Published online Dec 15, 2025. doi: 10.4239/wjd.v16.i12.108609
Revised: August 13, 2025
Accepted: October 23, 2025
Published online: December 15, 2025
Processing time: 241 Days and 3.9 Hours
A recent study in the World Journal of Diabetes by Yang et al explored how Rheb1 signaling influenced pancreatic β cell fate and its potential as a therapeutic target. This invited commentary by a senior diabetes researcher discussed the findings of Yang et al in the context of current knowledge on β cell biology, providing critical insight into the role of Rheb1 in β cell survival and function and the prospects for diabetes treatment. Key outcomes of the study were interpreted alongside est
Core Tip: Rheb1 is a critical regulator of pancreatic β cell survival and insulin secretion, acting through the mechanistic target of rapamycin complex 1 pathway to support β cell mass and function. This invited commentary evaluated how Rheb1-driven signaling modulates β cell fate and assesses its promise as a therapeutic target in diabetes. While enhancing Rheb1 activity could preserve or expand functional β cell mass, careful modulation is required to avoid unintended effects such as α cell overactivity. Understanding the dualistic nature of Rheb1 signaling in islet cell populations will be key to harnessing this pathway for future diabetes treatments.
- Citation: Karimkhani H. Rheb1 signaling and the fate of pancreatic β cells: Toward a new frontier in diabetes therapy. World J Diabetes 2025; 16(12): 108609
- URL: https://www.wjgnet.com/1948-9358/full/v16/i12/108609.htm
- DOI: https://dx.doi.org/10.4239/wjd.v16.i12.108609
The study by Yang et al[1] recently published in the World Journal of Diabetes provides compelling new insights into the role of Rheb1 signaling in pancreatic β cells. The authors examined how manipulating Rheb1, a small GTPase upstream of the mechanistic target of rapamycin complex (mTORC1) pathway, can influence β cell fate decisions, potentially offering novel therapeutic avenues for diabetes. This work is significant given the central importance of functional β cell mass in the pathogenesis of both type 1 and type 2 diabetes. Loss of insulin-producing β cells through apoptosis or functional decline underlies the progressive failure of glycemic control in diabetes. Strategies to safeguard or replenish β cells are therefore highly sought after in diabetes research. In this context the focus of Yang et al[1] on Rheb1 signaling as a lever to modulate β cell fate is timely and thought-provoking.
The study under discussion built on growing evidence that nutrient-sensing pathways, particularly the Rheb1-mTORC1 signaling axis, are critical regulators of β cell physiology. Rheb1 is known to be abundantly expressed in pancreatic islets[2,3] and to positively regulate mTORC1 activity, a pathway integral to cell growth, proliferation, and metabolism[4]. In murine models β cell-specific knockout of Rheb1 results in marked reductions in β cell size and mass due to suppressed β cell proliferation and increased apoptosis[3]. Yang et al[1] demonstrated that embryonic loss of Rheb1 in β cells leads to diabetes in mice, underscoring that Rheb1 is essential for maintaining an adequate β cell population.
Interestingly, when Rheb1 was deleted in adult β cells (after normal development), β cell mass remained grossly unchanged, but insulin secretion in response to glucose was significantly impaired[3]. This indicates that Rheb1 has dual roles: (1) Sustaining β cell survival and replication over the long term; and (2) Acutely enabling β cells to sense glucose and secrete insulin. Mechanistically, Rheb1 promotes glucose-stimulated insulin secretion by upregulating glucose transporter expression (GLUT1 in human β cells, GLUT2 in mouse β cells) via mTORC1-dependent signaling[3]. These findings, corroborated by Yang et al[1], paint Rheb1 as a master regulator of β cell fate, one that impacts both the quantity of β cells and their quality of function[1].
Yang et al[1] extended this paradigm by exploring the therapeutic potential of modulating Rheb1. The manuscript posits that enhancing Rheb1 signaling in pancreatic β cells could be harnessed to treat diabetes, essentially by preventing β cell loss or even promoting β cell regeneration. This idea is supported by prior observations that augmenting mTORC1 activity (to which Rheb1 is key) can drive β cell expansion in states of increased metabolic demand. For instance, nutrient and growth factor signals that activate mTORC1 are known to contribute to compensatory β cell hyperplasia in insulin-resistant conditions. Conversely, systemic mTORC1 inhibitors (like rapamycin used in transplant medicine) often induce glucose intolerance or diabetes as a side effect due in part to β cell dysfunction caused by mTORC1 suppression[3,5-7]. The convergence of these insights with the findings from Yang et al[1] highlights a fundamental concept: Adequate Rheb1/mTORC1 signaling is requisite for β cell health, and its strategic upregulation might counteract the β cell failure central to diabetes. Indeed, Rheb1 has been identified as a promising target for diabetes therapy in recent literature.
If Rheb1 signaling is to be leveraged in diabetes treatment, several implications and challenges warrant careful discussion. First, specificity of the intervention is crucial. Rheb1 is expressed in multiple cell types within the islets of Langerhans, including α cells (glucagon-producing cells). While bolstering Rheb1 in β cells is desirable for enhancing insulin output, inadvertent activation of Rheb1 in α cells could be counterproductive. Lubaczeuski et al[8] recently showed that inducing Rheb1/mTORC1 hyperactivation in α cells led to hypersecretion of glucagon and an increase in α cell mass. This condition of hyperglucagonemia can aggravate hyperglycemia and counterbalance the benefits of increased insulin. Yang et al[1] rightly acknowledges this concern and suggests that any therapeutic strategy must target β cells selectively. Gene therapy or advanced drug delivery systems might be needed to confine Rheb1 modulation to β cells. Future research could explore β cell-specific promoters or targeting ligands that activate Rheb1 only in the intended cells, thereby minimizing off-target effects on α cells or other tissues[1,9-11]. Among delivery tools, adeno-associated virus serotype 8 has demonstrated high tropism for pancreatic islets in rodents. Use of the rat insulin promoter or human insulin promoter elements can restrict expression to β cells. Alternatively, ligand-decorated lipid nanoparticles targeting surface proteins (e.g., GLP1R) could provide non-viral specificity[12-14].
Second, the timing and degree of Rheb1 activation require optimization. β cell failure in type 2 diabetes develops in the context of chronic metabolic stress (glucotoxicity, lipotoxicity, and inflammatory signals). An open question is whether boosting Rheb1 signaling in already stressed β cells will rescue them or potentially push them into overdrive. Chronic mTORC1 hyperactivation can in some settings lead to cellular stress by inhibiting autophagic pathways and altering feedback loops in metabolism. Thus, how much and when to stimulate Rheb1 is a critical matter. Transient or moderate enhancement of Rheb1 might promote β cell survival and function, whereas excessive or sustained activation could have diminishing returns or even adverse effects[11]. Preclinical studies could employ inducible Rheb1 upregulation in diabetic animal models to determine the optimal window and magnitude of intervention to a balance in which β cells are supported but not exhausted. Additionally, combination therapies may be envisioned. For example, coupling Rheb1 pathway activation with agents that mitigate oxidative or ER stress in β cells could be used to create a more permissive environment for β cell regeneration[15-17].
Third, translating Rheb1 modulation into a practical therapy will likely involve innovative pharmacological strategies. Direct agonists of Rheb1 are not currently available since Rheb1 is a small GTP-binding protein normally regulated by upstream inhibitors (TSC1/TSC2 complex) and activators. One approach could be targeting the inhibitory TSC1/TSC2 complex in β cells. Inhibiting TSC1/2 would release Rheb1 from repression, thus activating mTORC1. Small-molecule inhibitors of TSC2 or compounds that disrupt the TSC-Rheb interaction might simulate Rheb1 activation[18,19]. Another approach might utilize incretin hormones or growth factors that naturally enhance β cell mTORC1 signaling. For instance, insulin/IGF-1 signaling and glucagon-like peptide-1 (GLP-1) agonists can activate PI3K-Akt pathways upstream of mTORC1, potentially boosting Rheb1 activity indirectly. It would be intriguing to investigate whether part of the known β cell protective effects of GLP-1 analogues or modern antidiabetic drugs (like GLP-1 receptor agonists and possibly SGLT2 inhibitors through metabolic remodeling) involve modulation of the Rheb1/mTORC1 axis[20-22]. AMPK, a key energy sensor, antagonizes mTORC1 and is downregulated in diabetic β cells, potentially tipping the Rheb1 balance. Similarly, Rheb1-mediated mTORC1 signaling intersects with Notch1, a pathway implicated in β cell dedifferentiation. Dual modulation of these axes may enhance therapeutic specificity[23-26].
Fourth, to strengthen the translational relevance of the proposed strategies, future research should focus on validating the selective activation of Rheb1 using β cell-specific delivery systems such as adeno-associated virus-mediated gene delivery or ligand-conjugated nanoparticles. In vitro β cell models or human islet organoids could serve as platforms to assess the impact of targeted Rheb1 modulation on insulin secretion and β cell proliferation[3]. In vivo models should be used to evaluate systemic safety with particular attention to α cell activity and chronic mTORC1 overactivation. Additionally, combining Rheb1 activation with GLP-1 analogs could be tested for synergistic effects. A comprehensive risk-benefit framework should guide future studies, considering both therapeutic potential and unintended con
Finally, rigorous safety and efficacy assessments will be paramount. As with any intervention on a fundamental growth pathway, there is a theoretical risk of neoplastic transformation if cell proliferation is overstimulated. Pancreatic β cells have a low baseline proliferative rate in adults and forcing them to divide carries uncertainties. Long-term studies in animal models will be needed to ensure that enhancing Rheb1 does not predispose to islet cell tumorigenesis or other proliferative lesions[29,30]. Preclinical safety studies should assess tumor suppressor gene expression (e.g., p53, p16^INK4a) and conduct metabolomic profiling to detect dysregulated anabolic flux. Carcinogenicity trials with extended follow-up in Rheb1-activated models are essential before human translation[31,32]. Moreover, the metabolic con
Rheb1 signaling has emerged as a crucial nexus point in the control of pancreatic β cell fate, bridging nutrient-sensing pathways with the maintenance of β cell mass and function. Yang et al[1] makes a strong case that modulating Rheb1 activity can be a modus operandi to preserve and possibly restore the insulin-producing cell population in diabetes. This concept opens a new frontier in diabetes therapy, one that goes beyond managing blood sugar levels and aims to correct the underlying β cell deficiency. The commentary presented here underscores that while the prospects of targeting Rheb1 are exciting, a nuanced approach is essential. Future research should focus on developing cell-specific Rheb1 modulators, understanding the long-term effects of Rheb1 activation, and integrating this strategy with existing treatments. If these challenges can be met, Rheb1-based interventions might ultimately transform the landscape of diabetes care by offering patients therapies that protect and rejuvenate their own pancreatic β cells. To enhance translational relevance, future studies should incorporate human islet models such as pseudo-islets or pancreatic organoids. Single-cell RNA-sequencing of these systems following Rheb1 modulation may reveal subpopulation-specific responses and refine target selection.
| 1. | Yang Y, Song WJ, Zhang JJ. Ras homolog enriched in brain 1 regulates β cell mass and β cell function via mTORC1/AMPK/Notch1 pathways. World J Diabetes. 2025;16:104973. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 2] [Reference Citation Analysis (4)] |
| 2. | Lu Q, Wang M, Gui Y, Hou Q, Gu M, Liang Y, Xiao B, Zhao AZ, Dai C. Rheb1 protects against cisplatin-induced tubular cell death and acute kidney injury via maintaining mitochondrial homeostasis. Cell Death Dis. 2020;11:364. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 12] [Cited by in RCA: 50] [Article Influence: 10.0] [Reference Citation Analysis (0)] |
| 3. | Yang Y, Cai Z, Pan Z, Liu F, Li D, Ji Y, Zhong J, Luo H, Hu S, Song L, Yu S, Li T, Li J, Ma X, Zhang W, Zhou Z, Liu F, Zhang J. Rheb1 promotes glucose-stimulated insulin secretion in human and mouse β-cells by upregulating GLUT expression. Metabolism. 2021;123:154863. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 4] [Cited by in RCA: 13] [Article Influence: 3.3] [Reference Citation Analysis (0)] |
| 4. | Jain C, Ansarullah, Bilekova S, Lickert H. Targeting pancreatic β cells for diabetes treatment. Nat Metab. 2022;4:1097-1108. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 30] [Article Influence: 10.0] [Reference Citation Analysis (0)] |
| 5. | Heard JJ, Fong V, Bathaie SZ, Tamanoi F. Recent progress in the study of the Rheb family GTPases. Cell Signal. 2014;26:1950-1957. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 49] [Cited by in RCA: 56] [Article Influence: 5.1] [Reference Citation Analysis (0)] |
| 6. | Hamada S, Hara K, Hamada T, Yasuda H, Moriyama H, Nakayama R, Nagata M, Yokono K. Upregulation of the mammalian target of rapamycin complex 1 pathway by Ras homolog enriched in brain in pancreatic beta-cells leads to increased beta-cell mass and prevention of hyperglycemia. Diabetes. 2009;58:1321-1332. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 70] [Cited by in RCA: 70] [Article Influence: 4.4] [Reference Citation Analysis (0)] |
| 7. | Mattis KK, Krentz NAJ, Metzendorf C, Abaitua F, Spigelman AF, Sun H, Ikle JM, Thaman S, Rottner AK, Bautista A, Mazzaferro E, Perez-Alcantara M, Manning Fox JE, Torres JM, Wesolowska-Andersen A, Yu GZ, Mahajan A, Larsson A, MacDonald PE, Davies B, den Hoed M, Gloyn AL. Loss of RREB1 in pancreatic beta cells reduces cellular insulin content and affects endocrine cell gene expression. Diabetologia. 2023;66:674-694. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 8] [Cited by in RCA: 14] [Article Influence: 7.0] [Reference Citation Analysis (0)] |
| 8. | Lubaczeuski C, Bozadjieva-Kramer N, Louzada RA, Gittes GK, Leibowitz G, Bernal-Mizrachi E. Time-dependent effects of endogenous hyperglucagonemia on glucose homeostasis and hepatic glucagon action. JCI Insight. 2023;8:e162255. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 1] [Cited by in RCA: 4] [Article Influence: 2.0] [Reference Citation Analysis (0)] |
| 9. | Li MY, Liu LZ, Xin Q, Zhou J, Zhang X, Zhang R, Wu Z, Yi J, Dong M. Downregulation of mTORC1 and Mcl-1 by lipid-oversupply contributes to islet β-cell apoptosis and dysfunction. Biochim Biophys Acta Mol Cell Biol Lipids. 2023;1868:159332. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 2] [Reference Citation Analysis (0)] |
| 10. | So WY, Han W. Gene therapy targeting key beta cell regulators as a potential intervention for diabetes. EMBO Mol Med. 2024;16:1490-1494. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 6] [Article Influence: 6.0] [Reference Citation Analysis (0)] |
| 11. | Sionov RV, Ahdut-HaCohen R. A Supportive Role of Mesenchymal Stem Cells on Insulin-Producing Langerhans Islets with a Specific Emphasis on The Secretome. Biomedicines. 2023;11:2558. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Reference Citation Analysis (0)] |
| 12. | Flores RR, Zhou L, Robbins PD. Expression of IL-2 in β cells by AAV8 gene transfer in pre-diabetic NOD mice prevents diabetes through activation of FoxP3-positive regulatory T cells. Gene Ther. 2014;21:715-722. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 13] [Cited by in RCA: 14] [Article Influence: 1.3] [Reference Citation Analysis (0)] |
| 13. | Wang X, Olmsted-Davis E, Davis A, Liu S, Li Z, Yang J, Brunicardi FC. Specific targeting of pancreatic islet cells in vivo by insulin-promoter-driven adenoviral conjugated reporter genes. World J Surg. 2006;30:1543-1552. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 8] [Cited by in RCA: 7] [Article Influence: 0.4] [Reference Citation Analysis (0)] |
| 14. | Ramzy A, Tudurí E, Glavas MM, Baker RK, Mojibian M, Fox JK, O'Dwyer SM, Dai D, Hu X, Denroche HC, Edeer N, Gray SL, Verchere CB, Johnson JD, Kieffer TJ. AAV8 Ins1-Cre can produce efficient β-cell recombination but requires consideration of off-target effects. Sci Rep. 2020;10:10518. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 4] [Cited by in RCA: 11] [Article Influence: 2.2] [Reference Citation Analysis (0)] |
| 15. | Ghasemi Gojani E, Rai S, Norouzkhani F, Shujat S, Wang B, Li D, Kovalchuk O, Kovalchuk I. Targeting β-Cell Plasticity: A Promising Approach for Diabetes Treatment. Curr Issues Mol Biol. 2024;46:7621-7667. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 6] [Reference Citation Analysis (0)] |
| 16. | Eguchi N, Toribio AJ, Alexander M, Xu I, Whaley DL, Hernandez LF, Dafoe D, Ichii H. Dysregulation of β-Cell Proliferation in Diabetes: Possibilities of Combination Therapy in the Development of a Comprehensive Treatment. Biomedicines. 2022;10:472. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 3] [Cited by in RCA: 11] [Article Influence: 3.7] [Reference Citation Analysis (0)] |
| 17. | Lee YH, Kim HJ, Kim JA, Jang Y, Moon EJ, Kim HJ. 1473-P: Combination Therapy with Dual-Function Peptide S1K and Insulin Reduces Blood Glucose Levels in Mouse Models of Type 1 Diabetes through Beta-Cell Regeneration and Immunosuppression. Diabetes. 2022;71:1473-P. [DOI] [Full Text] |
| 18. | Shams R, Ito Y, Miyatake H. Development of an RHEB-Targeting Peptide To Inhibit mTORC1 Kinase Activity. ACS Omega. 2022;7:23479-23486. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 4] [Reference Citation Analysis (0)] |
| 19. | Mahoney SJ, Narayan S, Molz L, Berstler LA, Kang SA, Vlasuk GP, Saiah E. A small molecule inhibitor of Rheb selectively targets mTORC1 signaling. Nat Commun. 2018;9:548. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 66] [Cited by in RCA: 66] [Article Influence: 9.4] [Reference Citation Analysis (0)] |
| 20. | Zhang X, Cao C, Zheng F, Liu C, Tian X. Therapeutic Potential of GLP-1 Receptor Agonists in Diabetes and Cardiovascular Disease: Mechanisms and Clinical Implications. Cardiovasc Drugs Ther. 2025. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 7] [Reference Citation Analysis (0)] |
| 21. | Zhou J, Wu J, Zheng F, Jin M, Li H. Glucagon-like peptide-1 analog-mediated protection against cholesterol-induced apoptosis via mammalian target of rapamycin activation in pancreatic βTC-6 cells -1mTORβTC-6. J Diabetes. 2015;7:231-239. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 16] [Cited by in RCA: 26] [Article Influence: 2.6] [Reference Citation Analysis (0)] |
| 22. | Delrue C, Speeckaert MM. Mechanistic Pathways and Clinical Implications of GLP-1 Receptor Agonists in Type 1 Diabetes Management. Int J Mol Sci. 2024;25:9351. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 10] [Reference Citation Analysis (0)] |
| 23. | Kimball SR. Interaction between the AMP-activated protein kinase and mTOR signaling pathways. Med Sci Sports Exerc. 2006;38:1958-1964. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 73] [Cited by in RCA: 78] [Article Influence: 4.3] [Reference Citation Analysis (0)] |
| 24. | Szkudelski T, Szkudelska K. The relevance of AMP-activated protein kinase in insulin-secreting β cells: a potential target for improving β cell function? J Physiol Biochem. 2019;75:423-432. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 13] [Cited by in RCA: 35] [Article Influence: 5.8] [Reference Citation Analysis (0)] |
| 25. | Haythorne E, Lloyd M, Walsby-Tickle J, Tarasov AI, Sandbrink J, Portillo I, Exposito RT, Sachse G, Cyranka M, Rohm M, Rorsman P, McCullagh J, Ashcroft FM. Altered glycolysis triggers impaired mitochondrial metabolism and mTORC1 activation in diabetic β-cells. Nat Commun. 2022;13:6754. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 63] [Cited by in RCA: 68] [Article Influence: 22.7] [Reference Citation Analysis (0)] |
| 26. | Jaafar R, Tran S, Shah AN, Sun G, Valdearcos M, Marchetti P, Masini M, Swisa A, Giacometti S, Bernal-Mizrachi E, Matveyenko A, Hebrok M, Dor Y, Rutter GA, Koliwad SK, Bhushan A. mTORC1 to AMPK switching underlies β-cell metabolic plasticity during maturation and diabetes. J Clin Invest. 2019;129:4124-4137. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 79] [Cited by in RCA: 102] [Article Influence: 17.0] [Reference Citation Analysis (0)] |
| 27. | Bottino R, Lemarchand P, Trucco M, Giannoukakis N. Gene- and cell-based therapeutics for type I diabetes mellitus. Gene Ther. 2003;10:875-889. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 48] [Cited by in RCA: 46] [Article Influence: 2.1] [Reference Citation Analysis (0)] |
| 28. | Liou AP, Khanna N, Wainer J, Reese R, West J, Caplan J, Rajagopalan H. AAV-Mediated Pancreatic Gene Therapy for Type-2 Diabetes. Metabolism. 2023;142:155476. [DOI] [Full Text] |
| 29. | Lee S, Zhang J, Saravanakumar S, Flisher MF, Grimm DR, van der Meulen T, Huising MO. Virgin β-Cells at the Neogenic Niche Proliferate Normally and Mature Slowly. Diabetes. 2021;70:1070-1083. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 4] [Cited by in RCA: 18] [Article Influence: 4.5] [Reference Citation Analysis (0)] |
| 30. | Zhang X, Gaspard JP, Mizukami Y, Li J, Graeme-Cook F, Chung DC. Overexpression of cyclin D1 in pancreatic beta-cells in vivo results in islet hyperplasia without hypoglycemia. Diabetes. 2005;54:712-719. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 57] [Cited by in RCA: 56] [Article Influence: 2.8] [Reference Citation Analysis (0)] |
| 31. | Singh M, Murriel CL, Johnson L. Genetically engineered mouse models: closing the gap between preclinical data and trial outcomes. Cancer Res. 2012;72:2695-2700. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 97] [Cited by in RCA: 95] [Article Influence: 7.3] [Reference Citation Analysis (0)] |
| 32. | Boyd JA, Barrett JC. Tumor suppressor genes: possible functions in the negative regulation of cell proliferation. Mol Carcinog. 1990;3:325-329. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 23] [Cited by in RCA: 19] [Article Influence: 0.5] [Reference Citation Analysis (0)] |
| 33. | Kim WH, Lee JW, Suh YH, Hong SH, Choi JS, Lim JH, Song JH, Gao B, Jung MH. Exposure to chronic high glucose induces beta-cell apoptosis through decreased interaction of glucokinase with mitochondria: downregulation of glucokinase in pancreatic beta-cells. Diabetes. 2005;54:2602-2611. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 110] [Cited by in RCA: 121] [Article Influence: 6.1] [Reference Citation Analysis (0)] |
| 34. | Cheruiyot A, Hollister-Lock J, Sullivan B, Pan H, Dreyfuss JM, Bonner-Weir S, Schaffer JE. Sustained hyperglycemia specifically targets translation of mRNAs for insulin secretion. J Clin Invest. 2023;134:e173280. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 8] [Cited by in RCA: 7] [Article Influence: 3.5] [Reference Citation Analysis (0)] |
| 35. | Rahman SMN, Giacca A. β-Cell Stress Pathways in Diabetes: Potential Targets for Therapy? Endocrinology. 2022;164:bqac211. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 2] [Reference Citation Analysis (0)] |
