1
|
Ntetsika T, Catrina SB, Markaki I. Understanding the link between type 2 diabetes mellitus and Parkinson's disease: role of brain insulin resistance. Neural Regen Res 2025; 20:3113-3123. [PMID: 39715083 PMCID: PMC11881720 DOI: 10.4103/nrr.nrr-d-23-01910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/22/2024] [Accepted: 03/03/2024] [Indexed: 12/25/2024] Open
Abstract
Type 2 diabetes mellitus and Parkinson's disease are chronic diseases linked to a growing pandemic that affects older adults and causes significant socio-economic burden. Epidemiological data supporting a close relationship between these two aging-related diseases have resulted in the investigation of shared pathophysiological molecular mechanisms. Impaired insulin signaling in the brain has gained increasing attention during the last decade and has been suggested to contribute to the development of Parkinson's disease through the dysregulation of several pathological processes. The contribution of type 2 diabetes mellitus and insulin resistance in neurodegeneration in Parkinson's disease, with emphasis on brain insulin resistance, is extensively discussed in this article and new therapeutic strategies targeting this pathological link are presented and reviewed.
Collapse
Affiliation(s)
- Theodora Ntetsika
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Sergiu-Bogdan Catrina
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
- Center for Diabetes, Academic Specialist Center, Stockholm, Sweden
| | - Ioanna Markaki
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
- Center for Neurology, Academic Specialist Center, Stockholm, Sweden
| |
Collapse
|
2
|
Wu Z, Zhang Y, Zhou C, Zhang G, He L, Tang M. Unraveling the role of HIF-1α in allergic rhinitis: A key regulator of epithelial barrier integrity via PI3K pathway. Tissue Cell 2025; 95:102898. [PMID: 40187003 DOI: 10.1016/j.tice.2025.102898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/13/2025] [Accepted: 03/28/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Allergic rhinitis (AR) ranks among the most prevalent nasal disorders worldwide. Epithelial cells are the initial physiological barrier against allergen entry, and play a vital protective role. The precise role of hypoxia-inducible factor 1-alpha (HIF-1α) inhibitors in nasal epithelial cell injury in AR is still unknown, despite their confirmed association with nasal inflammation in AR models. METHODS An interleukin-13 (IL-13)-induced AR cell model has been employed to investigate how HIF-1α inhibition impacts nasal epithelial cells (JME/CF15). Cell viability, inflammatory cytokines, mucosal remodeling factors, and the tight junction protein zonula occludens-1 (ZO-1) were evaluated using cell counting kit-8, enzyme-linked immunosorbent assay, Western blot, and immunofluorescence. The influences of phosphoinositide 3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) pathways have been examined. RESULTS PX-478 (a HIF-1α inhibitor) alleviated IL-13-induced epithelial barrier dysfunction by upregulating ZO-1 and reducing levels of inflammatory and remodeling factors. Mechanistically, HIF-1α activated the PI3K/MEK signaling pathway, exacerbating epithelial barrier disruption and inflammatory responses. Knockdown of HIF-1α suppressed PI3K pathway activation, mitigating inflammation and restoring barrier integrity. However, these protective effects were reversed by a PI3K agonist. CONCLUSIONS HIF-1α aggravates AR by promoting inflammation, mucosal remodeling, and epithelial barrier dysfunction via PI3K pathway activation. This finding not only enriches our understanding of AR pathophysiology but also highlights HIF-1α and its downstream signaling pathways as prospective therapeutic targets for AR.
Collapse
Affiliation(s)
- Zhuo Wu
- Department of Otolaryngology Head and Neck surgery, Women and Children's Hospital of Ningbo University, China
| | - Yongbo Zhang
- Department of Otolaryngology Head and Neck surgery, Women and Children's Hospital of Ningbo University, China
| | - Changzeng Zhou
- Department of Otolaryngology Head and Neck surgery, Women and Children's Hospital of Ningbo University, China
| | - Guxuan Zhang
- Department of Otolaryngology Head and Neck surgery, Women and Children's Hospital of Ningbo University, China
| | - Lei He
- Department of Pediatric Internal Medicine, Women and Children's Hospital of Ningbo University, China
| | - Ming Tang
- Department of Otolaryngology Head and Neck surgery, Women and Children's Hospital of Ningbo University, China.
| |
Collapse
|
3
|
Tozihi M, Nourazarian A, Yousefi H, Dehghan G. Methylglyoxal-induced neuronal dysfunction: Linking diabetes to Alzheimer's disease through cytoskeletal disruption. Eur J Pharmacol 2025; 998:177526. [PMID: 40107340 DOI: 10.1016/j.ejphar.2025.177526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 03/15/2025] [Accepted: 03/17/2025] [Indexed: 03/22/2025]
Abstract
This study investigates how methylglyoxal affects Alzheimer's disease, which is common in patients with diabetes mellitus. Using SH-SY5Y cells as a model of AD, we investigated the effects of MGO on cell viability, morphology, inflammation, and stress responses. Exposure to MGO induces cytotoxicity, inflammation and oxidative stress that contribute to AD in diabetic patients. We analyzed how MGO (150-900 μM) affects SH-SY5Y cells and its effects on cell survival, gene expression, cytoskeletal integrity, stress indicators, and Aβ42 accumulation (dose- and time-dependent). MGO dramatically affected cell viability depending on the dose and exposure time. Cell death occurred via intrinsic (BAX, CASP9) and extrinsic (FAS, FASLG) apoptotic pathways. Markers related to insulin signaling such as INSR, IRS1, IRS2, SLC2A4, etc. were downregulated, whereas markers of inflammation such as TNF-α, IL-6 and oxidative markers such as HMOX1, G6PD, etc. were upregulated with MGO (P < 0.001). Changes in MAP2 and TUBB3 expression were associated with cytoskeletal damage (P < 0.01). High levels of Aβ42 and low SOD activity confirmed that oxidative stress was induced. LPS treatment exacerbated these effects (P < 0.01). The results highlight the possible role of MGO in cognitive decline associated with diabetes and suggest the need for novel treatment against MGO-related neurotoxicity.
Collapse
Affiliation(s)
- Majid Tozihi
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Alireza Nourazarian
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran.
| | - Hadi Yousefi
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran
| | - Gholamreza Dehghan
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
| |
Collapse
|
4
|
Matboli M, Khaled A, Ahmed MF, Ahmed MY, Khaled R, Elmakromy GM, Ghani AMA, El-Shafei MM, Abdelhalim MRM, Gwad AMAE. Machine learning-based stratification of prediabetes and type 2 diabetes progression. Diabetol Metab Syndr 2025; 17:227. [PMID: 40533788 PMCID: PMC12175357 DOI: 10.1186/s13098-025-01786-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 06/01/2025] [Indexed: 06/22/2025] Open
Abstract
BACKGROUND Diabetes mellitus, a global health concern with severe complications, demands early detection and precise staging for effective management. Machine learning approaches, combined with bioinformatics, offer promising avenues for enhancing diagnostic accuracy and identifying key biomarkers. METHODS This study employed a multi-class classification framework to classify patients across four health states: healthy, prediabetes, type 2 Diabetes Mellitus (T2DM) without complications, and T2DM with complications. Three models were developed using molecular markers, biochemical markers, and a combined model of both. Five machine learning classifiers were applied: Random Forest (RF), Extra Tree Classifier, Quadratic Discriminant Analysis, Naïve Bayes, and Light Gradient Boosting Machine. To improve the robustness and precision of the classification, Recursive Feature Elimination with Cross-Validation (RFECV) and a fivefold cross-validation were used. The multi-class classification approach enabled effective discrimination between the four diabetes stages. RESULTS The top contributing features identified for the combined model through RFECV included three molecular markers-miR342, NFKB1, and miR636-and two biochemical markers the albumin-to-creatinine ratio and HDLc, indicating their strong association with diabetes progression. The Extra Trees Classifier achieved the highest performance across all models, with an AUC value of 0.9985 (95% CI: [0.994-1.000]). This classifier outperformed other models, demonstrating its robustness and applicability for precise diabetes staging. CONCLUSION These findings underscore the value of integrating machine learning with molecular and biochemical markers for the accurate classification of diabetes stages, supporting a potential shift toward more personalized diabetes management.
Collapse
Affiliation(s)
- Marwa Matboli
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Ain Shams University, Cairo, 11566, Egypt.
| | - Abdelrahman Khaled
- Bioinformatics Group, Center of Informatics Sciences (CIS), School of Information Technology and Computer Sciences, Nile University, Giza, Egypt
| | - Manar Fouad Ahmed
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Ain Shams University, Cairo, 11566, Egypt
| | - Manar Yehia Ahmed
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Ain Shams University, Cairo, 11566, Egypt
| | - Radwa Khaled
- Biotechnology Department, Faculty of Science, Cairo University, Cairo, 11566, Egypt
| | - Gena M Elmakromy
- Endocrinology & Diabetes Mellitus Unit, Department of Internal Medicine, Badr University in Cairo, Badr, Egypt
| | | | - Marwa M El-Shafei
- Pathology Department, Faculty of Oral and Dental Medicine, Misr International University, Cairo, Egypt
| | | | - Asmaa Mohamed Abd El Gwad
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Ain Shams University, Cairo, 11566, Egypt
| |
Collapse
|
5
|
Nurkolis F, Romano R, Santini A. Nature's blueprint for sugar metabolism: translating bee and ant strategies into human diabetes therapies. Diabetol Metab Syndr 2025; 17:200. [PMID: 40483521 PMCID: PMC12144844 DOI: 10.1186/s13098-025-01755-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2025] [Accepted: 05/21/2025] [Indexed: 06/11/2025] Open
Abstract
Despite advances in pharmacological treatments, diabetes mellitus remains a significant global health challenge, characterized by chronic hyperglycemia and associated metabolic dysfunctions. Effective and sustainable glycemic control remains elusive, prompting exploration into unconventional sources of metabolic insights. Social insects, in particular bees and ants, exhibit remarkable physiological adaptations enabling them to thrive on carbohydrate-rich diets without developing metabolic disorders typical in humans. This review investigates the bees and ants metabolic strategies to avoid metabolic disorders like diabetes, focusing on their enzymatic pathways such as trehalose metabolism, specialized hormonal regulation involving insulin-like peptides, adipokinetic hormones, and genetic and epigenetic mechanisms underpinning their metabolic resilience. By systematically comparing these insect adaptations with human metabolic systems, the proposed study identifies potential translational applications, including engineered probiotics, gene-editing approaches, and bioactive compounds for diabetes management. Furthermore, it explores technical, ethical, and ecological considerations for translating insect-derived metabolic mechanisms into human therapies. Highlighting both opportunities and challenges, this review emphasizes the need for interdisciplinary research to responsibly integrate nature-inspired solutions into modern diabetes care.
Collapse
Affiliation(s)
- Fahrul Nurkolis
- Master of Basic Medical Science, Faculty of Medicine, Universitas Airlangga, Surabaya, 60131, Indonesia.
- Medical Research Center of Indonesia, Surabaya, 60281, Indonesia.
- State Islamic University of Sunan Kalijaga (UIN Sunan Kalijaga), Yogyakarta, 55281, Indonesia.
| | - Raffaele Romano
- Department of Agricultural Sciences, University of Napoli Federico II, Via Università 100, 80055, Portici, Italy.
| | - Antonello Santini
- Department of Pharmacy, University of Napoli Federico II, Via Domenico Montesano, 49, 80131, Naples, Italy.
| |
Collapse
|
6
|
Pattanaik SK, Anil PM, Jena S, Rath D. Interlinking diabetes and Alzheimer's disease: A pathway through medicinal plant-based treatments. JOURNAL OF ETHNOPHARMACOLOGY 2025; 351:120092. [PMID: 40484255 DOI: 10.1016/j.jep.2025.120092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 05/07/2025] [Accepted: 06/02/2025] [Indexed: 06/16/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Indian traditional medicine has long utilized medicinal plants for the management of chronic diseases such as Diabetes mellitus (DM) and related neurological complications, Alzheimer's disease (AD). The growing global burden of DM and its associated complications continues to rise; hence, it seems essential to explore effective, targeted therapies to mitigate their progression. Plant-based therapeutics have garnered significant attention for their safety, efficacy and ability to modulate multiple biological pathways. Indigenous medicinal plants, such as Sesbania grandiflora (L.) Poir., Moringa oleifera Lam., Centella asiatica (L.) Urb., Psidium guajava (L.), Aegle marmelos (L.) Corrêa, and Catharanthus roseus (L.) G. Don has been historically employed in ethnomedicine such as classical Ayurvedic texts, scientific literature and has a comprehensive and synergistic approach to address symptoms associated with DM and cognitive decline. AIM OF THE STUDY This review explores the interwined pathophysiological pathways between DM and AD, highlighting the potential of medicinal plants through (pre-clinical and clinical evidence), bridging the therapeutic gap. Additionally, we also discussed the currently used conventional antidiabetic drug that has been employed for managing AD. RESULTS In this study, six ayurvedic plants with dual activity against DM and AD are thoroughly reviewed with historical context, preclinical and clinical context. The plant's secondary metabolites have demonstrated significant antidiabetic and neuroprotective activities by regulating glucose metabolism, reducing oxidative stress, preventing amyloid-beta accumulation, etc. CONCLUSION: DM and AD share pathophysiology and multifaceted causes, requiring multi-targeted herbal remedies. The selected six Ayurveda medicinal plants showcase the dual benefits for both diseases. The obstacles, such as stability, pharmacokinetics, and safety, remain substantial barriers; addressing these challenges could constrain the clinical translation. This review demands further research to address the challenges to facilitate the effective integration of traditional medicinal knowledge with contemporary practice.
Collapse
Affiliation(s)
- Sandesh Kumar Pattanaik
- Department of Pharmacology, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan Deemed to be University, Bhubaneswar, Odisha, 751003, India.
| | - Prajapati Manisha Anil
- School of Pharmacy, Sharda University, Knowledge Park-3, Greater Noida, U.P, 201306, India.
| | - Sudipta Jena
- Centre of Biotechnology, Siksha 'O' Anusandhan Deemed to be University, Bhubaneswar, Odisha, 751003, India.
| | - Diptirani Rath
- Department of Pharmacology, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan Deemed to be University, Bhubaneswar, Odisha, 751003, India.
| |
Collapse
|
7
|
Pomares-Bri I, Roca M, Borrás F, Wabitsch M, Lahoz A, Micol V, Herranz-López M. Polyphenols reverse hyperglycemia-induced adipocyte dysfunction: A Metabolomic and Lipidomic study of efficacy. Food Res Int 2025; 211:116453. [PMID: 40356124 DOI: 10.1016/j.foodres.2025.116453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 03/20/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025]
Abstract
Hyperglycemia leads to metabolic dysfunction in human adipocytes, characterized by decreased AKT phosphorylation, downregulation of glycolysis, TCA cycle, and amino acid metabolism, as well as altered lipid profiles. This study aimed to elucidate these metabolic alterations and evaluate the potential therapeutic effects of selected polyphenols. Comprehensive metabolic profiling revealed profound disruptions, including impaired carbon metabolism, amino acids, and lipids associated with obesity. Importantly, treatment with polyphenols, particularly verbascoside and ferulic acid, effectively mitigated these metabolic disturbances, restoring adipocyte homeostasis. The polyphenols increased metabolites from carbon metabolism and amino acids, improving glycolysis, the TCA cycle, and related pathways. They also modulated lipid profiles that are negatively associated with obesity and related diseases. These findings provide valuable insights into the metabolic pathways underlying adipocyte dysfunction in hyperglycemia and highlight the therapeutic potential of polyphenols in ameliorating metabolic disorders.
Collapse
Affiliation(s)
- Irene Pomares-Bri
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche, Universidad Miguel Hernández (UMH), 03202 Elche, Spain
| | - Marta Roca
- Analytical Unit, Medical Research Institute-Hospital La Fe, Av. Fernando Abril Martorell 106, Valencia, 46026, Spain
| | - Fernando Borrás
- Statistics and Operative Research Department, UMH, Avda, Universidad s/n, 03202, Elche, Spain
| | - Martin Wabitsch
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Endocrinology and Diabetes, Ulm University Medical Center Ulm, Ulm, Germany
| | - Agustín Lahoz
- Biomarkers and Precision Medicine Unit, Health Research Institute-Hospital La Fe, 46026, Valencia, Spain
| | - Vicente Micol
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche, Universidad Miguel Hernández (UMH), 03202 Elche, Spain.; CIBER: CB12/03/30038, Fisiopatología de la Obesidad y la Nutrición, CIBERobn, Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain.
| | - María Herranz-López
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche, Universidad Miguel Hernández (UMH), 03202 Elche, Spain
| |
Collapse
|
8
|
Foroozanmehr B, Hemmati MA, Yaribeygi H, Karav S, Jamialahmadi T, Sahebkar A. Parkinson's disease and brain insulin signaling: Mechanisms and potential role of GLP-1 mimetics. Brain Res 2025; 1862:149738. [PMID: 40449678 DOI: 10.1016/j.brainres.2025.149738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 03/21/2025] [Accepted: 05/28/2025] [Indexed: 06/03/2025]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder characterized primarily by the degeneration of dopaminergic neurons in the substantia nigra pars compacta. The pathophysiology of PD is complex and multifactorial involving genetic factors, oxidative stress, mitochondrial dysfunction, impaired protein clearance, and neuroinflammation but recent evidence emphasizes the role of impaired brain insulin signaling. Insulin is a metabolic hormone with extensive effects on metabolic substrates but recent studies have demonstrated that it is also involved in central signaling pathways and induces different brain areas related to food craving, motor activities, cognitive abilities, and emotional feelings. Hence it has been suggested that induction of brain insulin sensitivity may be a promising treatment for PD. Glucagon-like peptide-1 (GLP-1) mimetics are a new-generation class of antidiabetics that normalize glucose homeostasis via several pathways. Recent studies suggest extra-glycemic benefits for GLP-1 mimetics against PD. GLP-1 mimetics can prevent or slow PD progression. Additionally, these agents can improve cognitive functions by improving brain insulin signaling pathways. In this review, we aim to highlight the role of brain insulin signaling in PD pathophysiology and discuss the possible benefits of GLP-1 mimetics in PD management.
Collapse
Affiliation(s)
- Behina Foroozanmehr
- Student Research Committee, Semnan University of Medical Sciences, Semnan, Iran
| | | | - Habib Yaribeygi
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Canakkale Onsekiz Mart University, Canakkale 17100, Turkey
| | - Tannaz Jamialahmadi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
9
|
Gouveia F, Pérez MC, Bicker J, Silva A, Santos AE, Pereira CF, Camins A, Falcão A, Cruz T, Ettcheto M, Fortuna A. Protective effects of irbesartan against neurodegeneration in APP/PS1 mice: Unraveling its triple anti-apoptotic, anti-inflammatory and anti-oxidant action. Biomed Pharmacother 2025; 188:118167. [PMID: 40414000 DOI: 10.1016/j.biopha.2025.118167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 05/07/2025] [Accepted: 05/09/2025] [Indexed: 05/27/2025] Open
Abstract
Alzheimer's disease (AD) involves a complex interplay between amyloid-β plaques, oxidative stress, neuroinflammation and apoptosis, suggesting that multi-target therapies may be more effective than single-target treatments. Angiotensin receptor blockers (ARBs) and angiotensin-converting enzyme inhibitors (ACEIs) have shown protective effects against AD in hypertensive patients. However, those poor blood-brain barrier (BBB) permeability exhibit limited efficacy in neurodegenerative disorders. This study investigated the neuroprotective potential of three ARBs and one ACEI using a combined in vitro and in vivo approach. In N2a and N2a-APPswe cell lines, irbesartan showed the most robust effects, notably increasing p-AKT and HMOX1 levels. Based on these results, irbesartan was selected for the following in vivo studies and administered intranasally (40 mg/kg) to APP/PS1 male mice for 10 weeks. Treated animals showed significant improvements in memory performance, as measured by the novel object recognition test and the Morris water maze. Additionally, irbesartan enhanced dendritic spine density, restored mitochondrial bioenergetics and BBB integrity, and reduced apoptotic markers. These effects were accompanied by a marked reduction in oxidative stress and neuroinflammation. Overall, these results support the potential of intranasal irbesartan as a promising multi-target therapeutic strategy for AD, capable of modulating key pathological features, including synaptic dysfunction, mitochondrial dysfunction, oxidative stress, apoptosis, and neuroinflammation.
Collapse
Affiliation(s)
- Filipa Gouveia
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; CIBIT/ICNAS - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal; Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain; Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
| | - Marina Carrasco Pérez
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain; Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain; Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Carlos III Health Institute, Madrid, Spain; Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Joana Bicker
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; CIBIT/ICNAS - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Ana Silva
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Armanda E Santos
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Laboratory of Biochemistry and Biology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Cláudia Fragão Pereira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Antoni Camins
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain; Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain; Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Carlos III Health Institute, Madrid, Spain; Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Amílcar Falcão
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; CIBIT/ICNAS - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Teresa Cruz
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Miren Ettcheto
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain; Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain; Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Carlos III Health Institute, Madrid, Spain; Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain.
| | - Ana Fortuna
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; CIBIT/ICNAS - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
10
|
Li M, Peng Y, Shi Y, Liu Y, Zhang J. Advancements in the study of DLK1 in the pathogenesis of diabetes. Life Sci 2025; 369:123535. [PMID: 40054732 DOI: 10.1016/j.lfs.2025.123535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 03/02/2025] [Accepted: 03/03/2025] [Indexed: 03/30/2025]
Abstract
DLK1, as a membrane-bound protein, has been extensively studied in the field of cancer research. As a ligand downstream of the Notch pathway, it broadly influences developmental and metabolic processes in the body. With deeper research, it has been found that DLK1 can induce the synthesis and secretion of insulin through the ERK and AKT pathways, playing a crucial role in the development of metabolic diseases. Diabetes mellitus (DM) is a chronic metabolic disorder characterized by insufficient insulin production by the pancreas or inadequate utilization of insulin by the body. This article aims to review the relationship between DLK1 and diabetes, recent research advancements, and to discuss future research directions and challenges.
Collapse
Affiliation(s)
- Min Li
- School of Bioengineering, Zunyi Medical University, Zhuhai 519041, China
| | - Yanqiu Peng
- School of Bioengineering, Zunyi Medical University, Zhuhai 519041, China
| | - Yuke Shi
- School of Bioengineering, Zunyi Medical University, Zhuhai 519041, China
| | - Yunfei Liu
- School of Bioengineering, Zunyi Medical University, Zhuhai 519041, China
| | - Jian Zhang
- School of Bioengineering, Zunyi Medical University, Zhuhai 519041, China.
| |
Collapse
|
11
|
Pilar Fernández-Figares Vicioso M, Riutord Sbert P, López-González ÁA, Ramírez-Manent JI, Del Barrio Fernández JL, Herrero MTV. Risk of Insulin Resistance: Comparison of the Commerce vs. Industry Sector and Associated Variables. Diseases 2025; 13:150. [PMID: 40422582 DOI: 10.3390/diseases13050150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Revised: 05/04/2025] [Accepted: 05/12/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND Insulin resistance (IR) is a key metabolic alteration that precedes type 2 diabetes and is closely linked to obesity and lifestyle factors. Occupational context may influence IR risk through variations in physical activity, diet, and socioeconomic determinants. OBJECTIVE To compare the risk of insulin resistance between workers in the commerce and industry sectors and identify associated sociodemographic and lifestyle factors, in order to improve their occupational health. METHODS This cross-sectional study analyzed data from 56,856 Spanish workers, assessing four IR-related indices: Triglyceride-Glucose Index (TyG), TyG-BMI (Triglyceride-Glucose Body Mass Index), Metabolic Score for Insulin Resistance (METS-IR), and the Single-Point Insulin Sensitivity Estimator (SPISE-IR). The analysis was stratified by sex and sector (commerce vs. industry) and included assessments of age, education level, physical activity, adherence to the Mediterranean diet, and smoking status. Multinomial logistic regressions were performed to determine the factors associated with high IR scores. RESULTS Across all IR indicators, industry workers-particularly men-presented higher mean values and greater prevalence of high-risk scores compared to those in commerce. Women showed lower values overall but also reflected sector-based differences. In both sexes, non-physical activity, non-adherence to the Mediterranean diet, and smoking were consistently associated with higher IR risk. Males exhibited significantly higher odds of elevated TyG (OR = 2.59, 95% CI: 2.41-2.78), while physical inactivity and poor diet emerged as the most powerful modifiable predictors across all scales (e.g., OR = 10.45 for TyG, OR = 12.33 for TyG-BMI). Industry sector was independently associated with higher odds of insulin resistance compared to commerce. CONCLUSIONS Insulin resistance is more prevalent among industrial workers, especially men and those with unhealthy lifestyles. Occupational health strategies should target sector-specific risk profiles, emphasizing physical activity and dietary interventions.
Collapse
Affiliation(s)
| | - Pere Riutord Sbert
- ADEMA-Health Group of IUNICS, University of Balearic Islands, 07122 Palma, Spain
| | - Ángel Arturo López-González
- Obesity and Metabolic Syndrome Group, Spanish Association of Specialists in Occupational Medicine, 28012 Madrid, Spain
- ADEMA-Health Group of IUNICS, University of Balearic Islands, 07122 Palma, Spain
| | - José Ignacio Ramírez-Manent
- ADEMA-Health Group of IUNICS, University of Balearic Islands, 07122 Palma, Spain
- Health Research Institute of the Balearic Islands (IDISBA), 07120 Palma, Spain
- Faculty of Medicine, University of Balearic Islands, 07122 Palma, Spain
| | | | - María Teófila Vicente Herrero
- Obesity and Metabolic Syndrome Group, Spanish Association of Specialists in Occupational Medicine, 28012 Madrid, Spain
- ADEMA-Health Group of IUNICS, University of Balearic Islands, 07122 Palma, Spain
| |
Collapse
|
12
|
Aliyu U, Toor SM, Abdalhakam I, Elrayess MA, Abou−Samra AB, Albagha OME. Evaluating indices of insulin resistance and estimating the prevalence of insulin resistance in a large biobank cohort. Front Endocrinol (Lausanne) 2025; 16:1591677. [PMID: 40421243 PMCID: PMC12104043 DOI: 10.3389/fendo.2025.1591677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Accepted: 04/11/2025] [Indexed: 05/28/2025] Open
Abstract
Introduction Insulin resistance (IR) is involved in the pathogenesis of various metabolic disorders. Several surrogate indices of IR have been proposed. We assessed the performance of seven clinically relevant indirect measures of IR and estimated the prevalence of IR in a large population-based cohort. Methods The study was conducted on fasting individuals from the Qatar biobank (QBB) participants (n = 7,875). Individuals were considered insulin sensitive (IS) if lean, not diagnosed with diabetes, no hypertriglyceridemia, and not on lipid-lowering drugs, while individuals with Type 2 diabetes (T2D) were considered insulin resistant (IR). Cut-offs were determined as the top or lowest quartile values in the IS participants. The performance of IR indices was based on area under the curve (AUC), sensitivity and specificity. Results The cut-off for HOMA-IR was determined at 1.878, HOMA2-IR (insulin); 1.128, HOMA2-IR (C-peptide); 1.307, QUICKI; 0.347, TyG; 8.281, McAi; 7.727 and 1.718 for TG/HDL. All IR indices analyzed yielded AUC values ranging from 0.83 to 0.92. TyG was the most robust measure for IR (AUC = 0.92, Sensitivity = 0.90, Specificity = 0.79). The overall prevalence of IR in Qatar was estimated at ~51 - 65%. Conclusions TyG index was the most robust index for determining IR in the Qatari population. The proposed cut-offs could serve as a reference in Middle Eastern populations for IR screening.
Collapse
Affiliation(s)
- Usama Aliyu
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Salman M. Toor
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | | | - Mohamed A. Elrayess
- Biomedical Research Center, QU Health, Qatar University, Doha, Qatar
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| | | | - Omar M. E. Albagha
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
13
|
Phochantachinda S, Photcharatinnakorn P, Chatchaisak D, Sakcamduang W, Chansawhang A, Buranasinsup S, Suemanotham N, Chantong B. Plasma-based proteomics analysis of molecular pathways in canine diabetes mellitus after astaxanthin supplementation. PLoS One 2025; 20:e0321509. [PMID: 40333882 PMCID: PMC12057883 DOI: 10.1371/journal.pone.0321509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 03/06/2025] [Indexed: 05/09/2025] Open
Abstract
The hyperglycemic state in diabetes mellitus induces oxidative stress and inflammation, contributing to diabetic tissue damage and associated complications. Astaxanthin, a potent antioxidant carotenoid, has been investigated for its potential to prevent and manage diabetes across various species; however, its effect on client-owned dogs remains poorly studied. This study explored the impact of astaxanthin supplementation on canine diabetes mellitus using a proteomics approach. A total of 18 client-owned dogs were enrolled: 6 dogs with diabetes mellitus and 12 clinically healthy dogs. The diabetic dogs received their standard treatment regimen along with daily oral supplementation of 12 mg of astaxanthin (1.5-2.4 mg/kg) for 90 days. Plasma samples were collected at the beginning and end of the study period for proteomics analysis. After astaxanthin supplementation, significant alterations in the expression of proteins associated with the complement system, coagulation cascade, JAK-STAT signaling, and protein kinase C signaling (all of which contribute to inflammation and oxidative stress) were observed. Astaxanthin exhibited potential for reducing diabetes-associated complications, such as insulin resistance, vascular dysfunction, nephropathy, and cardiac issues, even though it did not affect clinical parameters (hematology, plasma biochemistry, blood glucose, and serum fructosamine). These findings suggest that astaxanthin may be a valuable complementary therapy for managing diabetes-related complications in canines.
Collapse
Affiliation(s)
- Sataporn Phochantachinda
- Department of Clinical Sciences and Public Health, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
| | | | - Duangthip Chatchaisak
- Department of Clinical Sciences and Public Health, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
| | - Walasinee Sakcamduang
- Department of Clinical Sciences and Public Health, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
| | - Anchana Chansawhang
- The Center for Veterinary Diagnosis, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
| | - Shutipen Buranasinsup
- Department of Pre-Clinic and Applied Animal Science, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
| | - Namphung Suemanotham
- Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Boonrat Chantong
- Department of Pre-Clinic and Applied Animal Science, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
| |
Collapse
|
14
|
Sindhwani R, Bora KS, Hazra S. The dual challenge of diabesity: pathophysiology, management, and future directions. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4891-4912. [PMID: 39680103 DOI: 10.1007/s00210-024-03713-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/07/2024] [Indexed: 12/17/2024]
Abstract
Diabesity, the concurrent occurrence of obesity and type-2 diabetes mellitus (T2DM), represents a pressing global health challenge characterized by intricate pathophysiological mechanisms and a wide range of associated comorbidities. Central to its development are insulin resistance, metabolic syndrome, and chronic low-grade inflammation mediated by dysregulated adipokine secretion and systemic metabolic dysfunction. These mechanisms underpin the progression of diabesity and its complications, including cardiovascular disease and hypertension. Management strategies encompass lifestyle interventions focusing on tailored dietary modifications and structured physical activity, pharmacological treatments targeting both glycemic control and weight loss, and surgical interventions such as bariatric surgery, which have demonstrated efficacy in achieving durable outcomes. Clinical trials and meta-analyses underscore the comparative advantages of different treatment modalities in terms of efficacy, safety, and sustainability. Moreover, long-term follow-up studies emphasize the critical need for sustained multidisciplinary interventions to prevent relapse and enhance patient outcomes. Future advancements in management include exploring precision medicine approaches that integrate individual metabolic profiles, lifestyle factors, and emerging therapeutic innovations. A multidisciplinary approach combining advanced therapeutic strategies and patient-centered care remains pivotal for optimizing management and improving prognoses for individuals with diabesity. This review highlights the complex interplay between obesity and T2DM, offering comprehensive insights into their pathophysiology, clinical presentation, and management paradigms.
Collapse
Affiliation(s)
- Ritika Sindhwani
- University Institute of Pharma Sciences, Chandigarh University, Mohali, 140413, Punjab, India
| | - Kundan Singh Bora
- University Institute of Pharma Sciences, Chandigarh University, Mohali, 140413, Punjab, India.
| | - Subhajit Hazra
- University Institute of Pharma Sciences, Chandigarh University, Mohali, 140413, Punjab, India
| |
Collapse
|
15
|
Dong Z, She X, Ma J, Chen Q, Gao Y, Chen R, Qin H, Shen B, Gao H. The E3 Ligase NEDD4L Prevents Colorectal Cancer Liver Metastasis via Degradation of PRMT5 to Inhibit the AKT/mTOR Signaling Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2504704. [PMID: 40279519 DOI: 10.1002/advs.202504704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Indexed: 04/27/2025]
Abstract
Colorectal cancer is the second most common cause of cancer mortality worldwide, and liver metastasis is the major cause of death of patients with colorectal cancer. Dysfunctional E3 ligase activity has recently been shown to be associated with colorectal cancer. However, the key E3 ligases affecting colorectal cancer liver metastasis remain unknown. Therefore, an shRNA library targeting 156 E3 ubiquitin ligases has been used to perform an in vivo loss-of-function screen of a human colorectal cancer cell line in a mouse model of liver metastasis. The screen reveals that neural precursor cell expressed developmentally down-regulated gene 4-like (NEDD4L) knockdown promotes colorectal cancer liver metastasis. Mechanistic studies reveal that NEDD4L binds to the PPNAY motif in protein arginine methyltransferase 5 (PRMT5) and ubiquitinates PRMT5 to promote its degradation. PRMT5 degradation attenuates the arginine methylation of AKT1 to inhibit the AKT/mTOR signaling pathway. The effect of NEDD4L decreases colorectal cancer cell proliferation to suppress colonization. This study is the first to show that PRMT5 is a substrate of NEDD4L and reveals not only the metastasis-inhibiting function of NEDD4L but also a novel mechanism by which NEDD4L prevents colorectal cancer liver metastasis. These findings may provide a new preventive strategy for liver metastasis.
Collapse
Affiliation(s)
- Zhewen Dong
- Tongji University Cancer Center and Research Institute of Intestinal Diseases, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, P. R. China
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, P. R. China
| | - Xiaofei She
- Tongji University Cancer Center and Research Institute of Intestinal Diseases, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, P. R. China
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, P. R. China
| | - Junxian Ma
- Tongji University Cancer Center and Research Institute of Intestinal Diseases, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, P. R. China
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, P. R. China
| | - Qian Chen
- Tongji University Cancer Center and Research Institute of Intestinal Diseases, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, P. R. China
| | - Yaqun Gao
- Tongji University Cancer Center and Research Institute of Intestinal Diseases, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, P. R. China
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, P. R. China
| | - Ruiyan Chen
- Tongji University Cancer Center and Research Institute of Intestinal Diseases, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, P. R. China
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, P. R. China
| | - Huanlong Qin
- Tongji University Cancer Center and Research Institute of Intestinal Diseases, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, P. R. China
| | - Bing Shen
- Tongji University Cancer Center and Research Institute of Intestinal Diseases, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, P. R. China
- Department of Urology and Urologic Cancer Institute, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, P. R. China
| | - Hua Gao
- Tongji University Cancer Center and Research Institute of Intestinal Diseases, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, P. R. China
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, P. R. China
| |
Collapse
|
16
|
Roham PH, Yadav SS, Senthilnathan B, Potdar P, Roy S, Sharma S. Explaining Type 2 Diabetes with Transcriptomic Signatures of Pancreatic β-Cell Dysfunction and Death Induced by Human Islet Amyloid Polypeptide. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2025. [PMID: 40261698 DOI: 10.1089/omi.2024.0216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Amyloid deposits formed by misfolding and aggregation of human islet amyloid polypeptide (hIAPP) are one of the key pathophysiological features of type 2 diabetes mellitus (T2DM) and have been associated with the loss of function and viability of the pancreatic β-cells. The molecular processes by which hIAPP induces cytotoxicity in these cells are not well understood. To the best of our knowledge, this is the first report describing findings from the combined analysis of Affymetrix microarray and high-throughput sequencing (HTS) Gene Expression Omnibus (GEO) datasets of hIAPP-transgenic (Tg) mice islets. In brief, using GEO data, we compared in silico the pancreatic islets obtained from hIAPP-Tg and wild-type mice. Affymetrix microarray datasets (GSE84423, GSE85380, and GSE94672) and HTS datasets (GSE135276 and GSE148809) were chosen. Weighted gene coexpression network analysis was performed using GSE135276 to identify the coexpressed gene networks and establish a correlation pattern between gene modules and hIAPP overexpression under hyperglycemic conditions. Subsequently, we analyzed differential gene expression with the remaining datasets. Network analysis was performed to identify hub genes and the associated pathways using Cytoscape. Key findings from the present study include identification of seven hub genes, namely, Ins2, Agt, Jun, Fos, CD44, Igf1, and Ppar-γ, significantly involved in the process(es) of insulin synthesis and secretion, development of insulin resistance, oxidative stress, inflammation, mitophagy, and apoptosis. In conclusion, we propose that these hub genes can help explain T2DM pathogenesis and can be potentially utilized to develop therapeutic interventions targeting hIAPP for clinical management of T2DM.
Collapse
Affiliation(s)
- Pratiksha H Roham
- Department of Biotechnology, Savitribai Phule Pune University, Pune, India
| | | | | | - Pranjali Potdar
- Department of Biotechnology, Savitribai Phule Pune University, Pune, India
| | - Sujata Roy
- Department of Biotechnology, Rajalakshmi Engineering College, Thandalam, India
| | - Shilpy Sharma
- Department of Biotechnology, Savitribai Phule Pune University, Pune, India
| |
Collapse
|
17
|
Chen T, Zheng W, Zhang Y, Xu Q. The relationship between triglyceride-glucose index and serum neurofilament light chain: Findings from NHANES 2013-2014. PLoS One 2025; 20:e0321226. [PMID: 40208889 PMCID: PMC11984729 DOI: 10.1371/journal.pone.0321226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/03/2025] [Indexed: 04/12/2025] Open
Abstract
BACKGROUND The Triglyceride-Glucose (TyG) index has become a reliable indicator for evaluating the level of insulin resistance, a pivotal factor in both metabolic and neurodegenerative disorders. Serum neurofilament light chain (sNfL) serves as a responsive biomarker for detecting neuroaxonal injury. Despite this, the interplay between the TyG index and sNfL levels has not been sufficiently investigated. The aim of this research is to scrutinize the correlation between TyG index and sNfL levels across a substantial, population-based cohort. METHODS Our study involved an examination of the dataset from the 2013-2014 round of the National Health and Nutrition Examination Survey (NHANES), encompassing a total of 2029 enrolled subjects. The TyG index was calculated using fasting triglycerides and glucose levels. Multivariable linear regression models were conducted to evaluate the relationship between TyG index and sNfL levels, adjusting for potential confounders such as age, sex, race, BMI, hypertension, stroke, congestive heart failure, alcohol consumption and NHHR (Non-High-Density Lipoprotein Cholesterol to High-Density Lipoprotein Cholesterol Ratio). Nonlinear associations were investigated using regression models based on restricted cubic splines (RCS). RESULTS Both the unadjusted and adjusted regression analyses revealed a substantial positive correlation between the TyG index and ln-sNfL levels. After accounting for all covariates, each unit increase in the TyG index was associated with a 0.15 (95% CI: 0.02-0.27, p = 0.04) increase in ln-sNfL levels. RCS analysis revealed a nonlinear relationship, with a threshold around a TyG index value of 9.63, beyond which ln-sNfL levels increased more rapidly. The association was consistent across subgroups. CONCLUSION Our study links higher TyG index with increased sNfL levels, indicating insulin resistance's role in neuroaxonal injury. The nonlinear relationship implies a heightened risk of neurodegeneration beyond a certain insulin resistance threshold. This underscores the need for early metabolic interventions to prevent neurodegenerative processes.
Collapse
Affiliation(s)
- Tong Chen
- Department of Neurology, Xuzhou Central Hospital, Xuzhou, Jiangsu, China
| | - Wei Zheng
- Department of Neurology, Xuzhou Central Hospital, Xuzhou, Jiangsu, China
| | - Yan Zhang
- Department of Outpatient, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Qian Xu
- Department of Neurology, Xuzhou Central Hospital, Xuzhou, Jiangsu, China
| |
Collapse
|
18
|
Lan C, Li Y, Weng Z, Pan W, Lin W, Jiang Z, Yang L, Shen X. TLR4 mediates lipotoxic β-cell dysfunction by inhibiting the TMEM24/PI3K/AKT pathway. Acta Biochim Biophys Sin (Shanghai) 2025. [PMID: 40170616 DOI: 10.3724/abbs.2025045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2025] Open
Abstract
Immune imbalance is the core pathophysiological mechanism of the deterioration of β-cell function driven by lipid metabolism disorders. Toll-like receptor 4 (TLR4) inflammatory signaling is a key pathway that mediates lipotoxic injury in β-cells, but the underlying mechanism needs to be further elucidated. Transmembrane protein 24 (TMEM24) is a key transporter that regulates pulsatile insulin secretion, but its pathophysiology in lipotoxicity remains unclear. In this study, we investigate whether TLR4-mediated lipotoxicity is affected by the inhibition of TMEM24 expression. The PPI network shows that TLR4 is associated with both insulin secretion and ER stress proteins in islets from obese rats. Using in vitro lipotoxic β-cell models, we found that TMEM24 is the target signal of palmitic acid (PA)-induced insulin secretion impairment in islet β-cells, and TLR4 plays a mediating role in this process. Mechanistically, TLR4 mediates lipotoxicity by binding to TMEM24 and downregulating its protein expression to suppress PI3K/AKT signaling, leading to β-cell dysfunction. TLR4 knockout ameliorates islet function impairment through TMEM24/PI3K/AKT signaling in HFD-induced obese rats. Taken together, our results show that TLR4 mediates lipotoxicity in islet β-cells by inhibiting the TMEM24/PI3K/AKT pathway, and the mechanism of TLR4-mediated lipotoxicity is elucidated from the perspective of insulin vesicular secretion.
Collapse
Affiliation(s)
- Chao Lan
- Department of Endocrinology, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Department of Endocrinology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
- Clinical Research Center for Metabolic Diseases of Fujian Province, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Fujian Key Laboratory of Glycolipid and Bone Mineral Metabolism, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Diabetes Research Institute of Fujian Province, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Metabolic Diseases Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Yan Li
- Department of Endocrinology, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Zhiyan Weng
- Department of Endocrinology, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Wei Pan
- Department of Endocrinology, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Wanxin Lin
- Department of Endocrinology, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Zhen Jiang
- Department of Endocrinology, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Department of Endocrinology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
- Clinical Research Center for Metabolic Diseases of Fujian Province, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Fujian Key Laboratory of Glycolipid and Bone Mineral Metabolism, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Diabetes Research Institute of Fujian Province, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Metabolic Diseases Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Liyong Yang
- Department of Endocrinology, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Department of Endocrinology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
- Clinical Research Center for Metabolic Diseases of Fujian Province, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Fujian Key Laboratory of Glycolipid and Bone Mineral Metabolism, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Diabetes Research Institute of Fujian Province, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Metabolic Diseases Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Ximei Shen
- Department of Endocrinology, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Department of Endocrinology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
- Clinical Research Center for Metabolic Diseases of Fujian Province, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Fujian Key Laboratory of Glycolipid and Bone Mineral Metabolism, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Diabetes Research Institute of Fujian Province, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Metabolic Diseases Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| |
Collapse
|
19
|
Shi X, Xu L, Ren J, Jing L, Zhao X. Triglyceride-glucose index: a novel prognostic marker for sepsis-associated encephalopathy severity and outcomes. Front Neurol 2025; 16:1468419. [PMID: 40242624 PMCID: PMC12000067 DOI: 10.3389/fneur.2025.1468419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
Background Sepsis-associated encephalopathy (SAE) is a complex condition with variable outcomes. This study investigates the potential of the Triglyceride-glucose (TyG) index as a marker for disease severity and prognosis in SAE patients. Methods We conducted a retrospective cohort study using data from the Medical Information Mart for Intensive Care (MIMIC-IV) database. Patients with sepsis who were admitted to the intensive care unit (ICU) were categorized into two groups based on the occurrence of SAE. Key clinical outcomes were 90-day survival (primary outcome) and length of ICU and hospital stays, as well as the use of vasoactive medications (secondary outcomes). The TyG index was calculated, and its association with disease severity scores and patient outcomes was analyzed using statistical methods, including survival analysis, Cox regression, and correlation analyses. Results The study population's median age was 65.96 years, predominantly male (60.1%). Higher TyG index scores correlated with elevated clinical severity scores (APSIII, LODS, OASIS, SAPSII, and CCI) and increased ICU and hospital stay durations. TyG index categorization revealed significant differences in 90-day survival probabilities, with "high TyG" associated with a 25% increased mortality risk compared to "low TyG." Furthermore, TyG index showed a moderate positive correlation with ICU stay duration and use of norepinephrine and vasopressin, but not with dopamine and epinephrine use. Conclusion The TyG index is a significant independent predictor of disease severity and prognosis in SAE patients. High TyG levels correlate with worse clinical outcomes and increased mortality risk, suggesting its potential as a valuable tool in managing SAE.
Collapse
|
20
|
Shnayder NA, Pekarets NA, Pekarets NI, Dmitrenko DV, Grechkina VV, Petrova MM, Al-Zamil M, Nasyrova RF. MicroRNAs as Epigenetic Biomarkers of Pathogenetic Mechanisms of the Metabolic Syndrome Induced by Antiseizure Medications: Systematic Review. J Clin Med 2025; 14:2432. [PMID: 40217882 PMCID: PMC11989458 DOI: 10.3390/jcm14072432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/27/2025] [Accepted: 03/30/2025] [Indexed: 04/14/2025] Open
Abstract
Antiseizure medication (ASM) induced metabolic syndrome (AIMetS) is a common adverse drug reaction (ADR) of pharmacotherapy for epilepsy and psychiatric disorders. However, the sensitivity and specificity of blood biomarkers may be insufficient due to the influence of combined pathology, concomitant diseases, and the peculiarities of the metabolism of ASMs in patients with epilepsy. Methods: The presented results of experimental and clinical studies of microRNAs (miRs) as epigenetic biomarkers of MetS and AIMetS, which were entered into the different databases, were analyzed for the last decade (2014-2024). Results: A systematic review demonstrated that miRs can act as promising epigenetic biomarkers of key AIMetS domains. However, the results of the review demonstrated the variable role of various miRs and their paralogs in the pathogenesis of AIMetS. Therefore, as part of this study, an miRs signature was proposed that allows us to assess the risk of developing and the severity of AIMetS as low risk, medium risk, and high risk. Conclusions: The mechanisms of development and biomarkers of AIMetS are an actual problem of epileptology, which is still far from being resolved. The development of panels (signatures) of epigenetic biomarkers of this widespread ADR may help to increase the safety of pharmacotherapy of epilepsy. However, to increase the sensitivity and specificity of circulating miRs in the blood as biomarkers of AIMetS, it is necessary to conduct "bridge" studies in order to replicate the results of preclinical and clinical studies into real clinical practice.
Collapse
Affiliation(s)
- Natalia A. Shnayder
- Institute of Personalized Psychiatry and Neurology, V.M. Bekhterev National Medical Research Center for Psychiatry and Neurology, 3 Bekhterev St., 192019 St. Petersburg, Russia; (N.A.P.); (V.V.G.); (R.F.N.)
- Shared Core Facilities “Molecular and Cell Technologies”, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 1 Partizan Zheleznyak St., 660022 Krasnoyarsk, Russia; (D.V.D.); (M.M.P.)
| | - Nikolai A. Pekarets
- Institute of Personalized Psychiatry and Neurology, V.M. Bekhterev National Medical Research Center for Psychiatry and Neurology, 3 Bekhterev St., 192019 St. Petersburg, Russia; (N.A.P.); (V.V.G.); (R.F.N.)
| | - Natalia I. Pekarets
- Department of Psychiatry and Clinical Psychology, Irkutsk State Medical University, 1 Krasny Vosstaniya St., 664003 Irkutsk, Russia;
| | - Diana V. Dmitrenko
- Shared Core Facilities “Molecular and Cell Technologies”, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 1 Partizan Zheleznyak St., 660022 Krasnoyarsk, Russia; (D.V.D.); (M.M.P.)
| | - Violetta V. Grechkina
- Institute of Personalized Psychiatry and Neurology, V.M. Bekhterev National Medical Research Center for Psychiatry and Neurology, 3 Bekhterev St., 192019 St. Petersburg, Russia; (N.A.P.); (V.V.G.); (R.F.N.)
| | - Marina M. Petrova
- Shared Core Facilities “Molecular and Cell Technologies”, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 1 Partizan Zheleznyak St., 660022 Krasnoyarsk, Russia; (D.V.D.); (M.M.P.)
| | - Mustafa Al-Zamil
- Department of Physiotherapy, Faculty of Continuing Medical Education, Peoples’ Friendship University of Russia, 117198 Moscow, Russia;
| | - Regina F. Nasyrova
- Institute of Personalized Psychiatry and Neurology, V.M. Bekhterev National Medical Research Center for Psychiatry and Neurology, 3 Bekhterev St., 192019 St. Petersburg, Russia; (N.A.P.); (V.V.G.); (R.F.N.)
- Department of Psychiatry, General and Clinical Psychology, Tula State University, 92 Lenin Ave., 300012 Tula, Russia
| |
Collapse
|
21
|
Sorial MES, Abdelghany RM, El Sayed NSED. Modulation of the cognitive impairment associated with Alzheimer's disease by valproic acid: possible drug repurposing. Inflammopharmacology 2025; 33:2083-2094. [PMID: 40108007 PMCID: PMC11991970 DOI: 10.1007/s10787-025-01695-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 02/05/2025] [Indexed: 03/22/2025]
Abstract
Sporadic Alzheimer's disease is a progressive neurodegenerative disorder affecting the central nervous system. Its main two hallmarks are extracellular deposition of aggregated amyloid beta resulting in senile plaques and intracellular hyperphosphorylated tau proteins forming neuro-fibrillary tangles. As those processes are promoted by the glycogen synthase kinase-3 enzyme, GSK3 inhibitors may be of therapeutic value in SAD. GSK3 is also inhibited by the action of insulin on insulin signaling. Insulin receptor desensitization in the brain is hypothesized to cause inhibition of insulin signaling pathway that ultimately causes cognitive deficits seen in SAD. In extant research, induction of cognitive impairment is achieved by intracerebroventricular injection of streptozotocin-a diabetogenic compound that causes desensitization to insulin receptors in the brain leading to the appearance of most of the SAD signs and symptoms. Valproic acid -a histone deacetylase inhibitor and anti-epileptic drug-has been recently studied in the management of SAD as a possible GSK3 inhibitor. Accordingly, the aim of the present study is to explore the role of multiple VPA doses on the downstream effects of the insulin signaling pathway in ICV STZ-injected mice and suggest a possible mechanism of VPA action. ICV STZ-injected mice showed deficiency in short- and long-term memory as well as increased anxiety, as established by open field test, Modified Y-maze, Morris water maze, and elevated plus maze neurobehavioral tests.
Collapse
Affiliation(s)
- Mirna Ezzat Sedrak Sorial
- Pharmacology and Toxicology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo-GUC, Cairo, Egypt.
| | - Ragwa Mansour Abdelghany
- Pharmacology and Toxicology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo-GUC, Cairo, Egypt
| | | |
Collapse
|
22
|
Liu S, Liu T, Li J, Hong J, Moosavi-Movahedi AA, Wei J. Type 2 Diabetes Mellitus Exacerbates Pathological Processes of Parkinson's Disease: Insights from Signaling Pathways Mediated by Insulin Receptors. Neurosci Bull 2025; 41:676-690. [PMID: 39754628 PMCID: PMC11978575 DOI: 10.1007/s12264-024-01342-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 10/15/2024] [Indexed: 01/06/2025] Open
Abstract
Parkinson's disease (PD), a chronic and common neurodegenerative disease, is characterized by the progressive loss of dopaminergic neurons in the dense part of the substantia nigra and abnormal aggregation of alpha-synuclein. Type 2 diabetes mellitus (T2DM) is a metabolic disease characterized by chronic insulin resistance and deficiency in insulin secretion. Extensive evidence has confirmed shared pathogenic mechanisms underlying PD and T2DM, such as oxidative stress caused by insulin resistance, mitochondrial dysfunction, inflammation, and disorders of energy metabolism. Conventional drugs for treating T2DM, such as metformin and glucagon-like peptide-1 receptor agonists, affect nerve repair. Even drugs for treating PD, such as levodopa, can affect insulin secretion. This review summarizes the relationship between PD and T2DM and related therapeutic drugs from the perspective of insulin signaling pathways in the brain.
Collapse
Affiliation(s)
- Shufen Liu
- Center for Translational Neuromedicine and Neurology, School of Life Sciences, Institute for Brain Sciences Research, Henan University, Huaihe Hospital of Henan University, Kaifeng, 475004, China
- School of Life Sciences, Institute for Brain Sciences Research, Henan University, Kaifeng, 475004, China
| | - Tingting Liu
- Center for Translational Neuromedicine and Neurology, School of Life Sciences, Institute for Brain Sciences Research, Henan University, Huaihe Hospital of Henan University, Kaifeng, 475004, China
- School of Life Sciences, Institute for Brain Sciences Research, Henan University, Kaifeng, 475004, China
| | - Jingwen Li
- School of Life Sciences, Institute for Brain Sciences Research, Henan University, Kaifeng, 475004, China
| | - Jun Hong
- School of Life Sciences, Institute for Brain Sciences Research, Henan University, Kaifeng, 475004, China
| | | | - Jianshe Wei
- Center for Translational Neuromedicine and Neurology, School of Life Sciences, Institute for Brain Sciences Research, Henan University, Huaihe Hospital of Henan University, Kaifeng, 475004, China.
- School of Life Sciences, Institute for Brain Sciences Research, Henan University, Kaifeng, 475004, China.
| |
Collapse
|
23
|
Rai V, Deepu V, Agrawal DK. Targeting RAGE-signaling pathways in the repair of rotator-cuff injury. Mol Cell Biochem 2025; 480:2539-2554. [PMID: 39395136 PMCID: PMC11961478 DOI: 10.1007/s11010-024-05132-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 10/01/2024] [Indexed: 10/14/2024]
Abstract
Rotator cuff injury (RCI) is a common musculoskeletal problem that can have a significant impact on the quality of life and functional abilities of those affected. Novel therapies, including proteomics-based, stem cells, platelet-rich plasma, and exosomes, are being developed to promote rotator-cuff healing. The receptor for advanced glycation end-products (RAGE) is a multifunctional receptor that is expressed on several cell types and is implicated in several physiologic and pathological processes, such as tissue repair, inflammation, and degeneration. Because of its capacity to bind with a variety of ligands and initiate signaling pathways that lead to inflammatory responses in RCI, RAGE plays a crucial role in inflammation. In this critical review article, we discussed the role of RAGE-mediated persistent inflammation in RCI followed by novel factors including PKCs, TIRAP, DIAPH1, and factors related to muscle injury with their therapeutic potential in RCI. These factors involve various aspects of muscle injury and signaling and the possibility of targeting these factors to improve the clinical outcomes in RCI still needs further investigation.
Collapse
Affiliation(s)
- Vikrant Rai
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA
| | - Vinitha Deepu
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA.
| |
Collapse
|
24
|
Cognacq G, Attwood JE, DeLuca GC. Traumatic Brain Injury and Alzheimer's Disease: A Shared Neurovascular Hypothesis. Neurosci Insights 2025; 20:26331055251323292. [PMID: 40124421 PMCID: PMC11926848 DOI: 10.1177/26331055251323292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 02/10/2025] [Indexed: 03/25/2025] Open
Abstract
Traumatic brain injury (TBI) is a modifiable risk factor for Alzheimer's disease (AD). TBI and AD share several histopathological hallmarks: namely, beta-amyloid aggregation, tau hyperphosphorylation, and plasma protein infiltration. The relative contributions of these proteinopathies and their interplay in the pathogenesis of both conditions remains unclear although important differences are emerging. This review synthesises emerging evidence for the critical role of the neurovascular unit in mediating protein accumulation and neurotoxicity in both TBI and AD. We propose a shared pathogenic cascade centred on a neurovascular unit, in which increased blood-brain barrier permeability induces a series of noxious mechanisms leading to neuronal loss, synaptic dysfunction and ultimately cognitive dysfunction in both conditions. We explore the application of this hypothesis to outstanding research questions and potential treatments for TBI and AD, as well as other neurodegenerative and neuroinflammatory conditions. Limitations of this hypothesis, including the challenges of establishing a causal relationship between neurovascular damage and proteinopathies, are also discussed.
Collapse
Affiliation(s)
- Gabrielle Cognacq
- John Radcliffe Hospital, University of Oxford, Headley Way, Oxford, Oxfordshire, UK
| | - Jonathan E Attwood
- Nuffield Department of Clinical Neurosciences, Level 6 West Wing, John Radcliffe Hospital, Headley Way, Oxford, Oxfordshire, UK
| | - Gabriele C DeLuca
- Nuffield Department of Clinical Neurosciences, Level 6 West Wing, John Radcliffe Hospital, Headley Way, Oxford, Oxfordshire, UK
| |
Collapse
|
25
|
Kılınç GE, Vergi Y. Nutritional Approach to Diabetic Sarcopenia: A Comprehensive Review. Curr Nutr Rep 2025; 14:48. [PMID: 40106009 PMCID: PMC11922993 DOI: 10.1007/s13668-025-00637-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2025] [Indexed: 03/22/2025]
Abstract
PURPOSE OF THE REVIEW The aim of this review is to discuss and evaluate diabetic sarcopenia (DS) and its relationship with nutrition by discussing the mechanisms of diabetic sarcopenia in detail and comprehensively reviewing the literature. RECENT FINDINGS Type 2 diabetes (T2DM) affects approximately 25% of people aged 50 years and over and indicates a significant the cost of health for the elderly. Nutrition is an important part of these treatment approaches, and in this review, the literature was comprehensively reviewed, focusing on understanding the mechanisms of DS and discussing its relationship with nutrition. A comprehensive search was conducted on Web of Science, Google Scholar, Scopus, Science Direct, and PubMed from inception up to July 2024. The aim of nutritional treatment for DS is to improve muscle mass, muscle strength and physical performance while improving diabetes-related metabolic risk and glucose levels. In this context, it is important to determine energy intake in individuals with DS according to calorie intake exceeding 30 kcal/kg. For these individuals, a protein intake of at least 1-1.2 g/kg/day is recommended, with an emphasis on the number and timing of meals and a nutritional pattern rich in branched chain amino acids (BCAA). In addition, it is important to adopt a diet rich in antioxidants and to choose diet patterns that contain sufficient levels of macro and micronutrients. The Mediterranean diet model can be a good diet option for individuals with DS. Comprehensive studies in this field are needed so that clinicians can make specific dietary recommendations for DS.
Collapse
Affiliation(s)
- Gül Eda Kılınç
- Faculty of Health Sciences, Department of Nutrition and Dietetics, Ondokuz Mayıs University, Samsun, Turkey.
| | - Yeliz Vergi
- Faculty of Health Sciences, Department of Nutrition and Dietetics, Mersin University, Mersin, Turkey
| |
Collapse
|
26
|
Ahn B. Advances in Insulin Resistance-Molecular Mechanisms, Therapeutic Targets, and Future Directions. Int J Mol Sci 2025; 26:2574. [PMID: 40141215 PMCID: PMC11942056 DOI: 10.3390/ijms26062574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Accepted: 03/09/2025] [Indexed: 03/28/2025] Open
Abstract
The development of insulin resistance (IR) is characterized by a series of metabolic disturbances, including, but not limited to, impaired glucose uptake, increased blood sugar levels, and disrupted lipid metabolism [...].
Collapse
Affiliation(s)
- Byungyong Ahn
- Department of Food Science and Nutrition, University of Ulsan, Ulsan 44610, Republic of Korea; ; Tel.: +82-52-259-2373; Fax: +82-52-259-1698
- Basic-Clinical Convergence Research Institute, University of Ulsan, Ulsan 44610, Republic of Korea
| |
Collapse
|
27
|
Liu R, Liu H, Li R, Li C, Xiao H, Chen S. Identification and differential expression analysis of microRNAs in the liver and spleen tissues of Yunnan Zebu and Holstein cattle. Trop Anim Health Prod 2025; 57:96. [PMID: 40038148 DOI: 10.1007/s11250-025-04357-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 02/19/2025] [Indexed: 03/06/2025]
Abstract
Extensive research has shown that miRNAs play a crucial role in regulating biological processes within organisms. This study analyzed miRNAs differentially expressed and potentially associated with immune function and tissue-specific expression in Yunnan Zebu and Holstein cattle. To achieve this, 20 liver and spleen tissue samples from the two cattle breeds were collected for high-throughput miRNA sequencing, with the liver tissue as a reference. The findings revealed that bta-miR-122 and bta-miR-143 are the most abundantly expressed known miRNAs in the liver and spleen tissues of Yunnan Zebu and Holstein cattle, respectively. Additionally, AC_000181.1_27195 and AC_000168.1_14386 emerged as the most highly expressed novel miRNAs in these tissues, exhibiting significant homology with human hsa-miR-3591-3p and hsa-miR-126-3p, respectively. Six differentially expressed miRNAs were selected for validation using stem-loop RT-qPCR, and the results were largely consistent with the high-throughput sequencing data. Functional enrichment analysis of the target genes of these known miRNAs revealed their association with critical metabolic and immune-related pathways in cattle, such as the insulin pathway, mTOR signaling, IFN-γ pathway, and signaling mechanisms mediated by IL5 and IL3. These results enriched the miRNA database for liver and spleen tissues of Yunnan Zebu and Holstein cattle.
Collapse
Affiliation(s)
- Rong Liu
- School of Ecology and Environmental Science, Yunnan University, Kunming, 650500, China
- School of Life Sciences, Yunnan University, Kunming, 650500, China
| | - Hesong Liu
- School of Life Sciences, Yunnan University, Kunming, 650500, China
| | - Rong Li
- College of Life Science, Yunnan Normal University, Kunming, 650500, China
| | - Chunqing Li
- School of Ecology and Environmental Science, Yunnan University, Kunming, 650500, China
| | - Heng Xiao
- School of Ecology and Environmental Science, Yunnan University, Kunming, 650500, China
| | - Shanyuan Chen
- School of Ecology and Environmental Science, Yunnan University, Kunming, 650500, China.
| |
Collapse
|
28
|
Zhang X, Zhang Z. Insulin receptor tyrosine kinase substrate in health and disease (Review). Mol Med Rep 2025; 31:72. [PMID: 39930824 PMCID: PMC11795247 DOI: 10.3892/mmr.2025.13437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 12/19/2024] [Indexed: 02/14/2025] Open
Abstract
Insulin receptor (IR) tyrosine kinase substrate (IRTKS) was first identified >20 years ago as a tyrosine‑phosphorylated IR substrate and subsequently characterized as a protein containing an inverse‑Bin‑amphiphysin‑Rvs domain. Subsequent research has shown that IRTKS functions as a scaffold protein with multiple domains, which results in diverse functions in a variety of cell activities. For example, IRTKS plays roles in regulating the formation of membrane protrusions; triggering pathogen‑driven actin assembly; modulating insulin signaling, antiviral immunity and embryonic development; and promoting tumor occurrence and progression. It is also a candidate forensic biomarker of hypothermia. Nevertheless, a systematic summary of the biological functions of IRTKS and its underlying molecular mechanism is lacking. Therefore, the present review provides a comprehensive summary of the latest advancements in IRTKS research, thereby establishing a framework for understanding the contribution of IRTKS to diverse cell processes.
Collapse
Affiliation(s)
- Xueyan Zhang
- Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Zhewen Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
29
|
He J, Li X, Yu H, Xu C, Tian R, Zhou P, Yin Z. Inflammation-induced PFKFB3-mediated glycolysis promoting myometrium contraction through the PI3K-Akt-mTOR pathway in preterm birth mice. Am J Physiol Cell Physiol 2025; 328:C895-C907. [PMID: 39907705 DOI: 10.1152/ajpcell.00704.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/21/2024] [Accepted: 01/06/2025] [Indexed: 02/06/2025]
Abstract
Inflammation is a significant risk factor for preterm birth. Inflammation enhances glycolytic processes in various cell types and contributes to the development of myometrial contractions. However, the potential of inflammation to activate glycolysis in pregnant murine uterine smooth muscle cells (mUSMCs) and its role in promoting inflammatory preterm birth remain unexplored. In this study, lipopolysaccharide was employed to establish both cell and animal inflammation models. We found that inflammation of mUSMCs during late pregnancy could initiate glycolysis and promote cell contraction. Subsequently, the inhibition of glycolysis using the glycolysis inhibitor 2-deoxyglucose can reverse inflammation-induced cell contraction. The expression of 6-phosphofructokinase 2 kinase (PFKFB3) was significantly upregulated in mUSMCs following lipopolysaccharide stimulation. In addition, lactate accumulation and enhanced contraction were observed. Inhibition of PFKFB3 reversed the lactate accumulation and enhanced contraction induced by inflammation. We also found that inflammation activated the phosphatidylinositol 3-kinase (PI3K)-protein kinase B (Akt)-mammalian target of the rapamycin (mTOR) pathway, leading to the upregulation of PFKFB3 expression. The PI3K-Akt pathway inhibitor LY294002 and the mTOR pathway inhibitor rapamycin effectively inhibited the upregulation of PFKFB3 protein expression, lactate production, and the enhancement of cell contraction induced by lipopolysaccharide. This study indicates that inflammation regulates PFKFB3 through the PI3K-Akt-mTOR pathway, which enhances the glycolytic process in pregnant mUSMCs, ultimately leading to myometrial contraction.NEW & NOTEWORTHY Expression of PFKFB3, a key enzyme in glycolysis, was significantly upregulated both in the mUSMCs and myometrium of mice during late pregnancy after lipopolysaccharide stimulation. Activation of the PI3K-Akt-mTOR pathway enhanced PFKFB3 expression, which is involved in the initiation of glycolysis. Inflammation-activated PFKFB3 via the PI3K-Akt-mTOR pathway, which enhances the cellular glycolytic process and thus promotes myometrium contraction in pregnancy.
Collapse
Affiliation(s)
- Jing He
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
- NHC Key Laboratory of the Study of Abnormal Gametes and the Reproductive Tract, Anhui Medical University, Hefei, People's Republic of China
- Department of Obstetrics and Gynecology, Anqing Medical Center of Anhui Medical University, Anqing, People's Republic of China
| | - Xuan Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
- NHC Key Laboratory of the Study of Abnormal Gametes and the Reproductive Tract, Anhui Medical University, Hefei, People's Republic of China
- Anhui Province Key Laboratory of Reproductive Disorders and Obstetrics and Gynaecology Diseases, Hefei, People's Republic of China
| | - Huihui Yu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
- NHC Key Laboratory of the Study of Abnormal Gametes and the Reproductive Tract, Anhui Medical University, Hefei, People's Republic of China
- Anhui Province Key Laboratory of Reproductive Disorders and Obstetrics and Gynaecology Diseases, Hefei, People's Republic of China
| | - Chenyi Xu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
- Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, Hefei, People's Republic of China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, People's Republic of China
| | - Ruixian Tian
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
- Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, Hefei, People's Republic of China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, People's Republic of China
| | - Ping Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
- NHC Key Laboratory of the Study of Abnormal Gametes and the Reproductive Tract, Anhui Medical University, Hefei, People's Republic of China
- Anhui Province Key Laboratory of Reproductive Disorders and Obstetrics and Gynaecology Diseases, Hefei, People's Republic of China
| | - Zongzhi Yin
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
- NHC Key Laboratory of the Study of Abnormal Gametes and the Reproductive Tract, Anhui Medical University, Hefei, People's Republic of China
- Anhui Province Key Laboratory of Reproductive Disorders and Obstetrics and Gynaecology Diseases, Hefei, People's Republic of China
- Center for Big Data and Population Health of IHM, Hefei, People's Republic of China
| |
Collapse
|
30
|
Huang S, Lu Y, Fang W, Huang Y, Li Q, Xu Z. Neurodegenerative diseases and neuroinflammation-induced apoptosis. Open Life Sci 2025; 20:20221051. [PMID: 40026360 PMCID: PMC11868719 DOI: 10.1515/biol-2022-1051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 11/30/2024] [Accepted: 12/27/2024] [Indexed: 03/05/2025] Open
Abstract
Neuroinflammation represents a critical pathway in the brain for the clearance of foreign bodies and the maintenance of homeostasis. When the neuroinflammatory process is dysregulate, such as the over-activation of microglia, which results in the excessive accumulation of free oxygen and inflammatory factors in the brain, among other factors, it can lead to an imbalance in homeostasis and the development of various diseases. Recent research has indicated that the development of numerous neurodegenerative diseases is closely associated with neuroinflammation. The pathogenesis of neuroinflammation in the brain is intricate, involving alterations in numerous genes and proteins, as well as the activation and inhibition of signaling pathways. Furthermore, excessive inflammation can result in neuronal cell apoptosis, which can further exacerbate the extent of the disease. This article presents a summary of recent studies on the relationship between neuronal apoptosis caused by excessive neuroinflammation and neurodegenerative diseases. The aim is to identify the link between the two and to provide new ideas and targets for exploring the pathogenesis, as well as the prevention and treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Shi Huang
- School of Clinical Medicine, Wannan Medical College, 241002, Wuhu, Anhui, China
| | - Yaxin Lu
- School of Pharmaceutical Sciences, Wannan Medical College,
241002, Wuhu, Anhui, China
| | - Wanzhen Fang
- School of Stomatology, Wannan Medical College,
241002, Wuhu, Anhui, China
| | - Yanjiao Huang
- Human Anatomy Experimental Training Center, School of Basic Medical Science, Wannan Medical College, 241002, Wuhu, Anhui, China
| | - Qiang Li
- Human Anatomy Experimental Training Center, School of Basic Medical Science, Wannan Medical College, 241002, Wuhu, Anhui, China
| | - Zhiliang Xu
- Department of Human Anatomy, School of Basic Medical Science, Wannan Medical College, 241002, Wuhu, Anhui, China
- Anhui Province Key Laboratory of Basic Research and Translation of Aging-Related Diseases, Wannan Medical College, Wuhu, 241002, Anhui, China
| |
Collapse
|
31
|
Promsan S, Pengrattanachot N, Phengpol N, Sutthasupha P, Thongnak LO, Jaikumkao K, Lungkaphin A. Agomelatine Mitigates Kidney Damage in Obese Insulin-Resistant Rats by Inhibiting Inflammation and Necroptosis via the TNF-α/NF-ĸB/p-RIPK3 Pathway. Int J Mol Sci 2025; 26:1940. [PMID: 40076566 PMCID: PMC11900133 DOI: 10.3390/ijms26051940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/20/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Obesity is a risk factor for chronic kidney disease. The expansion of adipose tissues in obesity induces insulin resistance and low-grade systemic inflammation, promoting kidney damage. Our previous studies have demonstrated that agomelatine (AGOM) exerts renoprotective effects in experimental models of obesity and insulin resistance through various mechanisms, including the attenuation of ER stress and oxidative stress. This study aimed to further explore the effects of agomelatine on renal inflammation, insulin signaling, and necroptosis in obese, insulin-resistant rats. Obesity was induced in rats with a high-fat diet for 16 weeks, followed by 4 weeks of treatment with 20 mg kg-1 day-1 of AGOM or 10 mg kg-1 day-1 of pioglitazone (PIO). The results showed that insulin resistance was improved after treatment with AGOM and PIO, as demonstrated by the reduction in fasting plasma glucose, insulin, and HOMA-IR. Both treatments restored the levels of renal insulin signaling proteins. Moreover, AGOM inhibited TNFα, TNFR1, NF-ĸB, COX2, and IL1β, which attenuated the necroptosis-related proteins RIPK3 and MLKL. AGOM also prevented kidney DNA fragmentation, as detected by the TUNEL assay. In an obese condition, the level of the tight junction protein claudin-1 (CLDN1) was enhanced after being treated with AGOM. In conclusion, the novel mechanisms associated with AGOM and involved in limiting kidney injury were the inhibition of the TNFα/NF-ĸB/p-RIPK3 pathway and a reduction in inflammation and necroptosis. This suggested that AGOM could be an effective treatment for inhibiting kidney dysfunction in cases of obesity and insulin resistance. These findings open new avenues for the management of renal dysfunction, with implications for personalized medicine.
Collapse
Affiliation(s)
- Sasivimon Promsan
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Intravaroros Road, Chiang Mai 50200, Thailand; (S.P.); (N.P.); (N.P.); (P.S.)
- Division of Physiology, School of Medical Science, University of Phayao, Phayao 56000, Thailand
| | - Nattavadee Pengrattanachot
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Intravaroros Road, Chiang Mai 50200, Thailand; (S.P.); (N.P.); (N.P.); (P.S.)
| | - Nichakorn Phengpol
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Intravaroros Road, Chiang Mai 50200, Thailand; (S.P.); (N.P.); (N.P.); (P.S.)
| | - Prempree Sutthasupha
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Intravaroros Road, Chiang Mai 50200, Thailand; (S.P.); (N.P.); (N.P.); (P.S.)
| | - La-ongdao Thongnak
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok 10210, Thailand;
| | - Krit Jaikumkao
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Anusorn Lungkaphin
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Intravaroros Road, Chiang Mai 50200, Thailand; (S.P.); (N.P.); (N.P.); (P.S.)
- Functional Foods for Health and Disease, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Functional Food Research Center for Well-Being, Multidisciplinary Research Institute, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
32
|
Samavarchitehrani A, Mercantepe F, Behnoush AH, Klisic A. Exploring the TyG Index and the Homeostasis Model Assessment of Insulin Resistance as Insulin Resistance Markers: Implications for Fibromyalgia Management and Understanding-A Narrative Review. Diagnostics (Basel) 2025; 15:494. [PMID: 40002645 PMCID: PMC11854647 DOI: 10.3390/diagnostics15040494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/09/2025] [Accepted: 02/09/2025] [Indexed: 02/27/2025] Open
Abstract
Fibromyalgia (FM) is a chronic musculoskeletal disease with a higher prevalence among women. To date, there has been no definitive laboratory or imaging assessment for FM, and hence, the diagnosis criteria for FM remained based on subjective assessment of symptoms with high overlap with other rheumatological disorders. Many patients with FM suffer from metabolic disorders leading to insulin resistance (IR). There have been several methods to assess IR, among which the Homeostasis Model Assessment of Insulin Resistance (HOMA-IR) and the triglyceride-glucose (TyG) index have been used more frequently, with the latter being more available and cost-effective. As higher IR has been reported for patients with FM with various mechanisms, in this review, we sought to investigate the association between IR and FM using the current evidence. One of the possible underlying mechanisms of this association might be mitochondrial dysfunction and oxidative stress observed in IR conditions and its role in FM. Studies have also shown that IR indices are higher in patients with FM, compared to healthy controls, while higher HOMA-IR levels were also reported for higher severities of FM based on Fibromyalgia Impact Questionnaire-Revised (FIQR) scores. While these findings suggest the possible involvement of IR in FM pathophysiology and add to the value of IR measurement in FM clinical assessment, further large-scale studies are needed to establish a definitive causal association between them.
Collapse
Affiliation(s)
| | - Filiz Mercantepe
- Department of Endocrinology and Metabolism, Faculty of Medicine, Recep Tayyip Erdogan University, Rize 53100, Turkey;
| | | | - Aleksandra Klisic
- Faculty of Medicine, University of Montenegro, 81000 Podgorica, Montenegro;
- Center for Laboratory Diagnostics, Primary Health Care Center, 81000 Podgorica, Montenegro
| |
Collapse
|
33
|
Qiang M, Chen Z, Liu H, Dong J, Gong K, Zhang X, Huo P, Zhu J, Shao Y, Ma J, Zhang B, Liu W, Tang M. Targeting the PI3K/AKT/mTOR pathway in lung cancer: mechanisms and therapeutic targeting. Front Pharmacol 2025; 16:1516583. [PMID: 40041495 PMCID: PMC11877449 DOI: 10.3389/fphar.2025.1516583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 01/27/2025] [Indexed: 03/06/2025] Open
Abstract
Owing to its high mortality rate, lung cancer (LC) remains the most common cancer worldwide, with the highest malignancy diagnosis rate. The phosphatidylinositol-3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signaling (PAM) pathway is a critical intracellular pathway involved in various cellular functions and regulates numerous cellular processes, including growth, survival, proliferation, metabolism, apoptosis, invasion, and angiogenesis. This review aims to highlight preclinical and clinical studies focusing on the PAM signaling pathway in LC and underscore the potential of natural products targeting it. Additionally, this review synthesizes the existing literature and discusses combination therapy and future directions for LC treatment while acknowledging the ongoing challenges in the field. Continuous development of novel therapeutic agents, technologies, and precision medicine offers an increasingly optimistic outlook for the treatment of LC.
Collapse
Affiliation(s)
- Min Qiang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
- College of Clinical Medicine, Jilin University, Changchun, China
| | - Zhe Chen
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Hongyang Liu
- College of Clinical Medicine, Jilin University, Changchun, China
| | - Junxue Dong
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Kejian Gong
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Xinjun Zhang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Peng Huo
- Laboratory of Infection Oncology, Institute of Clinical Molecular Biology, Christian-Albrechts-Universität zu Kiel and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Jingjun Zhu
- Department of Thoracic and Cardiovascular Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yifeng Shao
- Department of General Surgery, Capital Institute of Pediatrics’ Children’s Hospital, Beijing, China
| | - Jinazun Ma
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Bowei Zhang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Wei Liu
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Mingbo Tang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
34
|
He M, Zhang Y, Zhai Y, Li Y, Yang G, Yu S, Xiao H, Song Y. Trilobatin regulates glucose metabolism by ameliorating oxidative stress and insulin resistance in vivo and in vitro. J Pharm Pharmacol 2025; 77:236-248. [PMID: 38642915 DOI: 10.1093/jpp/rgae035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 03/15/2024] [Indexed: 04/22/2024]
Abstract
OBJECTIVES Trilobatin, a glycosylated dihydrochalcone, has been reported to have anti-diabetic properties. However, the underlying mechanism remains unexplained. METHODS In this investigation, the regulation of trilobatin on glucose metabolism of insulin resistance (IR)-HepG2 cells and streptozocin (STZ)-induced mice and its mechanism were evaluated. KEY FINDINGS Different doses of trilobatin (5, 10 and 20 μM) increased glucose consumption, glycogen content, hexokinase (HK), and pyruvate kinase (PK) activity in IR-HepG2 cells. Among them, the HK and PK activity in IR-HepG2 cells treated with 20 μM trilobatin were 1.84 and 2.05 times than those of the IR-group. The overeating, body and tissue weight, insulin levels, liver damage, and lipid accumulation of STZ-induced mice were improved after feeding with different doses of trilobatin (10, 50, and 100 mg/kg/d) for 4 weeks. Compared with STZ-induced mice, fasting blood glucose decreased by 61.11% and fasting insulin (FINS) increased by 48.6% after feeding trilobatin (100 mg/kg/d). Meanwhile, data from quantitative real-time polymerase chain reaction (qRT-PCR) revealed trilobatin ameliorated glycogen synthesis via the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/glycogen synthase kinase-3β (GSK-3β) signaling pathway in IR-HepG2 cells and in STZ-induced mice. Furthermore, in vitro and in vivo experiments showed that trilobatin ameliorated oxidative stress by regulating the mRNA expression of nuclear erythroid-2 related factor 2 (Nrf2)/kelch-like ECH associated protein-1 (Keap-1) pathway as well as heme oxygenase-1 (HO-1) and NAD(P)H: quinone oxidoreductase-1 (NQO-1). CONCLUSIONS Our research reveals a novel pharmacological activity of trilobatin: regulating glucose metabolism through PI3K/Akt/GSK-3β and Nrf2/Keap-1 signaling pathways, improving insulin resistance and reducing oxidative stress. Trilobatin can be used as a reliable drug resource for the treatment of glucose metabolism disorders.
Collapse
Affiliation(s)
- Ming He
- School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, Shandong 255049, China
| | - Yuqing Zhang
- School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, Shandong 255049, China
| | - Yuhan Zhai
- School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, Shandong 255049, China
| | - Yaping Li
- School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, Shandong 255049, China
| | - Guorui Yang
- School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, Shandong 255049, China
| | - Shaoxuan Yu
- School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, Shandong 255049, China
| | - Haifang Xiao
- School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, Shandong 255049, China
| | - Yuanda Song
- School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, Shandong 255049, China
| |
Collapse
|
35
|
Abdulla A, Sadida HQ, Jerobin J, Elfaki I, Mir R, Mirza S, Singh M, Macha MA, Uddin S, Fakhro K, Bhat AA, Akil ASAS. Unraveling molecular interconnections and identifying potential therapeutic targets of significance in obesity-cancer link. JOURNAL OF THE NATIONAL CANCER CENTER 2025; 5:8-27. [PMID: 40040878 PMCID: PMC11873641 DOI: 10.1016/j.jncc.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/16/2024] [Accepted: 11/11/2024] [Indexed: 03/06/2025] Open
Abstract
Obesity, a global health concern, is associated with severe health issues like type 2 diabetes, heart disease, and respiratory complications. It also increases the risk of various cancers, including melanoma, endometrial, prostate, pancreatic, esophageal adenocarcinoma, colorectal carcinoma, renal adenocarcinoma, and pre-and post-menopausal breast cancer. Obesity-induced cellular changes, such as impaired CD8+ T cell function, dyslipidemia, hypercholesterolemia, insulin resistance, mild hyperglycemia, and fluctuating levels of leptin, resistin, adiponectin, and IL-6, contribute to cancer development by promoting inflammation and creating a tumor-promoting microenvironment rich in adipocytes. Adipocytes release leptin, a pro-inflammatory substance that stimulates cancer cell proliferation, inflammation, and invasion, altering the tumor cell metabolic pathway. Adiponectin, an insulin-sensitizing adipokine, is typically downregulated in obese individuals. It has antiproliferative, proapoptotic, and antiangiogenic properties, making it a potential cancer treatment. This narrative review offers a comprehensive examination of the molecular interconnections between obesity and cancer, drawing on an extensive, though non-systematic, survey of the recent literature. This approach allows us to integrate and synthesize findings from various studies, offering a cohesive perspective on emerging themes and potential therapeutic targets. The review explores the metabolic disturbances, cellular alterations, inflammatory responses, and shifts in the tumor microenvironment that contribute to the obesity-cancer link. Finally, it discusses potential therapeutic strategies aimed at disrupting these connections, offering valuable insights into future research directions and the development of targeted interventions.
Collapse
Affiliation(s)
- Alanoud Abdulla
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Research Program, Sidra Medicine, Doha, Qatar
| | - Hana Q. Sadida
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Research Program, Sidra Medicine, Doha, Qatar
| | - Jayakumar Jerobin
- Qatar Metabolic Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Imadeldin Elfaki
- Department of Biochemistry, Faculty of Science, University of Tabuk, Tabuk, Saudi Arabia
| | - Rashid Mir
- Department of Medical Laboratory Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Sameer Mirza
- Department of Chemistry, College of Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Mayank Singh
- Department of Medical Oncology (Lab.), Dr. BRAIRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Muzafar A. Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Pulwama, Jammu and Kashmir, India
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Laboratory of Animal Research Center, Qatar University, Doha, Qatar
| | - Khalid Fakhro
- Department of Human Genetics, Sidra Medicine, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
- Department of Genetic Medicine, Weill Cornell Medicine, Doha, Qatar
| | - Ajaz A. Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Research Program, Sidra Medicine, Doha, Qatar
| | - Ammira S. Al-Shabeeb Akil
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Research Program, Sidra Medicine, Doha, Qatar
| |
Collapse
|
36
|
Ma X, Shi Y, Shang Z. Epiberberine Improves Hyperglycemia and Ameliorates Insulin Sensitivity in Type 2 Diabetic Mice. Nephrology (Carlton) 2025; 30:e14430. [PMID: 39888160 DOI: 10.1111/nep.14430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 12/26/2024] [Accepted: 12/31/2024] [Indexed: 02/01/2025]
Abstract
AIM Type 2 diabetes mellitus (T2DM) is a metabolic syndrome characterised by absolute or relative insufficiency of insulin secretion. The alkaloids from Rhizoma coptidis have potential hypoglycemic effects. Epiberberine (EPI), a protoberberine alkaloid extracted from Rhizome coptidis, has been found to regulate lipid metabolism. Our study aimed to investigate the antidiabetic effects of EPI on mice with T2DM, as well as its underlying mechanism. METHODS The T2DM model in mice was established using a combination of high-fat diet and streptozotocin. Animals were divided into the control, T2DM, EPI-low dose (50 mg/kg EPI), EPI-medium dose (100 mg/kg EPI), EPI-high dose (200 mg/kg EPI) and metformin (MTF) (200 mg/kg MTF) groups. Body weight, water/food intake, serum lipids, blood glucose tolerance, insulin sensitivity, histopathological alterations, insulin signalling pathway and inflammation-related pathways in each group were detected. RESULTS EPI significantly reduced blood glucose levels and water/food intake in T2DM mice. EPI reduced the levels of total cholesterol, total triglyceride, low-density lipoprotein cholesterol, aspartate aminotransferase and alanine aminotransferase, and elevated the levels of high-density lipoprotein cholesterol in serum. EPI effectively improved oral glucose tolerance, alleviated hepatic insulin resistance, decreased glycosylated haemoglobin levels and increased liver glycogen content. EPI ameliorated the histopathological alterations of skeletal muscle and liver in T2DM mice. EPI stimulated the insulin signalling pathway by increasing glucose transporter type 4 levels and activating insulin receptor substrate-1, phosphatidylinositol 3-kinase and protein kinase B in skeletal muscle and liver. EPI reduced the levels of proinflammatory cytokine in serum and inhibited the activation of mitogen-activated protein kinase signalling in skeletal muscle and liver of diabetic mice. CONCLUSION Overall, these data demonstrate that EPI alleviates the symptoms of T2DM, providing new insights into EPI as a therapeutic compound for the alleviation of T2DM.
Collapse
Affiliation(s)
- Xiaohong Ma
- Nephrology Department, Shenzhen Bao'an Authentic Tcm Therapy Hospital, Shenzhen, China
| | - Yufeng Shi
- Internal Medicine Department, Shenzhen Bao'an Authentic Tcm Therapy Hospital, Shenzhen, China
| | - Zhitao Shang
- Internal Medicine Department, Shenzhen Bao'an Authentic Tcm Therapy Hospital, Shenzhen, China
| |
Collapse
|
37
|
Caprara G, Pallavi R, Sanyal S, Pelicci PG. Dietary Restrictions and Cancer Prevention: State of the Art. Nutrients 2025; 17:503. [PMID: 39940361 PMCID: PMC11820753 DOI: 10.3390/nu17030503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/22/2025] [Accepted: 01/25/2025] [Indexed: 02/16/2025] Open
Abstract
Worldwide, almost 10 million cancer deaths occurred in 2022, a number that is expected to rise to 16.3 million by 2040. Primary prevention has long been acknowledged as a crucial approach to reducing cancer incidence. In fact, between 30 and 50 percent of all tumors are known to be preventable by eating a healthy diet, staying active, avoiding alcohol, smoking, and being overweight. Accordingly, many international organizations have created tumor prevention guidelines, which underlie the importance of following a diet that emphasizes eating plant-based foods while minimizing the consumption of red/processed meat, sugars, processed foods, and alcohol. However, further research is needed to define the relationship between the effect of specific diets or nutritional components on cancer prevention. Interestingly, reductions in food intake and dietetic restrictions can extend the lifespan of yeast, nematodes, flies, and rodents. Despite controversial results in humans, those approaches have the potential to ameliorate health via direct and indirect effects on specific signaling pathways involved in cancer onset. Here, we describe the latest knowledge on the cancer-preventive potential of dietary restrictions and the biochemical processes involved. Molecular, preclinical, and clinical studies evaluating the effects of different fasting strategies will also be reviewed.
Collapse
Affiliation(s)
- Greta Caprara
- Department of Experimental Oncology, European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 20139 Milan, Italy
| | - Rani Pallavi
- Department of Experimental Oncology, European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 20139 Milan, Italy
- Brien Holden Eye Research Centre, L. V. Prasad Eye Institute, Hyderabad 500034, India
- The Operation Eyesight Universal Institute for Eye Cancer, L. V. Prasad Eye Institute, Hyderabad 500034, India; (R.P.); (S.S.)
| | - Shalini Sanyal
- Brien Holden Eye Research Centre, L. V. Prasad Eye Institute, Hyderabad 500034, India
- The Operation Eyesight Universal Institute for Eye Cancer, L. V. Prasad Eye Institute, Hyderabad 500034, India; (R.P.); (S.S.)
| | - Pier Giuseppe Pelicci
- Department of Experimental Oncology, European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 20139 Milan, Italy
| |
Collapse
|
38
|
Tárraga Marcos PJ, López-González ÁA, Martínez-Almoyna Rifá E, Paublini Oliveira H, Martorell Sánchez C, Tárraga López PJ, Ramírez-Manent JI. Risk of Insulin Resistance in 44,939 Spanish Healthcare Workers: Association with Sociodemographic Variables and Healthy Habits. Diseases 2025; 13:33. [PMID: 39997040 PMCID: PMC11854469 DOI: 10.3390/diseases13020033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/23/2025] [Accepted: 01/27/2025] [Indexed: 02/26/2025] Open
Abstract
Introduction: Insulin resistance (IR) is a highly prevalent pathophysiological entity implicated in the development of a wide variety of metabolic, cardiovascular, and endocrine disorders. The aim of this study is to assess the association between sociodemographic variables and healthy habits with IR risk scales. Methodology: This dual study, incorporating both longitudinal-retrospective and cross-sectional designs, analyzed healthcare workers across four professional categories (physicians, nurses, healthcare technicians, and auxiliary personnel). It examined the association of age, sex, professional category, smoking status, physical activity, and adherence to the Mediterranean diet with elevated scores on insulin resistance risk scales. Results: All the variables analyzed were associated with the presence of elevated values of the IR scales, with age, sex, and physical activity showing the strongest association (reflected in the odds ratio values). Conclusions: The profile of an individual with a higher risk of presenting elevated values of the IR risk scales would be an elderly male auxiliary health worker who is a smoker and is physically inactive, with a low adherence to the Mediterranean diet.
Collapse
Affiliation(s)
| | - Ángel Arturo López-González
- ADEMA-Health Group of University, Institute of Health Sciences (IUNICS) of Balearic Islands, 07009 Palma, Spain; (E.M.-A.R.); (H.P.O.); (C.M.S.); (J.I.R.-M.)
- Faculty of Odontology, University School ADEMA-UIB, 07009 Palma, Spain
- Health Service of the Balearic Islands, 07120 Palma, Spain
| | - Emilio Martínez-Almoyna Rifá
- ADEMA-Health Group of University, Institute of Health Sciences (IUNICS) of Balearic Islands, 07009 Palma, Spain; (E.M.-A.R.); (H.P.O.); (C.M.S.); (J.I.R.-M.)
- Faculty of Odontology, University School ADEMA-UIB, 07009 Palma, Spain
| | - Hernán Paublini Oliveira
- ADEMA-Health Group of University, Institute of Health Sciences (IUNICS) of Balearic Islands, 07009 Palma, Spain; (E.M.-A.R.); (H.P.O.); (C.M.S.); (J.I.R.-M.)
- Faculty of Odontology, University School ADEMA-UIB, 07009 Palma, Spain
| | - Cristina Martorell Sánchez
- ADEMA-Health Group of University, Institute of Health Sciences (IUNICS) of Balearic Islands, 07009 Palma, Spain; (E.M.-A.R.); (H.P.O.); (C.M.S.); (J.I.R.-M.)
- Faculty of Odontology, University School ADEMA-UIB, 07009 Palma, Spain
| | | | - José Ignacio Ramírez-Manent
- ADEMA-Health Group of University, Institute of Health Sciences (IUNICS) of Balearic Islands, 07009 Palma, Spain; (E.M.-A.R.); (H.P.O.); (C.M.S.); (J.I.R.-M.)
- Health Service of the Balearic Islands, 07120 Palma, Spain
- Faculty of Medicine, Balearic Islands University, 07120 Palma, Spain
| |
Collapse
|
39
|
Prada E, Marchetti GB, Pires Marafon D, Mazzocchi A, Scuvera G, Pezzani L, Agostoni C, Milani D. The Epigenetic Machinery and Energy Expenditure: A Network to Be Revealed. Genes (Basel) 2025; 16:104. [PMID: 39858651 PMCID: PMC11764581 DOI: 10.3390/genes16010104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/08/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Mendelian disorders of the epigenetic machinery (MDEMs) include a large number of conditions caused by defective activity of a member of the epigenetic machinery. MDEMs are characterized by multiple congenital abnormalities, intellectual disability and abnormal growth. that can be variably up- or down-regulated. Background/Objectives: In several MDEMs, a predisposition to metabolic syndrome and obesity since childhood has been reported. Methods: To investigate the metabolic bases of this abnormal growth, we collected physical data from a heterogeneous pool of 38 patients affected by MDEMs. Thirty-five patients performed indirect calorimetry (as a measure of resting energy expenditure, REE) and blood tests to monitor plasmatic nutritional parameters. Conclusions: Although limited by a small-sized and heterogeneous sample, our study demonstrates a linear correlation between REE and physical parameters, OFC, height and weight, and observed a slight imbalance on several plasmatic spies of metabolic syndrome predisposition. Furthermore, we demonstrated a significantly higher REE in Sotos Syndrome type 1 patients compared to the controls, which resulted independent from height, suggesting that impaired metabolism in these patients may go beyond overgrowth.
Collapse
Affiliation(s)
- Elisabetta Prada
- Azienda Socio Sanitaria Territoriale Lariana, 22100 Como, Italy;
| | - Giulia Bruna Marchetti
- Fondazione IRCSS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (A.M.); (G.S.); (L.P.); (C.A.); (D.M.)
| | - Denise Pires Marafon
- Department of Public Health and Infectious Diseases, Specialization School in Medical Statistics and Biometry, Università Sapienza di Roma, 00185 Roma, Italy
| | - Alessandra Mazzocchi
- Fondazione IRCSS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (A.M.); (G.S.); (L.P.); (C.A.); (D.M.)
| | - Giulietta Scuvera
- Fondazione IRCSS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (A.M.); (G.S.); (L.P.); (C.A.); (D.M.)
| | - Lidia Pezzani
- Fondazione IRCSS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (A.M.); (G.S.); (L.P.); (C.A.); (D.M.)
- Laboratorio di Genetica Medica, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy
| | - Carlo Agostoni
- Fondazione IRCSS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (A.M.); (G.S.); (L.P.); (C.A.); (D.M.)
- Department of Clinical and Community Sciences, Università degli Studi di Milano, 20133 Milano, Italy
| | - Donatella Milani
- Fondazione IRCSS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (A.M.); (G.S.); (L.P.); (C.A.); (D.M.)
| |
Collapse
|
40
|
Zhan S, Wang J, Zhu M, Liu Y, Han F, Sun L, Wang Q, Huang Z. The Inhibitory Effects of NCT503 and Exogenous Serine on High-Selenium Induced Insulin Resistance in Mice. Nutrients 2025; 17:311. [PMID: 39861441 PMCID: PMC11767638 DOI: 10.3390/nu17020311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/12/2024] [Accepted: 12/17/2024] [Indexed: 01/27/2025] Open
Abstract
OBJECTIVE This study aims to identify whether the development of insulin resistance (IR) induced by high selenium (Se) is related to serine deficiency via the inhibition of the de novo serine synthesis pathway (SSP) by the administrations of 3-phosphoglycerate dehydrogenase (PHGDH) inhibitor (NCT503) or exogenous serine in mice. METHOD forty-eight male C57BL/6J mice were randomly divided into four groups: adequate-Se (0.1 mgSe/kg), high-Se (0.8 mgSe/kg), high-Se +serine (240 mg/kg/day), and high-Se +NCT503 (30 mg/kg, twice a week) for 5 months. The glucose tolerance test (GTT) and insulin tolerance test (ITT) were used to confirm the development of IR in mice with high-Se intake, and fasting blood glucose levels were measured monthly. The Se contents in plasma and tissues were detected by ICP-MS. The levels of insulin (INS), homocysteine (HCY), and serine in plasma were tested by ELISA. Western blot analyses were conducted to evaluate the protein expressions of glutathione peroxidase 1 (GPX1), selenoprotein P (SELENOP) and PHGDH, the PI3K-AKT-mTOR pathway, folate cycle (SHMT1, MTHFR), and methionine cycle (MS). RESULTS An IR model was developed in mice from the high-Se group with elevated fasting blood glucose and INS levels, impaired glucose tolerance, and reduced insulin sensitivity, but not in both the high-Se +serine group and the high-Se +NCT503 group. Compared with the high-Se and high-Se +serine groups, the expressions of GPX1 and SELENOP significantly decreased for the high-Se +NCT503 group in the liver, muscle, and pancreas tissues. The expression of PHGDH of high-Se group was significantly higher than that of the adequate-Se group in the liver (p < 0.05) and pancreas (p < 0.001). Also, the expected high expression of PHGDH was effectively inhibited in mice from the high-Se +serine group but not from the high-Se +NCT503 group. The expression of p-AKT (Ser-473) for the high-Se group was significantly lower than that of the adequate-Se group in the liver, muscle, and pancreas. CONCLUSIONS The IR induced by high-Se intake in the body has been confirmed to be partially due to serine deficiency, which led to the initiation of SSP to produce endogenous serine. The supplementations of exogenous serine or inhibitors of PHGDH in this metabolic pathway could be used for the intervention.
Collapse
Affiliation(s)
- Shuo Zhan
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China; (S.Z.); (J.W.); (M.Z.); (Y.L.); (F.H.); (L.S.)
| | - Jianrong Wang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China; (S.Z.); (J.W.); (M.Z.); (Y.L.); (F.H.); (L.S.)
| | - Mingyu Zhu
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China; (S.Z.); (J.W.); (M.Z.); (Y.L.); (F.H.); (L.S.)
| | - Yiqun Liu
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China; (S.Z.); (J.W.); (M.Z.); (Y.L.); (F.H.); (L.S.)
| | - Feng Han
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China; (S.Z.); (J.W.); (M.Z.); (Y.L.); (F.H.); (L.S.)
| | - Licui Sun
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China; (S.Z.); (J.W.); (M.Z.); (Y.L.); (F.H.); (L.S.)
| | - Qin Wang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China; (S.Z.); (J.W.); (M.Z.); (Y.L.); (F.H.); (L.S.)
| | - Zhenwu Huang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China; (S.Z.); (J.W.); (M.Z.); (Y.L.); (F.H.); (L.S.)
- Key Laboratory of Public Nutrition and Health, National Health Commission of the People’s Republic of China, Beijing 100050, China
| |
Collapse
|
41
|
Zhang Z, Guo J. Deciphering Oxidative Stress in Cardiovascular Disease Progression: A Blueprint for Mechanistic Understanding and Therapeutic Innovation. Antioxidants (Basel) 2024; 14:38. [PMID: 39857372 PMCID: PMC11759168 DOI: 10.3390/antiox14010038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/20/2024] [Accepted: 12/27/2024] [Indexed: 01/27/2025] Open
Abstract
Oxidative stress plays a pivotal role in the pathogenesis and progression of cardiovascular diseases (CVDs). This review focuses on the signaling pathways of oxidative stress during the development of CVDs, delving into the molecular regulatory networks underlying oxidative stress in various disease stages, particularly apoptosis, inflammation, fibrosis, and metabolic imbalance. By examining the dual roles of oxidative stress and the influences of sex differences on oxidative stress levels and cardiovascular disease susceptibility, this study offers a comprehensive understanding of the pathogenesis of cardiovascular diseases. The study integrates key findings from current research in three comprehensive ways. First, it outlines the major CVDs associated with oxidative stress and their respective signaling pathways, emphasizing oxidative stress's central role in cardiovascular pathology. Second, it summarizes the cardiovascular protective effects, mechanisms of action, and animal models of various antioxidants, offering insights into future drug development. Third, it discusses the applications, advantages, limitations, and potential molecular targets of gene therapy in CVDs, providing a foundation for novel therapeutic strategies. These tables underscore the systematic and integrative nature of this study while offering a theoretical basis for precision treatment for CVDs. A major contribution of this study is the systematic review of the differential effects of oxidative stress across different stages of CVDs, in addition to the proposal of innovative, multi-level intervention strategies, which open new avenues for precision treatment of the cardiovascular system.
Collapse
Affiliation(s)
- Zhaoshan Zhang
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, China
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Jiawei Guo
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, China
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| |
Collapse
|
42
|
Saha S, Ghosh S, Ghosh S, Nandi S, Nayak A. Unraveling the complexities of colorectal cancer and its promising therapies - An updated review. Int Immunopharmacol 2024; 143:113325. [PMID: 39405944 DOI: 10.1016/j.intimp.2024.113325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 10/30/2024]
Abstract
Colorectal cancer (CRC) continues to be a global health concern, necessitating further research into its complex biology and innovative treatment approaches. The etiology, pathogenesis, diagnosis, and treatment of colorectal cancer are summarized in this thorough review along with recent developments. The multifactorial nature of colorectal cancer is examined, including genetic predispositions, environmental factors, and lifestyle decisions. The focus is on deciphering the complex interactions between signaling pathways such as Wnt/β-catenin, MAPK, TGF-β as well as PI3K/AKT that participate in the onset, growth, and metastasis of CRC. There is a discussion of various diagnostic modalities that span from traditional colonoscopy to sophisticated molecular techniques like liquid biopsy and radiomics, emphasizing their functions in early identification, prognostication, and treatment stratification. The potential of artificial intelligence as well as machine learning algorithms in improving accuracy as well as efficiency in colorectal cancer diagnosis and management is also explored. Regarding therapy, the review provides a thorough overview of well-known treatments like radiation, chemotherapy, and surgery as well as delves into the newly-emerging areas of targeted therapies as well as immunotherapies. Immune checkpoint inhibitors as well as other molecularly targeted treatments, such as anti-epidermal growth factor receptor (anti-EGFR) as well as anti-vascular endothelial growth factor (anti-VEGF) monoclonal antibodies, show promise in improving the prognosis of colorectal cancer patients, in particular, those suffering from metastatic disease. This review focuses on giving readers a thorough understanding of colorectal cancer by considering its complexities, the present status of treatment, and potential future paths for therapeutic interventions. Through unraveling the intricate web of this disease, we can develop a more tailored and effective approach to treating CRC.
Collapse
Affiliation(s)
- Sayan Saha
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India
| | - Shreya Ghosh
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India
| | - Suman Ghosh
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India
| | - Sumit Nandi
- Department of Pharmacology, Gupta College of Technological Sciences, Asansol, West Bengal 713301, India
| | - Aditi Nayak
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India.
| |
Collapse
|
43
|
Yao W, Huo J, Ji J, Liu K, Tao P. Elucidating the role of gut microbiota metabolites in diabetes by employing network pharmacology. Mol Med 2024; 30:263. [PMID: 39707185 DOI: 10.1186/s10020-024-01033-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 12/06/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Extensive research has underscored the criticality of preserving diversity and equilibrium within the gut microbiota for optimal human health. However, the precise mechanisms by which the metabolites and targets of the gut microbiota exert their effects remain largely unexplored. This study utilizes a network pharmacology methodology to elucidate the intricate interplay between the microbiota, metabolites, and targets in the context of DM, thereby facilitating a more comprehensive comprehension of this multifaceted disease. METHODS In this study, we initially extracted metabolite information of gut microbiota metabolites from the gutMGene database. Subsequently, we employed the SEA and STP databases to discern targets that are intricately associated with these metabolites. Furthermore, we leveraged prominent databases such as Genecard, DisGeNET, and OMIM to identify targets related to diabetes. A protein-protein interaction (PPI) network was established to screen core targets. Additionally, we conducted comprehensive GO and KEGG enrichment analyses utilizing the DAVID database. Moreover, a network illustrating the relationship among microbiota-substrate-metabolite-target was established. RESULTS We identified a total of 48 overlapping targets between gut microbiota metabolites and diabetes. Subsequently, we selected IL6, AKT1 and PPARG as core targets for the treatment of diabetes. Through the construction of the MSMT comprehensive network, we discovered that the three core targets exert therapeutic effects on diabetes through interactions with 8 metabolites, 3 substrates, and 5 gut microbiota. Additionally, GO analysis revealed that gut microbiota metabolites primarily regulate oxidative stress, inflammation and cell proliferation. KEGG analysis results indicated that IL-17, PI3K/AKT, HIF-1, and VEGF are the main signaling pathways involved in DM. CONCLUSION Gut microbiota metabolites primarily exert their therapeutic effects on diabetes through the IL6, AKT1, and PPARG targets. The mechanisms of gut microbiota metabolites regulating DM might involve signaling pathways such as IL-17 pathways, HIF-1 pathways and VEGF pathways.
Collapse
Affiliation(s)
- Weiguo Yao
- Department of Nephrology, Jinshan District Central Hospital, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Jinlin Huo
- Institute of Precision Medicine, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Jing Ji
- Department of Emergency, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Kun Liu
- Department of Nephrology, Jinshan District Central Hospital, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Pengyu Tao
- Department of Nephrology, Seventh People's Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
44
|
Medina-Vera D, López-Gambero AJ, Verheul-Campos J, Navarro JA, Morelli L, Galeano P, Suárez J, Sanjuan C, Pacheco-Sánchez B, Rivera P, Pavon-Morón FJ, Rosell-Valle C, Fonseca FRD. Therapeutic Efficacy of the Inositol D-Pinitol as a Multi-Faceted Disease Modifier in the 5×FAD Humanized Mouse Model of Alzheimer's Amyloidosis. Nutrients 2024; 16:4186. [PMID: 39683582 DOI: 10.3390/nu16234186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES Alzheimer's disease (AD), a leading cause of dementia, lacks effective long-term treatments. Current therapies offer temporary relief or fail to halt its progression and are often inaccessible due to cost. AD involves multiple pathological processes, including amyloid beta (Aβ) deposition, insulin resistance, tau protein hyperphosphorylation, and systemic inflammation accelerated by gut microbiota dysbiosis originating from a leaky gut. Given this context, exploring alternative therapeutic interventions capable of addressing the multifaceted components of AD etiology is essential. METHODS This study suggests D-Pinitol (DPIN) as a potential treatment modifier for AD. DPIN, derived from carob pods, demonstrates insulin-sensitizing, tau hyperphosphorylation inhibition, and antioxidant properties. To test this hypothesis, we studied whether chronic oral administration of DPIN (200 mg/kg/day) could reverse the AD-like disease progression in the 5×FAD mice. RESULTS Results showed that treatment of 5×FAD mice with DPIN improved cognition, reduced hippocampal Aβ and hyperphosphorylated tau levels, increased insulin-degrading enzyme (IDE) expression, enhanced pro-cognitive hormone circulation (such as ghrelin and leptin), and normalized the PI3K/Akt insulin pathway. This enhancement may be mediated through the modulation of cyclin-dependent kinase 5 (CDK5). DPIN also protected the gut barrier and microbiota, reducing the pro-inflammatory impact of the leaky gut observed in 5×FAD mice. DPIN reduced bacterial lipopolysaccharide (LPS) and LPS-associated inflammation, as well as restored intestinal proteins such as Claudin-3. This effect was associated with a modulation of gut microbiota towards a more balanced bacterial composition. CONCLUSIONS These findings underscore DPIN's promise in mitigating cognitive decline in the early AD stages, positioning it as a potential disease modifier.
Collapse
Affiliation(s)
- Dina Medina-Vera
- Grupo de Neuropsicofarmacología, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Unidades Clínicas de Neurología y Salud Mental, 29010 Málaga, Spain
- Facultad de Ciencias, Universidad de Málaga, 29010 Málaga, Spain
- Unidad de Gestión Clínica del Corazón-CIBERCV (Enfermedades Cardiovasculares), Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
| | - Antonio J López-Gambero
- Grupo de Neuropsicofarmacología, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Unidades Clínicas de Neurología y Salud Mental, 29010 Málaga, Spain
- INSERM, Neurocentre Magendie, University of Bordeaux, 33000 Bordeaux, France
| | - Julia Verheul-Campos
- Grupo de Neuropsicofarmacología, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Unidades Clínicas de Neurología y Salud Mental, 29010 Málaga, Spain
| | - Juan A Navarro
- Grupo de Neuropsicofarmacología, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Unidades Clínicas de Neurología y Salud Mental, 29010 Málaga, Spain
- Facultad de Medicina, Universidad de Málaga, 29010 Málaga, Spain
| | - Laura Morelli
- Laboratory of Brain Aging and Neurodegeneration, Fundación Instituto Leloir (IIBBA-CONICET), Av. Patricias Argentinas 435, Ciudad Autónoma de Buenos Aires C1405BWE, Argentina
| | - Pablo Galeano
- Laboratory of Brain Aging and Neurodegeneration, Fundación Instituto Leloir (IIBBA-CONICET), Av. Patricias Argentinas 435, Ciudad Autónoma de Buenos Aires C1405BWE, Argentina
| | - Juan Suárez
- Grupo de Neuropsicofarmacología, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Unidades Clínicas de Neurología y Salud Mental, 29010 Málaga, Spain
- Departamento de Anatomía Humana, Medicina Legal e Historia de la Ciencia, Facultad de Medicina, Universidad de Málaga, 29071 Málaga, Spain
- Andalusian Network for Clinical and Translational Research in Neurology [NEURO-RECA], 29001 Málaga, Spain
| | - Carlos Sanjuan
- Euronutra S.L. Calle Johannes Kepler, 3, 29590 Málaga, Spain
| | - Beatriz Pacheco-Sánchez
- Grupo de Neuropsicofarmacología, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Unidades Clínicas de Neurología y Salud Mental, 29010 Málaga, Spain
| | - Patricia Rivera
- Grupo de Neuropsicofarmacología, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Unidades Clínicas de Neurología y Salud Mental, 29010 Málaga, Spain
| | - Francisco J Pavon-Morón
- Grupo de Neuropsicofarmacología, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Unidades Clínicas de Neurología y Salud Mental, 29010 Málaga, Spain
- Unidad de Gestión Clínica del Corazón-CIBERCV (Enfermedades Cardiovasculares), Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
| | - Cristina Rosell-Valle
- Grupo de Neuropsicofarmacología, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Unidades Clínicas de Neurología y Salud Mental, 29010 Málaga, Spain
| | - Fernando Rodríguez de Fonseca
- Grupo de Neuropsicofarmacología, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Unidades Clínicas de Neurología y Salud Mental, 29010 Málaga, Spain
- Andalusian Network for Clinical and Translational Research in Neurology [NEURO-RECA], 29001 Málaga, Spain
| |
Collapse
|
45
|
Wang C, Sun X, Liu X, Wang Y, Luo J, Yang X, Liu Y. Protective effects of betaine on the early fatty liver in laying hens through ameliorating lipid metabolism and oxidative stress. Front Nutr 2024; 11:1505357. [PMID: 39654538 PMCID: PMC11627039 DOI: 10.3389/fnut.2024.1505357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 11/11/2024] [Indexed: 12/12/2024] Open
Abstract
Introduction Fatty liver syndrome (FLS) is a prevalent nutritional and metabolic disease that mainly occurs in caged laying hens, causing substantial losses in the poultry industry. The study was carried out to explore the protective effect and potential mechanism of betaine on early FLS. Methods There were three groups: Con group (basal diet), FLS group (Dexamethasone injection + basal diet) and betaine group (Dexamethasone injection + basal diet with 8 g/kg betaine). Birds in FLS and betaine groups were treated with subcutaneous dexamethasone injection once a day at a dosage of 4.50 mg/kg body weight for 7 days. Results The results revealed that DXM treatment significantly increased the liver index, serum aspartate aminotransferase (AST), total protein (TP), total bilirubin (TBIL), total biliary acid (TBA), total cholesterol (TC), high density lipoprotein cholesterol (HDL-c), low density lipoprotein cholesterol (LDL-c), and glucose (GLU) (p < 0.05). Additionally, hepatic TC and TG levels were also elevated (p < 0.05). Meanwhile, H&E and oil red O staining showed that there were a large number of vacuoles and lipid droplets in the liver of hens in FLS group. Dietary betaine addition significantly alleviated the increasing of serum TBIL, TBA and hepatic TC caused by dexamethasone treatment (p < 0.05). There existed 1,083 up- and 996 down-regulated genes in FLS group when compared with the control, and there were 169 upregulation and 405 downregulation genes in BT group when compared with FLS group. A total of 37 differential expression genes (DEGs) were rescued by betaine addition, which were related to lipid metabolism and antioxidant functions including APOC3, APOA4, G0S2, ERG28, PLA2G3, GPX4 and SLC5A8. Serum metabolomics analysis showed that 151 differential metabolites were identified in FLS group when compared with the control. Dietary betaine addition could rescue the changes of metabolites partly such as chicoric acid, gamma-aminobutyric acid, linoleic acid, telmisartan, which were associated with anti-oxidative function. In addition, RT-PCR results showed that genes involved in lipid metabolism, such as ACC, FAS, SCD1, ELOVL6, SREBP1, GR, ATGL and MTTP were markedly upregulated at the mRNA level (p < 0.05). However, dietary supplementation with betaine can reversed the expression of these genes (p < 0.05). Importantly, dietary betaine supplementation could reverse increased lipid synthesis partly by regulating PI3K/AKT/SREBP and CEBPα pathways in the liver based on western blot results (p < 0.05). Conclusion Dexamethasone treatment could establish the early FLS model in laying hens with hepatic lipid accumulation and no inflammation, which could be attenuated by dietary betaine addition.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yanli Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| |
Collapse
|
46
|
Lu F, Zeng N, Xiao X, Wang X, Gong H, Lei H. Exploring the ceRNA network involving AGAP2-AS1 as a novel biomarker for preeclampsia. Sci Rep 2024; 14:27330. [PMID: 39521940 PMCID: PMC11550820 DOI: 10.1038/s41598-024-79224-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 11/07/2024] [Indexed: 11/16/2024] Open
Abstract
Preeclampsia (PE) is an important research subject in obstetrics. Nevertheless, the underlying mechanisms of PE remain elusive. PE-related expression datasets (GSE96983, GSE96984 and GSE24129) were downloaded from the Gene Expression Omnibus (GEO) database. Firstly, the differentially expressed messenger RNAs (DE-mRNAs), DE-microRNA (DE-miRNAs) and DE-long non-coding RNA (DE-lncRNAs) between PE and control cohorts were identified, and the ceRNA network was constructed. Then candidate hub genes were obtained through five algorithms by the protein-protein intersection (PPI) network of the mRNAs. Further, five hub genes were identified by receiver operating characteristic (ROC) curve and gene expression profiles: DAXX, EFNB1, NCOR2, RBBP4 and SOCS1. The function of 5 hub genes was analyzed and the interaction between drugs and hub genes was predicted. A total of 5 small molecule drugs were predicted, namely benzbromarone, 9,10-phenanthrenequinone, chembl312032, insulin and aldesleukin. AGAP2-AS1 was mainly located in exosome and cytoplasm. Agap2-as1-related regulatory subnetworks were extracted from ceRNA networks which included 41 mRNAs, 2 miRNAs and 1 lncRNA, including the regulated relationship pairs AGAP2-AS1-hsa-miR-497-5p-SRPRB, and AGAP2-AS1-hsa-miR-195-5p-RPL36. In summary, we constructed a competitive endogenous RNA (ceRNA) network to identify five potential biomarkers (DAXX, EFNB1, NCOR2, SOCS1 and RBBP4) of PE. The in-depth analysis of the AGAP2-AS1 regulatory network will help to uncover more important molecules closely related to PE and provide a scientific Reference.
Collapse
Affiliation(s)
- Fan Lu
- Department of Obstetrics, Affiliated Hospital of GuiZhou Medical University, Guiyang, Guizhou, China
| | - Ni Zeng
- Department of Hospital infection and control, Affiliated Hospital of GuiZhou Medical University, Guiyang, Guizhou, China
| | - Xiang Xiao
- Department of Obstetrics, Affiliated Hospital of GuiZhou Medical University, Guiyang, Guizhou, China
| | - Xingxing Wang
- Department of Obstetrics, Affiliated Hospital of GuiZhou Medical University, Guiyang, Guizhou, China
| | - Han Gong
- Department of Obstetrics, Affiliated Hospital of GuiZhou Medical University, Guiyang, Guizhou, China
| | - Houkang Lei
- Department of Obstetrics, Affiliated Hospital of GuiZhou Medical University, Guiyang, Guizhou, China.
| |
Collapse
|
47
|
Alnaser RI, Alassaf FA, Abed MN. Incretin-Based Therapies: A Promising Approach for Modulating Oxidative Stress and Insulin Resistance in Sarcopenia. J Bone Metab 2024; 31:251-263. [PMID: 39496297 DOI: 10.11005/jbm.24.739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 07/07/2024] [Indexed: 11/06/2024] Open
Abstract
BACKGROUND Recent studies have linked sarcopenia development to the hallmarks of diabetes, oxidative stress, and insulin resistance. The anti-oxidant and insulin sensitivityenhancing effects of incretin-based therapies may provide a promising option for the treatment of sarcopenia. This review aimed to unveil the role of oxidative stress and insulin resistance in the pathogenesis of sarcopenia and explore the potential benefits of incretin-based therapies in individuals with sarcopenia. METHODS PubMed, the Cochrane Library, and Google Scholar databases were searched by applying keywords relevant to the main topic, to identify articles that met our selection criteria. RESULTS Incretin-based therapies manifested anti-oxidant effects by increasing the anti-oxidant defense system and decreasing free radical generation or by indirectly minimizing glucotoxicity, which was mainly achieved by improving insulin signaling and glucose homeostasis. Likewise, these drugs exhibit insulin-sensitizing activities by increasing insulin secretion, transduction, and β-cell function or by reducing inflammation and lipotoxicity. CONCLUSIONS Incretin-based therapies, as modulators of oxidation and insulin resistance, may target the main pathophysiological factors of sarcopenia, thus providing a promising strategy for the treatment of this disease.
Collapse
Affiliation(s)
- Raniah I Alnaser
- Nineveh Health Directorate, Mosul, Iraq
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Mosul, Mosul, Iraq
| | - Fawaz A Alassaf
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Mosul, Mosul, Iraq
| | - Mohammed N Abed
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, Iraq
| |
Collapse
|
48
|
Torres-Torres J, Monroy-Muñoz IE, Perez-Duran J, Solis-Paredes JM, Camacho-Martinez ZA, Baca D, Espino-Y-Sosa S, Martinez-Portilla R, Rojas-Zepeda L, Borboa-Olivares H, Reyes-Muñoz E. Cellular and Molecular Pathophysiology of Gestational Diabetes. Int J Mol Sci 2024; 25:11641. [PMID: 39519193 PMCID: PMC11546748 DOI: 10.3390/ijms252111641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/25/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Gestational diabetes (GD) is a metabolic disorder characterized by glucose intolerance during pregnancy, significantly impacting maternal and fetal health. Its global prevalence is approximately 14%, with risk factors including obesity, family history of diabetes, advanced maternal age, and ethnicity, which are linked to cellular and molecular disruptions in glucose regulation and insulin resistance. GD is associated with short- and long-term complications for both the mother and the newborn. For mothers, GD increases the risk of developing type 2 diabetes, cardiovascular diseases, and metabolic syndrome. In the offspring, exposure to GD in utero predisposes them to obesity, glucose intolerance, and metabolic disorders later in life. This review aims to elucidate the complex cellular and molecular mechanisms underlying GD to inform the development of effective therapeutic strategies. A systematic review was conducted using medical subject headings (MeSH) terms related to GD's cellular and molecular pathophysiology. Inclusion criteria encompassed original studies, systematic reviews, and meta-analyses focusing on GD's impact on maternal and fetal health, adhering to PRISMA guidelines. Data extraction captured study characteristics, maternal and fetal outcomes, key findings, and conclusions. GD disrupts insulin signaling pathways, leading to impaired glucose uptake and insulin resistance. Mitochondrial dysfunction reduces ATP production and increases reactive oxygen species, exacerbating oxidative stress. Hormonal influences, chronic inflammation, and dysregulation of the mammalian target of rapamycin (mTOR) pathway further impair insulin signaling. Gut microbiota alterations, gene expression, and epigenetic modifications play significant roles in GD. Ferroptosis and placental dysfunction primarily contribute to intrauterine growth restriction. Conversely, fetal macrosomia arises from maternal hyperglycemia and subsequent fetal hyperinsulinemia, resulting in excessive fetal growth. The chronic inflammatory state and oxidative stress associated with GD exacerbate these complications, creating a hostile intrauterine environment. GD's complex pathophysiology involves multiple disruptions in insulin signaling, mitochondrial function, inflammation, and oxidative stress. Effective management requires early detection, preventive strategies, and international collaboration to standardize care and improve outcomes for mothers and babies.
Collapse
Affiliation(s)
- Johnatan Torres-Torres
- Department of Reproductive and Perinatal Health Research, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
- Obstetric and Gynecology Department, Hospital General de México Dr. Eduardo Liceaga, Mexico City 06720, Mexico
| | - Irma Eloisa Monroy-Muñoz
- Department of Reproductive and Perinatal Health Research, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| | - Javier Perez-Duran
- Department of Reproductive and Perinatal Health Research, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| | - Juan Mario Solis-Paredes
- Department of Reproductive and Perinatal Health Research, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| | | | - Deyanira Baca
- Obstetric and Gynecology Department, Hospital General de México Dr. Eduardo Liceaga, Mexico City 06720, Mexico
| | - Salvador Espino-Y-Sosa
- Department of Reproductive and Perinatal Health Research, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
- Centro de Investigacion en Ciencias de la Salud, Universidad Anahuac Mexico, Campus Norte, Huixquilucan 52786, Mexico
| | - Raigam Martinez-Portilla
- Department of Reproductive and Perinatal Health Research, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| | - Lourdes Rojas-Zepeda
- Maternal-Fetal Department, Instituto Materno Infantil del Estado de Mexico, Toluca 50170, Mexico
| | - Hector Borboa-Olivares
- Community Interventions Research Branch, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| | - Enrique Reyes-Muñoz
- Research Division, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| |
Collapse
|
49
|
Liu N, Cui X, Guo T, Wei X, Sun Y, Liu J, Zhang Y, Ma W, Yan W, Chen L. Baicalein Ameliorates Insulin Resistance of HFD/STZ Mice Through Activating PI3K/AKT Signal Pathway of Liver and Skeletal Muscle in a GLP-1R-Dependent Manner. Antioxidants (Basel) 2024; 13:1246. [PMID: 39456499 PMCID: PMC11505556 DOI: 10.3390/antiox13101246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/10/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Insulin resistance (IR) is the principal pathophysiological change occurring in diabetes mellitus (DM). Baicalein, a bioactive flavonoid primarily extracted from the medicinal plant Scutellaria baicalensis Georgi, has been shown in our previous research to be a potential natural glucagon-like peptide-1 receptor (GLP-1R) agonist. However, the exact therapeutic effect of baicalein on DM and its underlying mechanisms remain elusive. In this study, we investigated the therapeutic effects of baicalein on diabetes and sought to clarify its underlying molecular mechanisms. Our results demonstrated that baicalein improves hyperglycemic, hyperinsulinemic, and glucometabolic disorders in mice with induced diabetes via GLP-1R. This was confirmed by the finding that baicalein's effects on improving IR were largely diminished in mice with whole-body Glp1r ablation. Complementarily, network pharmacology analysis highlighted the pivotal involvement of the phosphatidylinositol-3-kinase (PI3K)/protein kinase B (AKT) insulin signaling pathway in the therapeutic actions of baicalein on IR. Our mechanism research significantly confirmed that baicalein mitigates hepatic and muscular IR through the PI3K/AKT signal pathway, both in vitro and in vivo. Furthermore, we demonstrated that baicalein enhances glucose uptake in skeletal muscle cells under IR conditions through the Ca2+/calmodulin-dependent protein kinase II (CaMKII)-adenosine 5'-monophosphate-activated protein kinase (AMPK)-glucose transporter 4 (GLUT4) signaling pathway in a GLP-1R-dependent manner. In conclusion, our findings confirm the therapeutic effects of baicalein on IR and reveal that it improves IR in liver and muscle tissues through the PI3K/AKT insulin signaling pathway in a GLP-1R dependent manner. Moreover, we clarified that baicalein enhances the glucose uptake in skeletal muscle tissue through the Ca2+/CaMKII-AMPK-GLUT4 signal pathway.
Collapse
Affiliation(s)
- Na Liu
- Department of Pharmacology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (N.L.); (X.C.); (T.G.); (X.W.); (Y.S.); (J.L.); (Y.Z.)
| | - Xin Cui
- Department of Pharmacology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (N.L.); (X.C.); (T.G.); (X.W.); (Y.S.); (J.L.); (Y.Z.)
| | - Tingli Guo
- Department of Pharmacology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (N.L.); (X.C.); (T.G.); (X.W.); (Y.S.); (J.L.); (Y.Z.)
| | - Xiaotong Wei
- Department of Pharmacology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (N.L.); (X.C.); (T.G.); (X.W.); (Y.S.); (J.L.); (Y.Z.)
| | - Yuzhuo Sun
- Department of Pharmacology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (N.L.); (X.C.); (T.G.); (X.W.); (Y.S.); (J.L.); (Y.Z.)
| | - Jieyun Liu
- Department of Pharmacology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (N.L.); (X.C.); (T.G.); (X.W.); (Y.S.); (J.L.); (Y.Z.)
| | - Yangyang Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (N.L.); (X.C.); (T.G.); (X.W.); (Y.S.); (J.L.); (Y.Z.)
| | - Weina Ma
- School of Pharmacy, Xi’an Jiaotong University, Xi’an 710049, China;
| | - Wenhui Yan
- Department of Pharmacology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (N.L.); (X.C.); (T.G.); (X.W.); (Y.S.); (J.L.); (Y.Z.)
| | - Lina Chen
- Department of Pharmacology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (N.L.); (X.C.); (T.G.); (X.W.); (Y.S.); (J.L.); (Y.Z.)
- Key Laboratory of Environment and Genes Related to Diseases (Xi’an Jiaotong University), Ministry of Education, Xi’an 710061, China
- Cardiometabolic Innovation Center, Ministry of Education, Xi’an 710061, China
- Department of Endocrinology and Second Department of Geriatrics, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| |
Collapse
|
50
|
Liu H, Yi X, You M, Yang H, Zhang S, Huang S, Xie L. Bulk-RNA and single-nuclei RNA seq analyses reveal the role of lactate metabolism-related genes in Alzheimer's disease. Metab Brain Dis 2024; 39:1469-1480. [PMID: 39136807 DOI: 10.1007/s11011-024-01396-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 07/19/2024] [Indexed: 10/29/2024]
Abstract
Dysfunctional lactate metabolism in the brain has been implicated in neuroinflammation, Aβ deposition, and cell disturbance, all of which play a significant role in the pathogenesis of Alzheimer's disease (AD). In this study, we aimed to investigate the lactate metabolism-related genes (LMRGs) in AD via an integrated bulk RNA and single-nuclei RNA sequencing (snRNA-seq) analysis, with a specific focus on microglia. We obtained 26 HC and 24 AD snRNA-seq samples originated from human prefrontal cortex in Gene Expression Omnibus (GEO) database and collected 873 LMRGs from three databases, namely MSigDB, The Human Protein Atlas and GeneCards. Bulk RNA was analyzed with LMRG characteristics in AD by using Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), the protein-protein interaction (PPI), CytoHubba-MCC, Support Vector Machine (SVM) algorithms analyses. Then we conducted the Receiver Operating Characteristic (ROC) curve, correlation, and connection network analyses for biomarkers. Their differential expression validation was performed using AlzData database. The single-nuclei RNA analysis of microglia was applied to identify hub genes and pathways using cell-cell communication analysis and high dimensional Weighted Gene Co-Expression Network Analysis (hdWGCNA). Support Vector Machine (SVM) algorithm showed an AUC of 0.967, a sensitivity of 93.30% and a specificity of 100.00%. Our analysis identified biomarkers with LMRG characteristics, namely INSR, CDKL1, and PNISR. ROC analysis revealed that each of these biomarkers exhibited excellent diagnostic potential, as evidenced by their respective area under the curve (AUC) values: INSR (AUC: 0.679), CDKL1 (AUC: 0.788), and PNISR (AUC: 0.724). Correlation analysis showed that biomarkers exhibited a positive correlation with each other. Connection network illustrated their shared biological processes: aging, phosphorylation, metabolic process, and apoptosis. Cell-cell communication analysis revealed that GALECTIN signaling pathway was exclusively expressed in AD microglia, and only LGALS9 exhibited significant overexpression. HdWGCNA identified FTH1 as a hub gene enriched in ferroptosis and mineral absorption pathways within microglia. The roles of INSR, CDKL1, PNISR, LGALS9, and FTH1 should be taken into account to enhance our understanding of lactate metabolism in the context of AD.
Collapse
Affiliation(s)
- Hanjie Liu
- Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, P.R. China
| | - Xiaohong Yi
- Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, P.R. China
| | - Maochun You
- Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, P.R. China.
| | - Hui Yang
- Chengdu Shuangliu Hospital of Traditional Chinese Medicine, Chengdu, 610200, Sichuan, P.R. China
| | - Siyu Zhang
- Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, P.R. China
| | - Sihan Huang
- Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, P.R. China
| | - Lushuang Xie
- Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, P.R. China.
| |
Collapse
|