Avicenna Journal of Medical Biochemistry. 12(1):39-46.
doi: 10.34172/ajmb.2527
Review Article
Emerging Therapeutics Targeting Cellular Stress Pathways to Mitigate End-Organ Damage in Type 1 Diabetes
Tamer A. Addissouky 1, 2, 3, * 
Author information:
1Medical Laboratories Techniques Department, AL-Mustaqbal University College, 51001, Hillah, Babil, Iraq
2New Burg El-Arab Hospital, Ministry of Health, Alexandria, Egypt
3Department of Biochemistry, Science Faculty, Menoufia University, Menoufia, Egypt
Abstract
Type 1 diabetes (T1D) is an autoimmune disease characterized by insulin deficiency and impaired glucose regulation. While daily insulin therapy is life-saving, many patients struggle to achieve optimal glycemic control, leading to microvascular complications affecting various organs, including the kidneys and the liver. This review aims to summarize the current state of knowledge regarding the pathogenesis of hepatic and renal complications in T1D, highlight recent advances in potential therapeutic targets, and provide evidence-based recommendations for mitigating end-organ damage. Chronic hyperglycemia drives diabetic complications through several interrelated mechanisms, including increased polyol pathway flux, advanced glycation end-product (AGE) formation, protein kinase C activation, and mitochondrial reactive oxygen species overproduction. In the liver, these processes contribute to non-alcoholic fatty liver disease, with up to 50% of T1D patients developing hepatic steatosis. Diabetic nephropathy, affecting 25%–40% of long-term T1D patients, is characterized by glomerular basement membrane thickening, mesangial expansion, and tubulointerstitial fibrosis. Recent innovations in T1D management include genomics and precision medicine approaches, gut microbiome modulation, nanomedicine, and artificial intelligence-driven glucose monitoring systems. Emerging immunotherapies aim to fundamentally modify the autoimmune response in T1D. Mitigating T1D complications requires intensive glycemic control, targeted pharmacotherapy, and lifestyle modifications. Emerging therapies and precision medicine approaches offer promising avenues. Ongoing research into molecular mechanisms remains crucial for developing novel interventions and improving long-term outcomes in T1D patients.
Keywords: Type 1 diabetes, Diabetic nephropathy, Diabetic liver disease, Oxidative stress, Fibrosis,
Copyright and License Information
© 2024 The Author(s); Published by Hamadan University of Medical Sciences.
This is an open-access article distributed under the terms of the Creative Commons Attribution License (
https://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any medium provided the original work is properly cited.
Please cite this article as follows: Addissouky TA. Emerging therapeutics targeting cellular stress pathways to mitigate end-organ damage in type 1 diabetes. Avicenna J Med Biochem. 2024; 12(1):39-46. doi:10.34172/ajmb.2527
Background
Type 1 diabetes (T1D) is an autoimmune disease characterized by T cell-mediated destruction of insulin-producing pancreatic beta cells, resulting in insulin deficiency and impaired glucose regulation. While daily insulin therapy is life-saving, various patients struggle to achieve optimal glycemic control. Hyperglycemia is associated with microvascular complications affecting the kidneys, eyes, nerves, and cardiovascular system. There is now recognition that the liver is also a target organ subject to injury in T1D (1-5).
Diabetic nephropathy, leading to progressive chronic kidney disease, is one of the most severe complications of T1D. It affects 25%–40% of patients with a diabetes duration over 20 years. Hyperglycemia triggers kidney damage through multiple pathways, including advanced glycation end-product (AGE) formation, polyol flux, oxidative stress (OS), and pro-inflammatory and profibrotic signal activation. This results in structural changes, including glomerular basement membrane thickening, hypertrophy, mesangial expansion, and tubulointerstitial fibrosis. Functionally, increased glomerular permeability and albuminuria occur early, followed by a declining glomerular filtration rate (6,7).
Diabetic liver disease refers to a spectrum of abnormalities, ranging from steatosis to steatohepatitis and cirrhosis. Up to 50% of T1D patients develop non-alcoholic fatty liver disease (NAFLD). Insulin deficiency promotes adipose lipolysis, increasing free fatty acid flux to the liver. Hyperglycemia also drives de novo lipogenesis while impairing mitochondrial fatty acid oxidation. This imbalance causes triglyceride accumulation in hepatocytes. A subset of patients progresses to non-alcoholic steatohepatitis (NASH) with inflammation and fibrosis, which can advance to cirrhosis. Chronic hyperglycemia promotes OS, hepatocyte death, and dysregulated repair mechanisms that contribute to liver injury. The pathophysiology underlying hepatic and renal complications in T1D is complex and multifactorial (8,9).
Chronic hyperglycemia is implicated as a major driver of diabetic complications through several interrelated mechanisms (Table 1). First, increased flux through the polyol pathway is associated with deleterious osmotic stress and OS. Second, the generation and accumulation of advanced glycation end-products (AGEs) trigger inflammatory pathways and fibrosis through receptor-mediated signaling cascades, such as the receptor for AGE (RAGE). Third, the activation of various protein kinase C isoforms by hyperglycemia alters gene transcription in pathways that promote fibrotic changes. Fourth, the mitochondrial overproduction of reactive oxygen species (ROS), induced by excess glucose metabolism, can damage proteins, lipids, and DNA. Finally, the repeated cycles of glycogen accumulation and removal in tissues such as the liver and kidneys induced by a sustained excess glucose supply create cellular stress (10,11). Overall, these multiple downstream effects of chronic hyperglycemia are understood to drive the micro- and macro-vascular complications associated with poorly controlled diabetes. Hyperglycemia also dysregulates lipid metabolism, complement pathways, endothelial function, and gut microbial communities in ways that contribute to end-organ damage. Genetic and epigenetic factors influence individual predisposition and progression rate.
Table 1.
Mechanisms of Liver Injury in Type 1 Diabetes
Mechanism
|
Description
|
Hyperglycemia |
Increased glycolysis and mitochondrial ROS production impair hepatocyte function; AGE generation triggers inflammation and fibrosis via RAGE. |
Inflammation |
Elevated cytokines, such as TNF-α, IL-1β, and IL-6, activate hepatic macrophages and stellate cells to drive fibrosis. |
OS |
ROS overproduction from hyperglycemia damages cellular components and impairs NO availability. |
Dyslipidemia |
Insulin deficiency → adipose lipolysis → hepatic FFA accumulation; de novo lipogenesis and impaired mitochondrial FA oxidation. |
Note. ROS: Reactive oxygen species; AGE: Advanced glycation end-products; RAGE: Receptor for advanced glycation end-products; TNF-α: Tumor necrosis factor alpha; IL-1β: Interleukin-1 beta; NO: Nitric oxide; FFA: Free fatty acid; OS: Oxidative stress.
Aggressive glycemic control and pharmaceutical management targeting downstream pathways can mitigate complications, but additional therapies are needed in this regard. Ongoing research into the molecular drivers of injury and potential areas for intervention, such as inflammation, OS, and the microbiome, continues to be essential for improving the long-term prognosis of T1D (12-14). This review attempts to summarize the current state of knowledge concerning the pathogenesis of these microvascular complications, highlight the most recent advances in potential remedial targets, and present evidence-based suggestions for mitigating end-organ damage in patients with T1D. It is noteworthy that the focus on emerging combination approaches makes this review unique.
Pathogenesis of Hepatic Effects in Type 1 Diabetes
While the renal complications of poorly controlled T1D are well established, there is growing recognition that the liver is also a target organ subject to injury. The mechanisms linking T1D to liver damage include hyperglycemia, inflammation, and OS. Hyperglycemia appears to be a key driver of hepatic complications in T1D. Elevated blood glucose causes increased glycolysis and mitochondrial overproduction of ROS. ROS activate profibrogenic pathways and impair hepatocyte function. Hyperglycemia also promotes the generation of AGEs. The binding of AGEs to RAGE triggers OS and inflammation. Additionally, hyperglycemia alters lipid metabolism, leading to hepatic steatosis (15,16).
Inflammation significantly contributes to hepatic pathology in T1D. Levels of proinflammatory cytokines such as tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1β), and IL-6 are elevated in T1D patients. Chronic low-grade inflammation activates Kupffer and hepatic stellate cells, promoting fibrosis. Hyperglycemia-induced mitochondrial dysfunction and endoplasmic reticulum stress further feed into inflammatory pathways (17-19). OS is both a consequence and a driver of high blood glucose and inflammation in T1D. ROS generation outpaces antioxidant defenses, modifying lipids, proteins, and DNA. This oxidative damage impairs hepatocyte function and activates hepatic stellate cells to deposit collagen. Nitric oxide bioavailability is also reduced, contributing to the vasoconstriction of hepatic sinusoids (20,21).
NAFLD is the most common hepatic manifestation of T1D. It encompasses a spectrum ranging from simple steatosis to NASH, with the risk of progression to cirrhosis. Up to 50% of T1D patients have NAFLD, with a higher prevalence in older individuals with longer disease duration. Insulin deficiency leads to upregulated lipolysis and increased free fatty acid delivery to the liver. Hyperglycemia also drives de novo lipogenesis through activating carbohydrate response element-binding proteins. Mitochondrial dysfunction impairs fatty acid oxidation. This imbalance between synthesis, uptake, and oxidation causes excessive triglyceride accumulation in hepatocytes (22-25).
NASH represents more advanced NAFLD with hepatocellular injury, inflammation, and fibrosis. The mechanisms linking T1D to NASH progression are multifactorial. Hyperglycemia induces OS and hepatocyte death. Proinflammatory cytokines activate Kupffer cells and recruit lymphocytes to the liver. The activation of hepatic stellate cells increases collagen deposition, while impaired matrix degradation leads to fibrosis. There is emerging evidence that the gut microbiome may contribute to NASH through the release of lipopolysaccharides and other pathogen-associated molecular patterns. Small intestinal bacterial overgrowth and increased gut permeability are the features of T1D that may drive hepatic inflammation (26-29).
Uncontrolled T1D accelerates the progression of chronic liver injury to cirrhosis. The repeated cycles of necroinflammation lead to disruption of the normal hepatic architecture and collagen scarring. Patients with T1D have a 3–4-fold higher risk of cirrhosis compared to those without diabetes. Cirrhosis, in turn, increases the risk of liver failure and hepatocellular carcinoma. The duration of diabetes and the degree of glycemic control are major determinants of fibrosis progression. Additional factors such as obesity, hypertension, and genetic polymorphisms likely modify susceptibility to advanced fibrosis in T1D (30-37).
Relationship Between Type 1 Diabetes and Glomerulonephritis
Diabetic nephropathy leading to chronic kidney disease is a major complication of poorly controlled T1D. While diabetic nephropathy classically manifests as nodular glomerulosclerosis on biopsy, some patients develop other patterns of injury resembling primary GN. T1D can be complicated by or contribute to multiple histological forms of GN through mechanisms linked to the metabolic and immune effects of hyperglycemia.
At a molecular level, hyperglycemia induces kidney damage through several interconnected pathways. Increased glucose flux through the polyol pathway accumulates sorbitol in cells, causing osmotic stress. The accumulation of AGEs triggers inflammation and fibrosis through receptors such as RAGE. Protein kinase C activation and hexosamine pathway flux alter gene expression profiles. The mitochondrial overproduction of ROS is both a consequence and a driver of high glucose levels (38-41). In general, these mechanisms promote glomerular damage in diabetic nephropathy. Several types of GN are associated with T1D, including some that may be secondary to hyperglycemia-induced injury (Table 2).Membranous nephropathy is characterized by glomerular basement membrane thickening and is one of the most common findings in T1D patients biopsied for proteinuria. Hyperglycemia may directly induce changes in podocytes and alter basement membrane components through non-enzymatic glycation. Increased vascular endothelial growth factor levels can disrupt the filtration barrier. Autoantibodies against podocyte antigens such as M-type phospholipase A2 receptor are also described in T1D (42,43).
Table 2.
Types of GN Associated With Type 1 Diabetes
Type
|
Key Features
|
Potential Role of T1D
|
Membranous nephropathy |
Glomerular basement membrane thickening |
Direct effects of hyperglycemia on podocytes, the glomerular basement membrane |
Mesangiocapillary GN |
Mesangial expansion, complement deposition |
Hyperglycemia-induced mesangial matrix expansion, complement activation |
Post-infectious GN |
Proliferation, crescents, and immune deposits |
Increased infections in T1D; hyperglycemia impairs immune responses |
IgA nephropathy |
Mesangial IgA deposits |
Impact of hyperglycemia on IgA; gut permeability driving autoimmunity |
Rapidly progressive GN |
Necrosis, crescents, and rapid functional decline |
Unclear; potential contributions from hyperglycemia, shared genetic risks |
Note. GN: Glomerulonephritis; IgA: Immunoglobulin A.
Post-infectious glomerulonephritis (GN) follows bacterial infections such as streptococcal pharyngitis and manifests with proliferation, crescents, and deposits. T1D patients have increased susceptibility to certain infections, which may predispose them to this immune-mediated complication. Episodes of poor glycemic control also impair neutrophil function, leading to dysfunctional immune responses to pathogens. Immunoglobulin A (IgA) nephropathy demonstrates mesangial IgA deposition with variable proliferation and scarring. The association with T1D is likely multifactorial. Hyperglycemia may directly impact IgA production and glycosylation. Increased mucosal permeability in T1D could enable food or bacterial antigens to trigger nephritogenic IgA. Shared immunogenetic risks, such as IL-10 polymorphisms, may predispose to both diseases (44).
Rapidly progressive GN has features of necrosis, crescents, and a rapid decline in kidney function. The causes include anti-neutrophil cytoplasmic antibody vasculitis, anti-glomerular basement membrane disease, and immune complex deposition. The role of T1D is controversial, though shared genetic and environmental risk factors are proposed. Hyperglycemia directly causes OS and endothelial dysfunction that could contribute to vascular inflammation (45,46).
Recent Innovations in Treating Type 1 Diabetes
The treatment of T1D requires lifelong insulin replacement therapy and careful blood glucose monitoring. However, the complex pathophysiology of T1D offers opportunities for innovative therapies beyond insulin aimed at preserving beta cell function, modulating autoimmunity, and improving glycemic control. Ongoing research is leveraging new tools in genomics, gut microbiota modulation, nanomedicine, herbal medicine, artificial intelligence (AI), and immunotherapy (47,48).
Genome, omics, and precision medicine approaches are uncovering new molecular pathways and treatment targets for T1D (Table 3). Large-scale genetic studies have identified over 60 genomic loci associated with T1D risk. Many of them contain immune-related genes, highlighting the importance of pancreatic autoimmunity. Other implicated genes regulate beta-cell function and survival. Next-generation sequencing enables high-resolution examination of the genetic and epigenetic heterogeneity in T1D. Omics technologies, including transcriptomics, proteomics, and metabolomics, provide additional layers of molecular data. For example, circulating microRNAs show promise as biomarkers of beta-cell stress and early islet autoimmunity. The goal is to precisely define disease subtypes and match individuals with customized treatment regimens (49,50).
Table 3.
Emerging Therapeutics for Type 1 Diabetes Complications
Approach
|
Examples
|
Mechanisms
|
Genomics and precision medicine |
- Gene association studies, next-generation sequencing, and omics technologies |
- Identifying molecular pathways, defining disease subtypes, and enabling customized treatment |
Gut microbiome modulation |
- Probiotics, prebiotics, antibiotics, and fecal microbiota transplantation |
- Correcting dysbiosis, modulating immunity, and repairing barrier defects |
Nanomedicine |
- Encapsulate insulin, immunosuppressants, glucose-responsive release, and targeted delivery |
- Improving oral bioavailability, enhancing cell/tissue specificity, and reducing off-target effects |
Modulating the gut microbiome is an active area of research for preventing and managing T1D. Alterations in the intestinal microbiota are detectable years before the onset of autoimmunity. The transfer of gut microbes from humans with T1D can trigger autoimmune diabetes in mice. Probiotics, prebiotics, antibiotics, and dietary changes are being studied to correct dysbiosis and support immune regulation. Fecal microbiota transplantation may hold potential. Additionally, strategies to repair intestinal barrier defects may limit the translocation of inflammatory bacterial products. Minimizing microbiome disruptions early in life could lower the risk of autoimmunity. Ongoing research is defining optimal therapeutic approaches (51-53).
Nanomedicine utilizes nanoparticles (NPs) for targeted drug delivery in T1D. Encapsulating insulin or immunosuppressants in NPs helps overcome barriers to oral administration and protects payloads from degradation. NPs can be engineered to selectively release insulin in response to glucose. Coating NPs with ligands targets them to inflamed islets or immune cells, improving bioavailability while reducing off-target effects. Additionally, NPs are being developed for the intracellular delivery of immunomodulatory genes or RNA interference constructs. Further innovation in nanotechnology could enable cell-specific therapies (54-60). Traditional herbal medicines and Chinese formulations are also being studied for T1D. Compounds such as curcumin and berberine exhibit anti-inflammatory, antioxidant, and immunomodulatory bioactivities with preclinical efficacy in diabetes models. Other natural products, such as resveratrol and epigallocatechin gallate, found in green tea (EGCG), protect beta cells from cytokine-induced apoptosis in vitro. Clinical trials are ongoing, but high-quality evidence in humans remains limited. Safety considerations regarding drug interactions exist. Nevertheless, certain herbs and supplements may emerge as useful adjuvants (61-64).
AI and machine learning tools are transforming glucose monitoring and insulin dosing for T1D patients. Closed-loop systems integrate continuous glucose monitors with insulin pumps by using algorithms to automate insulin delivery. Data analytics identify personalized patterns and tailor therapy. Machine learning can even forecast eventual glycemic deterioration and enable earlier intervention. Beyond automation, AI may gain the ability to replicate clinician decision-making for fully autonomous care. With further refinement, AI-driven platforms could revolutionize diabetes self-management (65,67). Novel immunotherapies aim to fundamentally modify the autoimmune response in T1D. Agents targeting T cells, B cells, dendritic cells, and immune signaling pathways are under development. For example, teplizumab delays T1D onset by impairing pathogenic T cells. Abatacept inhibits T cell activation, and rituximab depletes autoantibody-producing B cells. Antigen-specific immunotherapy utilizes peptides or modified autoantigens to induce tolerance. Combination approaches are also being tested to synergistically rebalance immunity. Though challenges exist, immunotherapy holds hope for durable remission (68-70).
Recommendations for Protecting the Liver and Kidneys in Patients With Type 1 Diabetes
T1D confers an increased risk for liver disease and diabetic nephropathy, leading to chronic kidney disease. While complete prevention of these complications is difficult, optimizing glycemic control and using specific pharmaceuticals and lifestyle approaches can help protect organ function (71,72). Intensive glycemic control is foundational for minimizing liver and kidney damage in T1D. The Diabetes Control and Complications Trial clearly demonstrated that maintaining hemoglobin A1c levels < 7% greatly reduced microvascular complications (73).
For the liver, improved glycemic control reduces the risk of progression from simple steatosis to steatohepatitis and fibrosis. In the kidneys, tight glucose control significantly decreases proteinuria and slows a decline in the glomerular filtration rate. Real-time continuous glucose monitoring enables patients to spend more time in the target glycemic range. Islet transplantation and advanced insulin delivery systems, such as closed-loop pumps, also improve control (74-79).
Pharmaceutical management is multifaceted. Firstly, angiotensin-converting enzyme inhibitors and angiotensin receptor blockers are critical. By blocking the renin-angiotensin-aldosterone system (RAAS), they reduce proteinuria, blood pressure, inflammation, and fibrosis. Developed RAAS inhibitors are recommended for both renal and hepatic protection in diabetes. Recently, sodium-glucose cotransporter-2 (SGLT2) inhibitors, such as empagliflozin, have emerged as novel agents to reduce complications. These inhibitors lower glucose by increasing urinary excretion, thereby reducing glomerular hyperfiltration and tubular stress. They may also have direct anti-inflammatory and antifibrotic effects. Dual RAAS and SGLT2 blockades demonstrate synergy in slowing diabetic kidney disease. The benefits for liver outcomes are being investigated (80-83).
Additionally, dyslipidemia management with statins and triglyceride-lowering agents is important. Statins reduce cardiovascular risk and may attenuate fatty liver disease. Fibrates such as gemfibrozil primarily lower triglycerides, which likely contributes to their efficacy in diabetic kidney disease trials. Omega-3 fatty acids show potential for NAFLD and diabetic nephropathy, based on small studies. Multidrug regimens are often needed for optimal treatment (84).
Lifestyle measures are crucial adjuvants in diabetes care. Weight loss in obesity provides benefits, as a higher body mass index is an independent risk factor for complications (Table 4). Smoking cessation is critical, given the synergistic adverse effects of smoking and diabetes on vessels. A Mediterranean-style diet high in polyunsaturated fats, fiber, and antioxidants such as flavonoids reduces inflammation and dyslipidemia. Aerobic and resistance exercise improves glycemic control, insulin sensitivity, and cardiovascular health. Stress management techniques such as yoga lower blood pressure and cortisol levels. Such lifestyle modifications synergize with pharmaceutical management (85-87).
Table 4.
Recommendations for Hepatorenal Protection in Type 1 Diabetes
Modality
|
Specific Examples
|
Benefits
|
Glycemic control |
- Intensive insulin therapy, continuous glucose monitors, insulin pumps, and islet transplants |
- Reducing microvascular complications |
Pharmaceuticals |
- ACEi/ARBs, SGLT2 inhibitors, statins, and fibrates |
- Decreasing proteinuria and fibrosis
- Improving lipid profile |
Lifestyle |
- Weight loss, smoking cessation, Mediterranean diet, and exercise |
- Reducing inflammation
- Improving insulin sensitivity - Lowering cardiovascular risk |
Note. ACEi: Angiotensin-converting enzyme inhibitor; ARB: Angiotensin receptor blocker; SGLT2: Sodium-glucose cotransporter-2
Conclusion
Poor glycemic control promotes oxidative damage, inflammation, and fibrosis through interconnected mechanisms, including the accumulation of sorbitol and AGEs, protein kinase C activation, and mitochondrial ROS generation. In the liver, this manifests as NAFLD and a risk for progression to cirrhosis. In the kidneys, injury to glomerular structures leads to proteinuria and declining filtration. Emerging omics, nanotechnology, microbiome, AI, and immunotherapy approaches offer opportunities for earlier diagnosis, improved monitoring, and treatments that target root molecular pathways. However, intensive glycemic control remains the foundation for minimizing end-organ damage. Further research into the complex pathophysiology underlying diabetic microvascular complications is critical for developing combination therapies that preserve hepatic and renal function in T1D.
Recommendations
To protect the liver and kidneys in patients with T1D, clinicians should focus on optimizing glycemic control through continuous glucose monitoring, insulin pumps, and islet transplantation when feasible. Angiotensin system inhibitors, SGLT2 inhibitors, statins, and triglyceride-lowering agents should be utilized for pharmaceutical management. Lifestyle measures, such as weight loss, smoking cessation, a Mediterranean diet, and exercise, have synergistic benefits. Future research should continue to uncover disease subtypes and personalized therapy approaches through omics while also exploring the potential of emerging nanotechnology, microbiome modulation, natural products, and immunotherapy. Using these evidence-based combination strategies can help mitigate diabetic nephropathy and liver disease.
Acknowledgements
The author thanks all the researchers for their studies. Moreover, we are grateful to the editors, reviewers, and readers of this journal.
Competing Interests
None declared.
Ethical Approval
Not applicable.
Funding
The corresponding author supplied all study materials. There was no further funding for this study.
References
- Popoviciu MS, Kaka N, Sethi Y, Patel N, Chopra H, Cavalu S. Type 1 diabetes mellitus and autoimmune diseases: a critical review of the association and the application of personalized medicine. J Pers Med 2023; 13(3):422. doi: 10.3390/jpm13030422 [Crossref] [ Google Scholar]
- Addissouky TA, El Tantawy El Sayed I, Ali MM, Wang Y, El Baz A, Elarabany N. Shaping the future of cardiac wellness: exploring revolutionary approaches in disease management and prevention. J Clin Cardiol 2024; 5(1):6-29. doi: 10.33696/cardiology.5.048 [Crossref] [ Google Scholar]
- Addissouky TA, Ali MM, El Tantawy El Sayed I, Wang Y. Type 1 diabetes mellitus: retrospect and prospect. Bull Natl Res Cent 2024; 48(1):42. doi: 10.1186/s42269-024-01197-z [Crossref] [ Google Scholar]
- Kropp M, Golubnitschaja O, Mazurakova A, Koklesova L, Sargheini N, Vo TK. Diabetic retinopathy as the leading cause of blindness and early predictor of cascading complications-risks and mitigation. EPMA J 2023; 14(1):21-42. doi: 10.1007/s13167-023-00314-8 [Crossref] [ Google Scholar]
- Reynolds L, Luo Z, Singh K. Diabetic complications and prospective immunotherapy. Front Immunol 2023; 14:1219598. doi: 10.3389/fimmu.2023.1219598 [Crossref] [ Google Scholar]
- Jin Q, Liu T, Qiao Y, Liu D, Yang L, Mao H. Oxidative stress and inflammation in diabetic nephropathy: role of polyphenols. Front Immunol 2023; 14:1185317. doi: 10.3389/fimmu.2023.1185317 [Crossref] [ Google Scholar]
- Antar SA, Ashour NA, Sharaky M, Khattab M, Ashour NA, Zaid RT. Diabetes mellitus: classification, mediators, and complications; A gate to identify potential targets for the development of new effective treatments. Biomed Pharmacother 2023; 168:115734. doi: 10.1016/j.biopha.2023.115734 [Crossref] [ Google Scholar]
- Mobasheri L, Ahadi M, Beheshti Namdar A, Alavi MS, Bemidinezhad A, Moshirian Farahi SM. Pathophysiology of diabetic hepatopathy and molecular mechanisms underlying the hepatoprotective effects of phytochemicals. Biomed Pharmacother 2023; 167:115502. doi: 10.1016/j.biopha.2023.115502 [Crossref] [ Google Scholar]
- Niranjan S, Phillips BE, Giannoukakis N. Uncoupling hepatic insulin resistance - hepatic inflammation to improve insulin sensitivity and to prevent impaired metabolism-associated fatty liver disease in type 2 diabetes. Front Endocrinol (Lausanne) 2023; 14:1193373. doi: 10.3389/fendo.2023.1193373 [Crossref] [ Google Scholar]
- Ramatchandirin B, Pearah A, He L. Regulation of liver glucose and lipid metabolism by transcriptional factors and coactivators. Life (Basel) 2023; 13(2):515. doi: 10.3390/life13020515 [Crossref] [ Google Scholar]
- Addissouky TA, Ali MM, El Tantawy El Sayed I, Wang Y. Revolutionary innovations in diabetes research: from biomarkers to genomic medicine. Iran J Diabetes Obes 2023;15(4):228-42 10.18502/ijdo.v15i4.14556.
- Yi M, Cruz Cisneros L, Cho EJ, Alexander M, Kimelman FA, Swentek L. Nrf2 pathway and oxidative stress as a common target for treatment of diabetes and its comorbidities. Int J Mol Sci 2024; 25(2):821. doi: 10.3390/ijms25020821 [Crossref] [ Google Scholar]
- Martemucci G, Fracchiolla G, Muraglia M, Tardugno R, Dibenedetto RS, D’Alessandro AG. Metabolic syndrome: a narrative review from the oxidative stress to the management of related diseases. Antioxidants (Basel) 2023; 12(12):2091. doi: 10.3390/antiox12122091 [Crossref] [ Google Scholar]
- Jin Q, Liu T, Qiao Y, Liu D, Yang L, Mao H. Oxidative stress and inflammation in diabetic nephropathy: role of polyphenols. Front Immunol 2023; 14:1185317. doi: 10.3389/fimmu.2023.1185317 [Crossref] [ Google Scholar]
- Atawia RT, Batori RK, Jordan CR, Kennard S, Antonova G, Bruder-Nascimento T. Type 1 diabetes impairs endothelium-dependent relaxation via increasing endothelial cell glycolysis through advanced glycation end products, PFKFB3, and Nox1-mediated mechanisms. Hypertension 2023; 80(10):2059-71. doi: 10.1161/hypertensionaha.123.21341 [Crossref] [ Google Scholar]
- Zhang Y. The essential role of glutamine metabolism in diabetic cardiomyopathy: a review. Medicine (Baltimore) 2023; 102(47):e36299. doi: 10.1097/md.0000000000036299 [Crossref] [ Google Scholar]
- Wołoszyn-Durkiewicz A, Iwaszkiewicz-Grześ D, Świętoń D, Kujawa MJ, Jankowska A, Durawa A. The complex network of cytokines and chemokines in pediatric patients with long-standing type 1 diabetes. Int J Mol Sci 2024; 25(3):1565. doi: 10.3390/ijms25031565 [Crossref] [ Google Scholar]
- Klimontov VV, Mavlianova KR, Orlov NB, Semenova JF, Korbut AI. Serum cytokines and growth factors in subjects with type 1 diabetes: associations with time in ranges and glucose variability. Biomedicines 2023; 11(10):2843. doi: 10.3390/biomedicines11102843 [Crossref] [ Google Scholar]
- Dos Santos Haber JF, Barbalho SM, Sgarbi JA, de Argollo Haber RS, de Labio RW, Laurindo LF. The relationship between type 1 diabetes mellitus, TNF-α, and IL-10 gene expression. Biomedicines 2023; 11(4):1120. doi: 10.3390/biomedicines11041120 [Crossref] [ Google Scholar]
- Addissouky TA, El Tantawy El Sayed I, Ali MM, Wang Y, El Baz A, Elarabany N. Oxidative stress and inflammation: elucidating mechanisms of smoking-attributable pathology for therapeutic targeting. Bull Natl Res Cent 2024; 48(1):16. doi: 10.1186/s42269-024-01174-6 [Crossref] [ Google Scholar]
- Addissouky TA, El Tantawy El Sayed I, Ali MM, Wang Y, El Baz A, Khalil AA. Molecular pathways in sepsis pathogenesis: recent advances and therapeutic avenues. J Cell Immunol 2024; 5(6):174-83. doi: 10.33696/immunology.5.183 [Crossref] [ Google Scholar]
- Bae J, Lee BW. Significance of diabetic kidney disease biomarkers in predicting metabolic-associated fatty liver disease. Biomedicines 2023; 11(7):1928. doi: 10.3390/biomedicines11071928 [Crossref] [ Google Scholar]
- Addissouky TA, El Agroudy AE, El-Torgoman AM, El Tantawy El Sayed I, Ibrahim EM. Efficiency of alternative markers to assess liver fibrosis levels in viral hepatitis B patients. Biomed Res 2019; 30(2):351-6. doi: 10.35841/biomedicalresearch.30-19-107 [Crossref] [ Google Scholar]
- Xu M, Wu T, Li Z, Xin G. Influence of genetically predicted autoimmune diseases on NAFLD. Front Immunol 2023; 14:1229570. doi: 10.3389/fimmu.2023.1229570 [Crossref] [ Google Scholar]
- Addissouky TA. Detecting liver fibrosis by recent reliable biomarkers in viral hepatitis patients. Am J Clin Pathol 2019; 152:S85. doi: 10.1093/ajcp/aqz117.000 [Crossref] [ Google Scholar]
- Ata F, Khan AA, Khamees I, Mohammed BZ, Barjas HH, Muthanna B. Differential evolution of diabetic ketoacidosis in adults with pre-existent versus newly diagnosed type 1 and type 2 diabetes mellitus. BMC Endocr Disord 2023; 23(1):193. doi: 10.1186/s12902-023-01446-8 [Crossref] [ Google Scholar]
- Addissouky TA, El Tantawy El Sayed I, Ali MM, Wang Y, El Baz A, Khalil AA. Latest advances in hepatocellular carcinoma management and prevention through advanced technologies. Egypt Liver J 2024; 14(1):2. doi: 10.1186/s43066-023-00306-3 [Crossref] [ Google Scholar]
- Addissouky TA, Ali MM, El Tantawy El Sayed I, Wang Y, El Baz A, Elarabany N. Preclinical promise and clinical challenges for innovative therapies targeting liver fibrogenesis. Arch Gastroenterol Res 2023; 4(1):14-23. doi: 10.33696/Gastroenterology.4.044 [Crossref] [ Google Scholar]
- Fu Y, Li S, Xiao Y, Liu G, Fang J. A metabolite perspective on the involvement of the gut microbiota in type 2 diabetes. Int J Mol Sci 2023; 24(19):14991. doi: 10.3390/ijms241914991 [Crossref] [ Google Scholar]
- Lin J, Moradi E, Salenius K, Lehtipuro S, Häkkinen T, Laiho JE. Distinct transcriptomic profiles in children prior to the appearance of type 1 diabetes-linked islet autoantibodies and following enterovirus infection. Nat Commun 2023; 14(1):7630. doi: 10.1038/s41467-023-42763-9 [Crossref] [ Google Scholar]
- Addissouky TA, Ali MM, El Tantawy El Sayed I, Wang Y. Transforming screening, risk stratification, and treatment optimization in chronic liver disease through data science and translational innovation. The Indonesian Journal of Gastroenterology Hepatology and Digestive Endoscopy 2024; 25(1):53-62. doi: 10.24871/251202453-62 [Crossref] [ Google Scholar]
- Hammad SM, Lopes-Virella MF. Circulating sphingolipids in insulin resistance, diabetes and associated complications. Int J Mol Sci 2023; 24(18):14015. doi: 10.3390/ijms241814015 [Crossref] [ Google Scholar]
- Addissouky TA, El-Agroudy AE, El-Torgoman AM, El Tantawy El Sayed I. Efficacy of biomarkers in detecting fibrosis levels of liver diseases. World J Med Sci 2019; 16(1):11-8. doi: 10.5829/idosi.wjms.2019.11.18 [Crossref] [ Google Scholar]
- Biondi G, Marrano N, Borrelli A, Rella M, D’Oria R, Genchi VA. The p66(Shc) redox protein and the emerging complications of diabetes. Int J Mol Sci 2023; 25(1):108. doi: 10.3390/ijms25010108 [Crossref] [ Google Scholar]
- Addissouky TA, El Tantawy El Sayed I, Ali MM, Alubiady MH, Wang Y. Schisandra chinensis in liver disease: exploring the mechanisms and therapeutic promise of an ancient Chinese botanical. Arch Pharmacol Ther 2024; 6(1):27-33. doi: 10.33696/Pharmacol.6.052 [Crossref] [ Google Scholar]
- Cook TW, Wilstermann AM, Mitchell JT, Arnold NE, Rajasekaran S, Bupp CP. Understanding insulin in the age of precision medicine and big data: under-explored nature of genomics. Biomolecules 2023; 13(2):257. doi: 10.3390/biom13020257 [Crossref] [ Google Scholar]
- Addissouky T, Ali MM, El Tantawy El Sayed I, Alubiady MH. Realizing the promise of artificial intelligence in hepatocellular carcinoma through opportunities and recommendations for responsible translation. J Online Inform 2024; 9(1):70-9. doi: 10.15575/join.v9i1.1297 [Crossref] [ Google Scholar]
- Esposito P, Picciotto D, Cappadona F, Costigliolo F, Russo E, Macciò L. Multifaceted relationship between diabetes and kidney diseases: beyond diabetes. World J Diabetes 2023; 14(10):1450-62. doi: 10.4239/wjd.v14.i10.1450 [Crossref] [ Google Scholar]
- Addissouky TA, Khalil AA, El Agroudy AE. Assessment of potential biomarkers for early detection and management of glomerulonephritis patients with diabetic diseases. Am J Clin Pathol 2023; 160(Suppl 1):S18-9. doi: 10.1093/ajcp/aqad150.040 [Crossref] [ Google Scholar]
- Tommerdahl KL, Kendrick J, Nelson RG, Bjornstad P. Youth versus adult-onset type 2 diabetic kidney disease: insights into currently known structural differences and the potential underlying mechanisms. Clin Sci (Lond) 2022; 136(21):1471-83. doi: 10.1042/cs20210627 [Crossref] [ Google Scholar]
- Tserga A, Saulnier-Blache JS, Palamaris K, Pouloudi D, Gakiopoulou H, Zoidakis J. Complement cascade proteins correlate with fibrosis and inflammation in early-stage type 1 diabetic kidney disease in the Ins2Akita mouse model. Int J Mol Sci 2024; 25(3):1387. doi: 10.3390/ijms25031387 [Crossref] [ Google Scholar]
- Li X, Zhang Y, Xing X, Li M, Liu Y, Xu A. Podocyte injury of diabetic nephropathy: novel mechanism discovery and therapeutic prospects. Biomed Pharmacother 2023; 168:115670. doi: 10.1016/j.biopha.2023.115670 [Crossref] [ Google Scholar]
- Karachaliou CE, Livaniou E. Immunosensors for autoimmune-disease-related biomarkers: a literature review. Sensors (Basel) 2023; 23(15):6770. doi: 10.3390/s23156770 [Crossref] [ Google Scholar]
- Mohammadi F, Yadegar A, Mardani M, Ayati A, Abolhassani H, Rezaei N. Organ-based clues for diagnosis of inborn errors of immunity: a practical guide for clinicians. Immun Inflamm Dis 2023; 11(4):e833. doi: 10.1002/iid3.833 [Crossref] [ Google Scholar]
- Juin SK, Ouseph R, Gondim DD, Jala VR, Sen U. Diabetic nephropathy and gaseous modulators. Antioxidants (Basel) 2023; 12(5):1088. doi: 10.3390/antiox12051088 [Crossref] [ Google Scholar]
- Michalek DA, Onengut-Gumuscu S, Repaske DR, Rich SS. Precision medicine in type 1 diabetes. J Indian Inst Sci 2023; 103(1):335-51. doi: 10.1007/s41745-023-00356-x [Crossref] [ Google Scholar]
- Felton JL, Griffin KJ, Oram RA, Speake C, Long SA, Onengut-Gumuscu S. Disease-modifying therapies and features linked to treatment response in type 1 diabetes prevention: a systematic review. Commun Med (Lond) 2023; 3(1):130. doi: 10.1038/s43856-023-00357-y [Crossref] [ Google Scholar]
- Morales JF, Muse R, Podichetty JT, Burton J, David S, Lang P. Disease progression joint model predicts time to type 1 diabetes onset: optimizing future type 1 diabetes prevention studies. CPT Pharmacometrics Syst Pharmacol 2023; 12(7):1016-28. doi: 10.1002/psp4.12973 [Crossref] [ Google Scholar]
- Tobias DK, Merino J, Ahmad A, Aiken C, Benham JL, Bodhini D. Second international consensus report on gaps and opportunities for the clinical translation of precision diabetes medicine. Nat Med 2023; 29(10):2438-57. doi: 10.1038/s41591-023-02502-5 [Crossref] [ Google Scholar]
- Peng X, Rao G, Li X, Tong N, Tian Y, Fu X. Preclinical models for type 1 diabetes mellitus - a practical approach for research. Int J Med Sci 2023; 20(12):1644-61. doi: 10.7150/ijms.86566 [Crossref] [ Google Scholar]
- Rosell-Mases E, Santiago A, Corral-Pujol M, Yáñez F, Varela E, Egia-Mendikute L. Mutual modulation of gut microbiota and the immune system in type 1 diabetes models. Nat Commun 2023; 14(1):7770. doi: 10.1038/s41467-023-43652-x [Crossref] [ Google Scholar]
- Yang B, Xiong Z, Lin M, Yang Y, Chen Y, Zeng J. Astragalus polysaccharides alleviate type 1 diabetes via modulating gut microbiota in mice. Int J Biol Macromol 2023; 234:123767. doi: 10.1016/j.ijbiomac.2023.123767 [Crossref] [ Google Scholar]
- Luo M, Sun M, Wang T, Zhang S, Song X, Liu X. Gut microbiota and type 1 diabetes: a two-sample bidirectional Mendelian randomization study. Front Cell Infect Microbiol 2023; 13:1163898. doi: 10.3389/fcimb.2023.1163898 [Crossref] [ Google Scholar]
- Addissouky TA, Ali MM, El Tantawy El Sayed I, Wang Y. Recent advances in diagnosing and treating Helicobacter pylori through botanical extracts and advanced technologies. Arch Pharmacol Ther 2023; 5(1):53-66. doi: 10.33696/Pharmacol.4.045 [Crossref] [ Google Scholar]
- Rahman M. Magnetic resonance imaging and iron-oxide nanoparticles in the era of personalized medicine. Nanotheranostics 2023; 7(4):424-49. doi: 10.7150/ntno.86467 [Crossref] [ Google Scholar]
- Addissouky TA, Ali MM, El Tantawy El Sayed I, Wang Y, El Baz A, Elarabany N. Risk factors, etiology, pathology, and diagnostic methods for acute kidney injury: a review study. Res Mol Med 2022; 10(4):193-204. doi: 10.32598/rmm.10.4.1265.1 [Crossref] [ Google Scholar]
- Addissouky TA, Khalil AA, El Agroudy AE. Assessing the efficacy of a modified triple drug regimen supplemented with mastic gum in the eradication of Helicobacter pylori infection. Am J Clin Pathol 2023; 160(Suppl 1):S19. doi: 10.1093/ajcp/aqad150.041 [Crossref] [ Google Scholar]
- Chu DT, Thi HV, Nguyen TT, Vu TD, Thi YV, Mani I. Nanotechnology and nucleic acid nanoparticles for treatment of metabolic disorders. OpenNano 2023; 13:100181. doi: 10.1016/j.onano.2023.100181 [Crossref] [ Google Scholar]
- Addissouky TA, Wang Y, El Tantawy El Sayed I, El Baz A, Ali MM, Khalil AA. Recent trends in Helicobacter pylori management: harnessing the power of AI and other advanced approaches. Beni Suef Univ J Basic Appl Sci 2023; 12(1):80. doi: 10.1186/s43088-023-00417-1 [Crossref] [ Google Scholar]
- Hunt NJ, Lockwood GP, Heffernan SJ, Daymond J, Ngu M, Narayanan RK. Oral nanotherapeutic formulation of insulin with reduced episodes of hypoglycaemia. Nat Nanotechnol 2024; 19(4):534-44. doi: 10.1038/s41565-023-01565-2 [Crossref] [ Google Scholar]
- Yang S, Yang H, Zhang Y. Yao-Shan of traditional Chinese medicine: an old story for metabolic health. Front Pharmacol 2023; 14:1194026. doi: 10.3389/fphar.2023.1194026 [Crossref] [ Google Scholar]
- Elmi GR, Anum K, Saleem K, Fareed R, Noreen S, Wei H. Evaluation of clinical trials of ethnomedicine used for the treatment of diabetes: a systematic review. Front Pharmacol 2023; 14:1176618. doi: 10.3389/fphar.2023.1176618 [Crossref] [ Google Scholar]
- Gang R, Matsabisa M, Okello D, Kang Y. Ethnomedicine and ethnopharmacology of medicinal plants used in the treatment of diabetes mellitus in Uganda. Appl Biol Chem 2023; 66(1):39. doi: 10.1186/s13765-023-00797-z [Crossref] [ Google Scholar]
- Huo JL, Feng Q, Pan S, Fu WJ, Liu Z, Liu Z. Diabetic cardiomyopathy: early diagnostic biomarkers, pathogenetic mechanisms, and therapeutic interventions. Cell Death Discov 2023; 9(1):256. doi: 10.1038/s41420-023-01553-4 [Crossref] [ Google Scholar]
- Cambuli VM, Baroni MG. Intelligent insulin vs. artificial intelligence for type 1 diabetes: will the real winner please stand up?. Int J Mol Sci 2023; 24(17):13139. doi: 10.3390/ijms241713139 [Crossref] [ Google Scholar]
- Ihalapathirana A, Chalkou K, Siirtola P, Tamminen S, Chandra G, Benkert P. Explainable artificial intelligence to predict clinical outcomes in type 1 diabetes and relapsing-remitting multiple sclerosis adult patients. Inform Med Unlocked 2023; 42:101349. doi: 10.1016/j.imu.2023.101349 [Crossref] [ Google Scholar]
- Guan Z, Li H, Liu R, Cai C, Liu Y, Li J. Artificial intelligence in diabetes management: advancements, opportunities, and challenges. Cell Rep Med 2023; 4(10):101213. doi: 10.1016/j.xcrm.2023.101213 [Crossref] [ Google Scholar]
- Huang M, Chen W, Wang M, Huang Y, Liu H, Ming Y. Advanced delivery strategies for immunotherapy in type I diabetes mellitus. BioDrugs 2023; 37(3):331-52. doi: 10.1007/s40259-023-00594-6 [Crossref] [ Google Scholar]
- Singh A, Afshan N, Singh A, Singh SK, Yadav S, Kumar M. Recent trends and advances in type 1 diabetes therapeutics: a comprehensive review. Eur J Cell Biol 2023; 102(2):151329. doi: 10.1016/j.ejcb.2023.151329 [Crossref] [ Google Scholar]
- Pande AK, Dutta D, Singla R. Prevention of type 1 diabetes: current perspective. Indian J Endocrinol Metab 2023; 27(4):277-85. doi: 10.4103/ijem.ijem_78_23 [Crossref] [ Google Scholar]
- Carvalho F, Lahlou RA, Pires P, Salgado M, Silva LR. Natural functional beverages as an approach to manage diabetes. Int J Mol Sci 2023; 24(23):16977. doi: 10.3390/ijms242316977 [Crossref] [ Google Scholar]
- Owusu BA, Ofori-Boateng P, Doku DT. Coping and adaptation strategies among young persons living with type 1 diabetes and their caregivers: textual and photovoice analyses. BMC Public Health 2023; 23(1):1684. doi: 10.1186/s12889-023-16573-z [Crossref] [ Google Scholar]
- Addissouky TA, Ali MM, El Tantawy El Sayed I, Wang Y. Emerging advanced approaches for diagnosis and inhibition of liver fibrogenesis. Egypt J Intern Med 2024; 36(1):19. doi: 10.1186/s43162-024-00283-y [Crossref] [ Google Scholar]
- Alassaf A, Gharaibeh L, Ibrahim S, Alkhalaileh S, Odeh R. Effect on glycemic control of an early intensive dietary structured education program for newly diagnosed children with type 1 diabetes in Jordan. Pediatr Diabetes 2023; 2023(1):7258136. doi: 10.1155/2023/7258136 [Crossref] [ Google Scholar]
- Addissouky TA, El Agroudy AE, Khalil AA. Developing a novel non-invasive serum-based diagnostic test for early detection of colorectal cancer. Am J Clin Pathol 2023; 160(Suppl 1):S17. doi: 10.1093/ajcp/aqad150.037 [Crossref] [ Google Scholar]
- Bruttomesso D, Irace C, Pozzilli P. A sub-analysis of the SAGE study in Italy indicates good glycemic control in type 1 diabetes. Nutr Metab Cardiovasc Dis 2023; 33(3):631-9. doi: 10.1016/j.numecd.2022.11.008 [Crossref] [ Google Scholar]
- Addissouky TA, El Tantawy El Sayed I, Ali MM, Wang Y, El Baz A, Khalil AA. Can vaccines stop cancer before it starts? Assessing the promise of prophylactic immunization against high-risk preneoplastic lesions. J Cell Immunol 2023; 5(4):127-40. doi: 10.33696/immunology.5.178 [Crossref] [ Google Scholar]
- Pi L, Shi X, Wang Z, Zhou Z. Effect of smartphone apps on glycemic control in young patients with type 1 diabetes: a meta-analysis. Front Public Health 2023; 11:1074946. doi: 10.3389/fpubh.2023.1074946 [Crossref] [ Google Scholar]
- Addissouky TA, Khalil AA. Detecting lung cancer stages earlier by appropriate markers rather than biopsy and other techniques. Am J Clin Pathol 2020; 154(Suppl 1):S146-7. doi: 10.1093/ajcp/aqaa161.320 [Crossref] [ Google Scholar]
- Shiel EV, Hemingway S, Burton K, King N. Self-management of type 1 diabetes in young adults: is it impeded by aspects of everyday life? A scoping review. Diabetes Metab Syndr 2023; 17(12):102918. doi: 10.1016/j.dsx.2023.102918 [Crossref] [ Google Scholar]
- Xie X, Wu C, Hao Y, Wang T, Yang Y, Cai P. Benefits and risks of drug combination therapy for diabetes mellitus and its complications: a comprehensive review. Front Endocrinol (Lausanne) 2023; 14:1301093. doi: 10.3389/fendo.2023.1301093 [Crossref] [ Google Scholar]
- Turton JL, Brinkworth GD, Parker HM, Lim D, Lee K, Rush A. Effects of a low-carbohydrate diet in adults with type 1 diabetes management: a single arm non-randomised clinical trial. PLoS One 2023; 18(7):e0288440. doi: 10.1371/journal.pone.0288440 [Crossref] [ Google Scholar]
- Maffei P, Bettini S, Busetto L, Dassie F. SGLT2 inhibitors in the management of type 1 diabetes (T1D): an update on current evidence and recommendations. Diabetes Metab Syndr Obes 2023; 16:3579-98. doi: 10.2147/dmso.s240903 [Crossref] [ Google Scholar]
- Varkevisser RD, Birnie E, Mul D, van Dijk PR, Aanstoot HJ, Wolffenbuttel BH. Type 1 diabetes management: room for improvement. J Diabetes 2023; 15(3):255-63. doi: 10.1111/1753-0407.13368 [Crossref] [ Google Scholar]
- Ogrotis I, Koufakis T, Kotsa K. Changes in the global epidemiology of type 1 diabetes in an evolving landscape of environmental factors: causes, challenges, and opportunities. Medicina (Kaunas) 2023; 59(4):668. doi: 10.3390/medicina59040668 [Crossref] [ Google Scholar]
- Clerc A. Nutrition education to type 1 diabetes patients: few changes over the time. Front Clin Diabetes Healthc 2023; 4:1243237. doi: 10.3389/fcdhc.2023.1243237 [Crossref] [ Google Scholar]
- Kueh MTW, Chew NWS, Al-Ozairi E, Le Roux CW. The emergence of obesity in type 1 diabetes. Int J Obes (Lond) 2024; 48(3):289-301. doi: 10.1038/s41366-023-01429-8 [Crossref] [ Google Scholar]