liraglutide effects on beta-cell, insulin sensitivity and glucose effectiveness in patients with stable coronary artery disease and newly diagnosed type 2 diabetes.
        ,
        • Mari A.
        • Del Prato S.
        • Ludvik B.
        • Milicevic Z.
        • de la Peña A.
        • Shurzinske L.
        • et al.
        Differential effects of once-weekly glucagon-like peptide-1 receptor agonist dulaglutide and metformin on pancreatic β-cell and insulin sensitivity during a standardized test meal in patients with type 2 diabetes.
        ,
        • Kapitza C.
        • Dahl K.
        • Jacobsen J.B.
        • Axelsen M.B.
        • Flint A.
        Effects of semaglutide on beta cell function and glycaemic control in participants with type 2 diabetes: a randomised, double-blind, placebo-controlled trial.
        ]. They also protected islet β-cell function by reducing body weight and visceral fat and promoting brown remodeling of white fat [ [
        • Chinese Society of Endocrinology, Chinese Diabetes Society
        Consensus recommendations on utilizing glucagon-like peptide-1(GLP-1) receptor agonists in the treatment of type 2 diabetes mellitus.
        ].
      • 2.
        SGLT2i: SGLT2i plays a hypoglycemic role by inhibiting the reabsorption of glucose by SGLT2 in the proximal convoluted tubule of the kidney and promoting the excretion of urine glucose. This drug indirectly protects islet β-cell function by reducing body weight, lowering blood pressure, and improving insulin sensitivity, thus relieving oxidative stress and inhibiting chronic inflammation in islet β-cells. Furthermore, SGLT2i promoted GLP-1 secretion of α cells, induced the transformation of α cells into β cells, and activated the differentiation of endocrine precursor cells into β cells [
        • Kangli W.
        • Tianpei H.
        • Rui W.
        Protective effects and potential mechanism of the SGLT2 inhibitor on islet β cells.
        ]. A small-scale study found that the incremental area under the C-peptide curve increased by approximately 61% ± 10%, and the insulin secretion/insulin resistance index increased by 112% ± 20% in T2DM patients after two weeks of empagliflozin treatment [
        • Al Jobori H.
        • Daniele G.
        • Adams J.
        • Cersosimo E.
        • Solis-Herrera C.
        • Triplitt C.
        • et al.
        Empagliflozin treatment is associated with improved β-cell function in type 2 diabetes mellitus.
        ]. Randomized controlled trials found that 26 weeks of canagliflozin treatment increased HOMA-β by 9.9% ± 2.0% (100 mg/d) and 20% ± 2.0% (300 mg/d) compared to baseline values, significantly decreased the ratio of proinsulin/insulin, and increased the area under the C-peptide curve in the mixed meal tolerance test [
        • Stenlöf K.
        • Cefalu W.T.
        • Kim K.A.
        • Alba M.
        • Usiskin K.
        • Tong C.
        • et al.
        Efficacy and safety of canagliflozin monotherapy in subjects with type 2 diabetes mellitus inadequately controlled with diet and exercise.
        ]. Dapagliflozin treatment for 24 weeks increased HOMA-β by 17.0% (95% CI: 12.7 - 21.4%) compared to baseline values in T2DM patients [
        • Ekholm E.
        • Hansen L.
        • Johnsson E.
        • Iqbal N.
        • Carlsson B.
        • Chen H.
        • et al.
        Combined treatment with saxagliptin plus dapagliflozin reduces insulin levels by increased insulin clearance and improves β-cell function.
        ].
      • 3.
        Dipeptidyl peptidase IV inhibitor (DPP-4i): DPP-4i plays a role in regulating blood glucose by inhibiting the degradation of endogenous GLP-1, and its protection of islet β-cell function may mainly depend on GLP-1 [
        • Chen X.
        • Lv X.
        • Yang G.
        • Lu D.
        • Piao C.
        • Zhang X.
        • et al.
        Polyethylene glycol loxenatide injections added to metformin effectively improve glycemic control and exhibit favorable safety in type 2 diabetic patients.
        ]. DPP-4 is expressed in human islet β-cells, and its inhibitor MK-0626 could partially prevent the toxic effects of cytokines on nondiabetic β-cells, reduce apoptosis, and improve the ultrastructural defects of β-cells in T2DM patients [
        • Bugliani M.
        • Syed F.
        • Paula F.M.M.
        • Omar B.A.
        • Suleiman M.
        • Mossuto S.
        • et al.
        DPP-4 is expressed in human pancreatic beta cells and its direct inhibition improves beta cell function and survival in type 2 diabetes.
        ]. Clinical studies have found that different types of DPP4i, including sitagliptin, saxagliptin, and linagliptin, reduced the ratio of proinsulin/insulin and increased HOMA-β in T2DM patients [
        • Charbonnel B.
        • Karasik A.
        • Liu J.
        • Wu M.
        • Meininger G.
        Efficacy and safety of the dipeptidyl peptidase-4 inhibitor sitagliptin added to ongoing metformin therapy in patients with type 2 diabetes inadequately controlled with metformin alone.
        ]. Linagliptin plus metformin treatment significantly improved β-cell function in T2DM patients compared to metformin treatment alone [
        • Del Prato S.
        • Barnett A.H.
        • Huisman H.
        • Neubacher D.
        • Woerle H.J.
        • Dugi K.A.
        Effect of linagliptin monotherapy on glycaemic control and markers of β-cell function in patients with inadequately controlled type 2 diabetes: a randomized controlled trial.
        ].
      • 4.
        Glucokinase activator: Dorzagliatin, a novel dual-acting glucokinase activator, improved glycemic control by promoting insulin and GLP-1 secretion and inhibiting glucagon release in a glucose concentration-dependent manner. Animal experiments have shown that Dorzagliatin treatment significantly increased the number of insulin-positive cells and partially restored islet β-cell function [
        • Wang P.
        • Liu H.
        • Chen L.
        • Duan Y.
        • Chen Q.
        • Xi S.
        Effects of a novel glucokinase activator, HMS5552, on glucose metabolism in a rat model of type 2 diabetes mellitus.
        ]. A small-scale exploratory study reported that 28 days of Dorzagliatin treatment resulted in improvement of β-cell function as measured by HOMA-β, which increased by 36.31% and 40.59%, and by dynamic state parameter, ΔC30/ΔG30, which increased by 24.66% and 167.67%, for twice- and once-a-day groups, compared to the baseline value in T2DM patients [
        • Zhu X.X.
        • Zhu D.L.
        • Li X.Y.
        • Li Y.L.
        • Jin X.W.
        • Hu T.X.
        • et al.
        Dorzagliatin (HMS5552), a novel dual-acting glucokinase activator, improves glycaemic control and pancreatic β-cell function in patients with type 2 diabetes: a 28-day treatment study using biomarker-guided patient selection.
        ]. Randomized controlled trials revealed that Dorzagliatin treatment improved the glucose disposal index and homeostasis model assessment of insulin resistance index (HOMA-IR) significantly [
        • Zhu D.
        • Gan S.
        • Liu Y.
        • Ma J.
        • Dong X.
        • Song W.
        • et al.
        Dorzagliatin monotherapy in Chinese patients with type 2 diabetes: a dose-ranging, randomised, double-blind, placebo-controlled, phase 2 study.
        ]. Two registered clinical phase 3 trials have shown that Dorzagliatin treatment significantly improved the islet β-cell function index (HOMA2-β) of patients with newly diagnosed with T2DM and those with metformin treatment incapable of reaching euglycemia [
        • Zhu D.
        • Li X.
        • Ma J.
        • Zeng J.
        • Gan S.
        • Dong X.
        • et al.
        Dorzagliatin in drug-naïve patients with type 2 diabetes: a randomized, double-blind, placebo-controlled phase 3 trial.
        ,
        • Yang W.
        • Zhu D.
        • Gan S.
        • Dong X.
        • Su J.
        • Li W.
        • et al.
        Dorzagliatin add-on therapy to metformin in patients with type 2 diabetes: a randomized, double-blind, placebo-controlled phase 3 trial.
        ].
      • 5.
        Peroxisome proliferator-activated receptor (PPAR) agonist: PPAR includes three subtypes, namely α, γ, and δ. PPARγ participates directly in the regulation of islet β-cell function. In vitro studies have shown that PPARγ activation could reduce the apoptosis of β-cells induced by high glucose [
        • Zhang X.
        • Tong N.
        • Li Y.
        Concurrently using rosiglitazone prevents glucosamine-induced islet beta-cell apoptosis and dysfunction.
        ], and PPARδ activation could alleviate mitochondrial swelling in β-cells induced by palmitic acid and reduce β-cell apoptosis [
        • Wan J.
        • Jiang L.
        • Lü Q.
        • Ke L.
        • Li X.
        • Tong N.
        Activation of PPARdelta up-regulates fatty acid oxidation and energy uncoupling genes of mitochondria and reduces palmitate-induced apoptosis in pancreatic beta-cells.
        ]. Animal studies have reported that Chiglitazar sodium, a pan-agonist of PPAR, inhibited islet cell fibrosis, reduced lipid deposition in islets, and increased islet volume effectively [
        • Li P.P.
        • Shan S.
        • Chen Y.T.
        • Ning Z.Q.
        • Sun S.J.
        • Liu Q.
        • et al.
        The PPARalpha/gamma dual agonist chiglitazar improves insulin resistance and dyslipidemia in MSG obese rats.
        ]. The pan-agonist of PPAR was superior to the PPARγ agonist in terms of clinical efficacy because it exhibited both insulin-sensitizing and lipid-lowering effects. Two randomized controlled trials demonstrated that Chiglitazar sodium significantly reduced fasting plasma insulin, HOMA-IR, and free fatty acid levels, and increased HOMA-β compared to placebo or sitagliptin in T2DM patients [
        • Ji L.
        • Song W.
        • Fang H.
        • Li W.
        • Geng J.
        • Wang Y.
        • et al.
        Efficacy and safety of chiglitazar, a novel peroxisome proliferator-activated receptor pan-agonist, in patients with type 2 diabetes: a randomized, double-blind, placebo-controlled, phase 3 trial (CMAP).
        ,
        • Jia W.
        • Ma J.
        • Miao H.
        • Wang C.
        • Wang X.
        • Li Q.
        • et al.
        Chiglitazar monotherapy with sitagliptin as an active comparator in patients with type 2 diabetes: a randomized, double-blind, phase 3 trial (CMAS).
        ].
      III. Alternative protection strategies
      Relieving chronic nonspecific low-level inflammation, blocking the local renin-angiotensin-aldosterone system, and improving microcirculation in the islets are helpful strategies in protecting islet β-cell function.
      Short-term treatment with anakinra, a recombinant human lL-1 receptor antagonist, decreased the proinsulin/insulin ratio and significantly increased the secretion of C-peptide in T2DM patients; after 39 weeks of anakinra withdrawal, β-cell function continued to improve to some extent [
      • Larsen C.M.
      • Faulenbach M.
      • Vaag A.
      • Ehses J.A.
      • Donath M.Y.
      • Mandrup-Poulsen T.
      Sustained effects of interleukin-1 receptor antagonist treatment in type 2 diabetes.
      ]. Another clinical study demonstrated that first-phase insulin secretion in patients with impaired glucose tolerance increased by 20% after 4 weeks of anakinra treatment [
      • van Poppel P.C.
      • van Asseldonk E.J.
      • Holst J.J.
      • Vilsbøll T.
      • Netea M.G.
      • Tack C.J.
      The interleukin-1 receptor antagonist anakinra improves first-phase insulin secretion and insulinogenic index in subjects with impaired glucose tolerance.
      ]. A meta-analysis showed that canakinumab, an anti-IL-1β antibody, increased the 4-h postprandial C-peptide area under the curve and slightly increased HOMA-β in T2DM patients [
      • Huang J.
      • Yang Y.
      • Hu R.
      • Chen L.
      Anti-interleukin-1 therapy has mild hypoglycaemic effect in type 2 diabetes.
      ]. Renin-angiotensin II (Ang II) and type 1 Ang II receptor (AT1) are widely distributed in the exocrine and endocrine secretion areas of the human pancreas and play roles in maintaining the physiological function of the pancreas [
      • Tahmasebi M.
      • Puddefoot J.R.
      • Inwang E.R.
      • Vinson G.P.
      The tissue renin-angiotensin system in human pancreas.
      ]. High levels of Ang II enhanced oxidative stress caused by hyperglycemia and lipidemia and induced apoptosis of β-cells. Aldosterone elicited β-cell dysfunction through a mineralocorticoid receptor-independent mechanism. Valsartan, an action blocker of Ang II, improved the insulin secretion ability of islet β-cells in T2DM patients [
      • Luther J.M.
      • Brown N.J.
      The renin-angiotensin-aldosterone system and glucose homeostasis.
      ,
      • van der Zijl N.J.
      • Moors C.C.
      • Goossens G.H.
      • Hermans M.M.
      • Blaak E.E.
      • Diamant M.
      Valsartan improves {beta}-cell function and insulin sensitivity in subjects with impaired glucose metabolism: a randomized controlled trial.
      ]. In addition, animal experiments showed that recombinant human tissue kallikrein-1, a drug for improving microcirculation, significantly increased fasting C-peptide levels in autoimmune diabetic mice [
      • Maneva-Radicheva L.
      • Amatya C.
      • Parker C.
      • Ellefson J.
      • Radichev I.
      • Raghavan A.
      • et al.
      Autoimmune diabetes is suppressed by treatment with recombinant human tissue Kallikrein-1.
      ] and improved islet β-cell function in obese diabetic mice [
      • Kolodka T.
      • Charles M.L.
      • Raghavan A.
      • Radichev I.A.
      • Amatya C.
      • Ellefson J.
      • et al.
      Preclinical characterization of recombinant human tissue kallikrein-1 as a novel treatment for type 2 diabetes mellitus.
      ].
      The possible damage to islet β-cell function due to some drugs in clinical practice should be noted, and the risk-benefit ratio of these drugs should be carefully weighed.
      • 1.
        Glucocorticoid (GC): Insulin resistance induced by short-term systemic application of GC is often accompanied by the compensatory proliferation of β-cells. Long-term GC treatment may directly interfere with the expression of molecules necessary for glucose metabolism, decrease the transcription of insulin genes, enhance α-adrenergic signals to β-cells, and increase β-cell apoptosis [
        • Fichna M.
        • Fichna P.
        Glucocorticoids and beta-cell function.
        ].
      • 2.
        Statins: Long-term use of statins is associated with an increased risk of developing diabetes mellitus. This side effect does not vary with the water or fat solubility, half-life, and metabolic enzymes of statins but is closely related to treatment intensity of statins [
        • Shah R.V.
        • Goldfine A.B.
        Statins and risk of new-onset diabetes mellitus.
        ,
        • Preiss D.
        • Seshasai S.R.
        • Welsh P.
        • Murphy S.A.
        • Ho J.E.
        • Waters D.D.
        • et al.
        Risk of incident diabetes with intensive-dose compared with moderate-dose statin therapy: a meta-analysis.
        ,
        • Rajpathak S.N.
        • Kumbhani D.J.
        • Crandall J.
        • Barzilai N.
        • Alderman M.
        • Ridker P.M.
        Statin therapy and risk of developing type 2 diabetes: a meta-analysis.
        ]. Statins inhibit the influx of calcium ions induced by glucose, prevent the synthesis of coenzyme Q10 in β-cells, and decrease the intracellular concentration of isoprene, thus inducing insulin secretion dysfunction [
        • Galicia-Garcia U.
        • Jebari S.
        • Larrea-Sebal A.
        • Uribe K.B.
        • Siddiqi H.
        • Ostolaza H.
        • et al.
        Statin treatment-induced development of type 2 diabetes: from clinical evidence to mechanistic insights.
        ].
      • 3.
        Immune checkpoint inhibitors (ICPis): Clinically applied ICPis mainly include programmed death receptor 1 (PD-1) inhibitors, programmed death ligand 1 (PD-L1) inhibitors, and cytotoxic T lymphocyte-associated antigen (CTLA-4) inhibitors. ICPis are used for the treatment of unresectable or metastatic melanoma, metastatic non-small cell lung cancer, metastatic small cell lung cancer, advanced renal cell carcinoma, refractory classical Hodgkin's lymphoma, and other tumors. The incidence of diabetes mellitus associated with ICPis therapy (mainly with PD-1/PD-L1 inhibitors) is approximately 1%. Most patients have one or more islet autoantibody. The onset of diabetic ketoacidosis is attributed to rapid and complete deterioration of functional islet β-cell mass caused by ICPis [
        • Stamatouli A.M.
        • Quandt Z.
        • Perdigoto A.L.
        • Clark P.L.
        • Kluger H.
        • Weiss S.A.
        • et al.
        Collateral damage: insulin-dependent diabetes induced with checkpoint inhibitors.
        ].
      • 4.
        Thiazide diuretics: Thiazide diuretics cause potassium loss in cells, thus disturbing the polarization state of β-cells and leading to insulin secretion dysfunction. Low concentrations of hydrochlorothiazide directly inhibited glucose-stimulated insulin secretion in isolated islets [
        • Sandström P.E.
        Inhibition by hydrochlorothiazide of insulin release and calcium influx in mouse pancreatic beta-cells.
        ].
      • 5.
        Beta receptor blockers: Beta receptor blockers can inhibit insulin secretion by blocking beta 2 receptor signal transmission in β-cells [
        • Lacey R.J.
        • Cable H.C.
        • James R.F.
        • London N.J.
        • Scarpello J.H.
        • Morgan N.G.
        Concentration-dependent effects of adrenaline on the profile of insulin secretion from isolated human islets of Langerhans.
        ].

      5. Summary and prospects

      The onset and progression of T2DM are closely related to islet β-cell dysfunction, and accurate assessment of islet β-cell function contributes to the typing diagnosis of diabetes mellitus and development of individualized treatment regimens. Although many methods can be used to assess islet β-cell function, each method has its own merits and drawbacks, and a single method cannot be a “one size fits all” approach for different populations. Thus, these methods can be used alone or in combination depending on the purpose of β-cell function assessment and the subject's glucose tolerance stage. Antidiabetic regimens and some concomitant drugs have different effects on islet β-cell function. Early intensive insulin therapy and metabolic surgery substantially improved islet β-cell function in newly diagnosed T2DM patients and obese individuals with T2DM, respectively; however, it is still difficult to completely reverse T2DM, and the beneficial effects on islet β-cell function gradually disappear after treatment withdrawal. In the future, it will be necessary to establish a standardized assessment method for islet β-cell function and clarify the roles of related genes and signaling pathways in islet β-cell dysfunction. Precise intervention measures should be implemented for maximal restoration of islet functional β-cells by stimulating endogenous β-cell regeneration.

      Funding

      None.

      Author contributions

      Jian Zhu, Junfeng Han, Liehua Liu, Yu Liu, Wen Xu, Xiaomu Li, Lin Yang, Yong Gu, and Wei Tang wrote this manuscript. Jian Zhu and Junfeng Han revised this manuscript. Jianhua MA, Dalong Zhu, and Yiming Mu conceived and supervised the writing of this manuscript. All authors have discussed, read, and approved the manuscript.

      Declaration of Competing Interest

      All authors declare no conflict of interests.

      References

        • Chinese Diabetes Society
        Guideline for the prevention and treatment of type 2 diabetes mellitus in China (2020 edition).
        Chin J Diabetes Mellitus. 2021; 13: 315-409
        • Gao Z.
        • Yan W.
        • Fang Z.
        • Zhang Z.
        • Yuan L.
        • Wang X.
        • et al.
        Annual decline in β-cell function in patients with type 2 diabetes in China.
        Diabetes Metab Res Rev. 2021; 37e3364
        • Xiang K.
        Special types of diabetes mellitus. [M].
        Shanghai scientific & technical publishes, Shanghai2011
        • Hannon T.S.
        • Kahn S.E.
        • Utzschneider K.M.
        • Buchanan T.A.
        • Nadeau K.J.
        • Zeitler P.S.
        • et al.
        Review of methods for measuring β-cell function: design considerations from the restoring insulin secretion (RISE) consortium.
        Diabetes Obes Metab. 2018; 20: 14-24
        • Eisenbarth G.S.
        Type I diabetes mellitus. A chronic autoimmune disease.
        N Engl J Med. 1986; 314: 1360-1368
        • DeFronzo R.A.
        Pathogenesis of type 2 diabetes mellitus.
        Med Clin North Am. 2004; 88: ix
        • Yang Y.
        • Kim J.W.
        • Park H.S.
        • Lee E.Y.
        • Yoon K.H.
        Pancreatic stellate cells in the islets as a novel target to preserve the pancreatic β-cell mass and function.
        J Diabetes Investig. 2020; 11: 268-280
        • Ying W.
        • Fu W.
        • Lee Y.S.
        • Olefsky J.M.
        The role of macrophages in obesity-associated islet inflammation and β-cell abnormalities.
        Nat Rev Endocrinol. 2020; 16: 81-90
        • Weir G.C.
        • Gaglia J.
        • Bonner-Weir S.
        Inadequate β-cell mass is essential for the pathogenesis of type 2 diabetes.
        Lancet Diabetes Endocrinol. 2020; 8: 249-256
        • Talchai C.
        • Xuan S.
        • Lin H.V.
        • Sussel L.
        • Accili D.
        Pancreatic β cell dedifferentiation as a mechanism of diabetic β cell failure.
        Cell. 2012; 150: 1223-1234
        • Tian Q.
        • Hong T.
        The pathophysiology of insulin secretory dysfunction and its therapeutic strategy.
        Chin J Diabetes. 2012; 20: 872-874
        • Weng J.
        • Li Y.
        • Xu W.
        • Shi L.
        • Zhang Q.
        • Zhu D.
        • et al.
        Effect of intensive insulin therapy on beta-cell function and glycaemic control in patients with newly diagnosed type 2 diabetes: a multicentre randomised parallel-group trial.
        Lancet. 2008; 371: 1753-1760
        • Zhu X.
        • Yan H.
        • Chang X.
        • Xia M.
        • Wang L.
        • Bian H.
        • et al.
        The value of arginine stimulation test in evaluating the first-phase insulin secretion and its guiding role for the treatment and its guiding role for the treatment of type 2diabetes mellitus.
        Chin J Clin Med. 2017; 24: 548-553
        • Bo S.
        • Cavallo-Perin P.
        • Gentile L.
        • Repetti E.
        • Pagano G.
        Relationship of residual beta-cell function, metabolic control and chronic complications in type 2 diabetes mellitus.
        Acta Diabetol. 2000; 37: 125-129
        • Tung T.H.
        • Shih H.C.
        • Tsai S.T.
        • Chou P.
        • Chen S.J.
        • Lee F.L.
        • et al.
        A community-based study of the relationship between insulin resistance/beta-cell dysfunction and diabetic retinopathy among type II diabetics in Kinmen.
        Taiwan Ophthalmic Epidemiol. 2007; 14: 148-154
        • Chen T.
        • Xu F.
        • Su J.B.
        • Wang X.Q.
        • Chen J.F.
        • Wu G.
        • et al.
        Glycemic variability in relation to oral disposition index in the subjects with different stages of glucose tolerance.
        Diabetol Metab Syndr. 2013; 5: 38
        • Fang F.S.
        • Cheng X.L.
        • Gong Y.P.
        • Wang L.C.
        • Li L.
        • Li J.
        • et al.
        Association between glycemic indices and beta cell function in patients with newly diagnosed type 2 diabetes.
        Curr Med Res Opin. 2014; 30: 1437-1440
        • Luo X.
        • Lu H.
        • Gao Y.
        • Lu J.
        • Gu C.
        • Jia W.
        Comparative study of serum insulin immunoassays and its clinical significance.
        Chin J Endocrinol Metab. 2009; 25: 622-625
        • Luo X.
        • Lu H.
        • Gao Y.
        • Lu J.
        • Gu C.
        • Jia W.
        Evaluation and clinical significance of four different immunoassays in detection of serum C-peptide.
        Label Immunoassays Clin Med. 2009; 16: 175-178
        • Matthews D.
        • Hosker J.
        • Rudenski A.
        • Naylor B.
        • Treacher D.
        • Turner R.
        Homeostasis model assessment: insulin resistance and beta-cell function from fasting plasma glucose and insulin concentrations in man.
        Diabetologia. 1985; 28: 412-419
        • Zhou Z.G.
        • Qi H.Y.
        • Chen X.Y.
        • Huang G.
        Replacement of insulin by fasting C-peptide in modified homeostasis model assessment to evaluate insulin resistance and islet βcell function.
        J Cent South Univ(Med Sci). 2004; 29: 419-423
        • Li G.W.
        • Yang W.Y.
        • Jiang Y.Y.
        • Hu Y.H.
        The possibility of (FINS ×FPG) / (PG2h + PG1h - 2FPG) being taken as an index of pancreaticβ cell insulin secretion in a population-based study.
        Chin J Intern Med. 2000; 39: 234-238
        • Pacini G.
        • Mari A.
        Methods for clinical assessment of insulin sensitivity and beta-cell function.
        Best Pract Res Clin Endocrinol Metab. 2003; 17: 305-322
        • Rayman G.
        • Clark P.
        • Schneider A.
        • Hales C.
        The first phase insulin response to intravenous glucose is highly reproducible.
        Diabetologia. 1990; 33: 631-634
        • DeFronzo R.
        • Tobin J.
        • Andres R.
        Glucose clamp technique: a method for quantifying insulin secretion and resistance.
        Am J Physiol. 1979; 237: E214-E223
        • Sjostrand M.
        • Carlson K.
        • Arnqvist H.
        • Gudbjörnsdottir S.
        • Landin-Olsson M.
        • Lindmark S.
        • et al.
        Assessment of beta-cell function in young patients with type 2 diabetes: arginine-stimulated insulin secretion may reflect beta-cell reserve.
        J Intern Med. 2014; 275: 39-48
        • Scheen A.
        • Castillo M.
        • Lefèbvre P.
        Assessment of residual insulin secretion in diabetic patients using the intravenous glucagon stimulatory test: methodological aspects and clinical applications.
        Diabetes Metab. 1996; 22: 397-406
        • Magkos F.
        • Fraterrigo G.
        • Yoshino J.
        • Luecking C.
        • Kirbach K.
        • Kelly S.C.
        • et al.
        Effects of moderate and subsequent progressive weight loss on metabolic function and adipose tissue biology in humans with obesity.
        Cell Metab. 2016; 23: 591-601
        • Taylor R.
        • Al-Mrabeh A.
        • Zhyzhneuskaya S.
        • Peters C.
        • Barnes A.C.
        • Aribisala B.S.
        • et al.
        Remission of human type 2 diabetes requires decrease in liver and pancreas fat content but is dependent upon capacity for β cell recovery.
        Cell Metab. 2018; 28e3
        • Li D.
        • Zou H.
        • Yin P.
        • Li W.
        • He J.
        • Wang S.
        • et al.
        Durability of glycaemic control in type 2 diabetes: a systematic review and meta-analysis for its association with body weight changes.
        Diabetes Obes Metab. 2021; 23: 208-217
        • Lim E.L.
        • Hollingsworth K.G.
        • Aribisala B.S.
        • Chen M.J.
        • Mathers J.C.
        • Taylor R.
        Reversal of type 2 diabetes: normalisation of beta cell function in association with decreased pancreas and liver triacylglycerol.
        Diabetologia. 2011; 54: 2506-2514
        • Speakman J.R.
        • Mitchell S.E.
        Caloric restriction.
        Mol Aspects Med. 2011; 32: 159-221
        • de Lemos E.T.
        • Oliveira J.
        • Pinheiro J.P.
        • Reis F.
        Regular physical exercise as a strategy to improve antioxidant and anti-inflammatory status: benefits in type 2 diabetes mellitus.
        Oxid Med Cell Longev. 2012; 2012741545
        • Sheng C.
        • Li F.
        • Lin Z.
        • Zhang M.
        • Yang P.
        • Bu L.
        • et al.
        Reversibility of β-cell-specific transcript factors expression by long-term caloric restriction in db/db mouse.
        J Diabetes Res. 2016; 2016: 6035046
        • Zhyzhneuskaya S.V.
        • Al-Mrabeh A.
        • Peters C.
        • Barnes A.
        • Aribisala B.
        • Hollingsworth K.G.
        • et al.
        Time course of normalization of functional β-cell capacity in the diabetes remission clinical trial after weight loss in type 2 diabetes.
        Diabetes Care. 2020; 43: 813-820
        • Al-Mrabeh A.
        • Hollingsworth K.G.
        • Steven S.
        • Taylor R.
        Morphology of the pancreas in type 2 diabetes: effect of weight loss with or without normalisation of insulin secretory capacity.
        Diabetologia. 2016; 59: 1753-1759
        • Cao W.
        • Fang Y.
        • Sun H.
        • Xiang P.
        • Liu C.
        • Wang K.
        Effects of exercise on islet β cell function.
        Int J Endocrinol Metab. 2020; 40: 27-30
        • Kong X.
        • Cai Y.
        • Xie F.
        Effect of the increased excise intersity on newly diagnosed obese T2DM paitients.
        Chin J Diabetes. 2012; 20: 663-666
        • Dela F.
        • von Linstow M.E.
        • Mikines K.J.
        • Galbo H.
        Physical training may enhance beta-cell function in type 2 diabetes.
        Am J Physiol Endocrinol Metab. 2004; 287: E1024-E1031
        • Berne C.
        A randomized study of orlistat in combination with a weight management programme in obese patients with type 2 diabetes treated with metformin.
        Diabet Med. 2005; 22: 612-618
        • Jia X.
        • Xu J.
        • Liu Y.
        • Shen J.
        • Chen Y.
        Clinical trial of orlistat capsules combined with metformin tablets in the treatment of obese type 2 diabetes mellitus.
        Chin J Clin Pharmacol. 2020; 36: 756-759
        • Ottney A.
        Glucagon-like peptide-1 receptor agonists for weight loss in adult patients without diabetes.
        Am J Health Syst Pharm. 2013; 70: 2097-2103
        • Ard J.
        • Fitch A.
        • Fruh S.
        • Herman L.
        Weight loss and maintenance related to the mechanism of action of glucagon-like peptide 1 receptor agonists.
        Adv Ther. 2021; 38: 2821-2839
        • O’Neil P.M.
        • Birkenfeld A.L.
        • McGowan B.
        • Mosenzon O.
        • Pedersen S.D.
        • Wharton S.
        • et al.
        Efficacy and safety of semaglutide compared with liraglutide and placebo for weight loss in patients with obesity: a randomised, double-blind, placebo and active controlled, dose-ranging, phase 2 trial.
        Lancet. 2018; 392: 637-649
        • Anholm C.
        • Kumarathurai P.
        • Pedersen L.R.
        • Nielsen O.W.
        • Kristiansen O.P.
        • Fenger M.
        • et al.
        Liraglutide effects on beta-cell, insulin sensitivity and glucose effectiveness in patients with stable coronary artery disease and newly diagnosed type 2 diabetes.
        Diabetes Obes Metab. 2017; 19: 850-857
        • Mari A.
        • Del Prato S.
        • Ludvik B.
        • Milicevic Z.
        • de la Peña A.
        • Shurzinske L.
        • et al.
        Differential effects of once-weekly glucagon-like peptide-1 receptor agonist dulaglutide and metformin on pancreatic β-cell and insulin sensitivity during a standardized test meal in patients with type 2 diabetes.
        Diabetes Obes Metab. 2016; 18: 834-839
        • Kapitza C.
        • Dahl K.
        • Jacobsen J.B.
        • Axelsen M.B.
        • Flint A.
        Effects of semaglutide on beta cell function and glycaemic control in participants with type 2 diabetes: a randomised, double-blind, placebo-controlled trial.
        Diabetologia. 2017; 60: 1390-1399
        • Thyroid and Metabolic Surgery Group, Chinese Society of Surgery, Chinese Medical Association, Obesity and Diabetes Department of Chinese College of Surgeons, Chinese Medical Doctor Association
        Guidelines for surgical treatment of obesity and type 2 diabetes in China (2019 full version).
        Chin J Pract Surg. 2019; : 301-306
        • Wilding J.
        Weight loss is the major player in bariatric surgery benefits.
        Nat Med. 2020; 26: 1678-1679
        • Yoshino M.
        • Kayser B.D.
        • Yoshino J.
        • Stein R.I.
        • Reeds D.
        • Eagon J.C.
        • et al.
        Effects of diet versus gastric bypass on metabolic function in diabetes.
        N Engl J Med. 2020; 383: 721-732
        • Zhang Y.
        • Zhu C.
        • Wen X.
        • Wang X.
        • Li L.
        • Rampersad S.
        • et al.
        Laparoscopic sleeve gastrectomy improves body composition and alleviates insulin resistance in obesity related acanthosis nigricans.
        Lipids Health Dis. 2017; 16: 209
        • Jørgensen N.B.
        • Jacobsen S.H.
        • Dirksen C.
        • Bojsen-Møller K.N.
        • Naver L.
        • Hvolris L.
        • et al.
        Acute and long-term effects of roux-en-Y gastric bypass on glucose metabolism in subjects with type 2 diabetes and normal glucose tolerance.
        Am J Physiol Endocrinol Metab. 2012; 303: E122-E131
        • Polyzogopoulou E.V.
        • Kalfarentzos F.
        • Vagenakis A.G.
        • Alexandrides T.K.
        Restoration of euglycemia and normal acute insulin response to glucose in obese subjects with type 2 diabetes following bariatric surgery.
        Diabetes. 2003; 52: 1098-1103
        • Li Y.
        • Xu W.
        • Liao Z.
        • Yao B.
        • Chen X.
        • Huang Z.
        • et al.
        Induction of long-term glycemic control in newly diagnosed type 2 diabetic patients is associated with improvement of beta-cell function.
        Diabetes Care. 2004; 27: 2597-2602
        • Wang H.
        • Kuang J.
        • Xu M.
        • Gao Z.
        • Li Q.
        • Liu S.
        • et al.
        Predictors of long-term glycemic remission after 2-week intensive insulin treatment in newly diagnosed type 2 diabetes.
        J Clin Endocrinol Metab. 2019; 104: 2153-2162
        • Li Y.
        • Zeng L.
        • Shi L.
        • Zhu D.
        • Zhou Z.
        • Yan L.
        • et al.
        The effects of early intensive therapy on islet beta cell function and long-term glycemia control in newly diagnosed type 2 diabetic patients with different fasting plasma glucose levels.
        Chin J Intern Med. 2010; 49: 9-13
        • Liu J.
        • Tuladhar J.
        • Ke W.
        • Deng W.
        • Liu J.
        • Cao X.
        • et al.
        Effect on prognosis in type 2 diabetic patients of the changes of islet β-cell function during short-term continuous subcutaneous insulin infusion.
        Chin J Diabetes Mellitus. 2014; 6: 293-298
        • Liu L.
        • Yang S.
        • Liu J.
        • Li H.
        • Liu J.
        • Cao X.
        • et al.
        Fasting plasma glucose indicates reversibility of the acute insulin response after short-term intensive insulin therapy in patients with various duration of type 2 diabetes.
        J Diabetes Res. 2018; 2018: 9423965
        • Liu L.
        • Liu J.
        • Xu L.
        • Ke W.
        • Wan X.
        • Li H.
        • et al.
        Lower mean blood glucose during short-term intensive insulin therapy is associated with long-term glycemic remission in patients with newly diagnosed type 2 diabetes: evidence-based recommendations for standardization.
        J Diabetes Investig. 2018; 9: 908-916
        • Ding M.
        • Fang Q.H.
        • Cui Y.T.
        • Shen Q.L.
        • Liu Q.
        • Wang P.H.
        • et al.
        Liraglutide prevents β-cell apoptosis via inactivation of NOX2 and its related signaling pathway.
        J Diabetes Complications. 2019; 33: 267-277
        • Kimura T.
        • Obata A.
        • Shimoda M.
        • Hirukawa H.
        • Kanda-Kimura Y.
        • Nogami Y.
        • et al.
        Durability of protective effect of dulaglutide on pancreatic β-cells in diabetic mice: GLP-1 receptor expression is not reduced despite long-term dulaglutide exposure.
        Diabetes Metab. 2018; 44: 250-260
        • Cui Q.
        • Liao Y.
        • Jiang Y.
        • Huang X.
        • Tao W.
        • Zhou Q.
        • et al.
        Novel GLP-1 analog supaglutide improves glucose homeostasis in diabetic monkeys.
        J Endocrinol. 2021; 248: 145-154
        • Chowdhury A.I.
        • Bergsten P.
        GLP-1 analogue recovers impaired insulin secretion from human islets treated with palmitate via down-regulation of SOCS2.
        Mol Cell Endocrinol. 2017; 439: 194-202
        • Park Y.J.
        • Ao Z.
        • Kieffer T.J.
        • Chen H.
        • Safikhan N.
        • Thompson D.M.
        • et al.
        The glucagon-like peptide-1 receptor agonist exenatide restores impaired pro-islet amyloid polypeptide processing in cultured human islets: implications in type 2 diabetes and islet transplantation.
        Diabetologia. 2013; 56: 508-519
        • Son D.O.
        • Liu W.
        • Li X.
        • Prud’homme G.J.
        • Wang Q.
        Combined effect of GABA and glucagon-like peptide-1 receptor agonist on cytokine-induced apoptosis in pancreatic β-cell line and isolated human islets.
        J Diabetes. 2019; 11: 563-572
        • Toso C.
        • McCall M.
        • Emamaullee J.
        • Merani S.
        • Davis J.
        • Edgar R.
        • et al.
        Liraglutide, a long-acting human glucagon-like peptide 1 analogue, improves human islet survival in culture.
        Transpl Int. 2010; 23: 259-265
        • Rutti S.
        • Sauter N.S.
        • Bouzakri K.
        • Prazak R.
        • Halban P.A.
        • Donath M.Y.
        In vitro proliferation of adult human beta-cells.
        PLoS One. 2012; 7e35801
        • Chinese Society of Endocrinology, Chinese Diabetes Society
        Consensus recommendations on utilizing glucagon-like peptide-1(GLP-1) receptor agonists in the treatment of type 2 diabetes mellitus.
        Chin J Intern Med. 2020; 59: 836-846
        • Kangli W.
        • Tianpei H.
        • Rui W.
        Protective effects and potential mechanism of the SGLT2 inhibitor on islet β cells.
        Chin J Endocrinol Metab. 2021; 37: 297-300
        • Al Jobori H.
        • Daniele G.
        • Adams J.
        • Cersosimo E.
        • Solis-Herrera C.
        • Triplitt C.
        • et al.
        Empagliflozin treatment is associated with improved β-cell function in type 2 diabetes mellitus.
        J Clin Endocrinol Metab. 2018; 103: 1402-1407
        • Stenlöf K.
        • Cefalu W.T.
        • Kim K.A.
        • Alba M.
        • Usiskin K.
        • Tong C.
        • et al.
        Efficacy and safety of canagliflozin monotherapy in subjects with type 2 diabetes mellitus inadequately controlled with diet and exercise.
        Diabetes Obes Metab. 2013; 15: 372-382
        • Ekholm E.
        • Hansen L.
        • Johnsson E.
        • Iqbal N.
        • Carlsson B.
        • Chen H.
        • et al.
        Combined treatment with saxagliptin plus dapagliflozin reduces insulin levels by increased insulin clearance and improves β-cell function.
        Endocr Pract. 2017; 23: 258-265
        • Chen X.
        • Lv X.
        • Yang G.
        • Lu D.
        • Piao C.
        • Zhang X.
        • et al.
        Polyethylene glycol loxenatide injections added to metformin effectively improve glycemic control and exhibit favorable safety in type 2 diabetic patients.
        J Diabetes. 2017; 9: 158-167
        • Bugliani M.
        • Syed F.
        • Paula F.M.M.
        • Omar B.A.
        • Suleiman M.
        • Mossuto S.
        • et al.
        DPP-4 is expressed in human pancreatic beta cells and its direct inhibition improves beta cell function and survival in type 2 diabetes.
        Mol Cell Endocrinol. 2018; 473: 186-193
        • Charbonnel B.
        • Karasik A.
        • Liu J.
        • Wu M.
        • Meininger G.
        Efficacy and safety of the dipeptidyl peptidase-4 inhibitor sitagliptin added to ongoing metformin therapy in patients with type 2 diabetes inadequately controlled with metformin alone.
        Diabetes Care. 2006; 29: 2638-2643
        • Del Prato S.
        • Barnett A.H.
        • Huisman H.
        • Neubacher D.
        • Woerle H.J.
        • Dugi K.A.
        Effect of linagliptin monotherapy on glycaemic control and markers of β-cell function in patients with inadequately controlled type 2 diabetes: a randomized controlled trial.
        Diabetes Obes Metab. 2011; 13: 258-267
        • Wang P.
        • Liu H.
        • Chen L.
        • Duan Y.
        • Chen Q.
        • Xi S.
        Effects of a novel glucokinase activator, HMS5552, on glucose metabolism in a rat model of type 2 diabetes mellitus.
        J Diabetes Res. 2017; 2017: 5812607
        • Zhu X.X.
        • Zhu D.L.
        • Li X.Y.
        • Li Y.L.
        • Jin X.W.
        • Hu T.X.
        • et al.
        Dorzagliatin (HMS5552), a novel dual-acting glucokinase activator, improves glycaemic control and pancreatic β-cell function in patients with type 2 diabetes: a 28-day treatment study using biomarker-guided patient selection.
        Diabetes Obes Metab. 2018; 20: 2113-2120
        • Zhu D.
        • Gan S.
        • Liu Y.
        • Ma J.
        • Dong X.
        • Song W.
        • et al.
        Dorzagliatin monotherapy in Chinese patients with type 2 diabetes: a dose-ranging, randomised, double-blind, placebo-controlled, phase 2 study.
        Lancet Diabetes Endocrinol. 2018; 6: 627-636
        • Zhu D.
        • Li X.
        • Ma J.
        • Zeng J.
        • Gan S.
        • Dong X.
        • et al.
        Dorzagliatin in drug-naïve patients with type 2 diabetes: a randomized, double-blind, placebo-controlled phase 3 trial.
        Nat Med. 2022; 28: 965-973
        • Yang W.
        • Zhu D.
        • Gan S.
        • Dong X.
        • Su J.
        • Li W.
        • et al.
        Dorzagliatin add-on therapy to metformin in patients with type 2 diabetes: a randomized, double-blind, placebo-controlled phase 3 trial.
        Nat Med. 2022; 28: 974-981
        • Zhang X.
        • Tong N.
        • Li Y.
        Concurrently using rosiglitazone prevents glucosamine-induced islet beta-cell apoptosis and dysfunction.
        Cell Biochem Funct. 2010; 28: 509-514
        • Wan J.
        • Jiang L.
        • Lü Q.
        • Ke L.
        • Li X.
        • Tong N.
        Activation of PPARdelta up-regulates fatty acid oxidation and energy uncoupling genes of mitochondria and reduces palmitate-induced apoptosis in pancreatic beta-cells.
        Biochem Biophys Res Commun. 2010; 391: 1567-1572
        • Li P.P.
        • Shan S.
        • Chen Y.T.
        • Ning Z.Q.
        • Sun S.J.
        • Liu Q.
        • et al.
        The PPARalpha/gamma dual agonist chiglitazar improves insulin resistance and dyslipidemia in MSG obese rats.
        Br J Pharmacol. 2006; 148: 610-618
        • Ji L.
        • Song W.
        • Fang H.
        • Li W.
        • Geng J.
        • Wang Y.
        • et al.
        Efficacy and safety of chiglitazar, a novel peroxisome proliferator-activated receptor pan-agonist, in patients with type 2 diabetes: a randomized, double-blind, placebo-controlled, phase 3 trial (CMAP).
        Sci Bull. 2021; 66: 1571-1580
        • Jia W.
        • Ma J.
        • Miao H.
        • Wang C.
        • Wang X.
        • Li Q.
        • et al.
        Chiglitazar monotherapy with sitagliptin as an active comparator in patients with type 2 diabetes: a randomized, double-blind, phase 3 trial (CMAS).
        Sci Bull. 2021; 66: 1581-1590
        • Larsen C.M.
        • Faulenbach M.
        • Vaag A.
        • Ehses J.A.
        • Donath M.Y.
        • Mandrup-Poulsen T.
        Sustained effects of interleukin-1 receptor antagonist treatment in type 2 diabetes.
        Diabetes Care. 2009; 32: 1663-1668
        • van Poppel P.C.
        • van Asseldonk E.J.
        • Holst J.J.
        • Vilsbøll T.
        • Netea M.G.
        • Tack C.J.
        The interleukin-1 receptor antagonist anakinra improves first-phase insulin secretion and insulinogenic index in subjects with impaired glucose tolerance.
        Diabetes Obes Metab. 2014; 16: 1269-1273
        • Huang J.
        • Yang Y.
        • Hu R.
        • Chen L.
        Anti-interleukin-1 therapy has mild hypoglycaemic effect in type 2 diabetes.
        Diabetes Obes Metab. 2018; 20: 1024-1028
        • Tahmasebi M.
        • Puddefoot J.R.
        • Inwang E.R.
        • Vinson G.P.
        The tissue renin-angiotensin system in human pancreas.
        J Endocrinol. 1999; 161: 317-322
        • Luther J.M.
        • Brown N.J.
        The renin-angiotensin-aldosterone system and glucose homeostasis.
        Trends Pharmacol Sci. 2011; 32: 734-739
        • van der Zijl N.J.
        • Moors C.C.
        • Goossens G.H.
        • Hermans M.M.
        • Blaak E.E.
        • Diamant M.
        Valsartan improves {beta}-cell function and insulin sensitivity in subjects with impaired glucose metabolism: a randomized controlled trial.
        Diabetes Care. 2011; 34: 845-851
        • Maneva-Radicheva L.
        • Amatya C.
        • Parker C.
        • Ellefson J.
        • Radichev I.
        • Raghavan A.
        • et al.
        Autoimmune diabetes is suppressed by treatment with recombinant human tissue Kallikrein-1.
        PLoS One. 2014; 9e107213
        • Kolodka T.
        • Charles M.L.
        • Raghavan A.
        • Radichev I.A.
        • Amatya C.
        • Ellefson J.
        • et al.
        Preclinical characterization of recombinant human tissue kallikrein-1 as a novel treatment for type 2 diabetes mellitus.
        PLoS One. 2014; 9e103981
        • Fichna M.
        • Fichna P.
        Glucocorticoids and beta-cell function.
        Endokrynol Pol. 2017; 68: 568-573
        • Shah R.V.
        • Goldfine A.B.
        Statins and risk of new-onset diabetes mellitus.
        Circulation. 2012; 126: e282-e284
        • Preiss D.
        • Seshasai S.R.
        • Welsh P.
        • Murphy S.A.
        • Ho J.E.
        • Waters D.D.
        • et al.
        Risk of incident diabetes with intensive-dose compared with moderate-dose statin therapy: a meta-analysis.
        Jama. 2011; 305: 2556-2564
        • Rajpathak S.N.
        • Kumbhani D.J.
        • Crandall J.
        • Barzilai N.
        • Alderman M.
        • Ridker P.M.
        Statin therapy and risk of developing type 2 diabetes: a meta-analysis.
        Diabetes Care. 2009; 32: 1924-1929
        • Galicia-Garcia U.
        • Jebari S.
        • Larrea-Sebal A.
        • Uribe K.B.
        • Siddiqi H.
        • Ostolaza H.
        • et al.
        Statin treatment-induced development of type 2 diabetes: from clinical evidence to mechanistic insights.
        Int J Mol Sci. 2020; 21
        • Stamatouli A.M.
        • Quandt Z.
        • Perdigoto A.L.
        • Clark P.L.
        • Kluger H.
        • Weiss S.A.
        • et al.
        Collateral damage: insulin-dependent diabetes induced with checkpoint inhibitors.
        Diabetes. 2018; 67: 1471-1480
        • Sandström P.E.
        Inhibition by hydrochlorothiazide of insulin release and calcium influx in mouse pancreatic beta-cells.
        Br J Pharmacol. 1993; 110: 1359-1362
        • Lacey R.J.
        • Cable H.C.
        • James R.F.
        • London N.J.
        • Scarpello J.H.
        • Morgan N.G.
        Concentration-dependent effects of adrenaline on the profile of insulin secretion from isolated human islets of Langerhans.
        J Endocrinol. 1993; 138: 555-563