1
|
Thakur MR, Tupe RS. l-Arginine: A multifaceted regulator of diabetic cardiomyopathy. Biochem Biophys Res Commun 2025; 761:151720. [PMID: 40186920 DOI: 10.1016/j.bbrc.2025.151720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025]
Abstract
In diabetes mellitus, dysregulated glucose and lipid metabolism lead to diabetic cardiomyopathy (DCM) by imparting pathological myocardial remodeling and cellular injury. Accelerated glycation, oxidative stress, and activated inflammatory pathways culminate in cardiac fibrosis and hypertrophy in DCM. The regulatory effects of l-Arginine (L-Arg) have been elucidated in the pathological changes of DCM, including myocardial fibrosis, hypertrophy, and apoptosis, by inhibiting glycation and oxidative stress-induced inflammation. Disturbed L-Arg metabolism and decreased intracellular L-Arg pool are correlated with the progression of DCM; therefore, L-Arg supplementation has been prescribed for various cardiovascular dysfunctions. This review expands the therapeutic potential of L-Arg supplementation in DCM by elucidating its molecular mechanism of action and exploring potential clinical outcomes.
Collapse
Affiliation(s)
- Muskan R Thakur
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune, 412115, Maharashtra, India
| | - Rashmi S Tupe
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune, 412115, Maharashtra, India.
| |
Collapse
|
2
|
Dimeji IY, Abass KS, Audu NM, Ayodeji AS. L-Arginine and immune modulation: A pharmacological perspective on inflammation and autoimmune disorders. Eur J Pharmacol 2025; 997:177615. [PMID: 40216179 DOI: 10.1016/j.ejphar.2025.177615] [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: 02/23/2025] [Revised: 04/01/2025] [Accepted: 04/08/2025] [Indexed: 04/22/2025]
Abstract
L- Arginine (2-Amino-5-guanidinovaleric acid, L-Arg) is a semi-essential amino acid that is mainly produced within the urea cycle. It acts as a key precursor in the synthesis of proteins, urea, creatine, prolamines (including putrescine, spermine, and spermidine), proline, and nitric oxide (NO). WhenL-Arg is metabolized, it produces NO, glutamate, and prolamines, which all play important regulatory roles in various physiological functions. In addition to its metabolic roles,L-Arg significantly influences immune responses, especially in the context of inflammation and autoimmune diseases. It affects the activity of immune cells by modulating T-cell function, the polarization of macrophages, and the release of cytokines. Importantly,L-Arg plays a dual role in immune regulation, functioning as both an immunostimulatory and immunosuppressive agent depending on the specific cellular and biochemical environments. This review examines the immunopharmacological mechanisms of L-Arg, emphasizing its involvement in inflammatory responses and its potential therapeutic uses in autoimmune conditions like rheumatoid arthritis, multiple sclerosis, and inflammatory bowel disease. By influencing the pathways of nitric oxide synthase (NOS) and arginase (ARG), L-Arg helps maintain immune balance and contributes to the pathophysiology of diseases. Gaining a better understanding of the pharmacological effects of L-Arg on immune regulation could yield new perspectives on targeted treatments for immune-related diseases. Exploring its impact on immune signaling and metabolic pathways may result in novel therapeutic approaches for chronic inflammatory and autoimmune disorders.
Collapse
Affiliation(s)
- Igbayilola Yusuff Dimeji
- Department of Human Physiology, College of Medicine and Health Sciences, Baze University, Nigeria.
| | - Kasim Sakran Abass
- Department of Physiology, Biochemistry, and Pharmacology, College of Veterinary Medicine, University of Kirkuk, Kirkuk 36001, Iraq
| | - Ngabea Murtala Audu
- Department of Medicine Maitama District Hospital/ College of Medicine Baze University, Abuja, Nigeria
| | - Adekola Saheed Ayodeji
- Department of Chemical Pathology, Medical Laboratory Science Program, Faculty of Nursing and Allied Health Sciences, University of Abuja, Abuja, Nigeria.
| |
Collapse
|
3
|
Ali U, Makhdoom SI, Javed MU, Khan RA, Naveed M, Abbasi BH, Aziz T, Alshehri F, Al-Asmari F, Al-Joufi FA, Alwethaynani MS. Fenugreek seeds as a natural source of L-arginine-encapsulated lipid nanoparticles against diabetes. Sci Rep 2025; 15:7016. [PMID: 40016285 PMCID: PMC11868517 DOI: 10.1038/s41598-025-90675-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: 10/14/2024] [Accepted: 02/14/2025] [Indexed: 03/01/2025] Open
Abstract
Diabetes, affecting over 10.5% of the global population, leads to severe health complications and economic burdens, highlighting the urgent need for effective therapeutic approaches. Current treatments are often insufficient, prompting the exploration of novel therapeutic agents and delivery mechanisms. This study addresses this gap by investigating the roles of L-arginine (identified as a target drug candidate through network pharmacology) in diabetes management, while also evaluating lipid nanocarriers synthesized from fenugreek seed oil for improved drug delivery. Our docking analyses revealed L-arginine's strong interactions with diabetes-target genes (CYP1A2, CYP2C19, and NFKB), with multiple hydrogen bonds and binding energies ranging from - 7.2 to - 8.9 kcal/mol. Encapsulated L-arginine lipid nanoparticles were characterized using UV-Visible spectroscopy, showing absorbance peaks at 415 nm for simple nanoparticles and 521 nm for L-arginine-loaded nanoparticles. Scanning electron microscopy confirmed an average nanoparticle size of 100.2 nm, and zeta potential analysis indicated a neutral surface charge (- 9.37 mV). Antioxidative activity showed 84.44% inhibition with an IC50 value of 40.5 µg/mL The nanoparticles inhibited albumin denaturation by 81.10% and alpha-amylase by 89.30%, surpassing metformin (78.43% at 1000 µg/mL). Hemolysis percentage was minimal at 10.54%. These findings demonstrate the potential of L-arginine as an anti-diabetic agent and highlight the efficacy of lipid nanocarriers as innovative drug delivery systems, providing a foundation for advancing therapeutic interventions against diabetes.
Collapse
Affiliation(s)
- Urooj Ali
- Department of Biotechnology, Quaid-I-Azam University, Islamabad, 45320, Pakistan
- Faculty of Life and Environmental Sciences, School of Science, The University of Sydney, Camperdown, Sydney, NSW, 2050, Australia
| | - Syeda Izma Makhdoom
- Department of Biotechnology, Faculty of Science and Technology, University of Central Punjab, Lahore, 54590, Pakistan
| | - Muhammad Uzair Javed
- Department of Biotechnology, Quaid-I-Azam University, Islamabad, 45320, Pakistan
| | - Rafia Ali Khan
- Faculty of Pharmaceutical Sciences, University of Central Punjab, Lahore, 54590, Pakistan
| | - Muhammad Naveed
- Department of Biotechnology, Faculty of Science and Technology, University of Central Punjab, Lahore, 54590, Pakistan
| | - Bilal Haider Abbasi
- Department of Biotechnology, Quaid-I-Azam University, Islamabad, 45320, Pakistan.
- EA2106 Biomolécules et Biotechnologies Végétales, Université de Tours, Tours, 37000, France.
| | - Tariq Aziz
- Laboratory of Animal Health Food Hygiene and Quality, University of Ioannina, Arta, Greece.
| | - Fatma Alshehri
- Department of Biology, College of Sciences, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh, 11671, Saudi Arabia
| | - Fahad Al-Asmari
- Department of Food and Nutrition Sciences, College of Agricultural and Food Sciences, King Faisal University, Al Ahsa, 31982, Saudi Arabia
| | - Fakhria A Al-Joufi
- Department of Pharmacology, College of Pharmacy, Jouf University, Aljouf, 72341, Saudi Arabia
| | - Maher S Alwethaynani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Alquwayiyah, Riyadh, Saudi Arabia
| |
Collapse
|
4
|
Zhu Y, Yang P, Ren T, Chen Z, Tian H, Wang M, Zhang C. Integrated metabolomics and transcriptomics reveal the potential of hydroxy-alpha-sanshool in alleviating insulin resistance. Mol Med 2025; 31:76. [PMID: 39984861 PMCID: PMC11846303 DOI: 10.1186/s10020-025-01129-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 02/12/2025] [Indexed: 02/23/2025] Open
Abstract
Hydroxy-alpha-sanshool (HAS) has attracted attention because of its various biological activities, such as hypoglycemic, hypolipidemic, and antioxidant activities. In this study, we investigated the effects of HAS on insulin resistance (IR) and its mechanism. HAS reduced fasting blood glucose (FBG), promoted insulin (INS) secretion, significantly decreased levels of interleukin (IL)-1, IL-6, tumor necrosis factor (TNF)-α and monocyte chemoattractant protein-1 (MCP-1), and increased the IL-2 level in serum of IR model mice. HAS regulated the mRNA levels of protein kinase B (Akt), B-cell lymphoma extra-large (Bcl-xL), stearoyl-CoA desaturase-1 (SCD1), nuclear factor kappa B (NF-κB), and eukaryotic translation initiation factor 4E (eIF4E). Additionally, differentially abundant metabolites in IR model mice treated with HAS were involved in these signaling pathways including prion disease, choline metabolism in cancer, regulation of lipolysis in adipocytes and the pentose phosphate pathway and positively regulated betaine abundance. In conclusion, HAS activated the phosphatidylinositol-3 kinase (PI3K)/Akt insulin and NF-κB signaling pathways to maintain glucose homeostasis and regulate IR.
Collapse
Affiliation(s)
- Yuping Zhu
- School of Basic Medicine, Guizhou Medical University, Guiyang, 550025, China
| | - Pan Yang
- School of Basic Medicine, Guizhou Medical University, Guiyang, 550025, China
| | - Tingyuan Ren
- College of Brewing and Food Engineering, Guizhou University, Guiyang, 550025, China
| | - Zhuqi Chen
- School of Clinical Medicine, Guizhou Medical University, Guiyang, 550025, China
| | - Huanhuan Tian
- School of Clinical Medicine, Guizhou Medical University, Guiyang, 550025, China
| | - Mingfen Wang
- School of Clinical Medicine, Guizhou Medical University, Guiyang, 550025, China
| | - Chunlin Zhang
- School of Basic Medicine, Guizhou Medical University, Guiyang, 550025, China.
| |
Collapse
|
5
|
Mansouri RA, Aboubakr EM, Alshaibi HF, Ahmed AM. L-arginine administration exacerbates myocardial injury in diabetics via prooxidant and proinflammatory mechanisms along with myocardial structural disruption. World J Diabetes 2025; 16:100395. [PMID: 39959273 PMCID: PMC11718468 DOI: 10.4239/wjd.v16.i2.100395] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/02/2024] [Accepted: 11/25/2024] [Indexed: 12/30/2024] Open
Abstract
BACKGROUND L-arginine (L-Arg) is one of the most widely used amino acids in dietary and pharmacological products. However, the evidence on its usefulness and dose limitations, especially in diabetics is still controversial. AIM To investigate the effects of chronic administration of different doses of L-Arg on the cardiac muscle of type 2 diabetic rats. METHODS Of 96 male rats were divided into 8 groups as follows (n = 12): Control, 0.5 g/kg L-Arg, 1 g/kg L-Arg, 1.5 g/kg L-Arg, diabetic, diabetic + 0.5 g/kg L-Arg, diabetic + 1 g/kg L-Arg, and diabetic + 1.5 g/kg L-Arg; whereas L-Arg was orally administered for 3 months to all treated groups. RESULTS L-Arg produced a moderate upregulation of blood glucose levels to normal rats, but when given to diabetics a significant upregulation was observed, associated with increased nitric oxide, inflammatory cytokines, and malonaldehyde levels in diabetic rats treated with 1 g/kg L-Arg and 1.5 g/kg L-Arg. A substantial decrease in the antioxidant capacity, superoxide dismutase, catalase, glutathione peroxidase, reduced glutathione concentrations, and Nrf-2 tissue depletion were observed at 1 g/kg and 1.5 g/kg L-Arg diabetic treated groups, associated with myocardial injury, fibrosis, α-smooth muscle actin upregulation, and disruption of desmin cardiac myofilaments, and these effects were not noticeable at normal treated groups. On the other hand, L-Arg could significantly improve the lipid profile of diabetic rats and decrease their body weights. CONCLUSION L-Arg dose of 1 g/kg or more can exacerbates the diabetes injurious effects on the myocardium, while 0.5 g/kg dose can improve the lipid profile and decrease the body weight.
Collapse
Affiliation(s)
- Rasha A Mansouri
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 22254, Jeddah, Saudi Arabia
- College of Science and Humanities in Al-Kharj, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Esam M Aboubakr
- Department of Pharmacology and Toxicology, Faculty of Pharmacy-South Valley University, Qena 83523, Egypt
| | - Huda F Alshaibi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 22254, Jeddah, Saudi Arabia
- Stem Cell Unit, King Fahad Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Adel M Ahmed
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, South Valley University, Qena 83523, Egypt
| |
Collapse
|
6
|
Primavera R, Wang J, Buchwald P, Ganguly A, Patel S, Bettencourt L, Chetty S, Yarani R, Regmi S, Levitte S, Kevadiya B, Guindani M, Decuzzi P, Thakor AS. Controlled Nutrient Delivery to Pancreatic Islets Using Polydopamine-Coated Mesoporous Silica Nanoparticles. NANO LETTERS 2025; 25:939-950. [PMID: 39791700 DOI: 10.1021/acs.nanolett.4c03613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
In this study, we designed a nanoscale platform for sustained amino acid delivery to support transplanted pancreatic islets. The platform features mesoporous silica nanoparticles (MSNPs) loaded with glutamine (G), an essential amino acid required for islet survival and function, and coated with polydopamine (PD). We investigated various PD concentrations (0.5-2 mg/mL) and incubation times (0.5-2 h) to optimize G release, identifying that a PD concentration of 0.5 mg/mL incubated for 0.5 h yielded the best results to support islet viability and functionality ex vivo, particularly under inflammatory conditions. In syngeneic islet transplantation in STZ-diabetic mice, G alone provided only temporary benefits; however, PD-G-MSNPs significantly improved islet engraftment and function, with animals maintaining glycemic control for 30 days due to controlled G release. Our findings support the use of this nanoscale platform to provide essential nutrients like G to transplanted islets until they can establish their own blood and nutrient supply.
Collapse
Affiliation(s)
- Rosita Primavera
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Jing Wang
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Peter Buchwald
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
| | - Abantika Ganguly
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Shaini Patel
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Lili Bettencourt
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Shashank Chetty
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Reza Yarani
- Translational Type 1 Diabetes Research, Department of Clinical, Research, Steno Diabetes Center Copenhagen, Herlev 2730, Denmark
| | - Shobha Regmi
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Steven Levitte
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Bhavesh Kevadiya
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Michele Guindani
- Department of Biostatistics, Jonathan and Karin Fielding School of Public Health, University of California Los Angeles, Los Angeles, California 90095, United States
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, Genoa 16163, Italy
| | - Avnesh S Thakor
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| |
Collapse
|
7
|
Caterino M, Costanzo M, Castaldo A, Iacotucci P, Carnovale V, Ruoppolo M, Gelzo M, Castaldo G. Metabolomic profiling of saliva from cystic fibrosis patients. Sci Rep 2025; 15:479. [PMID: 39747338 PMCID: PMC11696459 DOI: 10.1038/s41598-024-84191-9] [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: 09/12/2024] [Accepted: 12/20/2024] [Indexed: 01/04/2025] Open
Abstract
The development of targeted therapies that correct the effect of mutations in patients with cystic fibrosis (CF) and the relevant heterogeneity of the clinical expression of the disease require biomarkers correlated to the severity of the disease useful for monitoring the therapeutic effects. We applied a targeted metabolomic approach by LC-MS/MS on saliva samples from 70 adult CF patients and 63 age/sex-matched controls to investigate alterations in metabolic pathways related to pancreatic insufficiency (PI), Pseudomonas aeruginosa (PA) colonization, CF liver disease (CFLD), and CF related diabetes (CFRD). Sixty salivary metabolites were differentially expressed, with 11 being less abundant and 49 more abundant in CF patients. Among these, the most relevant alterations involved salivary ADMA, N-acetylornithine, methionine and methionine sulfoxide levels. Furthermore, methionine was significantly lower in CF patients with PI and salivary histamine levels were significantly lower in patients colonized by PA. Moreover, ADMA as well as N-acetylornithine and methionine were significantly lower in CF patients with CFRD than in patients without CFRD. Finally, the levels of DOPA resulted significantly lower in saliva from patients with liver disease. Our study revealed an imbalance in arginine methylation and tryptophan pathway related to CFRD and PI as well as alterations in dopaminergic pathway and Krebs cycle related to CFLD. This study also highlights different salivary metabolites as new potential biomarkers in a non-invasive sample that could represent a useful tool for the stratification and management of CF patients.
Collapse
Affiliation(s)
- M Caterino
- CEINGE-Biotecnologie avanzate Franco Salvatore, Via G. Salvatore 486, Naples, 80145, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy
| | - M Costanzo
- CEINGE-Biotecnologie avanzate Franco Salvatore, Via G. Salvatore 486, Naples, 80145, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy
| | - A Castaldo
- SC di Pneumologia e UTSIR, AORN Santobono-Pausilipon, Naples, Italy
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli Federico II, Naples, Italy
| | - P Iacotucci
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli Federico II, Naples, Italy
| | - V Carnovale
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli Federico II, Naples, Italy
| | - M Ruoppolo
- CEINGE-Biotecnologie avanzate Franco Salvatore, Via G. Salvatore 486, Naples, 80145, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy
| | - M Gelzo
- CEINGE-Biotecnologie avanzate Franco Salvatore, Via G. Salvatore 486, Naples, 80145, Italy.
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy.
| | - G Castaldo
- CEINGE-Biotecnologie avanzate Franco Salvatore, Via G. Salvatore 486, Naples, 80145, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy
| |
Collapse
|
8
|
Fiordelisi A, Cerasuolo FA, Avvisato R, Buonaiuto A, Maisto M, Bianco A, D'Argenio V, Mone P, Perrino C, D'Apice S, Paolillo R, Pezone A, Varzideh F, Santulli G, Sorriento D, Iaccarino G, Gambardella J. L-Arginine supplementation as mitochondrial therapy in diabetic cardiomyopathy. Cardiovasc Diabetol 2024; 23:450. [PMID: 39707340 PMCID: PMC11662564 DOI: 10.1186/s12933-024-02490-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 10/23/2024] [Indexed: 12/23/2024] Open
Abstract
In patients with type II diabetes, the development of diabetic cardiomyopathy (DC) is associated with a high risk of mortality. Left ventricular hypertrophy, diastolic dysfunction, and exercise intolerance are the first signs of DC. The underlying mechanisms are not fully elucidated, and there is an urgent need for specific biomarkers and molecular targets for early diagnosis and treatment. Mitochondrial alterations play a key role in the development of DC, and microRNAs regulating mitochondrial function are emerging as potential biomarkers of metabolic stress in DC. L-Arginine (Arg) supplementation has been shown to be an effective strategy for improving mitochondrial function and energetics, with a significant impact on physical performance. The aim of the current study was to evaluate the effects of Arg supplementation on cardiac mitochondrial function, DC development, and relative phenotypes including exercise intolerance. We used db/db mice as a model of type II diabetes, chronically treated with Arg (1 mg/kg/day) for 12 weeks. Arg-treated db/db mice showed preserved diastolic function and left ventricular morphology compared with untreated diabetic mice. Arg supplementation also improved exercise tolerance and the propensity to physical activity. Mitochondrial respiration was significantly increased in cardiomyocytes isolated from treated db/db mice, as well as in diabetic cardiomyocytes treated with Arg in vitro. The improvement of cardiac mitochondrial function in db/db + Arg mice was associated with an increase in PGC-1-alpha levels, mitochondrial biogenesis, recycling, and antioxidant capacity. Arg treatment prevented the accumulation of circulating and cardiac miR-143 in db/db mice, which is an index of metabolic stress and activation of mitochondrial damage mechanisms. In conclusion, Arg supplementation is effective in preventing the development of DC, preserving diastolic function and exercise tolerance by improving mitochondrial fitness and homeostasis. Additionally, miR-143 could potentially be employed to monitor cardiac metabolic stress and the effects of Arg treatment in diabetes.
Collapse
MESH Headings
- Animals
- Diabetic Cardiomyopathies/metabolism
- Diabetic Cardiomyopathies/physiopathology
- Diabetic Cardiomyopathies/etiology
- Diabetic Cardiomyopathies/prevention & control
- Arginine/metabolism
- Dietary Supplements
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/drug effects
- Mitochondria, Heart/pathology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Male
- Ventricular Function, Left/drug effects
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/diagnosis
- Diabetes Mellitus, Type 2/metabolism
- Exercise Tolerance/drug effects
- Disease Models, Animal
- Mice, Inbred C57BL
- Energy Metabolism/drug effects
- MicroRNAs/metabolism
- MicroRNAs/genetics
- Mice
- Cells, Cultured
Collapse
Affiliation(s)
- Antonella Fiordelisi
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Naples, Italy
| | - Federica Andrea Cerasuolo
- Department of Statistics, Computer Science, Applications (DiSIA), University of Florence, Florence, Italy
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Roberta Avvisato
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Antonietta Buonaiuto
- Department of Statistics, Computer Science, Applications (DiSIA), University of Florence, Florence, Italy
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | | | - Antonio Bianco
- Department of Public Health, Federico II University, Naples, Italy
| | - Valeria D'Argenio
- Department of Human Sciences and Quality of Life Promotion, San Raffaele Open University, Rome, Italy
- CEINGE- Advanced Biotechnologies, Naples, Italy
| | - Pasquale Mone
- Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, Campobasso, Italy
- Casa di Cura Privata Montevergine, Mercogliano, Italy
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York City, NY, USA
| | - Cinzia Perrino
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Stefania D'Apice
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Roberta Paolillo
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Antonio Pezone
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York City, NY, USA
| | - Fahimeh Varzideh
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York City, NY, USA
- Department of Biology, Federico II University, Naples, Italy
| | - Gaetano Santulli
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York City, NY, USA
- Department of Molecular Pharmacology, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Neuroimmunology and Inflammation (INI), Fleischer Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, New York City, NY, USA
- International Translational Research and Medical Education (ITME) Consortium, Academic Research Unit, Naples, Italy
- Interdepartmental Center of Research on Hypertension and Related Conditions (CIRIAPA), Federico II University, Naples, Italy
| | - Daniela Sorriento
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
- Interdepartmental Center of Research on Hypertension and Related Conditions (CIRIAPA), Federico II University, Naples, Italy
| | - Guido Iaccarino
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
- Interdepartmental Center of Research on Hypertension and Related Conditions (CIRIAPA), Federico II University, Naples, Italy
| | - Jessica Gambardella
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy.
- International Translational Research and Medical Education (ITME) Consortium, Academic Research Unit, Naples, Italy.
- Interdepartmental Center of Research on Hypertension and Related Conditions (CIRIAPA), Federico II University, Naples, Italy.
| |
Collapse
|
9
|
Naik AR, Save SN, Sahoo SS, Yadav SS, Kumar A, Chugh J, Sharma S. Metabolic perturbations associated with hIAPP-induced insulin resistance in skeletal muscles: Implications to the development of type 2 diabetes. Int J Biochem Cell Biol 2024; 176:106665. [PMID: 39322038 DOI: 10.1016/j.biocel.2024.106665] [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: 04/23/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
The human islet amyloid polypeptide (hIAPP) tends to misfold and self-assemble to form amyloid fibrils, which has been associated with the loss of function and viability of pancreatic β-cells in type 2 diabetes mellitus (T2DM). The role of hIAPP in the development of insulin resistance (a hallmark of T2DM) in skeletal muscles - the major sites for glucose utilization - needs further investigation. Even though, insulin-resistant conditions have been known to stimulate hIAPP aggregation, the events that lead to the development of insulin resistance due to hIAPP aggregation in skeletal muscles remain unidentified. Here, we have attempted to identify metabolic perturbations in L6 myotubes that were exposed to increasing concentrations of recombinant hIAPP for different time durations. It was observed that hIAPP exposure was associated with increased mitochondrial and cellular ROS levels, loss in mitochondrial membrane potential and viability of the myotubes. Metabolomic investigations of hIAPP-treated myotubes revealed significant perturbations in o-phosphocholine, sn-glycero-3-phosphocholine and dimethylamine levels (p < 0.05). Therefore, we anticipate that defects in glycerophospholipid metabolism and the associated oxidative stress and membrane damage may play key roles in the development of insulin resistance due to protein misfolding in skeletal muscles. In summary, the perturbed metabolites and their pathways have not only the potential to be used as early biomarkers to predict the onset of insulin resistance and T2DM but also as therapeutic targets for the effective management of the same.
Collapse
Affiliation(s)
- Arya R Naik
- Department of Biotechnology, Savitribai Phule Pune University, Ganeshkhind Road, Pune 411007, India
| | - Shreyada N Save
- Department of Biotechnology, Savitribai Phule Pune University, Ganeshkhind Road, Pune 411007, India
| | - Soumya S Sahoo
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Dr. Homi Bhabha Road, Pashan, Pune 411008, India
| | - Saurabh S Yadav
- Department of Biotechnology, Savitribai Phule Pune University, Ganeshkhind Road, Pune 411007, India
| | - Ashutosh Kumar
- Department of Biosciences and Bioengineering, Indian institute of technology Bombay, Powai, Mumbai, Maharashtra 400076, India
| | - Jeetender Chugh
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Dr. Homi Bhabha Road, Pashan, Pune 411008, India
| | - Shilpy Sharma
- Department of Biotechnology, Savitribai Phule Pune University, Ganeshkhind Road, Pune 411007, India.
| |
Collapse
|
10
|
Wang YX, Pi JC, Yao YF, Peng XP, Li WJ, Xie MY. Hypoglycemic effects of white hyacinth bean polysaccharide on type 2 diabetes mellitus rats involvement with entero-insular axis and GLP-1 via metabolomics study. Int J Biol Macromol 2024; 281:136489. [PMID: 39393741 DOI: 10.1016/j.ijbiomac.2024.136489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 09/02/2024] [Accepted: 10/08/2024] [Indexed: 10/13/2024]
Abstract
The present study aimed to investigate the potential effects of white hyacinth bean polysaccharide (WHBP) against type 2 diabetic mellitus (T2DM) which was established by high-glucose/high-fat for 8 weeks, combined with a low-dose streptozotocin (STZ) injection. Our results showed that WHBP behaved the hypoglycemic effect by attenuating fasting blood glucose in vivo. WHBP-mediated anti-diabetic effects associated with the attenuation of insulin resistance and pancreatic impairment, as evidenced by the mitigation of pathological changes, inflammatory response and oxidative stress in the pancreas of T2DM rats. Meanwhile, gut protection was also shown during WHBP-mediated anti-diabetic effects, and glucagon-like peptide-1 (GLP-1), a mediator of the entero-insular axis, was observed to be elevated in both gut and pancreas of WHBP groups when compared to DM group, suggesting that hypoglycemic effects of WHBP were implicated in gut-pancreas interaction. Subsequently, untargeted metabolomics analysis performed by UPLC-QTOF/MS and showed that WHBP administration significantly adjusted the levels of 40 metabolites when compared to DM group. Further data concerning pathway analysis showed that WHBP administration significantly regulated the phenylalanine metabolism, tryptophan metabolism, arginine and proline, isoleucine metabolism, and glycerophospholipid metabolism in T2DM rats. Together, our results suggested that WHBP performed hypoglycemic effects and pancreatic protection linked to entero-insular axis involvement with GLP-1 and reversed metabolic disturbances in T2DM rats.
Collapse
Affiliation(s)
- Yi-Xuan Wang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Jin-Chan Pi
- College of Future Technology, Nanchang University, Nanchang 330031, China
| | - Yu-Fei Yao
- Department of Critical Care Medicine, The Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang 330006, China
| | - Xiao-Ping Peng
- Department of Cardiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Wen-Juan Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China.
| | - Ming-Yong Xie
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| |
Collapse
|
11
|
Sheethal G, Verma A, Mall R, Parsa KV, Tokala RK, Bynigeri R, Pondugala PK, Vemula K, Sai Latha S, Sowpati DT, Singh SS, Rao GV, Talukdar R, Kanneganti TD, Reddy DN, Sasikala M. Modulation of Nuclear Receptor 4A1 Expression Improves Insulin Secretion in a Mouse Model of Chronic Pancreatitis. Pancreas 2024; 53:e760-e773. [PMID: 38710022 DOI: 10.1097/mpa.0000000000002370] [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] [Indexed: 05/08/2024]
Abstract
OBJECTIVES Diabetes secondary to chronic pancreatitis (CP) presents clinical challenges due to lack of understanding on factor(s) triggering insulin secretory defects. Therefore, we aimed to delineate the molecular mechanism of β-cell dysfunction in CP. MATERIALS AND METHODS Transcriptomic analysis was conducted to identify endocrine-specific receptor expression in mice and human CP on microarray. The identified receptor (NR4A1) was overexpressed in MIN6 cells using PEI linear transfection. RNA-Seq analysis of NR4A1-overexpressed (OE) MIN6 cells on NovaSeq6000 identified aberrant metabolic pathways. Upstream trigger for NR4A1OE was studied by InBio Discover and cytokine exposure, whereas downstream effect was examined by Fura2 AM-based fluorimetric and imaging studies. Mice with CP were treated with IFN-γ-neutralizing monoclonal antibodies to assess NR4A1 expression and insulin secretion. RESULTS Increased expression of NR4A1 associated with decreased insulin secretion in islets (humans: controls 9 ± 0.2, CP 3.7 ± 0.2, mice: controls 8.5 ± 0.2, CP 2.1 ± 0.1 μg/L). NR4A1OE in MIN6 cells (13.2 ± 0.1) showed reduction in insulin secretion (13 ± 5 to 0.2 ± 0.1 μg/mg protein per minute, P = 0.001) and downregulation of calcium and cAMP signaling pathways. IFN-γ was identified as upstream signal for NR4A1OE in MIN6. Mice treated with IFN-γ-neutralizing antibodies showed decreased NR4A1 expression 3.4 ± 0.11-fold ( P = 0.03), showed improved insulin secretion (4.4 ± 0.2-fold, P = 0.01), and associated with increased Ca 2+ levels (2.39 ± 0.06-fold, P = 0.009). CONCLUSIONS Modulating NR4A1 expression can be a promising therapeutic strategy to improve insulin secretion in CP.
Collapse
Affiliation(s)
| | - Archana Verma
- CSIR-CCMB, Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Raghvendra Mall
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN
| | - Kishore Vl Parsa
- Centre for Innovation in Molecular and Pharmaceutical Sciences, Dr Reddy's Institute of Life Sciences
| | | | - Ratnakar Bynigeri
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN
| | | | | | - S Sai Latha
- Centre for Innovation in Molecular and Pharmaceutical Sciences, Dr Reddy's Institute of Life Sciences
| | - Divya Tej Sowpati
- CSIR-CCMB, Centre for Cellular and Molecular Biology, Hyderabad, India
| | | | - G V Rao
- Asian Institute of Gastroenterology (AIG Hospitals), Hyderabad, India
| | - Rupjyoti Talukdar
- Asian Institute of Gastroenterology (AIG Hospitals), Hyderabad, India
| | | | - D Nageshwar Reddy
- Asian Institute of Gastroenterology (AIG Hospitals), Hyderabad, India
| | | |
Collapse
|
12
|
Barciszewska AM, Belter A, Barciszewski JF, Gawrońska I, Giel-Pietraszuk M, Naskręt-Barciszewska MZ. Mechanistic Insights on Metformin and Arginine Implementation as Repurposed Drugs in Glioblastoma Treatment. Int J Mol Sci 2024; 25:9460. [PMID: 39273414 PMCID: PMC11394688 DOI: 10.3390/ijms25179460] [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: 07/19/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/15/2024] Open
Abstract
As the most common and aggressive primary malignant brain tumor, glioblastoma is still lacking a satisfactory curative approach. The standard management consisting of gross total resection followed by radiotherapy and chemotherapy with temozolomide only prolongs patients' life moderately. In recent years, many therapeutics have failed to give a breakthrough in GBM treatment. In the search for new treatment solutions, we became interested in the repurposing of existing medicines, which have established safety profiles. We focused on the possible implementation of well-known drugs, metformin, and arginine. Metformin is widely used in diabetes treatment, but arginine is mainly a cardiovascular protective drug. We evaluated the effects of metformin and arginine on total DNA methylation, as well as the oxidative stress evoked by treatment with those agents. In glioblastoma cell lines, a decrease in 5-methylcytosine contents was observed with increasing drug concentration. When combined with temozolomide, both guanidines parallelly increased DNA methylation and decreased 8-oxo-deoxyguanosine contents. These effects can be explained by specific interactions of the guanidine group with m5CpG dinucleotide. We showed that metformin and arginine act on the epigenetic level, influencing the foreground and potent DNA regulatory mechanisms. Therefore, they can be used separately or in combination with temozolomide, in various stages of disease, depending on desired treatment effects.
Collapse
Affiliation(s)
- Anna-Maria Barciszewska
- Intraoperative Imaging Unit, Chair and Department of Neurosurgery and Neurotraumatology, Karol Marcinkowski University of Medical Sciences, Przybyszewskiego 49, 60-355 Poznan, Poland
- Department of Neurosurgery and Neurotraumatology, University Clinical Hospital, Przybyszewskiego 49, 60-355 Poznan, Poland
| | - Agnieszka Belter
- Institute of Bioorganic Chemistry of the Polish Academy of Sciences, Noskowskiego 12, 61-704 Poznan, Poland
| | - Jakub F Barciszewski
- Institute of Bioorganic Chemistry of the Polish Academy of Sciences, Noskowskiego 12, 61-704 Poznan, Poland
| | - Iwona Gawrońska
- Institute of Bioorganic Chemistry of the Polish Academy of Sciences, Noskowskiego 12, 61-704 Poznan, Poland
| | - Małgorzata Giel-Pietraszuk
- Institute of Bioorganic Chemistry of the Polish Academy of Sciences, Noskowskiego 12, 61-704 Poznan, Poland
| | | |
Collapse
|
13
|
Chu PH, Jenol MA, Phang LY, Ibrahim MF, Purkan P, Hadi S, Abd-Aziz S. Innovative approaches for amino acid production via consolidated bioprocessing of agricultural biomass. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:33303-33324. [PMID: 38710845 DOI: 10.1007/s11356-024-33534-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 04/27/2024] [Indexed: 05/08/2024]
Abstract
Agricultural plantations in Indonesia and Malaysia yield substantial waste, necessitating proper disposal to address environmental concerns. Yet, these wastes, rich in starch and lignocellulosic content, offer an opportunity for value-added product development, particularly amino acid production. Traditional methods often rely on costly commercial enzymes to convert biomass into fermentable sugars for amino acid production. An alternative, consolidated bioprocessing, enables the direct conversion of agricultural biomass into amino acids using selected microorganisms. This review provides a comprehensive assessment of the potential of agricultural biomass in Indonesia and Malaysia for amino acid production through consolidated bioprocessing. It explores suitable microorganisms and presents a case study on using Bacillus subtilis ATCC 6051 to produce 9.56 mg/mL of amino acids directly from pineapple plant stems. These findings contribute to the advancement of sustainable amino acid production methods using agricultural biomass especially in Indonesia and Malaysia through consolidated bioprocessing, reducing waste and enhancing environmental sustainability.
Collapse
Affiliation(s)
- Pei-Hsia Chu
- Department of Bioprocess Technology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Mohd Azwan Jenol
- Department of Bioprocess Technology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Lai-Yee Phang
- Department of Bioprocess Technology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Mohamad Faizal Ibrahim
- Department of Bioprocess Technology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Purkan Purkan
- Biochemistry Division, Department of Chemistry, Faculty of Science and Technology, Airlangga University, Jl. Mulyorejo, Surabaya, 60115, Indonesia
| | - Sofijan Hadi
- Biochemistry Division, Department of Chemistry, Faculty of Science and Technology, Airlangga University, Jl. Mulyorejo, Surabaya, 60115, Indonesia
| | - Suraini Abd-Aziz
- Department of Bioprocess Technology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
- Biochemistry Division, Department of Chemistry, Faculty of Science and Technology, Airlangga University, Jl. Mulyorejo, Surabaya, 60115, Indonesia.
| |
Collapse
|
14
|
Miler I, Rabasovic MD, Askrabic S, Stylianou A, Korac B, Korac A. Short-Term l-arginine Treatment Mitigates Early Damage of Dermal Collagen Induced by Diabetes. Bioengineering (Basel) 2024; 11:407. [PMID: 38671828 PMCID: PMC11048012 DOI: 10.3390/bioengineering11040407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/12/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
Changes in the structural properties of the skin due to collagen alterations are an important factor in diabetic skin complications. Using a combination of photonic methods as an optic diagnostic tool, we investigated the structural alteration in rat dermal collagen I in diabetes, and after short-term l-arginine treatment. The multiplex approach shows that in the early phase of diabetes, collagen fibers are partially damaged, resulting in the heterogeneity of fibers, e.g., "patchy patterns" of highly ordered/disordered fibers, while l-arginine treatment counteracts to some extent the conformational changes in collagen-induced by diabetes and mitigates the damage. Raman spectroscopy shows intense collagen conformational changes via amides I and II in diabetes, suggesting that diabetes-induced structural changes in collagen originate predominantly from individual collagen molecules rather than supramolecular structures. There is a clear increase in the amounts of newly synthesized proline and hydroxyproline after treatment with l-arginine, reflecting the changed collagen content. This suggests that it might be useful for treating and stopping collagen damage early on in diabetic skin. Our results demonstrate that l-arginine attenuates the early collagen I alteration caused by diabetes and that it could be used to treat and prevent collagen damage in diabetic skin at a very early stage.
Collapse
Affiliation(s)
- Irena Miler
- Center for Biosystems, BioSense Institute, University of Novi Sad, Dr Zorana Djindjica 1, 21000 Novi Sad, Serbia;
| | - Mihailo D. Rabasovic
- Institute of Physics Belgrade, National Institute of the Republic of Serbia, University of Belgrade, Pregrevica 118, 11000 Belgrade, Serbia; (S.A.)
| | - Sonja Askrabic
- Institute of Physics Belgrade, National Institute of the Republic of Serbia, University of Belgrade, Pregrevica 118, 11000 Belgrade, Serbia; (S.A.)
| | - Andreas Stylianou
- School of Science, European University Cyprus, 6 Diogenous Str., Egkomi, Nicosia 2404, Cyprus;
| | - Bato Korac
- Institute for Biological Research “Sinisa Stankovic”, National Institute of the Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11000 Belgrade, Serbia;
| | - Aleksandra Korac
- Center for Electron Microscopy, Faculty of Biology, University of Belgrade, Studentski trg 16, 11000 Belgrade, Serbia
| |
Collapse
|
15
|
Galal HM, Abdelhafez AT, Sayed MM, Gomaa WMS, Tohamy TA, Gomaa AMS, El-Metwally TH. Impact of L-Arginine on diabetes-induced neuropathy and myopathy: Roles of PAI-1, Irisin, oxidative stress, NF-κβ, autophagy and microRNA-29a. Tissue Cell 2024; 87:102342. [PMID: 38430848 DOI: 10.1016/j.tice.2024.102342] [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: 08/26/2023] [Revised: 02/11/2024] [Accepted: 02/26/2024] [Indexed: 03/05/2024]
Abstract
BACKGROUND T2DM is a chronic disorder with progressive neuromuscular alterations. L-arginine (ARG) is the most common semi-essential amino acid having several metabolic functions. AIM to investigate the impact of L-arginine in combating diabetic-induced neuromyopathy and its possible mechanisms. MATERIALS & METHODS 24 rats were divided into CON, CON+ARG, DC, DC+ARG. Behavioral tests, Body weight (BW), fasting blood glucose (FBG), insulin, total antioxidant capacity (TAC), malondialdehyde (MDA), plasminogen activator inhibitor-1 (PAI-1), and irisin were done. Creatine kinase-MM (CK-MM), interleukin 4 (IL-4), interleukin 6 (IL-6), TAC, MDA, expression of microRNA-29a mRNA & light chain 3 protein were determined in muscle. Histological and NF-κβ immunohistochemical expression in muscle and nerve were assessed. RESULTS ARG supplementation to diabetic rats improved altered behavior, significantly increased BW, insulin, TAC, irisin and Il-4, decreased levels of glucose, microRNA-29a, NF-κβ and LC3 expression, PAI-1, CK-MM and restored the normal histological appearance. CONCLUSIONS ARG supplementation potently alleviated diabetic-induced neuromuscular alterations.
Collapse
Affiliation(s)
- Heba M Galal
- Department of Medical Physiology, College of Medicine, Jouf University, Sakaka, Saudi Arabia; Medical Physiology Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Alaa T Abdelhafez
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt; Department of Basic Medical Sciences, Badr University, New Nasser City, West of Assiut, Assiut, Egypt.
| | - Manal M Sayed
- Histology and Cell Biology Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Walaa M S Gomaa
- Department of Nutrition and Clinical Nutrition, Faculty of Vet. Medicine, Assiut University, Assiut, Egypt
| | | | - Asmaa M S Gomaa
- Medical Physiology Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Tarek H El-Metwally
- Biochemistry Division, Department of Pathology, College of Medicine, Jouf University, Sakaka, Saudi Arabia; Medical Biochemistry Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
16
|
Baker CL, Seo KS, Park N, Rutter JK, Thornton JA, Pruett SB, Park JY. L-arginine supplementation abrogates hypoxia-induced virulence of Staphylococcus aureus in a murine diabetic pressure wound model. mSphere 2024; 9:e0077423. [PMID: 38426801 PMCID: PMC10964415 DOI: 10.1128/msphere.00774-23] [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: 12/12/2023] [Accepted: 01/24/2024] [Indexed: 03/02/2024] Open
Abstract
Diabetic foot ulcers (DFUs) are the most common complications of diabetes resulting from hyperglycemia leading to ischemic hypoxic tissue and nerve damage. Staphylococcus aureus is the most frequently isolated bacteria from DFUs and causes severe necrotic infections leading to amputations with a poor 5-year survival rate. However, very little is known about the mechanisms by which S. aureus dominantly colonizes and causes severe disease in DFUs. Herein, we utilized a pressure wound model in diabetic TALLYHO/JngJ mice to reproduce ischemic hypoxic tissue damage seen in DFUs and demonstrated that anaerobic fermentative growth of S. aureus significantly increased the virulence and the severity of disease by activating two-component regulatory systems leading to expression of virulence factors. Our in vitro studies showed that supplementation of nitrate as a terminal electron acceptor promotes anaerobic respiration and suppresses the expression of S. aureus virulence factors through inactivation of two-component regulatory systems, suggesting potential therapeutic benefits by promoting anaerobic nitrate respiration. Our in vivo studies revealed that dietary supplementation of L-arginine (L-Arg) significantly attenuated the severity of disease caused by S. aureus in the pressure wound model by providing nitrate. Collectively, these findings highlight the importance of anaerobic fermentative growth in S. aureus pathogenesis and the potential of dietary L-Arg supplementation as a therapeutic to prevent severe S. aureus infection in DFUs.IMPORTANCES. aureus is the most common cause of infection in DFUs, often resulting in lower-extremity amputation with a distressingly poor 5-year survival rate. Treatment for S. aureus infections has largely remained unchanged for decades and involves tissue debridement with antibiotic therapy. With high levels of conservative treatment failure, recurrence of ulcers, and antibiotic resistance, a new approach is necessary to prevent lower-extremity amputations. Nutritional aspects of DFU treatment have largely been overlooked as there has been contradictory clinical trial evidence, but very few in vitro and in vivo modelings of nutritional treatment studies have been performed. Here we demonstrate that dietary supplementation of L-Arg in a diabetic mouse model significantly reduced duration and severity of disease caused by S. aureus. These findings suggest that L-Arg supplementation could be useful as a potential preventive measure against severe S. aureus infections in DFUs.
Collapse
Affiliation(s)
- Carol L. Baker
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi, USA
| | - Keun Seok Seo
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi, USA
| | - Nogi Park
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi, USA
| | - Jaime K. Rutter
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi, USA
| | - Justin A. Thornton
- Department of Biological Sciences, College of Arts and Sciences, Mississippi State University, Mississippi State, Mississippi, USA
| | - Stephen B. Pruett
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi, USA
| | - Joo Youn Park
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi, USA
| |
Collapse
|
17
|
Abdi F, Mohammadzadeh M, Abbasalizad-Farhangi M. Dietary amino acid patterns and cardiometabolic risk factors among subjects with obesity; a cross-sectional study. BMC Endocr Disord 2024; 24:21. [PMID: 38355488 PMCID: PMC10865612 DOI: 10.1186/s12902-024-01549-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: 11/14/2023] [Accepted: 01/29/2024] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND The prevalence of obesity is a growing global public health concern. Certain dietary amino acids have been shown to have a potential therapeutic role in improving metabolic syndrome parameters and body composition in individuals with obesity. However, some amino acids have been linked to an increased risk of cardiometabolic disorders. This cross-sectional study aims to investigate the association between dietary amino acid patterns and cardiometabolic risk factors in individuals with obesity. METHODS This cross-sectional study included 335 participants with obesity (57.9% males and 41.5% females) from Tabriz and Tehran, Iran. The participants were between the ages of 20-50, with a body mass index (BMI) of 30 kg/m2 or higher, and free from certain medical conditions. The study examined participants' general characteristics, conducted anthropometric assessments, dietary assessments, and biochemical assessments. The study also used principal component analysis to identify amino acid intake patterns and determined the association between these patterns and cardiometabolic risk factors in individuals with obesity. RESULTS Upon adjusting for potential confounders, the study found that individuals in the third tertiles of pattern 1 and 2 were more likely to have lower LDL levels (OR = 0.99 and 95% CI (0.98-0.99)) for both. Additionally, a significant decrease in total cholesterol was observed in the third tertiles of pattern 2 in model II (OR = 0.99, 95% CI (0.98-0.99)). These findings suggest a potential cardioprotective effect of these amino acid patterns in managing cardiometabolic risk factors in individuals with obesity. CONCLUSIONS This study found that two identified amino acid patterns were associated with lower serum LDL and total cholesterol levels, while a third pattern was associated with higher serum triglycerides. The specific amino acids contributing to these patterns highlight the importance of targeted dietary interventions in managing cardiometabolic risk factors in individuals with obesity.
Collapse
Affiliation(s)
- Fatemeh Abdi
- Department of Community Nutrition, Faculty of Nutrition, Tabriz University of Medical Sciences, Attar Neyshabouri, Daneshgah Blv, Tabriz, Iran
| | - Milad Mohammadzadeh
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdieh Abbasalizad-Farhangi
- Department of Community Nutrition, Faculty of Nutrition, Tabriz University of Medical Sciences, Attar Neyshabouri, Daneshgah Blv, Tabriz, Iran.
| |
Collapse
|
18
|
Margara-Escudero HJ, Paz-Graniel I, García-Gavilán J, Ruiz-Canela M, Sun Q, Clish CB, Toledo E, Corella D, Estruch R, Ros E, Castañer O, Arós F, Fiol M, Guasch-Ferré M, Lapetra J, Razquin C, Dennis C, Deik A, Li J, Gómez-Gracia E, Babio N, Martínez-González MA, Hu FB, Salas-Salvadó J. Plasma metabolite profile of legume consumption and future risk of type 2 diabetes and cardiovascular disease. Cardiovasc Diabetol 2024; 23:38. [PMID: 38245716 PMCID: PMC10800064 DOI: 10.1186/s12933-023-02111-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/29/2023] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND Legume consumption has been linked to a reduced risk of type 2 diabetes (T2D) and cardiovascular disease (CVD), while the potential association between plasma metabolites associated with legume consumption and the risk of cardiometabolic diseases has never been explored. Therefore, we aimed to identify a metabolite signature of legume consumption, and subsequently investigate its potential association with the incidence of T2D and CVD. METHODS The current cross-sectional and longitudinal analysis was conducted in 1833 PREDIMED study participants (mean age 67 years, 57.6% women) with available baseline metabolomic data. A subset of these participants with 1-year follow-up metabolomics data (n = 1522) was used for internal validation. Plasma metabolites were assessed through liquid chromatography-tandem mass spectrometry. Cross-sectional associations between 382 different known metabolites and legume consumption were performed using elastic net regression. Associations between the identified metabolite profile and incident T2D and CVD were estimated using multivariable Cox regression models. RESULTS Specific metabolic signatures of legume consumption were identified, these included amino acids, cortisol, and various classes of lipid metabolites including diacylglycerols, triacylglycerols, plasmalogens, sphingomyelins and other metabolites. Among these identified metabolites, 22 were negatively and 18 were positively associated with legume consumption. After adjustment for recognized risk factors and legume consumption, the identified legume metabolite profile was inversely associated with T2D incidence (hazard ratio (HR) per 1 SD: 0.75, 95% CI 0.61-0.94; p = 0.017), but not with CVD incidence risk (1.01, 95% CI 0.86-1.19; p = 0.817) over the follow-up period. CONCLUSIONS This study identified a set of 40 metabolites associated with legume consumption and with a reduced risk of T2D development in a Mediterranean population at high risk of cardiovascular disease. TRIAL REGISTRATION ISRCTN35739639.
Collapse
Affiliation(s)
- Hernando J Margara-Escudero
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Alimentació, Nutrició, Desenvolupament i Salut Mental ANUT-DSM, Reus, Spain
- Alimentació, Nutrició, Desenvolupament i Salut Mental, Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Indira Paz-Graniel
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Alimentació, Nutrició, Desenvolupament i Salut Mental ANUT-DSM, Reus, Spain
- Alimentació, Nutrició, Desenvolupament i Salut Mental, Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de La Obesidad y La Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús García-Gavilán
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Alimentació, Nutrició, Desenvolupament i Salut Mental ANUT-DSM, Reus, Spain.
- Alimentació, Nutrició, Desenvolupament i Salut Mental, Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain.
- Centro de Investigación Biomédica en Red Fisiopatología de La Obesidad y La Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
| | - Miguel Ruiz-Canela
- Centro de Investigación Biomédica en Red Fisiopatología de La Obesidad y La Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Preventive Medicine and Public Health, University of Navarra, Instituto de Investigación Sanitario de Navarra (IdiSNA), Pamplona, Spain
| | - Qi Sun
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Clary B Clish
- The Broad Institute of Harvard and MIT, Boston, MA, 02142, USA
| | - Estefania Toledo
- Centro de Investigación Biomédica en Red Fisiopatología de La Obesidad y La Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Preventive Medicine and Public Health, University of Navarra, Instituto de Investigación Sanitario de Navarra (IdiSNA), Pamplona, Spain
- Navarra Institute for Health Research, IdiSNA, Pamplona, Navarre, Spain
| | - Dolores Corella
- Centro de Investigación Biomédica en Red Fisiopatología de La Obesidad y La Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Preventive Medicine, University of Valencia, Valencia, Spain
| | - Ramón Estruch
- Centro de Investigación Biomédica en Red Fisiopatología de La Obesidad y La Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Internal Medicine, Institut d'Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Emilio Ros
- Centro de Investigación Biomédica en Red Fisiopatología de La Obesidad y La Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Endocrinology and Nutrition, Institut d'Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Lipid Clinic, Hospital Clínic, Barcelona, Spain
| | - Olga Castañer
- Centro de Investigación Biomédica en Red (CIBERESP) de Epidemiología y Salud Pública, Instituto de Salud Carlos III, Madrid, Spain
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Research Institute, Barcelona, Spain
| | - Fernando Arós
- Centro de Investigación Biomédica en Red Fisiopatología de La Obesidad y La Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Cardiology, University Hospital of Alava, Vitoria, Spain
| | - Miquel Fiol
- Centro de Investigación Biomédica en Red Fisiopatología de La Obesidad y La Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Illes Balears Health Research Institute (IdISBa), Hospital Son Espases, Palma, Spain
| | - Marta Guasch-Ferré
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Public Health, Section of Epidemiology, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - José Lapetra
- Centro de Investigación Biomédica en Red Fisiopatología de La Obesidad y La Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Family Medicine, Research Unit, Distrito Sanitario Atención Primaria Sevilla, Seville, Spain
| | - Cristina Razquin
- Centro de Investigación Biomédica en Red Fisiopatología de La Obesidad y La Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Preventive Medicine and Public Health, University of Navarra, Instituto de Investigación Sanitario de Navarra (IdiSNA), Pamplona, Spain
| | - Courtney Dennis
- The Broad Institute of Harvard and MIT, Boston, MA, 02142, USA
| | - Amy Deik
- The Broad Institute of Harvard and MIT, Boston, MA, 02142, USA
| | - Jun Li
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Enrique Gómez-Gracia
- Preventive Medicine and Public Health Department, School of Medicine, University of Málaga, 29010, Malaga, Spain
- Biomedical Research Institute of Malaga-IBIMA Plataforma BIONAND, 29010, Malaga, Spain
| | - Nancy Babio
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Alimentació, Nutrició, Desenvolupament i Salut Mental ANUT-DSM, Reus, Spain.
- Alimentació, Nutrició, Desenvolupament i Salut Mental, Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain.
- Centro de Investigación Biomédica en Red Fisiopatología de La Obesidad y La Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
| | - Miguel A Martínez-González
- Department of Preventive Medicine and Public Health, University of Navarra, Instituto de Investigación Sanitario de Navarra (IdiSNA), Pamplona, Spain
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jordi Salas-Salvadó
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Alimentació, Nutrició, Desenvolupament i Salut Mental ANUT-DSM, Reus, Spain
- Alimentació, Nutrició, Desenvolupament i Salut Mental, Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de La Obesidad y La Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
19
|
Lopez-Pedrosa JM, Camprubi-Robles M, Guzman-Rolo G, Lopez-Gonzalez A, Garcia-Almeida JM, Sanz-Paris A, Rueda R. The Vicious Cycle of Type 2 Diabetes Mellitus and Skeletal Muscle Atrophy: Clinical, Biochemical, and Nutritional Bases. Nutrients 2024; 16:172. [PMID: 38202001 PMCID: PMC10780454 DOI: 10.3390/nu16010172] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 12/30/2023] [Accepted: 12/30/2023] [Indexed: 01/12/2024] Open
Abstract
Today, type 2 diabetes mellitus (T2DM) and skeletal muscle atrophy (SMA) have become increasingly common occurrences. Whether the onset of T2DM increases the risk of SMA or vice versa has long been under investigation. Both conditions are associated with negative changes in skeletal muscle health, which can, in turn, lead to impaired physical function, a lowered quality of life, and an increased risk of mortality. Poor nutrition can exacerbate both T2DM and SMA. T2DM and SMA are linked by a vicious cycle of events that reinforce and worsen each other. Muscle insulin resistance appears to be the pathophysiological link between T2DM and SMA. To explore this association, our review (i) compiles evidence on the clinical association between T2DM and SMA, (ii) reviews mechanisms underlying biochemical changes in the muscles of people with or at risk of T2DM and SMA, and (iii) examines how nutritional therapy and increased physical activity as muscle-targeted treatments benefit this population. Based on the evidence, we conclude that effective treatment of patients with T2DM-SMA depends on the restoration and maintenance of muscle mass. We thus propose that regular intake of key functional nutrients, along with guidance for physical activity, can help maintain euglycemia and improve muscle status in all patients with T2DM and SMA.
Collapse
Affiliation(s)
| | | | | | | | - Jose Manuel Garcia-Almeida
- Department of Endocrinology and Nutrition, Virgen de la Victoria Hospital (IBIMA), Malaga University, 29010 Malaga, Spain;
| | - Alejandro Sanz-Paris
- Nutrition Unit, Universitary Hospital Miguel Servet, Isabel the Catholic 1-3, 50009 Zaragoza, Spain;
| | - Ricardo Rueda
- Abbott Nutrition R&D, 18004 Granada, Spain; (M.C.-R.); (A.L.-G.); (R.R.)
| |
Collapse
|
20
|
Manzano M, Girón MD, Salto R, Burgio C, Reinoso A, Cabrera E, Rueda R, López-Pedrosa JM. Arginine and Lysine Supplementation Potentiates the Beneficial β-Hydroxy ß-Methyl Butyrate (HMB) Effects on Skeletal Muscle in a Rat Model of Diabetes. Nutrients 2023; 15:4706. [PMID: 38004100 PMCID: PMC10674618 DOI: 10.3390/nu15224706] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 11/02/2023] [Accepted: 11/04/2023] [Indexed: 11/26/2023] Open
Abstract
Skeletal muscle is the key tissue for maintaining protein and glucose homeostasis, having a profound impact on the development of diabetes. Diabetes causes deleterious changes in terms of loss of muscle mass, which will contribute to reduced glucose uptake and therefore progression of the disease. Nutritional approaches in diabetes have been directed to increase muscle glucose uptake, and improving protein turnover has been at least partially an oversight. In muscle, β-hydroxy β-methyl butyrate (HMB) promotes net protein synthesis, while arginine and lysine increase glucose uptake, albeit their effects on promoting protein synthesis are limited. This study evaluates if the combination of HMB, lysine, and arginine could prevent the loss of muscle mass and function, reducing the progression of diabetes. Therefore, the combination of these ingredients was tested in vitro and in vivo. In muscle cell cultures, the supplementation enhances glucose uptake and net protein synthesis due to an increase in the amount of GLUT4 transporter and stimulation of the insulin-dependent signaling pathway involving IRS-1 and Akt. In vivo, using a rat model of diabetes, the supplementation increases lean body mass and insulin sensitivity and decreases blood glucose and serum glycosylated hemoglobin. In treated animals, an increase in GLUT4, creatine kinase, and Akt phosphorylation was detected, demonstrating the synergic effects of the three ingredients. Our findings showed that nutritional formulations based on the combination of HMB, lysine, and arginine are effective, not only to control blood glucose levels but also to prevent skeletal muscle atrophy associated with the progression of diabetes.
Collapse
Affiliation(s)
- Manuel Manzano
- Abbott Nutrition R&D, E18004 Granada, Spain; (M.M.); (R.R.); (J.M.L.-P.)
| | - María D. Girón
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, E18071 Granada, Spain; (M.D.G.); (C.B.); (A.R.); (E.C.)
| | - Rafael Salto
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, E18071 Granada, Spain; (M.D.G.); (C.B.); (A.R.); (E.C.)
| | - Chiara Burgio
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, E18071 Granada, Spain; (M.D.G.); (C.B.); (A.R.); (E.C.)
| | - Antonio Reinoso
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, E18071 Granada, Spain; (M.D.G.); (C.B.); (A.R.); (E.C.)
| | - Elena Cabrera
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, E18071 Granada, Spain; (M.D.G.); (C.B.); (A.R.); (E.C.)
| | - Ricardo Rueda
- Abbott Nutrition R&D, E18004 Granada, Spain; (M.M.); (R.R.); (J.M.L.-P.)
| | | |
Collapse
|
21
|
Liu M, Shen J, Zhu X, Ju T, Willing BP, Wu X, Lu Q, Liu R. Peanut skin procyanidins reduce intestinal glucose transport protein expression, regulate serum metabolites and ameliorate hyperglycemia in diabetic mice. Food Res Int 2023; 173:113471. [PMID: 37803795 DOI: 10.1016/j.foodres.2023.113471] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/05/2023] [Accepted: 09/10/2023] [Indexed: 10/08/2023]
Abstract
One of diabetic characteristics is the postprandial hyperglycemia. Inhibiting glucose uptake may be beneficial for controlling postprandial blood glucose levels and regulating the glucose metabolism Peanut skin procyanidins (PSP) have shown a potential for lowering blood glucose; however, the underlying mechanism through which PSP regulate glucose metabolism remains unknown. In the current study, we investigated the effect of PSP on intestinal glucose transporters and serum metabolites using a mouse model of diabetic mice. Results showed that PSP improved glucose tolerance and systemic insulin sensitivity, which coincided with decreased expression of sodium-glucose cotransporter 1 and glucose transporter 2 in the intestinal epithelium induced by an activation of the phospholipase C β2/protein kinase C signaling pathway. Moreover, untargeted metabolomic analysis of serum samples revealed that PSP altered arachidonic acid, sphingolipid, glycerophospholipid, bile acids, and arginine metabolic pathways. The study provides new insight into the anti-diabetic mechanism of PSP and a basis for further research.
Collapse
Affiliation(s)
- Min Liu
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430000, China; Wuhan Engineering Research Center of Bee Products on Quality and Safety Control, Wuhan 430000, China
| | - Jinxin Shen
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430000, China; Wuhan Engineering Research Center of Bee Products on Quality and Safety Control, Wuhan 430000, China
| | - Xiaoling Zhu
- Hubei Provincial Institute for Food Supervision and Test, Wuhan 430070, China
| | - Tingting Ju
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - Benjamin P Willing
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - Xin Wu
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430000, China; Wuhan Engineering Research Center of Bee Products on Quality and Safety Control, Wuhan 430000, China
| | - Qun Lu
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430000, China; Wuhan Engineering Research Center of Bee Products on Quality and Safety Control, Wuhan 430000, China; Key Laboratory of Environment Correlative Dietology, Ministry of Education, Wuhan 430000, China
| | - Rui Liu
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430000, China; Wuhan Engineering Research Center of Bee Products on Quality and Safety Control, Wuhan 430000, China; Key Laboratory of Environment Correlative Dietology, Ministry of Education, Wuhan 430000, China; Key Laboratory of Urban Agriculture in Central China, Ministry of Agriculture and Rural Affairs, China.
| |
Collapse
|
22
|
Garibay-Nieto N, Pedraza-Escudero K, Omaña-Guzmán I, Garcés-Hernández MJ, Villanueva-Ortega E, Flores-Torres M, Pérez-Hernández JL, León-Hernández M, Laresgoiti-Servitje E, Palacios-González B, López-Alvarenga JC, Lisker-Melman M, Vadillo-Ortega F. Metabolomic Phenotype of Hepatic Steatosis and Fibrosis in Mexican Children Living with Obesity. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1785. [PMID: 37893503 PMCID: PMC10608521 DOI: 10.3390/medicina59101785] [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: 07/26/2023] [Revised: 09/13/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023]
Abstract
Background and Objectives: Metabolic-dysfunction-associated steatotic liver disease or MASLD is the main cause of chronic liver diseases in children, and it is estimated to affect 35% of children living with obesity. This study aimed to identify metabolic phenotypes associated with two advanced stages of MASLD (hepatic steatosis and hepatic steatosis plus fibrosis) in Mexican children with obesity. Materials and Methods: This is a cross-sectional analysis derived from a randomized clinical trial conducted in children and adolescents with obesity aged 8 to 16 years. Anthropometric and biochemical data were measured, and targeted metabolomic analyses were carried out using mass spectrometry. Liver steatosis and fibrosis were estimated using transient elastography (Fibroscan® Echosens, Paris, France). Three groups were studied: a non-MASLD group, an MASLD group, and a group for MASLD + fibrosis. A partial least squares discriminant analysis (PLS-DA) was performed to identify the discrimination between the study groups and to visualize the differences between their heatmaps; also, Variable Importance Projection (VIP) plots were graphed. A VIP score of >1.5 was considered to establish the importance of metabolites and biochemical parameters that characterized each group. Logistic regression models were constructed considering VIP scores of >1.5, and the receiver operating characteristic (ROC) curves were estimated to evaluate different combinations of variables. Results: The metabolic MASLD phenotype was associated with increased concentrations of ALT and decreased arginine, glycine, and acylcarnitine (AC) AC5:1, while MASLD + fibrosis, an advanced stage of MASLD, was associated with a phenotype characterized by increased concentrations of ALT, proline, and alanine and a decreased Matsuda Index. Conclusions: The metabolic MASLD phenotype changes as this metabolic dysfunction progresses. Understanding metabolic disturbances in MASLD would allow for early identification and the development of intervention strategies focused on limiting the progression of liver damage in children and adolescents.
Collapse
Affiliation(s)
- Nayely Garibay-Nieto
- Pediatric Obesity Clinic and Wellness Unit, General Hospital of Mexico, Mexico City 06720, Mexico; (N.G.-N.); (K.P.-E.); (I.O.-G.); (M.J.G.-H.); (E.V.-O.)
| | - Karen Pedraza-Escudero
- Pediatric Obesity Clinic and Wellness Unit, General Hospital of Mexico, Mexico City 06720, Mexico; (N.G.-N.); (K.P.-E.); (I.O.-G.); (M.J.G.-H.); (E.V.-O.)
| | - Isabel Omaña-Guzmán
- Pediatric Obesity Clinic and Wellness Unit, General Hospital of Mexico, Mexico City 06720, Mexico; (N.G.-N.); (K.P.-E.); (I.O.-G.); (M.J.G.-H.); (E.V.-O.)
| | - María José Garcés-Hernández
- Pediatric Obesity Clinic and Wellness Unit, General Hospital of Mexico, Mexico City 06720, Mexico; (N.G.-N.); (K.P.-E.); (I.O.-G.); (M.J.G.-H.); (E.V.-O.)
| | - Eréndira Villanueva-Ortega
- Pediatric Obesity Clinic and Wellness Unit, General Hospital of Mexico, Mexico City 06720, Mexico; (N.G.-N.); (K.P.-E.); (I.O.-G.); (M.J.G.-H.); (E.V.-O.)
| | - Mariana Flores-Torres
- Unidad de Vinculación de la Facultad de Medicina, UNAM, Instituto Nacional de Medicina Genómica, Mexico City 14610, Mexico;
| | - José Luis Pérez-Hernández
- Hepatology Clinic, Gastroenterology Department, General Hospital of Mexico, Mexico City 06720, Mexico;
| | | | | | - Berenice Palacios-González
- Laboratorio de Envejecimiento Saludable, Centro de Investigación Sobre el Envejecimiento, Instituto Nacional de Medicina Genómica, Mexico City 14610, Mexico;
| | - Juan Carlos López-Alvarenga
- Department of Population Health & Biostatistics, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA;
| | - Mauricio Lisker-Melman
- Division of Gastroenterology, Washington University School of Medicine, St. Louis, MO 63110, USA;
| | - Felipe Vadillo-Ortega
- Unidad de Vinculación de la Facultad de Medicina, UNAM, Instituto Nacional de Medicina Genómica, Mexico City 14610, Mexico;
| |
Collapse
|
23
|
Mone P, Wang X, Trimarco V, Santulli G. Re: Differential benefits of physical training associated or not with L-Arginine supplementation in rats with metabolic syndrome: Evaluation of cardiovascular, autonomic and metabolic parameters. Physiol Behav 2023; 269:114274. [PMID: 37331457 PMCID: PMC11594566 DOI: 10.1016/j.physbeh.2023.114274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 06/16/2023] [Indexed: 06/20/2023]
Affiliation(s)
- Pasquale Mone
- Department of Molecular Pharmacology, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Neuroimmunology and Inflammation (INI), Fleischer Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, New York City, NY
| | - Xujun Wang
- Department of Molecular Pharmacology, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Neuroimmunology and Inflammation (INI), Fleischer Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, New York City, NY
| | - Valentina Trimarco
- Department of Neuroscience, Reproductive Sciences and Dentistry, "Federico II" University, Naples, Italy
| | - Gaetano Santulli
- Department of Molecular Pharmacology, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Neuroimmunology and Inflammation (INI), Fleischer Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, New York City, NY; Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy; Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York City, NY.
| |
Collapse
|
24
|
Santulli G, Kansakar U, Varzideh F, Mone P, Jankauskas SS, Lombardi A. Functional Role of Taurine in Aging and Cardiovascular Health: An Updated Overview. Nutrients 2023; 15:4236. [PMID: 37836520 PMCID: PMC10574552 DOI: 10.3390/nu15194236] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/22/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
Taurine, a naturally occurring sulfur-containing amino acid, has attracted significant attention in recent years due to its potential health benefits. Found in various foods and often used in energy drinks and supplements, taurine has been studied extensively to understand its impact on human physiology. Determining its exact functional roles represents a complex and multifaceted topic. We provide an overview of the scientific literature and present an analysis of the effects of taurine on various aspects of human health, focusing on aging and cardiovascular pathophysiology, but also including athletic performance, metabolic regulation, and neurological function. Additionally, our report summarizes the current recommendations for taurine intake and addresses potential safety concerns. Evidence from both human and animal studies indicates that taurine may have beneficial cardiovascular effects, including blood pressure regulation, improved cardiac fitness, and enhanced vascular health. Its mechanisms of action and antioxidant properties make it also an intriguing candidate for potential anti-aging strategies.
Collapse
Affiliation(s)
- Gaetano Santulli
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA; (U.K.); (S.S.J.); (A.L.)
- Department of Molecular Pharmacology, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA; (F.V.); (P.M.)
| | - Urna Kansakar
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA; (U.K.); (S.S.J.); (A.L.)
| | - Fahimeh Varzideh
- Department of Molecular Pharmacology, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA; (F.V.); (P.M.)
| | - Pasquale Mone
- Department of Molecular Pharmacology, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA; (F.V.); (P.M.)
| | - Stanislovas S. Jankauskas
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA; (U.K.); (S.S.J.); (A.L.)
| | - Angela Lombardi
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA; (U.K.); (S.S.J.); (A.L.)
| |
Collapse
|
25
|
Gomchok D, Ge RL, Wuren T. Platelets in Renal Disease. Int J Mol Sci 2023; 24:14724. [PMID: 37834171 PMCID: PMC10572297 DOI: 10.3390/ijms241914724] [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: 08/31/2023] [Revised: 09/18/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Kidney disease is a major global health concern, affecting millions of people. Nephrologists have shown interest in platelets because of coagulation disorders caused by renal diseases. With a better understanding of platelets, it has been found that these anucleate and abundant blood cells not only play a role in hemostasis, but also have important functions in inflammation and immunity. Platelets are not only affected by kidney disease, but may also contribute to kidney disease progression by mediating inflammation and immune effects. This review summarizes the current evidence regarding platelet abnormalities in renal disease, and the multiple effects of platelets on kidney disease progression. The relationship between platelets and kidney disease is still being explored, and further research can provide mechanistic insights into the relationship between thrombosis, bleeding, and inflammation related to kidney disease, and elucidate targeted therapies for patients with kidney disease.
Collapse
Affiliation(s)
- Drolma Gomchok
- Research Center for High Altitude Medicine, School of Medicine, Qinghai University, Xining 810001, China; (D.G.); (R.-L.G.)
| | - Ri-Li Ge
- Research Center for High Altitude Medicine, School of Medicine, Qinghai University, Xining 810001, China; (D.G.); (R.-L.G.)
- Key Laboratory for Application for High Altitude Medicine, Qinghai University, Xining 810001, China
| | - Tana Wuren
- Research Center for High Altitude Medicine, School of Medicine, Qinghai University, Xining 810001, China; (D.G.); (R.-L.G.)
- Key Laboratory for Application for High Altitude Medicine, Qinghai University, Xining 810001, China
| |
Collapse
|
26
|
Forzano I, Avvisato R, Varzideh F, Jankauskas SS, Cioppa A, Mone P, Salemme L, Kansakar U, Tesorio T, Trimarco V, Santulli G. Correction: L-Arginine in diabetes: clinical and preclinical evidence. Cardiovasc Diabetol 2023; 22:117. [PMID: 37208744 DOI: 10.1186/s12933-023-01852-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/21/2023] Open
Affiliation(s)
- Imma Forzano
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Fleischer Institute for Diabetes Research (FIDAM), Einstein - Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein University College of Medicine, New York, USA
| | - Roberta Avvisato
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Fleischer Institute for Diabetes Research (FIDAM), Einstein - Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein University College of Medicine, New York, USA
| | - Fahimeh Varzideh
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Fleischer Institute for Diabetes Research (FIDAM), Einstein - Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein University College of Medicine, New York, USA
| | - Stanislovas S Jankauskas
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Fleischer Institute for Diabetes Research (FIDAM), Einstein - Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein University College of Medicine, New York, USA
| | - Angelo Cioppa
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Fleischer Institute for Diabetes Research (FIDAM), Einstein - Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein University College of Medicine, New York, USA
- Montevergine Clinic, Mercogliano, (AV), Italy
| | - Pasquale Mone
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Fleischer Institute for Diabetes Research (FIDAM), Einstein - Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein University College of Medicine, New York, USA
| | | | - Urna Kansakar
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Fleischer Institute for Diabetes Research (FIDAM), Einstein - Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein University College of Medicine, New York, USA
| | | | - Valentina Trimarco
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Fleischer Institute for Diabetes Research (FIDAM), Einstein - Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein University College of Medicine, New York, USA
- Department of Neuroscience, Reproductive Sciences and Dentistry, "Federico II" University, Naples, Italy
| | - Gaetano Santulli
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Fleischer Institute for Diabetes Research (FIDAM), Einstein - Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein University College of Medicine, New York, USA.
- Department of Molecular Pharmacology, Institute for Neuroimmunology and Inflammation (INI), Albert Einstein University College of Medicine, New York, USA.
| |
Collapse
|