β-cell

β 细胞
  • 文章类型: Journal Article
    这篇综述的目的是研究当前有关外分泌和内分泌胰腺之间相互作用的科学文献,特别是外分泌胰腺在犬糖尿病发病机制中的作用。由外分泌胰腺炎症引起的β细胞死亡被认为是狗糖尿病的一个未被认可的因素。多达30%的犬糖尿病患者在验尸时同时有胰腺炎的证据。目前对胰腺炎的诊断不精确,这两种疾病的治疗都有其自身的局限性:每天注射胰岛素来治疗糖尿病,这对利益相关者既有福利又有财务影响,和胰腺炎通过临床症状的治疗,如镇痛和止吐药,而不是有针对性的治疗根本原因。这篇综述将考虑外分泌性胰腺炎症对狗的胰腺β细胞损失和胰岛素缺乏性糖尿病有积极贡献的证据,并探索当前和潜在的未来诊断和治疗途径,以改善这些患者的预后。
    The purpose of this review is to examine the current scientific literature regarding the interplay between the exocrine and endocrine pancreas, specifically the role of the exocrine pancreas in the pathogenesis of canine diabetes mellitus. β-cell death caused by exocrine pancreatic inflammation is thought to be an under-recognised contributor to diabetes mellitus in dogs, with up to 30% of canine diabetic patients with concurrent evidence of pancreatitis at post-mortem examination. Current diagnostics for pancreatitis are imprecise, and treatments for both diseases individually have their own limitations: diabetes through daily insulin injections, which has both welfare and financial implications for the stakeholders, and pancreatitis through treatment of clinical signs, such as analgesia and anti-emetics, rather than targeted treatment of the underlying cause. This review will consider the evidence for exocrine pancreatic inflammation making an active contribution to pancreatic β-cell loss and insulin-deficiency diabetes in the dog and explore current and potential future diagnostic and treatment avenues to improve outcomes for these patients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    β-casomorphins(BCM)是具有阿片样物质特性的生物活性肽,通过蛋白水解消化乳中的β-酪蛋白而形成。当A1牛奶在小肠中消化时,由于β-酪蛋白第67位的组氨酸而形成BCM-7,与A2牛奶不同,在该位置具有脯氨酸并产生BCM-9。BCM-7被肠道中的二肽基肽酶-IV(DPP-IV)进一步降解为BCM-5。BCM-7的类阿片样物质活性负责引发信号传导途径,所述信号传导途径实现广泛的生理效应。我们研究的目的是找出BCM(BCM-7,BCM-9和BCM-5)对胰腺β细胞增殖的不同作用,胰岛素分泌,和来自正常(5.5mM)和高葡萄糖(27.5mM)浓度的β细胞(RIN-5F细胞系)的阿片类肽结合受体。我们的结果表明,BCM-7/9/5不影响β细胞的活力,扩散,和正常葡萄糖水平下的胰岛素分泌。然而,在较高的葡萄糖浓度下,BCM显着保护β细胞免受葡萄糖毒性,但不影响胰岛素分泌。有趣的是,在Mu-阿片样肽受体拮抗剂CTOP的存在下,BCM不能保护β细胞免受葡萄糖毒性。结果表明,BCM通过非阿片介导的途径保护β细胞免受葡萄糖毒性,因为BCM不调节mu的基因表达,κ和δ阿片样肽受体。
    Beta-casomorphins (BCMs) are the bio-active peptides having opioid properties which are formed by the proteolytic digestion of β-caseins in milk. BCM-7 forms when A1 milk is digested in the small intestine due to a histidine at the 67th position in β-casein, unlike A2 milk, which has proline at this position and produces BCM-9. BCM-7 has further degraded into BCM-5 by the dipeptidyl peptidase-IV (DPP-IV) enzyme in the intestine. The opioid-like activity of BCM-7 is responsible for eliciting signaling pathways which enable a wide range of physiological effects. The aim of our study was to find out the differential role of BCMs (BCM-7, BCM-9 and BCM-5) on pancreatic β-cell proliferation, insulin secretion, and opioid peptide binding receptors from β-cells (RIN-5F cell line) in normal (5.5 mM) and high glucose (27.5 mM) concentrations. Our results showed that BCM-7/9/5 did not affect β-cell viability, proliferation, and insulin secretion at normal glucose level. However, at higher glucose concentration, BCMs significantly protected β-cells from glucotoxicity but did not affect the insulin secretion. Interestingly, in the presence of Mu-opioid peptide receptor antagonist CTOP, BCMs did not protect β-cells from glucotoxicity. The results suggest that BCMs protect β-cells from glucotoxicity via non-opioid mediated pathways because BCMs did not modulate the gene expression of the mu, kappa and delta opioid peptide receptors.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    1型糖尿病(T1D)是一种常见的自身免疫性疾病,其中葡萄糖代谢失调是关键特征。对T1D的了解很少,并且需要改进的治疗剂。在T1D患者的多个组织中经常遇到缺氧,包括胰腺和糖尿病并发症的部位。缺氧诱导因子(HIF)-1,一种普遍存在的缺氧适应性反应的主要调节因子,在多个临床前T1D模型中,通过转录和非转录机制促进葡萄糖代谢并改变疾病进展。然而,胰腺β细胞和免疫细胞(T1D中的两种关键细胞类型)中的HIF-1激活如何最终影响疾病进展仍存在争议.我们讨论了我们对缺氧/HIF-1诱导的糖酵解在T1D中的作用的理解的最新进展,并探讨了靶向该途径的药物作为潜在的新疗法的可能用途。
    Type 1 diabetes (T1D) is a common autoimmune disease in which dysregulated glucose metabolism is a key feature. T1D is both poorly understood and in need of improved therapeutics. Hypoxia is frequently encountered in multiple tissues in T1D patients including the pancreas and sites of diabetic complications. Hypoxia-inducible factor (HIF)-1, a ubiquitous master regulator of the adaptive response to hypoxia, promotes glucose metabolism through transcriptional and non-transcriptional mechanisms and alters disease progression in multiple preclinical T1D models. However, how HIF-1 activation in β-cells of the pancreas and immune cells (two key cell types in T1D) ultimately affects disease progression remains controversial. We discuss recent advances in our understanding of the role of hypoxia/HIF-1-induced glycolysis in T1D and explore the possible use of drugs targeting this pathway as potential new therapeutics.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    在1型糖尿病(T1D)中,自身反应性免疫细胞浸润胰腺并分泌促炎细胞因子,从而引发产生胰岛素的胰岛β细胞的细胞死亡。蛋白激酶Cδ(PKCδ)在介导细胞因子诱导的β细胞死亡中起作用;然而,确切的机制还没有很好的理解。为了解决这个问题,我们使用了诱导型β细胞特异性PKCδKO小鼠以及PKCδ的小肽抑制剂。我们确定了PKCδ在介导细胞因子诱导的β细胞死亡中的作用,并表明抑制PKCδ可保护胰腺β细胞免受小鼠和人胰岛中细胞因子诱导的凋亡。我们确定细胞因子诱导了PKCδ的核易位和活性,并且细胞因子介导的胰岛细胞凋亡可能需要caspase-3裂解PKCδ。Further,细胞因子激活的PKCδ增加急性治疗的促凋亡Bax和长期治疗的JNK的活性。总的来说,我们的结果表明,PKCδ通过核转位介导细胞因子诱导的细胞凋亡,caspase-3的裂解,以及胰腺β细胞中促凋亡信号的上调。结合PKCδ抑制与δV1-1的保护作用,这项研究的结果将有助于开发新疗法,以预防或延迟β细胞死亡并保留T1D中的β细胞功能。
    In type 1 diabetes (T1D), autoreactive immune cells infiltrate the pancreas and secrete proinflammatory cytokines that initiate cell death in insulin producing islet β-cells. Protein kinase C δ (PKCδ) plays a role in mediating cytokine-induced β-cell death; however, the exact mechanisms are not well understood. To address this, we used an inducible β-cell specific PKCδ KO mouse as well as a small peptide inhibitor of PKCδ. We identified a role for PKCδ in mediating cytokine-induced β-cell death and have shown that inhibiting PKCδ protects pancreatic β-cells from cytokine-induced apoptosis in both mouse and human islets. We determined that cytokines induced nuclear translocation and activity of PKCδ and that caspase-3 cleavage of PKCδ may be required for cytokine-mediated islet apoptosis. Further, cytokine activated PKCδ increases activity both of proapoptotic Bax with acute treatment and C-Jun N-terminal kinase with prolonged treatment. Overall, our results suggest that PKCδ mediates cytokine-induced apoptosis via nuclear translocation, cleavage by caspase-3, and upregulation of proapoptotic signaling in pancreatic β-cells. Combined with the protective effects of PKCδ inhibition with δV1-1, the results of this study will aid in the development of novel therapies to prevent or delay β-cell death and preserve β-cell function in T1D.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    胰腺再生是在正常和病理条件下观察到的复杂过程。这篇综述的目的是全面了解成年胰腺中功能活跃的分泌胰岛素的β细胞群的出现。β细胞的更新受遗传和表观遗传因素的细胞来源之间的多方面相互作用支配。了解β细胞群的发育和异质性对于功能性β细胞再生至关重要。在妊娠和肥胖等情况下,胰腺β细胞的功能质量会增加。然而,在这些条件下能够增加自我繁殖的成熟β细胞群和出生后胰腺祖细胞的特异性标志物仍有待阐明。通过各种途径再生β细胞群的能力,包括预先存在的β细胞的增殖,β细胞新生,从祖细胞群体中分化出β细胞,非β细胞转分化为β细胞,揭示了识别细胞来源和功能性细胞更新诱导物的关键分子机制。这为确定特定的细胞来源和再生机制提供了机会,可以在临床上应用于治疗各种疾病,包括体外细胞技术,加深对不同生理条件下再生的认识。
    Pancreatic regeneration is a complex process observed in both normal and pathological conditions. The aim of this review is to provide a comprehensive understanding of the emergence of a functionally active population of insulin-secreting β-cells in the adult pancreas. The renewal of β-cells is governed by a multifaceted interaction between cellular sources of genetic and epigenetic factors. Understanding the development and heterogeneity of β-cell populations is crucial for functional β-cell regeneration. The functional mass of pancreatic β-cells increases in situations such as pregnancy and obesity. However, the specific markers of mature β-cell populations and postnatal pancreatic progenitors capable of increasing self-reproduction in these conditions remain to be elucidated. The capacity to regenerate the β-cell population through various pathways, including the proliferation of pre-existing β-cells, β-cell neogenesis, differentiation of β-cells from a population of progenitor cells, and transdifferentiation of non-β-cells into β-cells, reveals crucial molecular mechanisms for identifying cellular sources and inducers of functional cell renewal. This provides an opportunity to identify specific cellular sources and mechanisms of regeneration, which could have clinical applications in treating various pathologies, including in vitro cell-based technologies, and deepen our understanding of regeneration in different physiological conditions.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    目的:受体相互作用蛋白激酶1(RIPK1)协调肿瘤坏死因子(TNF)和其他细胞因子在细胞存活和细胞死亡之间的决定。而RIPK1的支架功能对于防止TNF诱导的细胞凋亡和坏死性凋亡至关重要,它的激酶活性是细胞坏死所必需的,部分是细胞凋亡所必需的。尽管TNF是一种与糖尿病中β细胞丢失相关的促炎细胞因子,TNF诱导β细胞死亡的机制尚不清楚。
    方法:这里,我们使用缺乏RIPK1的小鼠(Ripk1β-KO小鼠)或表达激酶死亡版本的RIPK1(Ripk1D138N小鼠),分析了RIPK1支架与激酶功能对β细胞死亡调节的贡献,分别。这些小鼠被链脲佐菌素攻击,自身免疫性糖尿病模型.此外,用高脂肪饮食进一步攻击Ripk1β-KO小鼠以诱导高血糖。对于机械学研究,胰岛接受了各种杀伤剂和致敏剂。
    结果:抑制RIPK1激酶活性(Ripk1D138N小鼠)不影响1型糖尿病模型中高血糖的发作和进展。此外,β细胞中RIPK1表达缺失不影响基础条件下的血糖正常或糖尿病挑战下的高血糖.离体,在没有RIPK1的情况下,原代胰岛对TNF诱导的凋亡和坏死不敏感。有趣的是,我们发现胰岛显示高水平的抗凋亡细胞FLICE抑制蛋白(cFLIP)和低水平的凋亡(Caspase-8)和坏死(RIPK3)成分.环己酰亚胺处理,这导致了cFLIP水平的降低,使原代胰岛对TNF诱导的细胞死亡敏感,而TNF诱导的细胞死亡被胱天蛋白酶抑制完全阻断。
    结论:与许多其他细胞类型不同(例如,上皮,和免疫),在生理条件或糖尿病挑战下,RIPK1对于β细胞中的细胞死亡调节不是必需的。此外,体内和体外证据表明,胰腺β细胞不会发生坏死,而主要是响应TNF的caspase依赖性死亡。最后,我们的结果表明β细胞具有独特的TNF-细胞毒性调节模式,该模式独立于RIPK1,并且可能高度依赖于cFLIP.
    OBJECTIVE: Receptor-interacting protein kinase 1 (RIPK1) orchestrates the decision between cell survival and cell death in response to tumor necrosis factor (TNF) and other cytokines. Whereas the scaffolding function of RIPK1 is crucial to prevent TNF-induced apoptosis and necroptosis, its kinase activity is required for necroptosis and partially for apoptosis. Although TNF is a proinflammatory cytokine associated with β-cell loss in diabetes, the mechanism by which TNF induces β-cell demise remains unclear.
    METHODS: Here, we dissected the contribution of RIPK1 scaffold versus kinase functions to β-cell death regulation using mice lacking RIPK1 specifically in β-cells (Ripk1β-KO mice) or expressing a kinase-dead version of RIPK1 (Ripk1D138N mice), respectively. These mice were challenged with streptozotocin, a model of autoimmune diabetes. Moreover, Ripk1β-KO mice were further challenged with a high-fat diet to induce hyperglycemia. For mechanistic studies, pancreatic islets were subjected to various killing and sensitising agents.
    RESULTS: Inhibition of RIPK1 kinase activity (Ripk1D138N mice) did not affect the onset and progression of hyperglycemia in a type 1 diabetes model. Moreover, the absence of RIPK1 expression in β-cells did not affect normoglycemia under basal conditions or hyperglycemia under diabetic challenges. Ex vivo, primary pancreatic islets are not sensitised to TNF-induced apoptosis and necroptosis in the absence of RIPK1. Intriguingly, we found that pancreatic islets display high levels of the antiapoptotic cellular FLICE-inhibitory protein (cFLIP) and low levels of apoptosis (Caspase-8) and necroptosis (RIPK3) components. Cycloheximide treatment, which led to a reduction in cFLIP levels, rendered primary islets sensitive to TNF-induced cell death which was fully blocked by caspase inhibition.
    CONCLUSIONS: Unlike in many other cell types (e.g., epithelial, and immune), RIPK1 is not required for cell death regulation in β-cells under physiological conditions or diabetic challenges. Moreover, in vivo and in vitro evidence suggest that pancreatic β-cells do not undergo necroptosis but mainly caspase-dependent death in response to TNF. Last, our results show that β-cells have a distinct mode of regulation of TNF-cytotoxicity that is independent of RIPK1 and that may be highly dependent on cFLIP.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    目的:生长激素(GH)是β细胞增殖的中枢调节因子,胰岛素分泌和敏感性。这项研究的目的是研究GH不敏感对胰腺β细胞组织形态学的影响以及对体内代谢的影响。
    方法:来自生长激素受体缺乏的猪的胰腺(GHR-KO,n=12)与野生型对照(WT,n=12),年龄为3和7-8.5个月。在各年龄组的GHR-KO(n=3)和WT(n=3)猪中,通过静脉内葡萄糖耐量试验(ivGTT)评估了胰岛素和葡萄糖耐量的体内分泌能力。
    结果:无偏定量立体分析显示,总β细胞体积显着减少(年轻和成年GHR-KO分别减少了83%和73%年龄匹配的WT猪;p<0.0001)和GHR-KO猪胰腺中β细胞的体积密度(年轻和成年GHR-KO猪减少42%和39%;p=0.0018)。GHR-KO猪表现出显著的,胰腺中分离的β细胞比例的年龄依赖性增加(年轻人为28%,成人GHR-KO为97%。年龄匹配的WT猪;p=0.0009)。尽管ivGTT的胰岛素分泌减少,GHR-KO猪维持正常的葡萄糖耐量。
    结论:GHR-KO猪的GH不敏感导致胰腺中β细胞体积和β细胞体积比例降低,导致胰岛素分泌能力下降。GHR-KO猪的胰腺中分离的β细胞的比例增加强调了对GH刺激的依赖性以实现适当的β细胞成熟。保持葡萄糖耐量并减少胰岛素分泌表明GH不敏感时对胰岛素的敏感性增强。
    OBJECTIVE: Growth hormone (GH) is a central regulator of β-cell proliferation, insulin secretion and sensitivity. Aim of this study was to investigate the effect of GH insensitivity on pancreatic β-cell histomorphology and consequences for metabolism in vivo.
    METHODS: Pancreata from pigs with growth hormone receptor deficiency (GHR-KO, n = 12) were analyzed by unbiased quantitative stereology in comparison to wild-type controls (WT, n = 12) at 3 and 7-8.5 months of age. In vivo secretion capacity for insulin and glucose tolerance were assessed by intravenous glucose tolerance tests (ivGTTs) in GHR-KO (n = 3) and WT (n = 3) pigs of the respective age groups.
    RESULTS: Unbiased quantitative stereological analyses revealed a significant reduction in total β-cell volume (83% and 73% reduction in young and adult GHR-KO vs. age-matched WT pigs; p < 0.0001) and volume density of β-cells in the pancreas of GHR-KO pigs (42% and 39% reduction in young and adult GHR-KO pigs; p = 0.0018). GHR-KO pigs displayed a significant, age-dependent increase in the proportion of isolated β-cells in the pancreas (28% in young and 97% in adult GHR-KO vs. age-matched WT pigs; p = 0.0009). Despite reduced insulin secretion in ivGTTs, GHR-KO pigs maintained normal glucose tolerance.
    CONCLUSIONS: GH insensitivity in GHR-KO pigs leads to decreased β-cell volume and volume proportion of β-cells in the pancreas, causing a reduced insulin secretion capacity. The increased proportion of isolated β-cells in the pancreas of GHR-KO pigs highlights the dependency on GH stimulation for proper β-cell maturation. Preserved glucose tolerance accomplished with decreased insulin secretion indicates enhanced sensitivity for insulin in GH insensitivity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    2型糖尿病(T2D)是一种常见的代谢疾病,原因是在胰岛素抵抗的背景下,胰腺β细胞的胰岛素分泌不足。胰岛分子病理学揭示了蛋白质错误折叠在β细胞功能障碍和胰岛淀粉样蛋白来源的胰岛淀粉样蛋白(IAPP)缺失中的作用。与胰岛素共表达和共分泌的蛋白质。错误折叠的IAPP的最毒性形式是存在于T2D的β细胞和人IAPP(hIAPP)转基因小鼠的β细胞中的胞内膜破坏性毒性寡聚体。先前的工作揭示了来自T2D和9-10周龄糖尿病前小鼠的胰岛中转录变化的高度重叠,所述小鼠转基因用于hIAPP,其中大多数变化是促存活适应,因此治疗指导有限。在这里,我们研究了较早年龄(6周)的hIAPP转基因小鼠的胰岛,以筛选在促生存信号传导优势之前的hIAPP毒性的潜在介质。我们发现了早期抑制胆固醇合成和运输以及异常的β细胞内胆固醇和脂质沉积,和受损的胆固醇运输到细胞膜。这些发现与T2D中β细胞中存在的相当的脂质沉积以及服用抑制胆固醇合成的药物的个体发展T2D的脆弱性增加一致。
    Type 2 diabetes (T2D) is a common metabolic disease due to insufficient insulin secretion by pancreatic β-cells in the context of insulin resistance. Islet molecular pathology reveals a role for protein misfolding in β-cell dysfunction and loss with islet amyloid derived from islet amyloid polypeptide (IAPP), a protein coexpressed and cosecreted with insulin. The most toxic form of misfolded IAPP is intracellular membrane disruptive toxic oligomers present in β-cells in T2D and in β-cells of mice transgenic for human IAPP (hIAPP). Prior work revealed a high degree of overlap of transcriptional changes in islets from T2D and prediabetic 9- to 10-wk-old mice transgenic for hIAPP with most changes being pro-survival adaptations and therefore of limited therapeutic guidance. Here, we investigated islets from hIAPP transgenic mice at an earlier age (6 wk) to screen for potential mediators of hIAPP toxicity that precede predominance of pro-survival signaling. We identified early suppression of cholesterol synthesis and trafficking along with aberrant intra-β-cell cholesterol and lipid deposits and impaired cholesterol trafficking to cell membranes. These findings align with comparable lipid deposits present in β-cells in T2D and increased vulnerability to develop T2D in individuals taking medications that suppress cholesterol synthesis.NEW & NOTEWORTHY β-Cell failure in type 2 diabetes (T2D) is characterized by β-cell misfolded protein stress due to the formation of toxic oligomers of islet amyloid polypeptide (IAPP). Most transcriptional changes in islets in T2D are pro-survival adaptations consistent with the slow progression of β-cell loss. In the present study, investigation of the islet transcriptional signatures in a mouse model of T2D expressing human IAPP revealed decreased cholesterol synthesis and trafficking as a plausible early mediator of IAPP toxicity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    在胰岛素抵抗和β细胞质量降低的情况下,β细胞工作量增加,发生在2型和1型糖尿病中,分别。在糖尿病的发病过程中,胰岛素产生和分泌的长期升高会导致β细胞内质网应激。内质网应激过程中β细胞Ca2+内质网的消耗激活了未折叠的蛋白反应,导致β细胞功能障碍。Ca2+ER参与许多对β细胞功能至关重要的途径,比如蛋白质加工,调节细胞器和胞质Ca2+处理,和调节脂质稳态。促进β细胞内质网应激和耗尽Ca2+内质网储存的突变与糖尿病相关或引起糖尿病(例如,ryanodine受体和胰岛素的突变)。因此,改善β细胞Ca2+ER处理和减少糖尿病条件下的ER应激可以保持β细胞功能并延缓或预防糖尿病的发作。这篇综述着重于控制β细胞Ca2ER的机制在糖尿病的发病过程中如何受到干扰并导致β细胞衰竭。
    The β-cell workload increases in the setting of insulin resistance and reduced β-cell mass, which occurs in type 2 and type 1 diabetes, respectively. The prolonged elevation of insulin production and secretion during the pathogenesis of diabetes results in β-cell ER stress. The depletion of β-cell Ca2+ER during ER stress activates the unfolded protein response, leading to β-cell dysfunction. Ca2+ER is involved in many pathways that are critical to β-cell function, such as protein processing, tuning organelle and cytosolic Ca2+ handling, and modulating lipid homeostasis. Mutations that promote β-cell ER stress and deplete Ca2+ER stores are associated with or cause diabetes (e.g., mutations in ryanodine receptors and insulin). Thus, improving β-cell Ca2+ER handling and reducing ER stress under diabetogenic conditions could preserve β-cell function and delay or prevent the onset of diabetes. This review focuses on how mechanisms that control β-cell Ca2+ER are perturbed during the pathogenesis of diabetes and contribute to β-cell failure.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    2型糖尿病(T2D)是一种多基因代谢疾病,其特征是外周组织中的胰岛素抵抗和胰腺的胰岛素分泌受损。虽然胰岛素产生和分泌的下降以前归因于产生胰岛素的β细胞的凋亡,最近的研究表明糖尿病患者的β细胞凋亡率相对较低。相反,β细胞主要经历去分化,在这个过程中,它们失去了专门的身份,并转变为无功能的内分泌祖细胞样细胞,最终导致β细胞衰竭。由于遗传因素和细胞应激的复杂相互作用,驱动β细胞去分化的潜在机制仍然难以捉摸。了解这些机制有可能为旨在逆转T2D中β细胞去分化的创新治疗方法提供信息。这篇综述探讨了β细胞去分化导致β细胞衰竭的拟议驱动因素,并讨论了能够逆转这一过程的当前干预措施,从而恢复β细胞的身份和功能。
    Type 2 diabetes (T2D) is a polygenic metabolic disorder characterized by insulin resistance in peripheral tissues and impaired insulin secretion by the pancreas. While the decline in insulin production and secretion was previously attributed to apoptosis of insulin-producing β-cells, recent studies indicate that β-cell apoptosis rates are relatively low in diabetes. Instead, β-cells primarily undergo dedifferentiation, a process where they lose their specialized identity and transition into non-functional endocrine progenitor-like cells, ultimately leading to β-cell failure. The underlying mechanisms driving β-cell dedifferentiation remain elusive due to the intricate interplay of genetic factors and cellular stress. Understanding these mechanisms holds the potential to inform innovative therapeutic approaches aimed at reversing β-cell dedifferentiation in T2D. This review explores the proposed drivers of β-cell dedifferentiation leading to β-cell failure, and discusses current interventions capable of reversing this process, thus restoring β-cell identity and function.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号