1
|
Jahdkaran M, Sistanizad M. From lipids to glucose: Investigating the role of dyslipidemia in the risk of insulin resistance. J Steroid Biochem Mol Biol 2025; 250:106744. [PMID: 40158704 DOI: 10.1016/j.jsbmb.2025.106744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 03/11/2025] [Accepted: 03/26/2025] [Indexed: 04/02/2025]
Abstract
Dyslipidemia is recognized as one of the most prevalent metabolic disorders and is frequently associated with other prevalent conditions, particularly diabetes mellitus. There appears to be a bidirectional connection between these two metabolic disorders. While considerable research has focused on how insulin resistance can lead to lipid abnormalities, the reverse relationship specifically, how dyslipidemia could assist in developing insulin resistance and diabetes mellitus has received relatively less attention. This review aims to comprehensively evaluate the mechanisms through which dyslipidemia can induce insulin resistance. Dyslipidemia is primarily classified into three main categories: hypercholesterolemia, hypertriglyceridemia, and low levels of HDL. These conditions may promote insulin resistance across multiple pathways, including the accumulation of lipid metabolites, dysfunction of pancreatic β-cells, increased reactive oxygen species, endoplasmic reticulum stress and inflammation, endothelial dysfunction, alterations in adiponectin levels, changes in bile acid composition and concentration, and dysbiosis of gut microbiota. However, further investigation is required to fully elucidate the cellular and molecular mechanisms underlying the relationship between lipid disorders and insulin resistance. Emphasizing such research could facilitate the development of therapeutic strategies targeting both conditions simultaneously.
Collapse
Affiliation(s)
- Mahtab Jahdkaran
- Prevention of Cardiovascular Disease Research Center, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Sistanizad
- Prevention of Cardiovascular Disease Research Center, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Liu Y, Yu Z, Wang X, Yuan MQ, Lu MJ, Gong MR, Li Q, Xia YB, Yang GH, Xu B, Litscher G, Xu TC. Neurophysiological mechanisms of electroacupuncture in regulating pancreatic function and adipose tissue expansion. World J Diabetes 2025; 16:101354. [DOI: 10.4239/wjd.v16.i5.101354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 02/12/2025] [Accepted: 03/14/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Electroacupuncture (EA) has been recognized for its beneficial effects on glucolipid metabolism, potentially through the regulation of sensory nerve coordination. The expandability of peripancreatic adipose tissue (PAT) is implicated in the transition from obesity to type 2 diabetes mellitus (T2DM). However, the specific pancreatic responses to EA require further elucidation.
AIM To investigate the influence of EA on pancreatic glucolipid reduction level in a high-fat diet (HFD) rat model.
METHODS To delineate the precise pathway through which EA mediates interactions between PAT and islets, we assessed the expression levels of NGF, TRPV1, insulin, as well as other proteins in the pancreas and PAT. This approach enabled us to identify the acupoints that are most conducive to optimizing glycolipid metabolism.
RESULTS The ST25, LI11 and ST37 groups attenuated HFD-induced obesity and insulin resistance (IR) to distinct degrees, with ST25 group having the greatest effect. EA at ST25 was found to modify the local regulatory influence of PAT on the pancreatic intrinsic nervous system. Specifically, EA at ST25 obviously activated the TRPV1-CGRP-islet beta cell pathway, contributing to the relief of glucolipid metabolic stress. The beneficial effects were abrogated following the chemical silencing of TRPV1 sensory afferents, confirming their indispensable role in EA-mediated regulation of islet and PAT function. Furthermore, in TRPV1 knockout mice, a reduction in PAT inflammation was observed, along with the recovery of islet beta cell function. EA at LI11 and ST37 demonstrated anti-inflammatory properties and helped ameliorate IR.
CONCLUSION The PAT ecological niche influenced the progression from obesity to T2DM through various immunometabolic pathways. EA at ST25 could regulate glucolipid metabolism via the TRPV1-CGRP-islet beta cell pathway.
Collapse
Affiliation(s)
- Yun Liu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Zhi Yu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Xuan Wang
- College of Traditional Chinese Medicine, Jiangsu Vocational College of Medicine, Yancheng 224000, Jiangsu Province, China
| | - Ming-Qian Yuan
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Meng-Jiang Lu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Mei-Rong Gong
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Qian Li
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - You-Bing Xia
- Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| | - Guan-Hu Yang
- Department of Specialty Medicine, Ohio University, Athens, OH 45701, United States
| | - Bin Xu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Gerhard Litscher
- High-Tech Acupuncture and Digital Chinese Medicine, Swiss University of Traditional Chinese Medicine, Bad Zurzach 5530, Switzerland
| | - Tian-Cheng Xu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| |
Collapse
|
3
|
Sánchez-Cardona Y, Cuartas-Gómez E, Echeverri-Cuartas CE, Arévalo-Enríquez K, Londoño López ME, Moreno-Castellanos N. Effects of neutralization and functionalization on chitosan/gelatin/polyvinyl alcohol scaffolds in insulin-producing cell culture. Int J Biol Macromol 2025; 305:140800. [PMID: 39924037 DOI: 10.1016/j.ijbiomac.2025.140800] [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: 09/20/2024] [Revised: 01/24/2025] [Accepted: 02/06/2025] [Indexed: 02/11/2025]
Abstract
Diabetes is a disease that affects the patient's quality of life. Although there are several studies of therapeutic alternatives, there is still no definitive cure. Polymeric scaffolds represent a promising therapeutic strategy to preserve cell mass through 3D cultures. The aim of this study is to explore an alternative polymeric scaffold based on a mix of chitosan (Chi), gelatin (Ge), and polyvinyl alcohol (PVA) functionalized with VEGF for the culture of insulin-producing cells. The scaffolds were obtained by freeze-thaw cycles and lyophilization, followed by neutralization and, functionalization with vascular endothelial growth factor (VEGF). Physicochemical characteristics, biocompatibility and functionality were evaluated. Scaffolds obtained had interconnected heterogeneous pores. The presence of functional groups confirmed the integration of all the components without significantly losing thermal stability and mass. The functionalized and neutralized scaffolds positively impacted the biocompatibility and insulin secretion. Cell respiration was sustained, and cell morphology demonstrated the formation of cell clusters. It can be concluded that neutralization and functionalization of the scaffolds combined with VEGF are necessary to improve biocompatibility and functionality. Moreover, all these characteristics generated encouraging results on the diffusion of nutrients and cell adhesion, which could be valuable in the translational application for diabetes treatment.
Collapse
Affiliation(s)
- Yesenia Sánchez-Cardona
- Grupo de Investigación en Ingeniería Biomédica EIA (GIBEC), Programa de Ingeniería Biomédica, Escuela de Ciencias de la Vida, Universidad EIA, km 2 + 200 Vía al Aeropuerto José María Córdova, Envigado 055428, Colombia
| | - Elías Cuartas-Gómez
- CICTA Research Group, Department of Basic Sciences, Medicine School, Health Faculty, Universidad Industrial de Santander, Cra 27 calle 9, Bucaramanga 680002, Colombia
| | - Claudia E Echeverri-Cuartas
- Grupo de Investigación en Ingeniería Biomédica EIA (GIBEC), Programa de Ingeniería Biomédica, Escuela de Ciencias de la Vida, Universidad EIA, km 2 + 200 Vía al Aeropuerto José María Córdova, Envigado 055428, Colombia
| | - Karol Arévalo-Enríquez
- Grupo de Investigación en Ingeniería Biomédica EIA (GIBEC), Programa de Ingeniería Biomédica, Escuela de Ciencias de la Vida, Universidad EIA, km 2 + 200 Vía al Aeropuerto José María Córdova, Envigado 055428, Colombia
| | - Marta E Londoño López
- Grupo de Investigación en Ingeniería Biomédica EIA (GIBEC), Programa de Ingeniería Biomédica, Escuela de Ciencias de la Vida, Universidad EIA, km 2 + 200 Vía al Aeropuerto José María Córdova, Envigado 055428, Colombia
| | - Natalia Moreno-Castellanos
- CICTA Research Group, Department of Basic Sciences, Medicine School, Health Faculty, Universidad Industrial de Santander, Cra 27 calle 9, Bucaramanga 680002, Colombia.
| |
Collapse
|
4
|
Ren T, Fan X, Wu Q, Wu Y, Sun X, Tong H. Structural insights and therapeutic potential of plant-based pectin as novel therapeutic for type 2 diabetes mellitus: A review. Int J Biol Macromol 2025; 307:141876. [PMID: 40064270 DOI: 10.1016/j.ijbiomac.2025.141876] [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: 12/21/2024] [Revised: 03/03/2025] [Accepted: 03/07/2025] [Indexed: 03/15/2025]
Abstract
Type 2 diabetes mellitus (T2DM) is a global health challenge with limited efficacy of current treatments, necessitating alternative therapies. Plant-derived pectin, composed of galacturonic acid and structural domains such as homogalacturonan, has shown promise as an anti-diabetic agent. Pectin exerts its therapeutic effects through multiple mechanisms, including enhancing β-cell function, regulating glucose metabolism, improving insulin sensitivity, inhibiting digestive enzymes, and restoring gut microbiota balance. Its bioactivity is influenced by physicochemical properties like molecular weight, degree of methylation, and structural complexity. This review explores the anti-diabetic potential of pectin, its structure-activity relationships, and mechanisms of action, providing insights for its development as a novel therapeutic agent in T2DM management.
Collapse
Affiliation(s)
- Ting Ren
- School of Pharmaceutical Sciences, Jilin Medical University, Jilin 132013, China
| | - Xinrong Fan
- Department of Durg Preparation, Lishui Hospital of Traditional Chinese Medicine, Lishui 323000, China
| | - Qifang Wu
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Yu Wu
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Xin Sun
- School of Pharmaceutical Sciences, Jilin Medical University, Jilin 132013, China.
| | - Haibin Tong
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China; State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Beijing 100700, China.
| |
Collapse
|
5
|
Siva B, Sherin S, Somabattni RA, Nanjappan SK. Integrative CYP450 and network pharmacology approach for the assessment of Corilagin's influence on Sitagliptin pharmacokinetics. Biochem Pharmacol 2025; 237:116960. [PMID: 40287102 DOI: 10.1016/j.bcp.2025.116960] [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: 11/07/2024] [Revised: 04/15/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Type 2 diabetes (T2D) is a complex metabolic disorder marked by elevated blood glucose levels and a high risk of cardiovascular complications. Sitagliptin (SIT), a widely prescribed Dipeptidyl Peptidase-4 (DPP-4) inhibitor, is commonly used for T2D and undergoes extensive metabolism primarily via CYP3A4. Corilagin (COR), a bioactive ellagitannin known for its antioxidant, anti-inflammatory, and anti-diabetic properties, is frequently used in traditional medicine but is not well-studied for its CYP450 metabolism. This study employed a network pharmacology and pharmacokinetics approach to evaluate COR's influence on SIT. A total of 45 overlapping anti-diabetic gene targets were identified, and pathway enrichment analysis highlighted insulin resistance, lipid metabolism, and HIF-1 signalling, among others, as potential therapeutic intersections. CYP3A4 and CYP2C8 inhibition assays showed IC50 values of 2.815 µM and 0.645 µM for SIT, 4.277 µM and 0.470 µM for COR, and 3.999 µM and 0.389 µM for their combination, respectively. CYP3A4 inhibition assays showed IC50 values of 2.815 µM for SIT, 4.277 µM for COR, and 3.999 µM for their combination, indicating COR's influence on SIT metabolism. These findings suggest that COR may alter SIT's pharmacokinetic profile via CYP3A4 modulation, warranting caution in their combined use to maintain therapeutic efficacy. A sensitive LC-MS-QTOF method was developed to quantify SIT and COR in rat plasma concurrently. Pharmacokinetic analysis revealed that COR co-administration significantly reduced SIT's bioavailability, decreasing Cmax by 5.8-fold and AUC by 14.96-fold and prolonging t1/2 by increasing 1.52-fold. This integrated approach provides insight into herb-drug interactions in diabetes treatment, emphasizing the need for tailored dosing strategies in clinical applications.
Collapse
Affiliation(s)
- Bhukya Siva
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER) Kolkata, Chunilal Bhawan, Maniktala, Kolkata 700054, West Bengal, India
| | - Sahla Sherin
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER) Kolkata, Chunilal Bhawan, Maniktala, Kolkata 700054, West Bengal, India
| | - Ravi Adinarayan Somabattni
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER) Kolkata, Chunilal Bhawan, Maniktala, Kolkata 700054, West Bengal, India
| | - Satheesh Kumar Nanjappan
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER) Kolkata, Chunilal Bhawan, Maniktala, Kolkata 700054, West Bengal, India.
| |
Collapse
|
6
|
Bal T. Scaffold-free endocrine tissue engineering: role of islet organization and implications in type 1 diabetes. BMC Endocr Disord 2025; 25:107. [PMID: 40259265 PMCID: PMC12010671 DOI: 10.1186/s12902-025-01919-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 01/17/2025] [Indexed: 04/23/2025] Open
Abstract
Type 1 diabetes (T1D) is a chronic hyperglycemia disorder emerging from beta-cell (insulin secreting cells of the pancreas) targeted autoimmunity. As the blood glucose levels significantly increase and the insulin secretion is gradually lost, the entire body suffers from the complications. Although various advances in the insulin analogs, blood glucose monitoring and insulin application practices have been achieved in the last few decades, a cure for the disease is not obtained. Alternatively, pancreas/islet transplantation is an attractive therapeutic approach based on the patient prognosis, yet this treatment is also limited mainly by donor shortage, life-long use of immunosuppressive drugs and risk of disease transmission. In research and clinics, such drawbacks are addressed by the endocrine tissue engineering of the pancreas. One arm of this engineering is scaffold-free models which often utilize highly developed cell-cell junctions, soluble factors and 3D arrangement of islets with the cellular heterogeneity to prepare the transplant formulations. In this review, taking T1D as a model autoimmune disease, techniques to produce so-called pseudoislets and their applications are studied in detail with the aim of understanding the role of mimicry and pointing out the promising efforts which can be translated from benchside to bedside to achieve exogenous insulin-free patient treatment. Likewise, these developments in the pseudoislet formation are tools for the research to elucidate underlying mechanisms in pancreas (patho)biology, as platforms to screen drugs and to introduce immunoisolation barrier-based hybrid strategies.
Collapse
Affiliation(s)
- Tugba Bal
- Department of Bioengineering, Faculty of Engineering and Natural Sciences, Uskudar University, Istanbul, 34662, Turkey.
| |
Collapse
|
7
|
Ou X, Yu Z, Pan C, Zheng X, Li D, Qiao Z, Zheng X. Paeoniflorin: a review of its pharmacology, pharmacokinetics and toxicity in diabetes. Front Pharmacol 2025; 16:1551368. [PMID: 40260393 PMCID: PMC12009869 DOI: 10.3389/fphar.2025.1551368] [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: 12/25/2024] [Accepted: 03/19/2025] [Indexed: 04/23/2025] Open
Abstract
The escalating global prevalence of diabetes underscores the urgency of addressing its treatment and associated complications. Paeoniflorin, a monoterpenoid glycoside compound, has garnered substantial attention in recent years owing to its potential therapeutic efficacy in diabetes management. Thus, this study aims to systematically overview the pharmacological effects, pharmacokinetics and toxicity of paeoniflorin in diabetes. Plenty of evidences have verified that paeoniflorin improves diabetes and its complication through reducing blood sugar, enhancing insulin sensitivity, regulating gut microbiota and autophagy, restoration of mitochondrial function, regulation of lipid metabolism, anti-inflammation, anti-oxidative stress, inhibition of apoptosis, immune regulation and so on. Paeoniflorin possess the characteristics of rapid absorption, wide distribution, rapid metabolism and renal excretion. Meanwhile, toxicity studies have suggested that paeoniflorin has low acute toxicity, minimal subacute and chronic toxicity, and no genotoxic or mutational toxic effects. In conclusion, this paper systematically elucidates the potential therapeutic application and safety profile of paeoniflorin in diabetes management.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xiaoyuan Zheng
- Pharmacy Department, Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing, China
| |
Collapse
|
8
|
Shen ZQ, Chiu WT, Kao CH, Chen YC, Chen LH, Teng TW, Hsiung SY, Tzeng TY, Tung CY, Juan CC, Tsai TF. Wolfram syndrome 2 gene (CISD2) deficiency disrupts Ca 2+-mediated insulin secretion in β-cells. Mol Metab 2025; 96:102140. [PMID: 40189101 PMCID: PMC12020879 DOI: 10.1016/j.molmet.2025.102140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 03/24/2025] [Accepted: 04/01/2025] [Indexed: 04/15/2025] Open
Abstract
OBJECTIVE Diabetes, characterized by childhood-onset, autoantibody-negativity and insulin-deficiency, is a major manifestation of Wolfram syndrome 2 (WFS2), which is caused by recessive mutations of CISD2. Nevertheless, the mechanism underlying β-cell dysfunction in WFS2 remains elusive. Here we delineate the essential role of CISD2 in β-cells. METHODS We use β-cell specific Cisd2 knockout (Cisd2KO) mice, a CRISPR-mediated Cisd2KO MIN6 β-cell line and transcriptomic analysis. RESULTS Four findings are pinpointed. Firstly, β-cell specific Cisd2KO in mice disrupts systemic glucose homeostasis via impairing β-granules synthesis and insulin secretion; hypertrophy of the β-islets and the presence of a loss of identity that affects certain β-cells. Secondly, Cisd2 deficiency leads to impairment of glucose-induced extracellular Ca2+ influx, which compromises Ca2+-mediated insulin secretory signaling, causing mitochondrial dysfunction and, thereby impairing insulin secretion in the MIN6-Cisd2KO β-cells. Thirdly, transcriptomic analysis of β-islets reveals that Cisd2 modulates proteostasis and ER stress, mitochondrial function, insulin secretion and vesicle transport. Finally, the activated state of two potential upstream regulators, Glis3 and Hnf1a, is significantly suppressed under Cisd2 deficiency; notably, their downstream target genes are deeply involved in β-cell function and identity. CONCLUSIONS These findings provide mechanistic insights and form a basis for developing therapeutics for the effective treatment of diabetes in WFS2 patients.
Collapse
Affiliation(s)
- Zhao-Qing Shen
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Wen-Tai Chiu
- Department of Biomedical Engineering, College of Engineering, National Cheng Kung University, Tainan 701, Taiwan
| | - Cheng-Heng Kao
- Center of General Education, Chang Gung University, Taoyuan 333, Taiwan
| | - Yu-Chen Chen
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Li-Hsien Chen
- Department of Pharmacology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Tsai-Wen Teng
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Shao-Yu Hsiung
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Tsai-Yu Tzeng
- The National Genomics Center for Clinical and Biotechnological Applications, Cancer and Immunology Research Center, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Chien-Yi Tung
- The National Genomics Center for Clinical and Biotechnological Applications, Cancer and Immunology Research Center, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Chi-Chang Juan
- Institutes of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Ting-Fen Tsai
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; Center for Healthy Longevity and Aging Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli 350, Taiwan.
| |
Collapse
|
9
|
Li S, Ye T, Hou Z, Wang Y, Hao Z, Chen J. FOXO6: A unique transcription factor in disease regulation and therapeutic potential. Pharmacol Res 2025; 214:107691. [PMID: 40058512 DOI: 10.1016/j.phrs.2025.107691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/21/2025] [Accepted: 03/06/2025] [Indexed: 03/15/2025]
Abstract
FOXO6, a unique member of the Forkhead box O (FOXO) transcription factor family, has emerged as a pivotal regulator in various physiological and pathological processes, including apoptosis, oxidative stress, autophagy, cell cycle control, and inflammation. Unlike other FOXO proteins, FOXO6 exhibits distinct regulatory mechanisms, particularly its inability to undergo classical nucleocytoplasmic shuttling. These unique properties suggest that FOXO6 may function through alternative pathways, positioning it as a novel research target. This review provides the first comprehensive review of FOXO6's biological functions and its roles in the progression of multiple diseases, such as cancer, metabolic disorders, neurodegenerative conditions, and cardiovascular dysfunction. We highlight FOXO6's interaction with critical signaling pathways, including PI3K/Akt, PPARγ, and TXNIP, and discuss its contributions to tumor progression, glucose and lipid metabolism, oxidative stress, and neuronal degeneration. Moreover, FOXO6's potential as a therapeutic target is explored, with particular emphasis on its ability to modulate drug resistance and its implications for disease treatment. Despite its promising therapeutic potential, the development of FOXO6-targeted therapies remains challenging due to overlapping functions within the FOXO family and the context-dependent nature of FOXO6's regulatory roles. This review underscores the need for further experimental and clinical studies to elucidate the molecular mechanisms underlying FOXO6's functions and to validate its application in disease prevention and treatment. By systematically analyzing current research, this review aims to provide a foundational reference for future studies on FOXO6, paving the way for novel therapeutic strategies targeting this unique transcription factor.
Collapse
Affiliation(s)
- Songzhe Li
- College of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ting Ye
- The Second Hospital Affiliated Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Zhitao Hou
- College of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yuqing Wang
- College of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zhihua Hao
- College of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jing Chen
- College of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, China.
| |
Collapse
|
10
|
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
|
11
|
Ito T, Ozaki Y, Tanaka A, Kanai Y. Carbohydrate intake and activation of gastric acid secretion decrease gastric estrogen secretion. Sci Rep 2025; 15:10818. [PMID: 40155468 PMCID: PMC11953264 DOI: 10.1038/s41598-025-95718-z] [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: 11/14/2024] [Accepted: 03/24/2025] [Indexed: 04/01/2025] Open
Abstract
Gastric parietal cells (gastric acid-secreting cells) secrete estrogen in response to blood lipid (triglycerides [TG] and fatty acids [FFA]) levels. Since estrogen helps reduce blood lipid levels, gastric estrogen would be important in lowering elevated blood lipid levels. However, gastric parietal cells use lipid-derived energy for estrogen production and acid secretion. Thus, postprandial changes in blood lipid levels and acid secretion could affect gastric estrogen production. Here, we show that blood estrogen and FFA decrease after meals, especially after carbohydrate intake. Postprandially decreased blood estrogen levels are partially restored by intravenous lipid injections. Hormones that activate and suppress gastric acid secretion decrease and increase estrogen production in isolated gastric gland epithelia, respectively. Suppression of gastric acid secretion increases blood estrogen levels in postprandial, but not in fasted, rats. Carbohydrate ingestion releases insulin, which lowers blood FFA levels, while lipid intake releases hormones that suppress gastric acid secretion, such as glucagon-like peptide-1 (GLP-1). Insulin and gastric estrogen directly enter the liver in high concentrations, enhancing and suppressing hepatic de novo lipogenesis, respectively. The more lipids ingested, the less lipogenesis is required. Therefore, we propose that ingested carbohydrates and lipids negatively and positively control gastric estrogen production, respectively, for proper hepatic lipogenesis.
Collapse
Affiliation(s)
- Takao Ito
- Department of Cell Biology and Anatomy, Graduate School of Medicine, Wakayama Medical University, Wakayama, Wakayama, Japan
| | - Yuichi Ozaki
- Department of Cardiovascular Medicine, Graduate School of Medicine, Wakayama Medical University, Wakayama, Wakayama, Japan
| | - Atsushi Tanaka
- Department of Cardiovascular Medicine, Graduate School of Medicine, Wakayama Medical University, Wakayama, Wakayama, Japan
| | - Yoshimitsu Kanai
- Department of Cell Biology and Anatomy, Graduate School of Medicine, Wakayama Medical University, Wakayama, Wakayama, Japan.
| |
Collapse
|
12
|
Delannoy P, Tolan DR, Lanaspa MA, San Millán I, Bae SY, Johnson RJ. Aldose reductase, fructose and fat production in the liver. Biochem J 2025; 482:295-307. [PMID: 40040471 DOI: 10.1042/bcj20240748] [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: 11/18/2024] [Revised: 01/06/2025] [Accepted: 01/16/2025] [Indexed: 03/06/2025]
Abstract
There is an increasing interest in the role of fructose as a major driver of non-alcoholic fatty liver disease (NAFLD), and it is linked closely with the intake of sugar. However, there has also been the recognition that fructose can be produced directly from intracellular glucose via the evolutionarily conserved polyol pathway whose access is governed by aldose reductase (AR). The purpose of this article is to review the biochemistry of AR and the role of the polyol pathway in opening fructose metabolism. This article provides a new perspective about AR and the other key enzymes surrounding the decision to divert glucose into the polyol pathway which suggests that the production of endogenous fructose may be of much greater significance than historically viewed. There are important aspects of the regulation of the polyol pathway and its committal step catalyzed by AR, which supports the notion that fructose-uric acid pathway is activated by elevated glucose with the downstream consequence of NAFLD and perhaps other chronic metabolic diseases.
Collapse
Affiliation(s)
- Peter Delannoy
- Orgins of Human Metabolic Disease, Phoneix, AZ,85016, U.S.A
| | - Dean R Tolan
- Department of Biology, Boston University, Boston, MA, U.S.A
| | - Miguel A Lanaspa
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Denver, Aurora, CO, U.S.A
| | - Iñigo San Millán
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, U.S.A
| | - So Young Bae
- Molecular Biology, Cell Biology, and Biochemistry Program, Boston University, Boston, U.S.A
| | - Richard J Johnson
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, U.S.A
| |
Collapse
|
13
|
Huber MK, Widener AE, Cuaycal AE, Smurlick D, Butterworth EA, Lenchik NI, Chen J, Beery M, Hiller H, Verney E, Kusmartseva I, Rupnik MS, Campbell-Thompson M, Gerling IC, Atkinson MA, Mathews CE, Phelps EA. Beta cell dysfunction occurs independently of insulitis in type 1 diabetes pathogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.29.630665. [PMID: 39763971 PMCID: PMC11703223 DOI: 10.1101/2024.12.29.630665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
The loss of insulin secretory function associated with type 1 diabetes (T1D) is attributed to the immune-mediated destruction of beta cells. Yet, at onset of T1D, patients often have a significant beta cell mass remaining while T cell infiltration of pancreatic islets is sporadic. Thus, we investigated the hypothesis that the remaining beta cells in T1D are largely dysfunctional using live human pancreas tissue slices prepared from organ donors with recently diagnosed T1D. Beta cells in slices from donors with T1D had significantly diminished Ca2+ mobilization and insulin secretion responses to glucose. Beta cell function was equally impaired in T cell-infiltrated and non-infiltrated islets. Fixed tissue staining and gene expression profiling of laser-capture microdissected islets revealed significant decreases of proteins and genes in the glucose stimulus secretion coupling pathway. From these data, we posit that functional defects occur in the remaining mass of beta cells during human T1D pathogenesis.
Collapse
Affiliation(s)
- Mollie K. Huber
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, United States
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Adrienne E. Widener
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Alexandra E. Cuaycal
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
- Department of Infectious Diseases and Immunology, UF College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - Dylan Smurlick
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Elizabeth A. Butterworth
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, United States
| | - Nataliya I. Lenchik
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Jing Chen
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, United States
| | - Maria Beery
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, United States
| | - Helmut Hiller
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, United States
| | - Ellen Verney
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, United States
| | - Irina Kusmartseva
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, United States
| | - Marjan Slak Rupnik
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Alma Mater Europaea University—European Center Maribor, Maribor, Slovenia
| | - Martha Campbell-Thompson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, United States
| | - Ivan C. Gerling
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Mark A. Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, United States
- Department of Pediatrics, University of Florida, Gainesville, FL, United States
| | - Clayton E. Mathews
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, United States
- Department of Infectious Diseases and Immunology, UF College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - Edward A. Phelps
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, United States
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| |
Collapse
|
14
|
Hogrebe NJ, Schmidt MD, Augsornworawat P, Gale SE, Shunkarova M, Millman JR. Depolymerizing F-actin accelerates the exit from pluripotency to enhance stem cell-derived islet differentiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.21.618465. [PMID: 39484596 PMCID: PMC11526947 DOI: 10.1101/2024.10.21.618465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
In this study, we demonstrate that cytoskeletal state at the onset of directed differentiation is critical for the specification of human pluripotent stem cells (hPSCs) to all three germ layers. In particular, a polymerized actin cytoskeleton facilitates directed ectoderm differentiation, while depolymerizing F-actin promotes mesendoderm lineages. Applying this concept to a stem cell-derived islet (SC-islet) differentiation protocol, we show that depolymerizing F-actin with latrunculin A (latA) during the first 24 hours of definitive endoderm formation facilitates rapid exit from pluripotency and alters Activin/Nodal, BMP, JNK-JUN, and WNT pathway signaling dynamics. These signaling changes influence downstream patterning of the gut tube, leading to improved pancreatic progenitor identity and decreased expression of markers associated with other endodermal lineages. Continued differentiation generates islets containing a higher percentage of β cells that exhibit improved maturation, insulin secretion, and ability to reverse hyperglycemia. Furthermore, this latA treatment reduces enterochromaffin cells in the final cell population and corrects differentiations from hPSC lines that otherwise fail to consistently produce pancreatic islets, highlighting the importance of cytoskeletal signaling at the onset of directed differentiation.
Collapse
Affiliation(s)
- Nathaniel J. Hogrebe
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Mason D. Schmidt
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Punn Augsornworawat
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Sarah E. Gale
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Mira Shunkarova
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Jeffrey R. Millman
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, 1 Brookings Drive, St. Louis, MO 63130, USA
| |
Collapse
|
15
|
Karami N, Taei A, Hassani SN, Alizadeh N, Eftekhari-Yazdi P, Hassani F. The effects of insulin-transferrin-selenium (ITS) and CHIR99021 on the development of pre-implantation human arrested embryos in vitro. Sci Rep 2025; 15:5006. [PMID: 39929940 PMCID: PMC11811121 DOI: 10.1038/s41598-025-89460-9] [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: 07/29/2024] [Accepted: 02/05/2025] [Indexed: 02/13/2025] Open
Abstract
Pre-implantation development arrest poses a significant challenge in infertility treatment cycles. This study aims to evaluate the effect of Insulin, Transferrin, Selenium (ITS), and CHIR99021 on arrested human embryos. Arrested human embryos were obtained from the Embryology Department of the Royan Institute. After determining optimal concentrations, the embryos were assigned to control, CHIR99021, and ITS groups and cultured for 48-72 h. The arrest rate significantly decreased in the ITS and CHIR99021 groups compared to the control group (P < 0.05). The developmental rate up to the pre-morula stage significantly increased in the CHIR99021 group compared to the control group (P < 0.05). Additionally, there were significant increases in the expression of SOX2 in the CHIR99021 group and CCNA2 in the ITS group compared to the control group (P < 0.05). Immunofluorescent staining confirmed the expression of NANOG protein in the experimental groups. GSK3 inhibition by CHIR99021 and the application of ITS can alleviate arrest in human embryos, promote cell cycle induction, and enable progression to the blastocyst stage. Comprehensive characterization of these blastocysts in future studies is crucial to support ITS and CHIR99021 probable application in culture systems, particularly for women of advanced maternal age and those experiencing severe male factor infertility.
Collapse
Affiliation(s)
- Narges Karami
- MSC., Faculty of Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Adeleh Taei
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Seyedeh Nafiseh Hassani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Nazanin Alizadeh
- MSC., Faculty of Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Poopak Eftekhari-Yazdi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, P.O. Box 16635-148, Tehran, Iran.
| | - Fatemeh Hassani
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, P.O. Box 16635-148, Tehran, Iran.
| |
Collapse
|
16
|
Li Y, Li RY, Zhu JY, Chen M, Mu WJ, Luo HY, Li S, Yan LJ, Yin MT, Li X, Chen HM, Guo L. Maternal exercise prevents metabolic disorders in offspring mice through SERPINA3C. Nat Metab 2025; 7:401-420. [PMID: 39891022 DOI: 10.1038/s42255-024-01213-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 12/17/2024] [Indexed: 02/03/2025]
Abstract
Maternal exercise can improve the metabolic health of the offspring. However, the molecular mechanisms underlying the beneficial effects of maternal exercise on the offspring remain unclear. Here, we show that maternal exercise during pregnancy alleviates high-fat diet (HFD)-induced adipose inflammation and glucose intolerance in offspring mice, accompanied by upregulation of the adipokine serine protease inhibitor A3C (SERPINA3C) both in maternal adipose tissues and the fetal circulation. Adipose SERPINA3C knockdown impairs, but its overexpression in dams mimics, maternal exercise-mediated metabolic benefits in HFD-fed offspring. Maternal SERPINA3C is transported into the fetal circulation and promotes Krüppel-like factor 4 (Klf4) gene promoter demethylation in fetal preadipocytes to increase KLF4 expression, which inhibits adipose inflammation in HFD-fed offspring mice. The SERPINA3C-cathepsin G-integrin β1 axis activates phosphatidylinositol 3-kinase signalling in preadipocytes. This promotes nuclear translocation of the p110β subunit to generate phosphatidylinositol 3,4,5-trisphosphate (PIP3) in the nucleus. O-linked β-N-acetylglucosamine (O-GlcNAc) transferase then binds to PIP3 to promote ten-eleven translocation methylcytosine dioxygenase 1 (TET1) O-GlcNAcylation, thereby enhancing TET1 activity to facilitate Klf4 gene promoter demethylation. These results provide mechanistic insights into maternal exercise-mediated improvement of offspring metabolism.
Collapse
Affiliation(s)
- Yang Li
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Ruo-Ying Li
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Jie-Ying Zhu
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Min Chen
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Wang-Jing Mu
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Hong-Yang Luo
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Shan Li
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Lin-Jing Yan
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Meng-Ting Yin
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Xin Li
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Hu-Min Chen
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Liang Guo
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China.
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China.
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China.
| |
Collapse
|
17
|
Tiwari RK, Rawat SG, Rai S, Kumar A. Stress regulatory hormones and cancer: the contribution of epinephrine and cancer therapeutic value of beta blockers. Endocrine 2025:10.1007/s12020-025-04161-7. [PMID: 39869294 DOI: 10.1007/s12020-025-04161-7] [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: 06/10/2024] [Accepted: 01/08/2025] [Indexed: 01/28/2025]
Abstract
The word "cancer" evokes myriad emotions, ranging from fear and despair to hope and determination. Cancer is aptly defined as a complex and multifaceted group of diseases that has unapologetically led to the loss of countless lives and affected innumerable families across the globe. The battle with cancer is not only a physical battle, but also an emotional, as well as a psychological skirmish for patients and for their loved ones. Cancer has been a part of our history, stories, and lives for centuries and has challenged the ingenuity of health and medical science, and the resilience of the human spirit. From the early days of surgery and radiation therapy to cutting-edge developments in chemotherapeutic agents, immunotherapy, and targeted treatments, the medical field continues to make significant headway in the fight against cancer. However, even after all these advancements, cancer is still among the leading cause of death globally. This urges us to understand the central hallmarks of neoplastic cells to identify novel molecular targets for the development of promising therapeutic approaches. Growing research suggests that stress mediators, including epinephrine, play a critical role in the development and progression of cancer by inducing neoplastic features through activating adrenergic receptors, particularly β-adrenoreceptors. Further, our experimental data has also shown that epinephrine mediates the growth of T-cell lymphoma by inducing proliferation, glycolysis, and apoptosis evasion via altering the expression levels of key regulators of these vital cellular processes. The beauty of receptor-based therapy lies in its precision and higher therapeutic value. Interestingly, the enhanced expression of β-adrenergic receptors (ADRBs), namely ADRB2 (β2-adrenoreceptor) and ADRB3 (β3-adrenoreceptor) has been noted in many cancers, such as breast, colon, gastric, pancreatic, and prostate and has been reported to play a pivotal role in facilitating cancer growth mainly by promoting proliferation, evasion of apoptosis, angiogenesis, invasion and metastasis, and chemoresistance. The present review article is an attempt to summarize the available findings which indicate a distinct relationship between stress hormones and cancer, with a special emphasis on epinephrine, considered as a key stress regulatory molecule. This article also discusses the possibility of using beta-blockers for cancer therapy.
Collapse
Affiliation(s)
- Rajan Kumar Tiwari
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
- School of Medicine and Health Sciences, The George Washington University, Washington DC, USA
| | - Shiv Govind Rawat
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
- MD Anderson Cancer Center, The University of Texas, Texas, USA
| | - Siddharth Rai
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Ajay Kumar
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India.
| |
Collapse
|
18
|
Hickey TE, Mudunuri U, Hempel HA, Kemp TJ, Roche NV, Talsania K, Sellers BA, Cherry JM, Pinto LA. Proteomic and serologic assessments of responses to mRNA-1273 and BNT162b2 vaccines in human recipient sera. Front Immunol 2025; 15:1502458. [PMID: 39931577 PMCID: PMC11808009 DOI: 10.3389/fimmu.2024.1502458] [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: 09/26/2024] [Accepted: 11/25/2024] [Indexed: 02/13/2025] Open
Abstract
Introduction The first vaccines approved against SARS-CoV-2, mRNA-1273 and BNT162b2, utilized mRNA platforms. However, little is known about the proteomic markers and pathways associated with host immune responses to mRNA vaccination. In this proof-of-concept study, sera from male and female vaccine recipients were evaluated for proteomic and immunologic responses 1-month and 6-months following homologous third vaccination. Methods An aptamer-based (7,289 marker) proteomic assay coupled with traditional serology was leveraged to generate a comprehensive evaluation of systemic responsiveness in 64 and 68 healthy recipients of mRNA-1273 and BNT162b2 vaccines, respectively. Results Sera from female recipients of mRNA-1273 showed upregulated indicators of inflammatory and immunological responses at 1-month post-third vaccination, and sera from female recipients of BNT162b2 demonstrated upregulated negative regulators of RNA sensors at 1-month. Sera from male recipients of mRNA-1273 showed no significant upregulation of pathways at 1-month post-third vaccination, though there were multiple significantly upregulated proteomic markers. Sera from male recipients of BNT162b2 demonstrated upregulated markers of immune response to doublestranded RNA and cell-cycle G(2)/M transition at 1-month. Random Forest analysis of proteomic data from pre-third-dose sera identified 85 markers used to develop a model predictive of robust or weaker IgG responses and antibody levels to SARS-CoV-2 spike protein at 6-months following boost; no specific markers were individually predictive of 6-month IgG response. Thirty markers that contributed most to the model were associated with complement cascade and activation; IL-17, TNFR pro-apoptotic, and PI3K signaling; and cell cycle progression. Discussion These results demonstrate the utility of proteomics to evaluate correlates or predictors of serological responses to SARS-CoV-2 vaccination.
Collapse
Affiliation(s)
- Thomas E. Hickey
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Uma Mudunuri
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Heidi A. Hempel
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Troy J. Kemp
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Nancy V. Roche
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Keyur Talsania
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Brian A. Sellers
- Center for Human Immunology, Inflammation and Autoimmunity, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - James M. Cherry
- Center for Human Immunology, Inflammation and Autoimmunity, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Ligia A. Pinto
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| |
Collapse
|
19
|
Chong S, Lin M, Chong D, Jensen S, Lau NS. A systematic review on gut microbiota in type 2 diabetes mellitus. Front Endocrinol (Lausanne) 2025; 15:1486793. [PMID: 39897957 PMCID: PMC11782031 DOI: 10.3389/fendo.2024.1486793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 12/18/2024] [Indexed: 02/04/2025] Open
Abstract
Aims/hypothesis The gut microbiota play crucial roles in the digestion and degradation of nutrients, synthesis of biological agents, development of the immune system, and maintenance of gastrointestinal integrity. Gut dysbiosis is thought to be associated with type 2 diabetes mellitus (T2DM), one of the world's fastest growing diseases. The aim of this systematic review is to identify differences in the composition and diversity of the gut microbiota in individuals with T2DM. Methods A systematic search was conducted to identify studies reporting on the difference in gut microbiota composition between individuals with T2DM and healthy controls. Relevant studies were evaluated, and their characteristics and results were extracted using a standardized data extraction form. The studies were assessed for risk of bias and their findings were reported narratively. Results 58 observational studies published between 2010 and 2024 were included. Beta diversity was commonly reported to be different between individuals with T2DM and healthy individuals. Genera Lactobacillus, Escherichia-Shigella, Enterococcus, Subdoligranulum and Fusobacteria were found to be positively associated; while Akkermansia, Bifidobacterium, Bacteroides, Roseburia, Faecalibacteirum and Prevotella were found to be negatively associated with T2DM. Conclusions This systematic review demonstrates a strong association between T2DM and gut dysbiosis, as evidenced by differential microbial abundances and altered diversity indices. Among these taxa, Escherichia-Shigella is consistently associated with T2DM, whereas Faecalibacterium prausnitzii appears to offer a protective effect against T2DM. However, the heterogeneity and observational nature of these studies preclude the establishment of causative relationships. Future research should incorporate age, diet and medication-matched controls, and include functional analysis of these gut microbes. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42023459937.
Collapse
Affiliation(s)
- Serena Chong
- South West Sydney Limb Preservation and Wound Research, Ingham Institute for Applied Medical Research, Sydney, NSW, Australia
- South West Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Mike Lin
- Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
- Garvan Institute of Research, Sydney, NSW, Australia
| | - Deborah Chong
- Animal Health Laboratory, Department of Natural Resources and Environment Tasmania, Tasmania, TAS, Australia
| | - Slade Jensen
- South West Sydney Limb Preservation and Wound Research, Ingham Institute for Applied Medical Research, Sydney, NSW, Australia
- Infectious Disease and Microbiology, Ingham Institute for Applied Medical Research, Sydney, NSW, Australia
- School of Medicine Antibiotic Resistance and Mobile Elements Groups, Ingham Institute for Applied Medical Research, Sydney, NSW, Australia
| | - Namson S. Lau
- South West Sydney Limb Preservation and Wound Research, Ingham Institute for Applied Medical Research, Sydney, NSW, Australia
- South West Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
- Liverpool Diabetes Collaboration, Ingham Institute of Applied Medical Research, Sydney, NSW, Australia
| |
Collapse
|
20
|
Bano S, More S, Mongad DS, Khalique A, Dhotre DP, Bhat MK, Seshadri V. Prolonged exposure to insulin might cause epigenetic alteration leading to insulin resistance. FEBS Open Bio 2025; 15:81-93. [PMID: 39471069 PMCID: PMC11705401 DOI: 10.1002/2211-5463.13891] [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: 09/09/2022] [Revised: 08/02/2024] [Accepted: 08/26/2024] [Indexed: 11/01/2024] Open
Abstract
Glucose homeostasis is maintained by insulin. Insulin resistance is caused by multiple factors including hereditary factors and diet. The molecular mechanism underlying insulin resistance (IR) is not completely understood. Hyperinsulinemia often precedes insulin resistance and Type 2 diabetes. We had previously shown that prolonged exposure of insulin-responsive cells to insulin in the absence of high levels of glucose led to insulin resistance. In the present study, we show that the underlying cause for the impaired insulin signalling is the defective PI3K/AKT pathway. The observed insulin resistance is likely due to epigenetic alterations, as it can be maintained for several generations even when insulin is not provided, and epigenetic modifiers can reverse it. We also show that liver cell line (BRL-3A) developed impaired insulin signalling upon prolonged exposure to insulin in the absence of high levels of glucose. Transcriptomic analysis of the insulin-sensitive and resistance cells uncover altered signalling networks involved in chromatin remodelling, Rho GTPases, and ubiquitination. Furthermore, trimethylation of histone H3 at lysine 4 (H3K4me3) is increased in insulin-resistant cells. We extended these studies to mice, and show that mice injected with low doses of insulin when fasting develop insulin resistance with impaired glucose tolerance and increased HOMA-IR index. Altogether, these findings suggest that dysregulated synthesis of insulin in the absence of glucose stimulus could lead to epigenetic alterations that may ultimately result in insulin resistance.
Collapse
Affiliation(s)
- Shehnaz Bano
- National Centre for Cell SciencePuneIndia
- Savitribai Phule Pune UniversityPuneIndia
| | - Shyam More
- National Centre for Cell SciencePuneIndia
- Savitribai Phule Pune UniversityPuneIndia
| | | | | | | | | | | |
Collapse
|
21
|
Yang Y, Wang TT, Xie HA, Hu PP, Li P. Experimental cell models of insulin resistance: overview and appraisal. Front Endocrinol (Lausanne) 2024; 15:1469565. [PMID: 39749015 PMCID: PMC11693592 DOI: 10.3389/fendo.2024.1469565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 12/02/2024] [Indexed: 01/04/2025] Open
Abstract
Insulin resistance, a key factor in the development of type 2 diabetes mellitus (T2DM), is defined as a defect in insulin-mediated control of glucose metabolism in tissues such as liver, fat and muscle. Insulin resistance is a driving force behind various metabolic diseases, such as T2DM, hyperlipidemia, hypertension, coronary heart disease and fatty liver. Therefore, improving insulin sensitivity can be considered as an effective strategy for the prevention and treatment of these complex metabolic diseases. Cell-based models are extensively employed for the study of pathological mechanisms and drug screening, particularly in relation to insulin resistance in T2DM. Currently, numerous methods are available for the establishment of in vitro insulin resistance models, a comprehensive review of these models is required and can serve as an excellent introduction or understanding for researchers undertaking studies in this filed. This review examines and discusses the primary methods for establishing and evaluating insulin resistance cell models. Furthermore, it highlights key issues and suggestions on cell selection, establishment, evaluation and drug screening of insulin resistance, thereby providing valuable references for the future research efforts.
Collapse
Affiliation(s)
- Ying Yang
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Research Laboratory for Drug Metabolism, Chongqing Medical University, Chongqing, China
| | - Ting-ting Wang
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Research Laboratory for Drug Metabolism, Chongqing Medical University, Chongqing, China
| | - Hu-ai Xie
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Research Laboratory for Drug Metabolism, Chongqing Medical University, Chongqing, China
| | - Ping Ping Hu
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Research Laboratory for Drug Metabolism, Chongqing Medical University, Chongqing, China
| | - Pan Li
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Research Laboratory for Drug Metabolism, Chongqing Medical University, Chongqing, China
| |
Collapse
|
22
|
Aldroubi BG, Najjar JA, Youssef TS, Rizk CE, Abuamreh BA, Aramouni K, Ghadieh HE, Najjar SM. Cell-specific regulation of insulin action and hepatic fibrosis by CEACAM1. METABOLISM AND TARGET ORGAN DAMAGE 2024; 4:34. [PMID: 39640841 PMCID: PMC11619085 DOI: 10.20517/mtod.2024.48] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
The incidence of metabolic dysfunction-associated steatotic liver disease (MASLD) has reached an epidemic rise worldwide. The disease is a constellation of a broad range of metabolic and histopathologic abnormalities. It begins with hepatic steatosis and progresses to metabolic dysfunction-associated steatohepatitis (MASH), including hepatic fibrosis, apoptosis, and cell injury. Despite ample research effort, the pathogenesis of the disease has not been fully delineated. Whereas insulin resistance is implicated in the early stages of the disease, its role in hepatic fibrosis remains controversial. We have focused our studies on the role of carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) in hepatocytes and endothelial cells in the metabolic and histopathological dysregulation in MASH. Patients with MASH exhibit lower hepatic CEACAM1 with a progressive decline in hepatocytes and endothelial cells as the fibrosis stage advances. In mice, conditional deletion of CEACAM1 in hepatocytes impairs insulin clearance to cause hyperinsulinemia-driven insulin resistance with steatohepatitis and hepatic fibrosis even when mice are fed a regular chow diet. In contrast, its conditional deletion in endothelial cells causes inflammation-driven hepatic fibrosis without adversely affecting metabolism (mice remain insulin-sensitive and do not develop hepatic steatosis). Thus, this review provides in vivo evidence that supports or discards the role of insulin resistance in liver injury and hepatic fibrosis.
Collapse
Affiliation(s)
- Basel G. Aldroubi
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - John A. Najjar
- Department of Pathology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Tya S. Youssef
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Al-Koura PO box 100 Tripoli, Kalhat, Lebanon
| | - Carl E. Rizk
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Al-Koura PO box 100 Tripoli, Kalhat, Lebanon
| | - Basil A.M. Abuamreh
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Karl Aramouni
- Department of Medicine, Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Hilda E. Ghadieh
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Al-Koura PO box 100 Tripoli, Kalhat, Lebanon
| | - Sonia M. Najjar
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 43614, USA
| |
Collapse
|
23
|
Yang B, Wang Q, Li Y, Zhang S, Sun Y, Wei Y, Jiang Q, Huang Y. Resveratrol inhibits white adipose deposition by the ESR1-mediated PI3K/AKT signaling pathway. Cell Signal 2024; 124:111448. [PMID: 39369759 DOI: 10.1016/j.cellsig.2024.111448] [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: 07/08/2024] [Revised: 09/20/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024]
Abstract
Excessive adipose accumulation is the primary cause of obesity. Resveratrol (RES), a natural polyphenolic compound, has garnered significant attention for its anti-obesity properties. However, the precise mechanisms by which RES influences fat deposition have not yet been explored. In this study, the aim was to identify the target proteins and associated pathways of RES in order to elucidate the mechanisms by which RES reduces fat deposition. In this study, mice were administered 400 mg/kg of RES via gavage for 12 weeks. We found that while 400 mg/kg RES had no impact on the growth of the mice, it significantly reduced the weight of various white adipose tissues, as well as the serum and liver concentrations of total cholesterol and triglycerides. Network pharmacology identified 15 potential targets of RES and highlighted the PI3K/AKT signaling pathway as a key pathway. Molecular docking and dynamic simulations suggested that ESR1 might be the target protein through which RES exerts its anti-fat deposition effects. In vitro experiments revealed that ESR1 promotes the proliferation and inhibits the differentiation of 3 T3-L1 adipocytes, and suppresses the PI3K/AKT signaling pathway. Silencing the ESR1 gene altered the ability of RES to inhibit cell differentiation via the PI3K/AKT pathway. Gene expression results in subcutaneous adipose tissue, epididymal fat tissue, and liver tissue of mice were consistent with observations in cells. In summary, RES reduces white fat deposition by directly targeting the ESR1 protein and inhibiting the PI3K/AKT signaling pathway. Our findings provide new insights into the potential use of RES in the prevention and treatment of obesity.
Collapse
Affiliation(s)
- Bao Yang
- College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China
| | - Qian Wang
- College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China
| | - Yin Li
- College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China
| | - Sanbao Zhang
- College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China; Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi University, Nanning 530004, Guangxi, China
| | - Yanjie Sun
- College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China
| | - Yangyang Wei
- College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China
| | - Qinyang Jiang
- College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China; Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi University, Nanning 530004, Guangxi, China.
| | - Yanna Huang
- College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China; Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi University, Nanning 530004, Guangxi, China.
| |
Collapse
|
24
|
Bednarczyk M, Dąbrowska-Szeja N, Łętowski D, Dzięgielewska-Gęsiak S, Waniczek D, Muc-Wierzgoń M. Relationship Between Dietary Nutrient Intake and Autophagy-Related Genes in Obese Humans: A Narrative Review. Nutrients 2024; 16:4003. [PMID: 39683397 PMCID: PMC11643440 DOI: 10.3390/nu16234003] [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: 09/24/2024] [Revised: 11/14/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Obesity is one of the world's major public health challenges. Its pathogenesis and comorbid metabolic disorders share common mechanisms, such as mitochondrial or endoplasmic reticulum dysfunction or oxidative stress, gut dysbiosis, chronic inflammation and altered autophagy. Numerous pro-autophagy dietary interventions are being investigated for their potential obesity-preventing or therapeutic effects. We summarize current data on the relationship between autophagy and obesity, and discuss various dietary interventions as regulators of autophagy-related genes in the prevention and ultimate treatment of obesity in humans, as available in scientific databases and published through July 2024. Lifestyle modifications (such as calorie restriction, intermittent fasting, physical exercise), including following a diet rich in flavonoids, antioxidants, specific fatty acids, specific amino acids and others, have shown a beneficial role in the induction of this process. The activation of autophagy through various nutritional interventions tends to elicit a consistent response, characterized by the induction of certain kinases (including AMPK, IKK, JNK1, TAK1, ULK1, and VPS34) or the suppression of others (like mTORC1), the deacetylation of proteins, and the alleviation of inhibitory interactions between BECN1 and members of the Bcl-2 family. Significant health/translational properties of many nutrients (nutraceuticals) can affect chronic disease risk through various mechanisms that include the activation or inhibition of autophagy. The role of nutritional intervention in the regulation of autophagy in obesity and its comorbidities is not yet clear, especially in obese individuals.
Collapse
Affiliation(s)
- Martyna Bednarczyk
- Department of Cancer Prevention, Faculty of Public Health, Medical University of Silesia in Katowice, 40-055 Katowice, Poland; (M.B.); (N.D.-S.); (D.Ł.)
| | - Nicola Dąbrowska-Szeja
- Department of Cancer Prevention, Faculty of Public Health, Medical University of Silesia in Katowice, 40-055 Katowice, Poland; (M.B.); (N.D.-S.); (D.Ł.)
| | - Dariusz Łętowski
- Department of Cancer Prevention, Faculty of Public Health, Medical University of Silesia in Katowice, 40-055 Katowice, Poland; (M.B.); (N.D.-S.); (D.Ł.)
| | - Sylwia Dzięgielewska-Gęsiak
- Department of Internal Diseases Propaedeutics and Emergency Medicine, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, 40-055 Katowice, Poland;
| | - Dariusz Waniczek
- Department of Oncological Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 40-055 Katowice, Poland;
| | - Małgorzata Muc-Wierzgoń
- Department of Internal Diseases Propaedeutics and Emergency Medicine, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, 40-055 Katowice, Poland;
| |
Collapse
|
25
|
Park JE, Yoo J, Han JS. HM-Chromanone Alleviates Hyperglycemia by Activating AMPK and PI3K/AKT Pathways in Mice Fed a High-Fat Diet. Nutrients 2024; 16:3972. [PMID: 39599757 PMCID: PMC11597832 DOI: 10.3390/nu16223972] [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: 09/20/2024] [Revised: 11/06/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024] Open
Abstract
OBJECTIVES We investigated potential antihyperglycemic effects of HM-chromanone (HMC), a homoisoflavonoid isolated from Portulaca oleracea, in mice fed a high-fat diet (HFD). METHODS Five-week-old male C57BL/6J mice (n = 24) were divided into three groups: controls, mice fed an HFD (11 weeks), and HFD-fed mice receiving HMC supplementation (8 weeks). Various analyses assessed liver and skeletal muscle proteins, pancreatic β-cell histology, blood glucose and HbA1c levels, and homeostatic index of insulin resistance (HOMA-IR). RESULTS HMC supplementation significantly reduced fasting blood glucose and postprandial blood glucose levels in HFD-fed mice. HbA1c and serum insulin levels reduced significantly, and HOMA-IR improved. Compensatory β-cell hyperplasia was reduced, and pancreatic β-cell function improved. AMP-activated protein kinase (AMPK) was significantly activated in skeletal muscle and liver tissues. IRS-1tyr612 expression increased significantly. PI3K activation and Akt phosphorylation in skeletal muscles improved insulin signaling. Forkhead box protein O1 phosphorylation increased through hepatic AMPK activation. Phosphoenolpyruvate carboxykinase and glucose-6-phosphatase expression was inhibited. Glycogen synthase kinase 3β phosphorylation increased. CONCLUSIONS HMC supplementation alleviated hyperglycemia by activating the AMPK and PI3K/Akt pathways in skeletal muscles and the AMPK pathway in the liver of HFD-fed mice.
Collapse
Affiliation(s)
- Jae-eun Park
- Department of Hotel Baking Technology, Busan Health University, Busan 49318, Republic of Korea;
| | - Jeong Yoo
- Department of Food Science and Nutrition, Kimchi Research Institute, Pusan National University, Busan 46241, Republic of Korea;
| | - Ji-sook Han
- Department of Food Science and Nutrition, Kimchi Research Institute, Pusan National University, Busan 46241, Republic of Korea;
| |
Collapse
|
26
|
Kim MJ, Park SL, Kim HJ, Sung BH, Sohn JH, Bae JH. Functional expression of recombinant insulins in Saccharomyces cerevisiae. Microb Cell Fact 2024; 23:302. [PMID: 39529045 PMCID: PMC11552327 DOI: 10.1186/s12934-024-02571-2] [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: 04/08/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Since 1982, recombinant insulin has been used as a substitute for pancreatic insulin from animals. However, increasing demand in medical and food industries warrants the development of more efficient production methods. In this study, we aimed to develop a novel and efficient method for insulin production using a yeast secretion system. METHODS Here, insulin C-peptide was replaced with a hydrophilic fusion partner (HL18) containing an affinity tag for the hypersecretion and easy purification of proinsulin. The HL18 fusion partner was then removed by in vitro processing with the Kex2 endoprotease (Kex2p), and authentic insulin was recovered via affinity chromatography. To improve the insulin functions, molecular chaperones of the host strain were reinforced via the constitutive expression of HAC1. RESULTS The developed method was successfully applied for the expression of cow, pig, and chicken insulins in yeast. Moreover, biological activity of recombinant insulins was confirmed by growth stimulation of cell line. CONCLUSIONS Therefore, replacement of the C-peptide of insulin with the HL18 fusion partner and use of Kex2p for in vitro processing of proinsulin guarantees the economic production of animal insulins in yeast.
Collapse
Affiliation(s)
- Mi-Jin Kim
- Synthetic Biology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-Ro, Yuseong-Gu, Daejeon, 34141, Republic of Korea
| | - Se-Lin Park
- Synthetic Biology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-Ro, Yuseong-Gu, Daejeon, 34141, Republic of Korea
| | - Hyun-Jin Kim
- Cellapy Bio Inc. Bio-Venture Center, #211, 125 Gwahak-Ro, Yuseong-Gu, Daejeon, 34141, Republic of Korea
| | - Bong Hyun Sung
- Synthetic Biology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-Ro, Yuseong-Gu, Daejeon, 34141, Republic of Korea
| | - Jung-Hoon Sohn
- Synthetic Biology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-Ro, Yuseong-Gu, Daejeon, 34141, Republic of Korea.
- Cellapy Bio Inc. Bio-Venture Center, #211, 125 Gwahak-Ro, Yuseong-Gu, Daejeon, 34141, Republic of Korea.
| | - Jung-Hoon Bae
- Synthetic Biology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-Ro, Yuseong-Gu, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
27
|
Kryska A, Depciuch J, Krysa M, Paja W, Wosiak A, Nicoś M, Budzynska B, Sroka-Bartnicka A. Lipids balance as a spectroscopy marker of diabetes. Analysis of FTIR spectra by 2D correlation and machine learning analyses. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 320:124653. [PMID: 38901232 DOI: 10.1016/j.saa.2024.124653] [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: 01/03/2024] [Revised: 05/28/2024] [Accepted: 06/11/2024] [Indexed: 06/22/2024]
Abstract
The number of people suffering from type 2 diabetes has rapidly increased. Taking into account, that elevated intracellular lipid concentrations, as well as their metabolism, are correlated with diminished insulin sensitivity, in this study we would like to show lipids spectroscopy markers of diabetes. For this purpose, serum collected from rats (animal model of diabetes) was analyzed using Fourier Transformed Infrared-Attenuated Total Reflection (FTIR-ATR) spectroscopy. Analyzed spectra showed that rats with diabetes presented higher concentration of phospholipids and cholesterol in comparison with non-diabetic rats. Moreover, the analysis of second (IInd) derivative spectra showed no structural changes in lipids. Machine learning methods showed higher accuracy for IInd derivative spectra (from 65 % to 89 %) than for absorbance FTIR spectra (53-65 %). Moreover, it was possible to identify significant wavelength intervals from IInd derivative spectra using random forest-based feature selection algorithm, which further increased the accuracy of the classification (up to 92 % for phospholipid region). Moreover decision tree based on the selected features showed, that peaks at 1016 cm-1 and 2936 cm-1 can be good candidates of lipids marker of diabetes.
Collapse
Affiliation(s)
- Adrianna Kryska
- Independent Unit of Spectroscopy and Chemical Imaging, Faculty of Biomedical Sciences, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Joanna Depciuch
- Institute of Nuclear Physics, Polish Academy of Sciences, Walerego Eljasza - Radzikowskiego 152, 31-342 Kraków, Poland; Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodźki 1, Lublin 20-093, Poland
| | - Mikolaj Krysa
- Independent Unit of Spectroscopy and Chemical Imaging, Faculty of Biomedical Sciences, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Wiesław Paja
- Institute of Computer Science, University of Rzeszow, Pigonia 1, 35-310 Rzeszów, Poland
| | - Agnieszka Wosiak
- Institute of Information Technology, Lodz University of Technology, Politechniki 8, 93-590 Łódź, Poland
| | - Marcin Nicoś
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Jaczewskiego 8, 20-090 Lublin, Poland
| | - Barbara Budzynska
- Independent Laboratory of Behavioral Studies, Faculty of Biomedical Sciences, Medical University of Lublin, Chodzki 4a, 20-093 Lublin, Poland
| | - Anna Sroka-Bartnicka
- Independent Unit of Spectroscopy and Chemical Imaging, Faculty of Biomedical Sciences, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland.
| |
Collapse
|
28
|
Duangjan C, Arpawong TE, Spatola BN, Curran SP. Hepatic WDR23 proteostasis mediates insulin homeostasis by regulating insulin-degrading enzyme capacity. GeroScience 2024; 46:4461-4478. [PMID: 38767782 PMCID: PMC11336002 DOI: 10.1007/s11357-024-01196-y] [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: 02/08/2024] [Accepted: 05/08/2024] [Indexed: 05/22/2024] Open
Abstract
Maintaining insulin homeostasis is critical for cellular and organismal metabolism. In the liver, insulin is degraded by the activity of the insulin-degrading enzyme (IDE). Here, we establish a hepatic regulatory axis for IDE through WDR23-proteostasis. Wdr23KO mice have increased IDE expression, reduced circulating insulin, and defective insulin responses. Genetically engineered human cell models lacking WDR23 also increase IDE expression and display dysregulated phosphorylation of insulin signaling cascade proteins, IRS-1, AKT2, MAPK, FoxO, and mTOR, similar to cells treated with insulin, which can be mitigated by chemical inhibition of IDE. Mechanistically, the cytoprotective transcription factor NRF2, a direct target of WDR23-Cul4 proteostasis, mediates the enhanced transcriptional expression of IDE when WDR23 is ablated. Moreover, an analysis of human genetic variation in WDR23 across a large naturally aging human cohort in the US Health and Retirement Study reveals a significant association of WDR23 with altered hemoglobin A1C (HbA1c) levels in older adults, supporting the use of WDR23 as a new molecular determinant of metabolic health in humans.
Collapse
Affiliation(s)
- Chatrawee Duangjan
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
| | - Thalida Em Arpawong
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
| | - Brett N Spatola
- Dornsife College of Letters, Arts, and Science, University of Southern California, Los Angeles, CA, 90089, USA
| | - Sean P Curran
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA.
| |
Collapse
|
29
|
Tan J, Li X, Dou N. Insulin Resistance Triggers Atherosclerosis: Caveolin 1 Cooperates with PKCzeta to Block Insulin Signaling in Vascular Endothelial Cells. Cardiovasc Drugs Ther 2024; 38:885-893. [PMID: 37289375 PMCID: PMC11438709 DOI: 10.1007/s10557-023-07477-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/26/2023] [Indexed: 06/09/2023]
Abstract
OBJECTIVE To date, therapies for endothelial dysfunction have primarily focused on ameliorating identified atherosclerosis (AS) risk factors rather than explicitly addressing endothelium-based mechanism. An in-depth exploration of the pathological mechanisms of endothelial injury was performed herein. METHODS Aortic caveolin 1 (Cav1) knockdown was achieved in mice using lentivirus, and AS was induced using a high-fat diet. Mouse body weight, blood glucose, insulin, lipid parameters, aortic plaque, endothelial injury, vascular nitric oxide synthase (eNOS), injury marker, and oxidative stress were examined. The effect of Cav1 knockdown on the content of PKCzeta and PI3K/Akt/eNOS pathway-related protein levels, as well as PKCzeta binding to Akt, was studied. ZIP, a PKCzeta inhibitor, was utilized to treat HUVECs in vitro, and the effect of ZIP on cell viability, inflammatory response, oxidative stress, and Akt activation was evaluated. RESULTS Cav1 knockdown had no significant effect on body weight or blood glucose in mice over an 8-week period, whereas drastically reduced insulin, lipid parameters, endothelial damage, E-selectin, and oxidative stress and elevated eNOS levels. Moreover, Cav1 knockdown triggered decreased PKCzeta enrichment and the activation of the PI3K/Akt/eNOS pathway. PKCzeta has a positive effect on cells without being coupled by Cav1, and ZIP had no marked influence on PKCzeta-Akt binding following Cav1/PKCzeta coupling. CONCLUSION Cav1/PKCzeta coupling antagonizes the activation of PI3K on Akt, leading to eNOS dysfunction, insulin resistance, and endothelial cell damage.
Collapse
Affiliation(s)
- Jingjing Tan
- Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, 200081, China
| | - Xiaoguang Li
- Department of Thyroid Breast and Vascular Surgery, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, 1279 Sanmen Road, Hongkou District, Shanghai, 200081, China
- Department of Vascular and Endovascular Surgery, Changzheng Hospital Affiliated to the Naval Medical University, Shanghai, 200003, China
| | - Ning Dou
- Department of Thyroid Breast and Vascular Surgery, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, 1279 Sanmen Road, Hongkou District, Shanghai, 200081, China.
| |
Collapse
|
30
|
Mandal AK, Sahoo A, Almalki WH, Almujri SS, Alhamyani A, Aodah A, Alruwaili NK, Abdul Kadir SZBS, Mandal RK, Almalki RA, Lal JA, Rahman M. Phytoactives for Obesity Management: Integrating Nanomedicine for Its Effective Delivery. Nutr Rev 2024:nuae136. [PMID: 39331591 DOI: 10.1093/nutrit/nuae136] [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] [Indexed: 09/29/2024] Open
Abstract
Obesity is a global health concern that requires urgent investigation and management. While synthetic anti-obesity medications are available, they come with a high risk of side-effects and variability in their efficacy. Therefore, natural compounds are increasingly being used to treat obesity worldwide. The proposition that naturally occurring compounds, mainly polyphenols, can be effective and safer for obesity management through food and nutrient fortification is strongly supported by extensive experimental research. This review focuses on the pathogenesis of obesity while reviewing the efficacy of an array of phytoactives used for obesity treatment. It details mechanisms such as enzyme inhibition, energy expenditure, appetite suppression, adipocyte differentiation, lipid metabolism, and modulation of gut microbiota. Comprehensive in vitro, in vivo, and preclinical studies underscore the promise of phytoactives in combating obesity, which have been thoroughly reviewed. However, challenges, such as poor bioavailability and metabolism, limit their potential. Advances in nanomedicines may overcome these constraints, offering a new avenue for enhancing the efficacy of phytoactives. Nonetheless, rigorous and targeted clinical trials are essential before applying phytoactives as a primary treatment for obesity.
Collapse
Affiliation(s)
- Ashok Kumar Mandal
- Department of Pharmacology, Faculty of Medicine, University Malaya, Kuala Lumpur 50603, Malaysia
| | - Ankit Sahoo
- Department of Pharmaceutical Sciences, Shalom Institute of Health & Allied Sciences, Sam Higginbottom University of Agriculture, Technology & Sciences, Allahabad, Uttar Pradesh 211007, India
| | - Waleed H Almalki
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Salem Salman Almujri
- Department of Pharmacology, College of Pharmacy, King Khalid University, Asir-Abha 61421, Saudi Arabia
| | - Abdulrahman Alhamyani
- Pharmaceuticals Chemistry Department, Faculty of Clinical Pharmacy, Al Baha University, Al Baha 65779, Saudi Arabia
| | - Alhussain Aodah
- College of Pharmacy, Prince Sattam bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| | - Nabil K Alruwaili
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakakah 72341, Saudi Arabia
| | | | | | - Rami A Almalki
- Clinical Pharmacy Unit, Pharmaceutical Care Department, King Faisal Hospital, Makkah Health Cluster, Makkah 24382, Saudi Arabia
| | - Jonathan A Lal
- Department of Molecular and Cellular Engineering, Jacob Institute of Biotechnology and Bioengineering, Sam Higginbottom University of Agriculture, Technology, and Sciences, Prayagraj, Uttar Pradesh 211007, India
| | - Mahfoozur Rahman
- Department of Pharmaceutical Sciences, Shalom Institute of Health & Allied Sciences, Sam Higginbottom University of Agriculture, Technology & Sciences, Allahabad, Uttar Pradesh 211007, India
| |
Collapse
|
31
|
Azar S, Maroun Abou Jaoude N, Kędzia A, Niechciał E. Barriers to Type 1 Diabetes Adherence in Adolescents. J Clin Med 2024; 13:5669. [PMID: 39407728 PMCID: PMC11477045 DOI: 10.3390/jcm13195669] [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: 07/26/2024] [Revised: 08/26/2024] [Accepted: 09/14/2024] [Indexed: 10/20/2024] Open
Abstract
Background: Adolescence is a particularly crucial period of physical, emotional, and social development and adaptation, rendering these formative years rather challenging for individuals with chronic conditions like type 1 diabetes (T1D). Despite rapid improvement in diabetes therapies, adolescents with T1D are characterized by poorer adherence to treatment regimens compared with other pediatric age groups. Insufficient adherence is strongly related to low diabetes control, increasing morbidity, and risk for premature mortality. This study aimed to provide a comprehensive overview of adolescents' most common barriers to T1D adherence, stressing the need for a deep and comprehensive understanding of these barriers. The complexity of these barriers is underscored by the diverse factors contributing to poor T1D adherence in adolescents. Methods: A narrative review was conducted, surveying four databases (PubMed, Scopus, EMBASE, and Web of Science) for full-text articles in the English language published up to June 2024. All studies related to barriers to T1D adherence in adolescents were considered. The literature was selected using selection and exclusion criteria and extracted and organized using Mendeley. Exclusion criteria included studies with insufficient data and non-peer-reviewed articles. This review revealed that adolescents face numerous obstacles to T1D adherence related to psychological factors, flux in family dynamics, perceived social pressures, therapy-related factors, transitioning responsibility, risk-taking behaviors, and pubertal changes. Conclusions: Navigating the adaptations to the different aspects of T1D, from treatment to complications and adolescents' personal growth, effectively requires a thorough understanding of the barriers of a treatment regimen that patients at this critical age face.
Collapse
Affiliation(s)
| | | | | | - Elżbieta Niechciał
- Department of Pediatric Diabetes, Clinical Auxology and Obesity, Poznan University of Medical Sciences, Szpitalna Street 27/33, 60-572 Poznan, Poland; (S.A.); (N.M.A.J.); (A.K.)
| |
Collapse
|
32
|
Ziaunys M, Mikalauskaite K, Sakalauskas A, Smirnovas V. Study of Insulin Aggregation and Fibril Structure under Different Environmental Conditions. Int J Mol Sci 2024; 25:9406. [PMID: 39273350 PMCID: PMC11395423 DOI: 10.3390/ijms25179406] [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: 08/07/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
Protein amyloid aggregation is linked with widespread and fatal neurodegenerative disorders as well as several amyloidoses. Insulin, a small polypeptide hormone, is associated with injection-site amyloidosis and is a popular model protein for in vitro studies of amyloid aggregation processes as well as in the search for potential anti-amyloid compounds. Despite hundreds of studies conducted with this specific protein, the procedures used have employed a vast array of different means of achieving fibril formation. These conditions include the use of different solution components, pH values, ionic strengths, and other additives. In turn, this variety of conditions results in the generation of fibrils with different structures, morphologies and stabilities, which severely limits the possibility of cross-study comparisons as well as result interpretations. In this work, we examine the condition-structure relationship of insulin amyloid aggregation under a range of commonly used pH and ionic strength conditions as well as solution components. We demonstrate the correlation between the reaction solution properties and the resulting aggregation kinetic parameters, aggregate secondary structures, morphologies, stabilities and dye-binding modes.
Collapse
Affiliation(s)
| | | | | | - Vytautas Smirnovas
- Institute of Biotechnology, Life Sciences Center, Vilnius University, LT-10257 Vilnius, Lithuania; (M.Z.); (K.M.); (A.S.)
| |
Collapse
|
33
|
Ferraz L, Barros M, Almeida K, Silva M, Bueno N. Effects of dietary supplementation in treatment and control of progression and complications of insulin-dependent diabetes mellitus: a systematic review with meta-analyses of randomized clinical trials. Braz J Med Biol Res 2024; 57:e13649. [PMID: 39194033 PMCID: PMC11349153 DOI: 10.1590/1414-431x2024e13649] [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: 03/07/2024] [Accepted: 06/19/2024] [Indexed: 08/29/2024] Open
Abstract
There is no safe and effective prevention for insulin-dependent diabetes (IDDM) mellitus, which makes it highly dependent on its treatment. This systematic review with meta-analyses of randomized clinical trials investigated the overall effects of dietary supplements of vitamins, minerals, trace elements, and non-essential compounds with antioxidant properties, fatty acids, and amino acids in IDDM. Searches of MEDLINE, Embase, CENTRAL, LILACS, The Grey Literature Report, and ClinicaTrials.gov, and citations from previous reviews were used to identify reports published through July 2023. The Risk of Bias 2 (RoB2) tool was used to analyze the risk of bias and GRADE was used to assess the quality of the results. Fifty-eight studies (n=3,044) were included in qualitative analyses and seventeen (n=723) in meta-analyses. Qualitative analyses showed few positive effects on some metabolic function markers, such as endothelial and renal function and lipid profile. Meta-analyses showed a positive effect of omega-3 on glycated hemoglobin (HbA1c) (RMD=-0.33; 95%CI: -0.53, -0.12, P=0.002; I2=0%; GRADE: low quality; 4 studies) and of vitamin D on fasting C-peptide (FCP) (RMD=0.05; 95%CI: 0.01, 0.9, P=0.023; I2=0%; GRADE: very low quality; 4 studies). Most studies showed bias concern or high risk of bias. A recommendation for dietary supplementation in IDDM cannot be made because of the few positive results within different interventions and markers, the serious risk of bias in the included studies, and the low quality of evidence from meta-analyses. The positive result of vitamin D on FCP is preliminary, requiring further investigation.
Collapse
Affiliation(s)
- L.C. Ferraz
- Laboratório de Nutrição e Metabolismo, Faculdade de Nutrição, Universidade Federal de Alagoas, Maceió, AL, Brasil
| | - M.D.R. Barros
- Laboratório de Nutrição e Metabolismo, Faculdade de Nutrição, Universidade Federal de Alagoas, Maceió, AL, Brasil
| | - K.M.M. Almeida
- Laboratório de Nutrição e Metabolismo, Faculdade de Nutrição, Universidade Federal de Alagoas, Maceió, AL, Brasil
| | - M.B.G. Silva
- Laboratório de Nutrição e Metabolismo, Faculdade de Nutrição, Universidade Federal de Alagoas, Maceió, AL, Brasil
| | - N.B. Bueno
- Laboratório de Nutrição e Metabolismo, Faculdade de Nutrição, Universidade Federal de Alagoas, Maceió, AL, Brasil
| |
Collapse
|
34
|
Mazzotta FA, Lucaccini Paoli L, Rizzi A, Tartaglione L, Leo ML, Popolla V, Barberio A, Viti L, Di Leo M, Pontecorvi A, Pitocco D. Unmet needs in the treatment of type 1 diabetes: why is it so difficult to achieve an improvement in metabolic control? Nutr Diabetes 2024; 14:58. [PMID: 39095349 PMCID: PMC11297181 DOI: 10.1038/s41387-024-00319-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 07/17/2024] [Accepted: 07/23/2024] [Indexed: 08/04/2024] Open
Abstract
The development of advanced diabetes technology has permitted persons with type 1 diabetes mellitus to improve metabolic control significantly, particularly with the development of advanced hybrid closed-loop systems which have improved the quality of life by reducing hypoglycemia, decreasing macroangiopathy and microangiopathy-related complications, ameliorating HbA1c and improving glycemic variability. Despite the progression made over the past few decades, there is still significant margin for improvement to be made in terms of attaining appropriate metabolic control. Various factors are responsible for poor glycemic control including inappropriate carbohydrate counting, repeated bouts of hypoglycemia, hypoglycemia unawareness, cutaneous manifestations due to localized insulin use and prolonged use of diabetes technology, psychosocial comorbidities such as eating disorders or 'diabulimia', the coexistence of insulin resistance among people with type 1 diabetes and the inability to mirror physiological endogenous pancreatic insulin secretion appropriately. Hence, the aim of this review is to highlight and overcome the barriers in attaining appropriate metabolic control among people with type 1 diabetes by driving research into adjunctive treatment for coexistent insulin resistance and developing new advanced diabetic technologies to preserve β cell function and mirror as much as possible endogenous pancreatic functions.
Collapse
Affiliation(s)
- Francesco Antonio Mazzotta
- Department of Endocrinology, Catholic University of the Sacred Heart, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Lorenzo Lucaccini Paoli
- Department of Endocrinology, Catholic University of the Sacred Heart, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.
| | - Alessandro Rizzi
- Diabetes Care Unit, Catholic University of the Sacred Heart, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Linda Tartaglione
- Diabetes Care Unit, Catholic University of the Sacred Heart, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Maria Laura Leo
- Department of Endocrinology, Catholic University of the Sacred Heart, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Valentina Popolla
- Diabetes Care Unit, Catholic University of the Sacred Heart, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Annarita Barberio
- Department of Internal Medicine, Catholic University of the Sacred Heart, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Luca Viti
- Diabetes Care Unit, Catholic University of the Sacred Heart, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Mauro Di Leo
- Diabetes Care Unit, Catholic University of the Sacred Heart, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Alfredo Pontecorvi
- Department of Endocrinology, Catholic University of the Sacred Heart, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Dario Pitocco
- Diabetes Care Unit, Catholic University of the Sacred Heart, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| |
Collapse
|
35
|
Barron MP, Vilseck JZ. A λ-Dynamics Investigation of Insulin Wakayama and Other A3 Variant Binding Affinities to the Insulin Receptor. J Chem Inf Model 2024; 64:5657-5670. [PMID: 38963805 PMCID: PMC11268370 DOI: 10.1021/acs.jcim.4c00662] [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: 04/19/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/06/2024]
Abstract
Insulin Wakayama is a clinical insulin variant where a conserved valine at the third residue on insulin's A chain (ValA3) is replaced with a leucine (LeuA3), weakening insulin receptor (IR) binding by 140-500-fold. This severe impact on binding from a subtle modification has posed an intriguing problem for decades. Although experimental investigations of natural and unnatural A3 mutations have highlighted the sensitivity of insulin-IR binding at this site, atomistic explanations of these binding trends have remained elusive. We investigate this problem computationally using λ-dynamics free energy calculations to model structural changes in response to perturbations of the ValA3 side chain and to calculate associated relative changes in binding free energy (ΔΔGbind). The Wakayama LeuA3 mutation and seven other A3 substitutions were studied in this work. The calculated ΔΔGbind results showed high agreement compared to experimental binding potencies with a Pearson correlation of 0.88 and a mean unsigned error of 0.68 kcal/mol. Extensive structural analyses of λ-dynamics trajectories revealed that critical interactions were disrupted between insulin and the insulin receptor as a result of the A3 mutations. This investigation also quantifies the effect that adding an A3 Cδ atom or losing an A3 Cγ atom has on insulin's binding affinity to the IR. Thus, λ-dynamics was able to successfully model the effects of mutations to insulin's A3 side chain on its protein-protein interactions with the IR and shed new light on a decades-old mystery: the exquisite sensitivity of hormone-receptor binding to a subtle modification of an invariant insulin residue.
Collapse
Affiliation(s)
- Monica P Barron
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Jonah Z Vilseck
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| |
Collapse
|
36
|
Ampofo E, Pack M, Wrublewsky S, Boewe AS, Spigelman AF, Koch H, MacDonald PE, Laschke MW, Montenarh M, Götz C. CK2 activity is crucial for proper glucagon expression. Diabetologia 2024; 67:1368-1385. [PMID: 38503901 PMCID: PMC11153270 DOI: 10.1007/s00125-024-06128-1] [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: 08/03/2023] [Accepted: 02/07/2024] [Indexed: 03/21/2024]
Abstract
AIMS/HYPOTHESIS Protein kinase CK2 acts as a negative regulator of insulin expression in pancreatic beta cells. This action is mainly mediated by phosphorylation of the transcription factor pancreatic and duodenal homeobox protein 1 (PDX1). In pancreatic alpha cells, PDX1 acts in a reciprocal fashion on glucagon (GCG) expression. Therefore, we hypothesised that CK2 might positively regulate GCG expression in pancreatic alpha cells. METHODS We suppressed CK2 kinase activity in αTC1 cells by two pharmacological inhibitors and by the CRISPR/Cas9 technique. Subsequently, we analysed GCG expression and secretion by real-time quantitative RT-PCR, western blot, luciferase assay, ELISA and DNA pull-down assays. We additionally studied paracrine effects on GCG secretion in pseudoislets, isolated murine islets and human islets. In vivo, we examined the effect of CK2 inhibition on blood glucose levels by systemic and alpha cell-specific CK2 inhibition. RESULTS We found that CK2 downregulation reduces GCG secretion in the murine alpha cell line αTC1 (e.g. from 1094±124 ng/l to 459±110 ng/l) by the use of the CK2-inhibitor SGC-CK2-1. This was due to a marked decrease in Gcg gene expression through alteration of the binding of paired box protein 6 (PAX6) and transcription factor MafB to the Gcg promoter. The analysis of the underlying mechanisms revealed that both transcription factors are displaced by PDX1. Ex vivo experiments in isolated murine islets and pseudoislets further demonstrated that CK2-mediated reduction in GCG secretion was only slightly affected by the higher insulin secretion after CK2 inhibition. The kidney capsule transplantation model showed the significance of CK2 for GCG expression and secretion in vivo. Finally, CK2 downregulation also reduced the GCG secretion in islets isolated from humans. CONCLUSIONS/INTERPRETATION These novel findings not only indicate an important function of protein kinase CK2 for proper GCG expression but also demonstrate that CK2 may be a promising target for the development of novel glucose-lowering drugs.
Collapse
Affiliation(s)
- Emmanuel Ampofo
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - Mandy Pack
- Medical Biochemistry and Molecular Biology, Saarland University, Homburg, Germany
| | - Selina Wrublewsky
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - Anne S Boewe
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - Aliya F Spigelman
- Department of Pharmacology and Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Hanna Koch
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - Patrick E MacDonald
- Department of Pharmacology and Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Matthias W Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - Mathias Montenarh
- Medical Biochemistry and Molecular Biology, Saarland University, Homburg, Germany
| | - Claudia Götz
- Medical Biochemistry and Molecular Biology, Saarland University, Homburg, Germany.
| |
Collapse
|
37
|
Luo Y, Luo D, Li M, Tang B. Insulin Resistance in Pediatric Obesity: From Mechanisms to Treatment Strategies. Pediatr Diabetes 2024; 2024:2298306. [PMID: 40302954 PMCID: PMC12016791 DOI: 10.1155/2024/2298306] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/07/2024] [Accepted: 06/15/2024] [Indexed: 05/02/2025] Open
Abstract
Insulin resistance, an increasingly prevalent characteristic among children and adolescents with obesity, is now recognized as a significant contributor to the development of type 2 diabetes mellitus (T2DM) and other metabolic diseases in individuals with obesity. Insulin resistance refers to a decrease in the sensitivity of peripheral tissues (primarily skeletal muscle, adipose tissue, and liver) to insulin, which is mainly characterized by impaired glucose uptake and utilization. Although the mechanisms underlying insulin resistance in children with obesity remain incompletely elucidated, several risk factors including lipid metabolism disorders, oxidative stress (OS), mitochondrial dysfunction, inflammation, and genetic factors have been identified as pivotal contributors to the pathogenesis of obesity-related insulin resistance. In this review, we comprehensively analyze relevant literature and studies to elucidate the underlying mechanisms of insulin resistance in childhood obesity. Additionally, we discuss treatment strategies for pediatric obesity from a perspective centered on improving insulin sensitivity, aiming to provide valuable insights for the prevention and management of pediatric obesity.
Collapse
Affiliation(s)
- Yu Luo
- Department of PediatricsSichuan Provincial People's HospitalSchool of MedicineUniversity of Electronic Science and Technology of China, Chengdu, China
| | - Dan Luo
- Department of PediatricsSchool of Medicine and Life Science of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Maojun Li
- Department of PediatricsSichuan Provincial People's HospitalSchool of MedicineUniversity of Electronic Science and Technology of China, Chengdu, China
| | - Binzhi Tang
- Department of PediatricsSichuan Provincial People's HospitalSchool of MedicineUniversity of Electronic Science and Technology of China, Chengdu, China
- Department of PediatricsSchool of Medicine and Life Science of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
38
|
Singh K, Das S, Sutradhar S, Howard J, Ray K. Insulin signaling accelerates the anterograde movement of Rab4 vesicles in axons through Klp98A/KIF16B recruitment via Vps34-PI3Kinase. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.24.590898. [PMID: 38895253 PMCID: PMC11185528 DOI: 10.1101/2024.04.24.590898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Rab4 GTPase organizes endosomal sorting essential for maintaining the balance between recycling and degradative pathways. Rab4 localizes to many cargos whose transport in neurons is critical for regulating neurotransmission and neuronal health. Furthermore, elevated Rab4 levels in the CNS are associated with synaptic atrophy and neurodegeneration in Drosophila and humans, respectively. However, how the transport of Rab4-associated vesicles is regulated in neurons remains unknown. Using in vivo time-lapse imaging of Drosophila larvae, we show that activation of insulin signaling via Dilp2 and dInR increases the anterograde velocity, run length, and flux of Rab4 vesicles in the axons. Molecularly, we show that activation of neuronal insulin signaling further activates Vps34, elevates the levels of PI(3)P on Rab4-associated vesicles, recruits Klp98A (a PI(3)P-binding kinesin-3 motor) and activates their anterograde transport. Together, these observations delineate the role of insulin signaling in regulating axonal transport and synaptic homeostasis.
Collapse
Affiliation(s)
- Kamaldeep Singh
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai - 400005, India
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT - 06520, United States
| | - Semanti Das
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai - 400005, India
| | - Sabyasachi Sutradhar
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT - 06520, United States
| | - Jonathon Howard
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT - 06520, United States
| | - Krishanu Ray
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai - 400005, India
- National Brain Research Centre, Manesar, Haryana – 122051, India
| |
Collapse
|
39
|
Orlowski J. Nuclear SMAD5 dances to a different tune in regulating insulin secretion. Cell Metab 2024; 36:1169-1171. [PMID: 38838638 DOI: 10.1016/j.cmet.2024.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 05/14/2024] [Accepted: 05/14/2024] [Indexed: 06/07/2024]
Abstract
In this issue of Cell Metabolism, Fang et al.1 report a novel pH-sensitive cellular signaling mechanism involving the transcription factor SMAD5 that regulates the vesicular secretion of insulin from pancreatic β cells in response to dietary challenges. Dysregulation of this pathway may contribute to metabolic disorders such as type 2 diabetes mellitus.
Collapse
Affiliation(s)
- John Orlowski
- Department of Physiology, McGill University, Bellini Life Sciences Bldg., Rm. 166, 3649 Promenade Sir-William-Osler, Montreal, QC H3G 0B1, Canada.
| |
Collapse
|
40
|
Kazmi I, Afzal M, Almalki WH, S RJ, Alzarea SI, Kumar A, Sinha A, Kukreti N, Ali H, Abida. From oncogenes to tumor suppressors: The dual role of ncRNAs in fibrosarcoma. Pathol Res Pract 2024; 258:155329. [PMID: 38692083 DOI: 10.1016/j.prp.2024.155329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/07/2024] [Accepted: 04/22/2024] [Indexed: 05/03/2024]
Abstract
Fibrosarcoma is a challenging cancer originating from fibrous tissues, marked by aggressive growth and limited treatment options. The discovery of non-coding RNAs (ncRNAs), including long non-coding RNAs (lncRNAs), microRNAs (miRNAs), and small interfering RNAs (siRNAs), has opened new pathways for understanding and treating this malignancy. These ncRNAs play crucial roles in gene regulation, cellular processes, and the tumor microenvironment. This review aims to explore the impact of ncRNAs on fibrosarcoma's pathogenesis, progression, and resistance to treatment, focusing on their mechanistic roles and therapeutic potential. A comprehensive review of literature from databases like PubMed and Google Scholar was conducted, focusing on the dysregulation of ncRNAs in fibrosarcoma, their contribution to tumor growth, metastasis, drug resistance, and their cellular pathway interactions. NcRNAs significantly influence fibrosarcoma, affecting cell proliferation, apoptosis, invasion, and angiogenesis. Their function as oncogenes or tumor suppressors makes them promising biomarkers and therapeutic targets. Understanding their interaction with the tumor microenvironment is essential for developing more effective treatments for fibrosarcoma. Targeting ncRNAs emerges as a promising strategy for fibrosarcoma therapy, offering hope to overcome the shortcomings of existing treatments. Further investigation is needed to clarify specific ncRNAs' roles in fibrosarcoma and to develop ncRNA-based therapies, highlighting the significance of ncRNAs in improving patient outcomes in this challenging cancer.
Collapse
Affiliation(s)
- Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia.
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Renuka Jyothi S
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, 72341, Sakaka, Al-Jouf, Saudi Arabia
| | - Ashwani Kumar
- Department of Pharmacy, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Aashna Sinha
- School of Applied and Life Sciences, Division of Research and Innovation Uttaranchal University, Dehradun, Uttarakhand, India
| | - Neelima Kukreti
- School of Pharmacy, Graphic Era Hill University, Dehradun 248007, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - Abida
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| |
Collapse
|
41
|
Haahr H, Cieslarová B, Hingst JR, Jiang S, Kristensen NR, Kupčová V, Nørgreen L, Wagner FDH, Ignatenko S. The Effect of Various Degrees of Renal or Hepatic Impairment on the Pharmacokinetic Properties of Once-Weekly Insulin Icodec. Clin Pharmacokinet 2024; 63:819-830. [PMID: 38722461 PMCID: PMC11222188 DOI: 10.1007/s40262-024-01375-2] [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] [Accepted: 02/13/2024] [Indexed: 07/04/2024]
Abstract
BACKGROUND AND OBJECTIVE Icodec is a once-weekly insulin being developed to provide basal insulin coverage in diabetes mellitus. This study evaluated the effects of renal or hepatic impairment on icodec pharmacokinetics. METHODS Two open-label, parallel-group, single-dose (1.5 U/kg subcutaneously) trials were conducted. In a renal impairment trial, 58 individuals were allocated to normal renal function (measured glomerular filtration rate ≥ 90 mL/min), mild (60 to < 90 mL/min), moderate (30 to < 60 mL/min) or severe (< 30 mL/min) renal impairment or end-stage renal disease. In a hepatic impairment trial, 25 individuals were allocated to normal hepatic function or mild (Child-Pugh Classification grade A), moderate (grade B) or severe (grade C) hepatic impairment. Blood was sampled frequently for a pharmacokinetic analysis until 35 days post-dose. RESULTS The shape of the icodec pharmacokinetic profile was not affected by renal or hepatic impairment. Total icodec exposure was greater for mild (estimated ratio [95% confidence interval]: 1.12 [1.01; 1.24]), moderate (1.24 [1.12; 1.37]) and severe (1.28 [1.16; 1.42]) renal impairment, and for end-stage renal disease (1.14 [1.03; 1.28]), compared with normal renal function. It was also greater for mild (1.13 [1.00; 1.28]) and moderate (1.15 [1.02; 1.29]) hepatic impairment versus normal hepatic function. There was no statistically significant difference between severe hepatic impairment and normal hepatic function. Serum albumin levels (range 2.7-5.1 g/dL) did not statistically significantly influence icodec exposure. CONCLUSIONS The clinical relevance of the slightly higher icodec exposure with renal or hepatic impairment is limited as icodec should be dosed according to individual need. No specific icodec dose adjustment is required in renal or hepatic impairment. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov identifiers: NCT03723785 and NCT04597697.
Collapse
Affiliation(s)
| | | | | | | | | | - Viera Kupčová
- Dérer's Hospital, Comenius University, Bratislava, Slovakia
| | | | | | | |
Collapse
|
42
|
Janssen JAMJL. Overnutrition, Hyperinsulinemia and Ectopic Fat: It Is Time for A Paradigm Shift in the Management of Type 2 Diabetes. Int J Mol Sci 2024; 25:5488. [PMID: 38791525 PMCID: PMC11121669 DOI: 10.3390/ijms25105488] [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: 04/15/2024] [Revised: 05/12/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
The worldwide incidence of prediabetes/type 2 has continued to rise the last 40 years. In the same period, the mean daily energy intake has increased, and the quality of food has significantly changed. The chronic exposure of pancreatic β-cells to calorie excess (excessive energy intake) and food additives may increase pancreatic insulin secretion, decrease insulin pulses and/or reduce hepatic insulin clearance, thereby causing chronic hyperinsulinemia and peripheral insulin resistance. Chronic calorie excess and hyperinsulinemia may promote lipogenesis, inhibit lipolysis and increase lipid storage in adipocytes. In addition, calorie excess and hyperinsulinemia can induce insulin resistance and contribute to progressive and excessive ectopic fat accumulation in the liver and pancreas by the conversion of excess calories into fat. The personal fat threshold hypothesis proposes that in susceptible individuals, excessive ectopic fat accumulation may eventually lead to hepatic insulin receptor resistance, the loss of pancreatic insulin secretion, hyperglycemia and the development of frank type 2 diabetes. Thus, type 2 diabetes seems (partly) to be caused by hyperinsulinemia-induced excess ectopic fat accumulation in the liver and pancreas. Increasing evidence further shows that interventions (hypocaloric diet and/or bariatric surgery), which remove ectopic fat in the liver and pancreas by introducing a negative energy balance, can normalize insulin secretion and glucose tolerance and induce the sustained biochemical remission of type 2 diabetes. This pathophysiological insight may have major implications and may cause a paradigm shift in the management of type 2 diabetes: avoiding/reducing ectopic fat accumulation in the liver and pancreas may both be essential to prevent and cure type 2 diabetes.
Collapse
Affiliation(s)
- Joseph A M J L Janssen
- Department of Internal Medicine, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
43
|
Rettig AR, Ganesh K. Cancer Cells Hijack Physiologic Metabolic Signals to Seed Liver Metastasis. Cancer Res 2024; 84:1548-1549. [PMID: 38502849 PMCID: PMC11157585 DOI: 10.1158/0008-5472.can-24-0835] [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: 03/12/2024] [Accepted: 03/14/2024] [Indexed: 03/21/2024]
Abstract
Metastasis arises from cancer cell-intrinsic adaptations and permissive tumor microenvironments (TME) that are distinct across different organs. Deciphering the mechanisms underpinning organotropism could provide novel preventive and therapeutic strategies for patients with cancer. Rogava and colleagues identified Pip4k2c as a driver of liver metastasis, acting by sensitizing cancer cells to insulin-dependent PI3K/AKT signaling, which could be reversed by dual pharmacologic inhibition of PI3K and SGLT2 or a ketogenic diet. The study highlights the importance of tumor microenvironment communication in the context of systemic physiology and points toward potential combination therapies.
Collapse
Affiliation(s)
- Andres R Rettig
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Karuna Ganesh
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
44
|
Duda-Madej A, Stecko J, Szymańska N, Miętkiewicz A, Szandruk-Bender M. Amyloid, Crohn's disease, and Alzheimer's disease - are they linked? Front Cell Infect Microbiol 2024; 14:1393809. [PMID: 38779559 PMCID: PMC11109451 DOI: 10.3389/fcimb.2024.1393809] [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: 02/29/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024] Open
Abstract
Crohn's disease (CD) is a chronic inflammatory disease that most frequently affects part of the distal ileum, but it may affect any part of the gastrointestinal tract. CD may also be related to systemic inflammation and extraintestinal manifestations. Alzheimer's disease (AD) is the most common neurodegenerative disease, gradually worsening behavioral and cognitive functions. Despite the meaningful progress, both diseases are still incurable and have a not fully explained, heterogeneous pathomechanism that includes immunological, microbiological, genetic, and environmental factors. Recently, emerging evidence indicates that chronic inflammatory condition corresponds to an increased risk of neurodegenerative diseases, and intestinal inflammation, including CD, increases the risk of AD. Even though it is now known that CD increases the risk of AD, the exact pathways connecting these two seemingly unrelated diseases remain still unclear. One of the key postulates is the gut-brain axis. There is increasing evidence that the gut microbiota with its proteins, DNA, and metabolites influence several processes related to the etiology of AD, including β-amyloid abnormality, Tau phosphorylation, and neuroinflammation. Considering the role of microbiota in both CD and AD pathology, in this review, we want to shed light on bacterial amyloids and their potential to influence cerebral amyloid aggregation and neuroinflammation and provide an overview of the current literature on amyloids as a potential linker between AD and CD.
Collapse
Affiliation(s)
- Anna Duda-Madej
- Department of Microbiology, Faculty of Medicine, Wroclaw Medical University, Wrocław, Poland
| | - Jakub Stecko
- Faculty of Medicine, Wroclaw Medical University, Wrocław, Poland
| | | | | | - Marta Szandruk-Bender
- Department of Pharmacology, Faculty of Medicine, Wroclaw Medical University, Wrocław, Poland
| |
Collapse
|
45
|
Rosenstock J, Juneja R, Beals JM, Moyers JS, Ilag L, McCrimmon RJ. The Basis for Weekly Insulin Therapy: Evolving Evidence With Insulin Icodec and Insulin Efsitora Alfa. Endocr Rev 2024; 45:379-413. [PMID: 38224978 PMCID: PMC11091825 DOI: 10.1210/endrev/bnad037] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Indexed: 01/17/2024]
Abstract
Basal insulin continues to be a vital part of therapy for many people with diabetes. First attempts to prolong the duration of insulin formulations were through the development of suspensions that required homogenization prior to injection. These insulins, which required once- or twice-daily injections, introduced wide variations in insulin exposure contributing to unpredictable effects on glycemia. Advances over the last 2 decades have resulted in long-acting, soluble basal insulin analogues with prolonged and less variable pharmacokinetic exposure, improving their efficacy and safety, notably by reducing nocturnal hypoglycemia. However, adherence and persistence with once-daily basal insulin treatment remains low for many reasons including hypoglycemia concerns and treatment burden. A soluble basal insulin with a longer and flatter exposure profile could reduce pharmacodynamic variability, potentially reducing hypoglycemia, have similar efficacy to once-daily basal insulins, simplify dosing regimens, and improve treatment adherence. Insulin icodec (Novo Nordisk) and insulin efsitora alfa (basal insulin Fc [BIF], Eli Lilly and Company) are 2 such insulins designed for once-weekly administration, which have the potential to provide a further advance in basal insulin replacement. Icodec and efsitora phase 2 clinical trials, as well as data from the phase 3 icodec program indicate that once-weekly insulins provide comparable glycemic control to once-daily analogues, with a similar risk of hypoglycemia. This manuscript details the technology used in the development of once-weekly basal insulins. It highlights the clinical rationale and potential benefits of these weekly insulins while also discussing the limitations and challenges these molecules could pose in clinical practice.
Collapse
Affiliation(s)
- Julio Rosenstock
- Velocity Clinical Research at Medical City,
Dallas, TX 75230, USA
| | - Rattan Juneja
- Lilly Diabetes and Obesity, Eli Lilly and Company,
Indianapolis, IN 46225, USA
| | - John M Beals
- Lilly Diabetes and Obesity, Eli Lilly and Company,
Indianapolis, IN 46225, USA
| | - Julie S Moyers
- Lilly Diabetes and Obesity, Eli Lilly and Company,
Indianapolis, IN 46225, USA
| | - Liza Ilag
- Lilly Diabetes and Obesity, Eli Lilly and Company,
Indianapolis, IN 46225, USA
| | - Rory J McCrimmon
- School of Medicine, University of Dundee, Dundee
DD1 9SY, Scotland, UK
| |
Collapse
|
46
|
Gignac T, Trépanier G, Pradeau M, Morissette A, Agrinier AL, Larose É, Marois J, Pilon G, Gagnon C, Vohl MC, Marette A, Carreau AM. Metabolic-associated fatty liver disease is characterized by a post-oral glucose load hyperinsulinemia in individuals with mild metabolic alterations. Am J Physiol Endocrinol Metab 2024; 326:E616-E625. [PMID: 38477665 DOI: 10.1152/ajpendo.00294.2023] [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: 09/07/2023] [Revised: 03/07/2024] [Accepted: 03/11/2024] [Indexed: 03/14/2024]
Abstract
Metabolic-associated fatty liver disease (MAFLD) has been identified as risk factor of incident type 2 diabetes (T2D), but the underlying postprandial mechanisms remain unclear. We compared the glucose metabolism, insulin resistance, insulin secretion, and insulin clearance post-oral glucose tolerance test (OGTT) between individuals with and without MAFLD. We included 50 individuals with a body mass index (BMI) between 25 and 40 kg/m2 and ≥1 metabolic alteration: increased fasting triglycerides or insulin, plasma glucose 5.5-6.9 mmol/L, or glycated hemoglobin 5.7-5.9%. Participants were grouped according to MAFLD status, defined as hepatic fat fraction (HFF) ≥5% on MRI. We used oral minimal model on a frequently sampled 3 h 75 g-OGTT to estimate insulin sensitivity, insulin secretion, and pancreatic β-cell function. Fifty percent of participants had MAFLD. Median age (IQR) [57 (45-65) vs. 57 (44-63) yr] and sex (60% vs. 56% female) were comparable between groups. Post-OGTT glucose concentrations did not differ between groups, whereas post-OGTT insulin concentrations were higher in the MAFLD group (P < 0.03). Individuals with MAFLD exhibited lower insulin clearance, insulin sensitivity, and first-phase pancreatic β-cell function. In all individuals, increased insulin incremental area under the curve and decreased insulin clearance were associated with HFF after adjusting for age, sex, and BMI (P < 0.02). Among individuals with metabolic alterations, the presence of MAFLD was characterized mainly by post-OGTT hyperinsulinemia and reduced insulin clearance while exhibiting lower first phase β-cell function and insulin sensitivity. This suggests that MAFLD is linked with impaired insulin metabolism that may precede T2D.NEW & NOTEWORTHY Using an oral glucose tolerance test, we found hyperinsulinemia, lower insulin sensitivity, lower insulin clearance, and lower first-phase pancreatic β-cell function in individuals with MAFLD. This may explain part of the increased risk of incident type 2 diabetes in this population. These data also highlight implications of hyperinsulinemia and impaired insulin clearance in the progression of MAFLD to type 2 diabetes.
Collapse
Affiliation(s)
- Théo Gignac
- Axe Endocrinologie et Néphrologie, Centre de Recherche du CHU de Québec-Université Laval, Québec, Quebec, Canada
- Département de Médecine, Faculté de Médecine, Université Laval, Québec, Quebec, Canada
| | - Gabrielle Trépanier
- Axe Endocrinologie et Néphrologie, Centre de Recherche du CHU de Québec-Université Laval, Québec, Quebec, Canada
- Département de Médecine, Faculté de Médecine, Université Laval, Québec, Quebec, Canada
| | - Marion Pradeau
- Axe Endocrinologie et Néphrologie, Centre de Recherche du CHU de Québec-Université Laval, Québec, Quebec, Canada
- Département de Médecine, Faculté de Médecine, Université Laval, Québec, Quebec, Canada
| | - Arianne Morissette
- Département de Médecine, Faculté de Médecine, Université Laval, Québec, Quebec, Canada
- Centre Nutrition, santé et société (NUTRISS), Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Université Laval, Québec, Quebec, Canada
- Axe Obésité, Diabète de type 2 et Métabolisme, Centre de recherche de l'IUCPQ-Université Laval, Québec, Quebec, Canada
| | - Anne-Laure Agrinier
- Département de Médecine, Faculté de Médecine, Université Laval, Québec, Quebec, Canada
- Centre Nutrition, santé et société (NUTRISS), Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Université Laval, Québec, Quebec, Canada
- Axe Obésité, Diabète de type 2 et Métabolisme, Centre de recherche de l'IUCPQ-Université Laval, Québec, Quebec, Canada
| | - Éric Larose
- Département de Médecine, Faculté de Médecine, Université Laval, Québec, Quebec, Canada
- Axe Obésité, Diabète de type 2 et Métabolisme, Centre de recherche de l'IUCPQ-Université Laval, Québec, Quebec, Canada
| | - Julie Marois
- Centre Nutrition, santé et société (NUTRISS), Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Université Laval, Québec, Quebec, Canada
| | - Geneviève Pilon
- Centre Nutrition, santé et société (NUTRISS), Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Université Laval, Québec, Quebec, Canada
- Axe Obésité, Diabète de type 2 et Métabolisme, Centre de recherche de l'IUCPQ-Université Laval, Québec, Quebec, Canada
| | - Claudia Gagnon
- Axe Endocrinologie et Néphrologie, Centre de Recherche du CHU de Québec-Université Laval, Québec, Quebec, Canada
- Département de Médecine, Faculté de Médecine, Université Laval, Québec, Quebec, Canada
- Centre Nutrition, santé et société (NUTRISS), Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Université Laval, Québec, Quebec, Canada
- Axe Obésité, Diabète de type 2 et Métabolisme, Centre de recherche de l'IUCPQ-Université Laval, Québec, Quebec, Canada
| | - Marie-Claude Vohl
- Centre Nutrition, santé et société (NUTRISS), Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Université Laval, Québec, Quebec, Canada
- École de nutrition, Faculté des sciences de l'agriculture et de l'alimentation, Université Laval, Québec, Quebec, Canada
| | - André Marette
- Centre Nutrition, santé et société (NUTRISS), Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Université Laval, Québec, Quebec, Canada
- Axe Obésité, Diabète de type 2 et Métabolisme, Centre de recherche de l'IUCPQ-Université Laval, Québec, Quebec, Canada
| | - Anne-Marie Carreau
- Axe Endocrinologie et Néphrologie, Centre de Recherche du CHU de Québec-Université Laval, Québec, Quebec, Canada
- Département de Médecine, Faculté de Médecine, Université Laval, Québec, Quebec, Canada
| |
Collapse
|
47
|
Hervé J, Haurogné K, Allard M, Sourice S, Lindenbaum P, Bach JM, Lieubeau B. Spontaneous Akt2 deficiency in a colony of NOD mice exhibiting early diabetes. Sci Rep 2024; 14:9100. [PMID: 38643275 PMCID: PMC11032318 DOI: 10.1038/s41598-024-60021-w] [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: 02/02/2024] [Accepted: 04/17/2024] [Indexed: 04/22/2024] Open
Abstract
Diabetes constitutes a major public health problem, with dramatic consequences for patients. Both genetic and environmental factors were shown to contribute to the different forms of the disease. The monogenic forms, found both in humans and in animal models, specially help to decipher the role of key genes in the physiopathology of the disease. Here, we describe the phenotype of early diabetes in a colony of NOD mice, with spontaneous invalidation of Akt2, that we called HYP. The HYP mice were characterised by a strong and chronic hyperglycaemia, beginning around the age of one month, especially in male mice. The phenotype was not the consequence of the acceleration of the autoimmune response, inherent to the NOD background. Interestingly, in HYP mice, we observed hyperinsulinemia before hyperglycaemia occurred. We did not find any difference in the pancreas' architecture of the NOD and HYP mice (islets' size and staining for insulin and glucagon) but we detected a lower insulin content in the pancreas of HYP mice compared to NOD mice. These results give new insights about the role played by Akt2 in glucose homeostasis and argue for the ß cell failure being the primary event in the course of diabetes.
Collapse
|
48
|
Bian H, Shao X, Cai W, Fu H. Understanding the Reversible Binding of a Multichain Protein-Protein Complex through Free-Energy Calculations. J Phys Chem B 2024; 128:3598-3604. [PMID: 38574232 DOI: 10.1021/acs.jpcb.4c00519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
We demonstrate that the binding affinity of a multichain protein-protein complex, insulin dimer, can be accurately predicted using a streamlined route of standard binding free-energy calculations. We find that chains A and C, which do not interact directly during binding, stabilize the insulin monomer structures and reduce the binding affinity of the two monomers, therefore enabling their reversible association. Notably, we confirm that although classical methods can estimate the binding affinity of the insulin dimer, conventional molecular dynamics, enhanced sampling algorithms, and classical geometrical routes of binding free-energy calculations may not fully capture certain aspects of the role played by the noninteracting chains in the binding dynamics. Therefore, this study not only elucidates the role of noninteracting chains in the reversible binding of the insulin dimer but also offers a methodological guide for investigating the reversible binding of multichain protein-protein complexes utilizing streamlined free-energy calculations.
Collapse
Affiliation(s)
- Hengwei Bian
- Research Center for Analytical Sciences, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
| | - Xueguang Shao
- Research Center for Analytical Sciences, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
- School of Materials Science and Engineering, Smart Sensing Interdisciplinary Science Center, Nankai University, Tianjin 300350, China
| | - Wensheng Cai
- Research Center for Analytical Sciences, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
- School of Materials Science and Engineering, Smart Sensing Interdisciplinary Science Center, Nankai University, Tianjin 300350, China
| | - Haohao Fu
- Research Center for Analytical Sciences, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
- School of Materials Science and Engineering, Smart Sensing Interdisciplinary Science Center, Nankai University, Tianjin 300350, China
| |
Collapse
|
49
|
Sloan L, Cheng AYY, Escalada J, Haluzík M, Mauricio D. The role of basal insulins in the treatment of people with type 2 diabetes and chronic kidney disease: A narrative review. Diabetes Obes Metab 2024; 26:1157-1170. [PMID: 38225217 DOI: 10.1111/dom.15425] [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: 05/25/2023] [Revised: 12/05/2023] [Accepted: 12/05/2023] [Indexed: 01/17/2024]
Abstract
The majority of cases of chronic kidney disease (CKD) worldwide are driven by the presence of type 2 diabetes (T2D), resulting in an increase in CKD rates over the past few decades. The existence of CKD alongside diabetes is associated with increased burden of cardiovascular disease and increased risk of death. Optimal glycaemic control is essential to prevent progression of CKD, but achieving glycaemic targets in people with CKD and diabetes can be challenging because of increased risk of hypoglycaemia and limitations on glucose-lowering therapeutic options. This review considers the challenges in management of T2D in people with impaired kidney function and assesses evidence for use of basal insulin analogues in people with CKD.
Collapse
Affiliation(s)
- Lance Sloan
- Texas Institute for Kidney and Endocrine Disorders, Lufkin, Texas, USA
- University of Texas Medical Branch, Galveston, Texas, USA
| | - Alice Y Y Cheng
- Trillium Health Partners & Unity Health, University of Toronto, Toronto, Ontario, Canada
| | - Javier Escalada
- Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
- Center for Biomedical Research in Physiopathology of Obesity and Nutrition (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Martin Haluzík
- Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Didac Mauricio
- Center for Biomedical Research on Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Barcelona, Spain
- Department of Endocrinology and Nutrition, CIBERDEM, Hospital de la Santa Creu i Sant Pau & Sant Pau Biomedical Research Institute (IIB Sant Pau), Barcelona, Spain
- Faculty of Medicine, University of Vic (UVIC/UCC), Vic, Spain
| |
Collapse
|
50
|
Glatz JFC, Heather LC, Luiken JJFP. CD36 as a gatekeeper of myocardial lipid metabolism and therapeutic target for metabolic disease. Physiol Rev 2024; 104:727-764. [PMID: 37882731 DOI: 10.1152/physrev.00011.2023] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 10/02/2023] [Accepted: 10/22/2023] [Indexed: 10/27/2023] Open
Abstract
The multifunctional membrane glycoprotein CD36 is expressed in different types of cells and plays a key regulatory role in cellular lipid metabolism, especially in cardiac muscle. CD36 facilitates the cellular uptake of long-chain fatty acids, mediates lipid signaling, and regulates storage and oxidation of lipids in various tissues with active lipid metabolism. CD36 deficiency leads to marked impairments in peripheral lipid metabolism, which consequently impact on the cellular utilization of multiple different fuels because of the integrated nature of metabolism. The functional presence of CD36 at the plasma membrane is regulated by its reversible subcellular recycling from and to endosomes and is under the control of mechanical, hormonal, and nutritional factors. Aberrations in this dynamic role of CD36 are causally associated with various metabolic diseases, in particular insulin resistance, diabetic cardiomyopathy, and cardiac hypertrophy. Recent research in cardiac muscle has disclosed the endosomal proton pump vacuolar-type H+-ATPase (v-ATPase) as a key enzyme regulating subcellular CD36 recycling and being the site of interaction between various substrates to determine cellular substrate preference. In addition, evidence is accumulating that interventions targeting CD36 directly or modulating its subcellular recycling are effective for the treatment of metabolic diseases. In conclusion, subcellular CD36 localization is the major adaptive regulator of cellular uptake and metabolism of long-chain fatty acids and appears a suitable target for metabolic modulation therapy to mend failing hearts.
Collapse
Affiliation(s)
- Jan F C Glatz
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Lisa C Heather
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, United Kingdom
| | - Joost J F P Luiken
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|