1
|
de Paulo Farias D, de Araújo FF, Villasante J, Fogliano V, Pastore GM. In vitro gastrointestinal digestion and gut microbiota fermentation of phenolic compounds from uvaia. Food Chem 2025; 477:143462. [PMID: 40043608 DOI: 10.1016/j.foodchem.2025.143462] [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: 10/23/2024] [Revised: 02/11/2025] [Accepted: 02/15/2025] [Indexed: 03/27/2025]
Abstract
Gastrointestinal digestion and gut microbiota fermentation can alter the bioaccessibility and bioactivity of phenolic compounds. This study assessed the effects of gastrointestinal digestion and gut microbiota fermentation on the bioaccessibility, bioactivity, and catabolism of phenolic compounds from uvaia (Eugenia pyriformis) seed and edible fraction (pulp + peel). The bioaccessibility of epigallocatechin, epicatechin, myricetin, and ferulic acid increased after the gastrointestinal digestion of the edible fraction, while seed digestion reduced epigallocatechin, procyanidin B2, and salicyl aldehyde levels. Acetate and butyrate production was higher from uvaia seed after 24-h fermentation (212.93 and 192.09 mg/L, respectively), while propionate production was higher from the edible fraction (63.37 mg/L). These findings suggest that gastrointestinal digestion influences the bioaccessibility and bioactivity of phenolic compounds in uvaia fractions. Additionally, the increased production of short-chain fatty acids points to a potential prebiotic effect, highlighting the potential of uvaia for developing intestinal health-promoting food products or supplements.
Collapse
Affiliation(s)
- David de Paulo Farias
- Bioflavors and Bioactive Compounds Laboratory, Department of Food Science, School of Food Engineering, University of Campinas, UNICAMP, Campinas, SP, 13083-862, 7, Brazil.
| | - Fábio Fernandes de Araújo
- Bioflavors and Bioactive Compounds Laboratory, Department of Food Science, School of Food Engineering, University of Campinas, UNICAMP, Campinas, SP, 13083-862, 7, Brazil
| | - Juliana Villasante
- Chemical Engineering Department, Universitat Politècnica de Catalunya, Av. Diagonal 647, 08028 Barcelona, Spain; Food Quality and Design Group, Department of Agrotechnology and Food Sciences, Wageningen University & Research, P.O. Box 17, 6700, AA Wageningen, the Netherlands
| | - Vincenzo Fogliano
- Food Quality and Design Group, Department of Agrotechnology and Food Sciences, Wageningen University & Research, P.O. Box 17, 6700, AA Wageningen, the Netherlands
| | - Glaucia Maria Pastore
- Bioflavors and Bioactive Compounds Laboratory, Department of Food Science, School of Food Engineering, University of Campinas, UNICAMP, Campinas, SP, 13083-862, 7, Brazil.
| |
Collapse
|
2
|
Kaushik AS, Agarwal V, Kumar N, Rehman M, Chaudhary R, Srivastava S, Srivastava S, Mishra V. Stimulation of auricular vagus nerve ameliorates chronic stress induced metabolic syndrome via activation of Sirtuin-6. Biochem Biophys Res Commun 2025; 756:151567. [PMID: 40056501 DOI: 10.1016/j.bbrc.2025.151567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/18/2025] [Accepted: 02/28/2025] [Indexed: 03/10/2025]
Abstract
Chronic stress is one of the potential causes of the progression of metabolic syndrome (MS). Chronic stress decreases the release of Sirtuin-6 (SIRT6), which regulates MS by controlling glucose, insulin, lipids, and hypertension. Vagus nerve stimulation (VNS) activates SIRT6 via the cholinergic anti-inflammatory pathway (CAP). However, the effectiveness of VNS therapy for treating MS induced by chronic stress has not yet been studied. In this study, we first validated a rat model of chronic unpredictable stress (CUS) and assessed the characteristic features of MS. The CUS rats were exposed to random stressors daily for 8 weeks. The stress response was then confirmed by behavioral alteration and elevated serum corticosterone levels in rats, as measured by various behavioral tests and an ELISA kit, respectively. The MS characteristics in CUS rats were assessed using measurements of fasting blood glucose (FBG), systolic blood pressure (SBP), lipid indices, insulin levels, and HOMA-IR. The stressed animals demonstrated a rise in FBG, SBP, and insulin along with altered lipid indices. After CUS, the rats were treated with VNS (6 Hz, 1.0 ms, 6 V, for 40 min × 14 days, alternatively), and their metabolic activity and vagal flow were assessed. Moreover, SIRT6 and AMP-activated protein kinase (AMPK) expression in rats was also assessed by immunohistochemistry and mRNA expression of liver and pancreatic tissue. SIRT6 and AMPK expression was decreased in CUS animals. Interestingly, VNS treatment attenuated CUS induced MS-associated parameters. These results indicate that VNS may be a beneficial complementary and non-pharmacological method for managing CUS-associated MS.
Collapse
Affiliation(s)
- Arjun Singh Kaushik
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, (A Central University), Vidya Vihar, Raebareli Road, Lucknow, 226 025, (U.P.), India
| | - Vipul Agarwal
- MIT College of Pharmacy, Ram Ganga Vihar Phase-II, Moradabad, 244001, (U.P.), India
| | - Neeraj Kumar
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, (A Central University), Vidya Vihar, Raebareli Road, Lucknow, 226 025, (U.P.), India
| | - Mujeeba Rehman
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, (A Central University), Vidya Vihar, Raebareli Road, Lucknow, 226 025, (U.P.), India
| | - Rishabh Chaudhary
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, (A Central University), Vidya Vihar, Raebareli Road, Lucknow, 226 025, (U.P.), India
| | - Siddhi Srivastava
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, (A Central University), Vidya Vihar, Raebareli Road, Lucknow, 226 025, (U.P.), India
| | - Sukriti Srivastava
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, (A Central University), Vidya Vihar, Raebareli Road, Lucknow, 226 025, (U.P.), India
| | - Vikas Mishra
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, (A Central University), Vidya Vihar, Raebareli Road, Lucknow, 226 025, (U.P.), India.
| |
Collapse
|
3
|
Carollo C, Sorce A, Cirafici E, Mulè G, Caimi G. Sirtuins and Resveratrol in Cardiorenal Diseases: A Narrative Review of Mechanisms and Therapeutic Potential. Nutrients 2025; 17:1212. [PMID: 40218970 PMCID: PMC11990745 DOI: 10.3390/nu17071212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 03/28/2025] [Accepted: 03/28/2025] [Indexed: 04/14/2025] Open
Abstract
Aging is a very complex process, and it has been linked with Sirtuins. Sirtuin enzymes are a family of deacetylases that are related to caloric restriction and aging by modulating energy metabolism, genomic stability, and stress resistance. Up to now, seven sirtuins have been recognized. This narrative review aimed to analyze the literature produced between January 2005 and March 2025 to evaluate the role of sirtuins in chronic kidney disease and, as heart and kidney diseases are strictly interrelated, to explore their role in heart diseases and cardio-renal cross-talk. A reciprocal relationship between CKD and aging seems to exist since CKD may contribute to premature biological aging of different organ systems. SIRTs are involved in the pathophysiology of renal diseases; their activation can delay the progression of several renal diseases. Notably, an increasing number of studies linked SIRTs with different CVDs. SIRTs affect the production of mitochondrial reactive oxygen species (ROS) by modulating mitochondrial function. The imbalance of SIRT levels may increase the vulnerability to CVDs. SIRTs are involved in the pathophysiological mechanisms of HFpEF (heart failure with preserved ejection fraction) through different signaling pathways. Fibrosis is the linkage mechanism between the heart and kidney in the development of cardio-renal diseases. Current studies on sirtuins, resveratrol, and cardiorenal disease highlight their potential therapeutic benefits in regulating blood pressure, kidney function, lipid profiles, and inflammation, making them a promising area of investigation for improving cardiovascular and renal health outcomes. However, significant gaps remain. The limited availability of highly selective and potent sirtuin modulators hampers their clinical translation, as most existing compounds exhibit poor bioavailability and suboptimal pharmacokinetic properties.
Collapse
Affiliation(s)
- Caterina Carollo
- Department of Health Promotion, Mother and Child Care, Internal and Specialistic Medicine, University of Palermo, 90127 Palermo, Italy (E.C.); (G.M.)
| | | | | | | | | |
Collapse
|
4
|
Sang P, Ma Y, Zhang X, Chen B, He F, Shen N, Zhao J. BMAL1 attenuates intervertebral disc degeneration by activating the SIRT1/PGC-1α pathway: evidence from vitro studies. Sci Rep 2025; 15:9651. [PMID: 40113885 PMCID: PMC11926130 DOI: 10.1038/s41598-025-94029-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 03/11/2025] [Indexed: 03/22/2025] Open
Abstract
To explore the potential effects and the corresponding mechanisms of brain and muscle arnt-like protein-1 (BMAL1) on the progression of intervertebral disc degeneration (IVDD) in vitro studies. The expression of BMAL1, SIRT1 and PINK1 were evaluated by the method of siRNA/pcDNA in the immortalized nucleus pulposus (NP) cells. The expression of SIRT1/PGC-1α pathway was assessed. The characteristics of NP cell, containing the activity and density, the level of apoptosis, inflammatory response, reactive oxygen species (ROS), senescence, and mitophagy were evaluated. The overexpression of BMAL1 was achieved with the pcDNA3.1, the expression of SIRT1 and PGC-1α were increased, the inflammatory response, the ROS, the level of apoptosis and senescence were decreased, however, the level of mitophagy, the activity and density of NP cell were enhanced. The BMAL1 inhibites the progression of IVDD by activating the SIRT1/PGC-1α pathway in the vitro studies.
Collapse
Affiliation(s)
- Peiming Sang
- Ningbo Medical Center LiHuiLi Hospital, Ningbo, Zhejiang, People's Republic of China
- The Affiliated LiHuiLi Hospital of Ningbo University, #57, Xingning Road, Yinzhou District, Ningbo, Zhejiang, People's Republic of China
| | - Yanyan Ma
- Ningbo Medical Center LiHuiLi Hospital, Ningbo, Zhejiang, People's Republic of China.
- The Affiliated LiHuiLi Hospital of Ningbo University, #57, Xingning Road, Yinzhou District, Ningbo, Zhejiang, People's Republic of China.
| | - Xie Zhang
- Ningbo Medical Center LiHuiLi Hospital, Ningbo, Zhejiang, People's Republic of China
- The Affiliated LiHuiLi Hospital of Ningbo University, #57, Xingning Road, Yinzhou District, Ningbo, Zhejiang, People's Republic of China
| | - Binhui Chen
- Ningbo Medical Center LiHuiLi Hospital, Ningbo, Zhejiang, People's Republic of China
- The Affiliated LiHuiLi Hospital of Ningbo University, #57, Xingning Road, Yinzhou District, Ningbo, Zhejiang, People's Republic of China
| | - Fan He
- Ningbo Medical Center LiHuiLi Hospital, Ningbo, Zhejiang, People's Republic of China
- The Affiliated LiHuiLi Hospital of Ningbo University, #57, Xingning Road, Yinzhou District, Ningbo, Zhejiang, People's Republic of China
| | - Neng Shen
- The NO. 3 Hospital of Yinzhou District, Ningbo, Zhejiang, People's Republic of China
| | - Jiangang Zhao
- The NO. 4 Hospital of Yuyao District, Ningbo, Zhejiang, People's Republic of China
| |
Collapse
|
5
|
Zeng S, Huang X, Qu S, Hu Q. Role and therapeutic considerations of SIRT1 in epilepsy. Neuroscience 2025; 568:109-115. [PMID: 39824342 DOI: 10.1016/j.neuroscience.2025.01.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 01/04/2025] [Accepted: 01/11/2025] [Indexed: 01/20/2025]
Abstract
Epilepsy is a primary study focus for scientists worldwide due to its prevalence and poor prognosis. Silent information regulator 1 (SIRT1), a nicotinamide adenine dinucleotide-dependent deacetylase, is becoming increasingly recognized for its critical role in the pathophysiology and progression of epilepsy. The treatment of epilepsy remains challenging despite the discovery of numerous factors that contribute to the development of several beneficial medications. In recent years, many microRNAs have been linked to the progression of epilepsy because they target SIRT1 mRNA. SIRT1, which protects from epilepsy, has been reported to be upregulated by several natural compounds and their derivatives. This review will summarize the latest findings about SIRT1's role in epilepsy. Results from the literature indicate that SIRT1 is a promising target for epilepsy therapy.
Collapse
Affiliation(s)
- Shasha Zeng
- The Second Affiliated Hospital, Department of Pediatrics, Hengyang Medical School, University of South China, Hengyang, Hunan 4210001, China
| | - Xiangyi Huang
- The Second Affiliated Hospital, Department of Function Examination, Hengyang Medical School, University of South China, Hengyang, Hunan 4210001, China
| | - Shunlin Qu
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| | - Qingpeng Hu
- The Second Affiliated Hospital, Department of Pediatrics, Hengyang Medical School, University of South China, Hengyang, Hunan 4210001, China.
| |
Collapse
|
6
|
Golubnitschaja O, Sargheini N, Bastert J. Mitochondria in cutaneous health, disease, ageing and rejuvenation-the 3PM-guided mitochondria-centric dermatology. EPMA J 2025; 16:1-15. [PMID: 39991093 PMCID: PMC11842662 DOI: 10.1007/s13167-025-00400-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 01/30/2025] [Indexed: 02/25/2025]
Abstract
Association of both intrinsic and extrinsic risk factors leading to accelerated skin ageing is reflected in excessive ROS production and ir/reversible mitochondrial injury and burnout, as abundantly demonstrated by accumulating research data. Due to the critical role of mitochondrial stress in the pathophysiology of skin ageing and disorders, maintained (primary care) and restored (secondary care) mitochondrial health, rejuvenation and homoeostasis are considered the most effective holistic approach to advance dermatological treatments based on systemic health-supportive and stimulating measures. Per evidence, an effective skin anti-ageing protection, wound healing and scarring quality - all strongly depend on the sustainable mitochondrial functionality and well-balanced homoeostasis. The latter can be objectively measured and, if necessary, restored in a systemic manner by pre- and rehabilitation algorithms tailored to individualised patient profiles. The entire spectrum of corresponding innovations in the area includes natural and systemic skin rejuvenation, aesthetic and reconstructive medicine, sustainable skin protection and targeted treatments of skin disorders. Contextually, mitochondria-centric dermatology is instrumental for advanced 3PM-guided approach which makes a good use of predictive multi-level diagnostics and targeted protection of skin against both - the health-to-disease transition and progression of relevant disorders. Cost-effective targeted protection and new treatment avenues focused on sustainable mitochondrial health and physiologic homoeostasis are proposed in the article including in-depth analysis of patient cases and exemplified 3PM-guided care with detailed mechanisms and corresponding expert recommendations presented.
Collapse
Affiliation(s)
- Olga Golubnitschaja
- Predictive, Preventive and Personalised (3P) Medicine, Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany
| | - Nafiseh Sargheini
- Max Planck Institute for Plant Breeding Research, Carl-Von-Linne-Weg 10, 50829 Cologne, Germany
| | - Janine Bastert
- Private Dermatological Clinic, Kirchheimer Str. 71, 70619 Stuttgart, Germany
| |
Collapse
|
7
|
Elmorsy EA, Elsisi HA, Alkhamiss AS, Alsoqih NS, Khodeir MM, Alsalloom AA, Almeman AA, Elghandour SR, Nadwa EH, Khalifa AK, Khaled BEA, Ramadan A, Kamal MM, Alsaeed TS, Alharbi MS, Abdel-Moneim AMH, Ellethy AT, Saber S. Activation of SIRT1 by SRT1720 alleviates dyslipidemia, improves insulin sensitivity and exhibits liver-protective effects in diabetic rats on a high-fat diet: New insights into the SIRT1/Nrf2/NFκB signaling pathway. Eur J Pharm Sci 2025; 206:107002. [PMID: 39778687 DOI: 10.1016/j.ejps.2025.107002] [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: 10/27/2024] [Revised: 11/29/2024] [Accepted: 01/05/2025] [Indexed: 01/11/2025]
Abstract
Insulin resistance and diabetes are associated with non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) conditions, which are distinguished by metabolic dysfunction, oxidative stress and inflammation. Sirtuin 1 (SIRT1), a NAD+-dependent deacetylase, is fundamental in regulating metabolic pathways, reducing inflammation, and improving antioxidant defenses. This is the first study to investigate the effects of SRT1720, a SIRT1 activator, in diabetic rats on a high-fat diet. SRT1720 significantly lowered fasting blood glucose and insulin levels and enhanced glucose tolerance and HOMA-IR and QUICKI scores, indicating increased insulin sensitivity. The treatment also reduced total cholesterol, triglycerides, and LDL levels, showing amelioration of dyslipidemia. Moreover, SRT1720 lowered markers of liver fibrosis, including TGF-β, TIMP-1, Col1a1, and hydroxyproline, and decreased inflammation by reducing NFκB activity and pro-inflammatory cytokines (TNF-α and IL-6). Furthermore, SRT1720 augmented Nrf2 activity and HO-1 levels. Consequently, the SRT1720's protective role improved liver function and histology and prolonged rats' survival. These functions were suppressed by the co-administration of the SIRT1 inhibitor EX527, confirming that the beneficial effects of SRT1720 are SIRT1-dependent. Correlation analyses uncovered that increased SIRT1 activity was strongly associated with decreased oxidative stress, inflammation, insulin resistance, and fibrosis markers. To conclude, our results find that SRT1720 represents a promising therapeutic strategy for managing Type 2 diabetes in NAFLD or NASH patients possibly through the modulation of the SIRT1/Nrf2/NFκB signaling pathwa. SRT1720 could potentially halt or reverse the progression of these conditions and associated complications and merits further investigations.
Collapse
Affiliation(s)
- Elsayed A Elmorsy
- Department of Pharmacology and Therapeutics, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia; Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt.
| | - Hossam A Elsisi
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraidah 51452, Saudi Arabia; Department of Clinical Pharmacology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt.
| | - Abdullah S Alkhamiss
- Department of Pathology, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia.
| | - Norah Suliman Alsoqih
- Department of Pediatrics, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia.
| | - Mostafa M Khodeir
- Department of Pathology, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia; Department of Pathology, Faculty of Medicine, Cairo University, Cairo 11562, Egypt.
| | - Abdulaziz A Alsalloom
- Department of Pathology, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia.
| | - Ahmad A Almeman
- Department of Pharmacology and Therapeutics, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia.
| | - Sahar R Elghandour
- Department of Anatomy and Histology, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia.
| | - Eman Hassan Nadwa
- Department of Pharmacology and Therapeutics, College of Medicine, Jouf University, Sakaka, Saudi Arabia; Department of Medical Pharmacology, Faculty of Medicine, Cairo University, Cairo 11562, Egypt.
| | - Amira Karam Khalifa
- Department of Medical Pharmacology, Faculty of Medicine, Cairo University, Cairo 11562, Egypt; Department of Medical Pharmacology, Nahda Faculty of Medicine, Beni Suef, 62521, Egypt.
| | - Bahaa Eldin Ali Khaled
- Department of Anatomy, College of Medicine, Jouf University, Sakaka, Saudi Arabia; Department of Anatomy and Embryology, Faculty of Medicine, Cairo University, Cairo 11562, Egypt.
| | - Asmaa Ramadan
- Department of Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Egypt.
| | - Manal M Kamal
- Department of Medical Physiology, Faculty of Medicine, Assiut University, Assiut, Egypt; Department of Physiology, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia.
| | - Thamir Saad Alsaeed
- Department of Biology and Immunology, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia.
| | - Mariam S Alharbi
- Department of Medicine, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia.
| | | | - Abousree T Ellethy
- Department of Basic Oral Sciences and Dental Education, Biochemistry Division, College of Dentistry, Qassim University, Buraidah 51452, Saudi Arabia.
| | - Sameh Saber
- Department of Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Egypt.
| |
Collapse
|
8
|
Sola-Sevilla N, Garmendia-Berges M, Mera-Delgado MC, Puerta E. Context-dependent role of sirtuin 2 in inflammation. Neural Regen Res 2025; 20:682-694. [PMID: 38886935 PMCID: PMC11433891 DOI: 10.4103/nrr.nrr-d-23-02063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/09/2024] [Accepted: 03/30/2024] [Indexed: 06/20/2024] Open
Abstract
Sirtuin 2 is a member of the sirtuin family nicotinamide adenine dinucleotide (NAD+)-dependent deacetylases, known for its regulatory role in different processes, including inflammation. In this context, sirtuin 2 has been involved in the modulation of key inflammatory signaling pathways and transcription factors by deacetylating specific targets, such as nuclear factor κB and nucleotide-binding oligomerization domain-leucine-rich-repeat and pyrin domain-containing protein 3 (NLRP3). However, whether sirtuin 2-mediated pathways induce a pro- or an anti-inflammatory response remains controversial. Sirtuin 2 has been implicated in promoting inflammation in conditions such as asthma and neurodegenerative diseases, suggesting that its inhibition in these conditions could be a potential therapeutic strategy. Conversely, arthritis and type 2 diabetes mellitus studies suggest that sirtuin 2 is essential at the peripheral level and, thus, its inhibition in these pathologies would not be recommended. Overall, the precise role of sirtuin 2 in inflammation appears to be context-dependent, and further investigation is needed to determine the specific molecular mechanisms and downstream targets through which sirtuin 2 influences inflammatory processes in various tissues and pathological conditions. The present review explores the involvement of sirtuin 2 in the inflammation associated with different pathologies to elucidate whether its pharmacological modulation could serve as an effective strategy for treating this prevalent symptom across various diseases.
Collapse
Affiliation(s)
- Noemí Sola-Sevilla
- Department of Pharmaceutical Sciences, Division of Pharmacology, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Maider Garmendia-Berges
- Department of Pharmaceutical Sciences, Division of Pharmacology, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
| | - MCarmen Mera-Delgado
- Department of Pharmaceutical Sciences, Division of Pharmacology, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
| | - Elena Puerta
- Department of Pharmaceutical Sciences, Division of Pharmacology, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| |
Collapse
|
9
|
Spagnuolo MC, Gottmann P, Sommer J, Borgmann SO, Strassburger K, Rathmann W, Zaharia OP, Trenkamp S, Wagner R, Icks A, Herder C, Roden M, Maalmi H. Three-protein signature is associated with baseline and persistently elevated or recurrent depressive symptoms in individuals with recent-onset diabetes. BMJ Open Diabetes Res Care 2025; 13:e004396. [PMID: 39965868 PMCID: PMC11836832 DOI: 10.1136/bmjdrc-2024-004396] [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: 06/11/2024] [Accepted: 01/05/2025] [Indexed: 02/20/2025] Open
Abstract
Depression is associated with diabetes, but the underlying causes remain unclear. To better understand depression in diabetes, this study investigated associations between 135 inflammatory and neurological protein biomarkers and depressive symptoms in individuals with diabetes.This cross-sectional study included 430 adults with a known diabetes duration <1 year from the German Diabetes Study (GDS), in whom biomarkers were measured in serum and depressive symptoms were evaluated at baseline and annually over 5 years using the Center for Epidemiological Studies Depression Scale (CES-D). Based on the information on depressive symptoms from the baseline and follow-up visits (n=305, ≥3 time points), we subdivided the sample into individuals with persistent or recurrent and transient or never depressive symptoms. We assessed the associations of each biomarker with baseline CES-D score (continuous) and persistent/recurrent depressive symptoms using multiple linear and logistic regression models, respectively.After adjustment for covariates, we identified a three-protein signature associated with baseline CES-D score and persistent/recurrent depressive symptoms. CUB domain-containing protein 1 (CDCP1) and NAD-dependent protein deacetylase sirtuin-2 (SIRT2) were positively associated with baseline (β 1.24 (95% CI 0.19 to 2.29); β 0.89 (95% CI 0.06 to 1.72)), respectively) and persistent/recurrent depressive symptoms (OR 1.58 (95% CI 1.08 to 2.31); OR 1.32 (95% CI 1.03 to 1.71), respectively), whereas leptin receptor (LEPR) was inversely associated with baseline (β -0.99 (95% CI -1.87 to -0.11)) and persistent/recurrent depressive symptoms (OR 0.70 (95% CI 0.49 to 0.99)). However, results were not significant after adjustment for multiple testing.In conclusion, the three-protein signature identified may provide insights into mechanisms underlying depressive symptoms in diabetes and might open new therapeutic avenues.The trial registration number of the study is NCT01055093.
Collapse
Affiliation(s)
- Maria C Spagnuolo
- Institute for Clinical Diabetology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), München, Germany
| | - Pascal Gottmann
- German Center for Diabetes Research (DZD), München, Germany
- Department of Experimental Diabetology, German Institute of Human Nutrition, Potsdam, Germany
| | - Jana Sommer
- German Center for Diabetes Research (DZD), München, Germany
- Institute for Health Services Research and Health Economics, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Sandra Olivia Borgmann
- German Center for Diabetes Research (DZD), München, Germany
- Institute for Health Services Research and Health Economics, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute for Health Services Research and Health Economics, Centre for Health and Society, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Klaus Strassburger
- German Center for Diabetes Research (DZD), München, Germany
- Institute for Biometrics and Epidemiology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Wolfgang Rathmann
- German Center for Diabetes Research (DZD), München, Germany
- Institute for Biometrics and Epidemiology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Oana Patricia Zaharia
- Institute for Clinical Diabetology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), München, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Sandra Trenkamp
- Institute for Clinical Diabetology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), München, Germany
| | - Robert Wagner
- Institute for Clinical Diabetology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), München, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Andrea Icks
- German Center for Diabetes Research (DZD), München, Germany
- Institute for Health Services Research and Health Economics, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute for Health Services Research and Health Economics, Centre for Health and Society, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Christian Herder
- Institute for Clinical Diabetology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), München, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), München, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Haifa Maalmi
- Institute for Clinical Diabetology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), München, Germany
| |
Collapse
|
10
|
Steyn SF. An Updated Bio-Behavioral Profile of the Flinders Sensitive Line Rat: Reviewing the Findings of the Past Decade. Pharmacol Res Perspect 2025; 13:e70058. [PMID: 39786312 PMCID: PMC11717001 DOI: 10.1002/prp2.70058] [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: 10/04/2024] [Revised: 12/09/2024] [Accepted: 12/22/2024] [Indexed: 01/12/2025] Open
Abstract
The Flinders sensitive line (FSL) rat is an accepted rodent model for depression that presents with strong face, construct, and predictive validity, thereby making it suitable to investigate novel antidepressant mechanisms. Despite the translatability of this model, available literature on this model has not been reviewed for more than ten years. The PubMed, ScienceDirect and Web of Science databases were searched for relevant articles between 2013 and 2024, with keywords relating to the Flinders line rat, and all findings relevant to treatment naïve animals, included. Following screening, 77 studies were included and used to create behavioral reference standards and calculate FSL favor ratios for the various behavioral parameters. The GRADE and SYRCLE risk of bias tools were used to scale the quality of these studies. Based on these results, FSL rats display reliable and reproducible depressive-like behavior in the forced swim test, together with hyperlocomotor activity across various behavioral tests. Despite reports of increased anhedonia, anxiety-like behavior, and cognitive dysfunction, the reviewed findings indicate that these parameters are comparable between strains. For the various neuro- and biological constructs, oxidative stress, energy production, and glutamatergic, noradrenergic and serotonergic neurotransmission received the most support for strain differences. Taken together, the FSL remains a reliable, popular, and translatable rodent model of depression, with strong face and construct validity. As for predictive validity, similar review approaches should be considered to establish whether the mentioned behavioral aspects and neurochemical constructs may be more sensitive (or resistant) to certain antidepressant strategies.
Collapse
Affiliation(s)
- Stephan F. Steyn
- Faculty of Health Sciences, Centre of Excellence for Pharmaceutical SciencesNorth‐West UniversityPotchefstroomSouth Africa
| |
Collapse
|
11
|
Ahmed R, Safa MR, Zahid ZI, Chowdhury MMI, Hasan AK, Mostaid MS, Reza HM. Association of SIRT1 rs3758391 Polymorphism With T2DM in Bangladeshi Population: Evidence From a Case-Control Study and Meta-Analysis. Health Sci Rep 2025; 8:e70495. [PMID: 39980831 PMCID: PMC11839489 DOI: 10.1002/hsr2.70495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 10/22/2024] [Accepted: 02/04/2025] [Indexed: 02/22/2025] Open
Abstract
Background and Aim Type 2 diabetes mellitus (T2DM) remains one of the major causes of morbidity and mortality worldwide, including Bangladesh. SIRT1, an NAD-dependent deacetylase, is involved in energy homeostasis and protects β-cells of the pancreas from oxidative stress. Single nucleotide polymorphisms (SNPs) in the SIRT1 gene have been found to be associated with T2DM in several populations, however, with conflicting results. The aim of this present case-control study, along with the meta-analysis, was to elucidate the association of rs3758391 polymorphism with the susceptibility to T2DM in Bangladeshi population. Methods 72 T2DM patients and 90 healthy controls were enrolled in our study and polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) was employed for genotyping the SNP. Odds ratio (OR) with 95% confidence interval (95% CI) was used to represent the association of SIRT1 polymorphism with T2DM. For the meta-analysis six studies were included and pooled odds ratio with 95% CI were calculated for six genetic models using the random effects model. Heterogeneity and publication bias was also calculated for each study. Results A significant association was found between rs3758391 polymorphism and increased risk of T2DM under codominant TT versus CC (OR = 3.88, 95% CI = 1.34-11.25, p = 0.012), recessive TT versus CC + CT (OR = 2.83, 95% CI = 1.12-7.09, p = 0.027) and allelic T versus C (OR = 1.67, 95% CI = 1.07-2.60, p = 0.024) genetic models. However, no significant association between rs3758391 and other biochemical and anthropometric parameters were found. Our meta-analysis showed no statistically significant association of this polymorphism. Conclusion We conclude that, polymorphism at rs3758391 of SIRT1 gene conferred an increased risk of T2DM in Bangladeshi population.
Collapse
Affiliation(s)
- Rezwana Ahmed
- Department of Pharmaceutical SciencesNorth South UniversityDhakaBangladesh
| | | | | | - Md. Mazharul Islam Chowdhury
- Department of Pharmaceutical SciencesNorth South UniversityDhakaBangladesh
- Appalachian College of PharmacyOakwoodGeorgiaUSA
| | | | - Md. Shaki Mostaid
- Department of Pharmaceutical SciencesNorth South UniversityDhakaBangladesh
| | - Hasan Mahmud Reza
- Department of Pharmaceutical SciencesNorth South UniversityDhakaBangladesh
| |
Collapse
|
12
|
Zgutka K, Tkacz M, Grabowska M, Mikołajek-Bedner W, Tarnowski M. Sirtuins and Their Implications in the Physiopathology of Gestational Diabetes Mellitus. Pharmaceuticals (Basel) 2025; 18:41. [PMID: 39861104 PMCID: PMC11768332 DOI: 10.3390/ph18010041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 12/23/2024] [Accepted: 12/27/2024] [Indexed: 01/27/2025] Open
Abstract
Gestational diabetes mellitus (GDM) imposes serious short- and long-term health problems for the mother and her child. An effective therapeutic that can reduce the incidence of GDM and improve long-term outcomes is a major research priority and is very important for public health. Unfortunately, despite numerous studies, the molecular mechanisms underlying GDM are not fully defined and require further study. Chronic low-grade inflammation, oxidative stress, and insulin resistance are central features of pregnancies complicated by GDM. There is evidence of the involvement of sirtuins, which are NAD+-dependent histone deacetylases, in energy metabolism and inflammation. Taking these facts into consideration, the role of sirtuins in the pathomechanism of GDM will be discussed.
Collapse
Affiliation(s)
- Katarzyna Zgutka
- Department of Physiology in Health Sciences, Faculty of Health Sciences, Pomeranian Medical University, 70-210 Szczecin, Poland
| | - Marta Tkacz
- Department of Physiology in Health Sciences, Faculty of Health Sciences, Pomeranian Medical University, 70-210 Szczecin, Poland
| | - Marta Grabowska
- Department of Histology and Developmental Biology, Faculty of Health Sciences, Pomeranian Medical University, 71-210 Szczecin, Poland
| | - Wioletta Mikołajek-Bedner
- Department of Obstetrics and Gynecology, Pomeranian Medical University in Szczecin, 70-204 Szczecin, Poland
| | - Maciej Tarnowski
- Department of Physiology in Health Sciences, Faculty of Health Sciences, Pomeranian Medical University, 70-210 Szczecin, Poland
| |
Collapse
|
13
|
Szkudelski T, Szkudelska K. The relevance of the heme oxygenase system in alleviating diabetes-related hormonal and metabolic disorders. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167552. [PMID: 39490940 DOI: 10.1016/j.bbadis.2024.167552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 11/05/2024]
Abstract
Heme oxygenase (HO) is an enzyme that catalyzes heme degradation. HO dysfunction is linked to various pathological conditions, including diabetes. Results of animal studies indicate that HO expression and activity are downregulated in experimentally induced diabetes. This is associated with severe hormonal and metabolic disturbances. However, these pathological changes have been shown to be reversed by therapy with HO activators. In animals with experimentally induced diabetes, HO was upregulated by genetic manipulation or by pharmacological activators such as hemin and cobalt protoporphyrin. Induction of HO alleviated elevated blood glucose levels and improved insulin action, among other effects. This effect resulted from beneficial changes in the main insulin-sensitive tissues, i.e., the skeletal muscle, the liver, and the adipose tissue. The action of HO activators was due to positive alterations in pivotal signaling molecules and regulatory enzymes. Furthermore, diabetes-related oxidative and inflammatory stress was reduced due to HO induction. HO upregulation was effective in various animal models of type 1 and type 2 diabetes. These data suggest the possibility of testing HO activators as a potential tool for alleviating hormonal and metabolic disorders in people with diabetes.
Collapse
Affiliation(s)
- Tomasz Szkudelski
- Department of Animal Physiology, Biochemistry and Biostructure, Poznan University of Life Sciences, Wolynska 35, 60-637 Poznan, Poland.
| | - Katarzyna Szkudelska
- Department of Animal Physiology, Biochemistry and Biostructure, Poznan University of Life Sciences, Wolynska 35, 60-637 Poznan, Poland.
| |
Collapse
|
14
|
Zhang Z, Ma C, Gao X, Wang C, Li Y, Yang C, Ma E, Cheng M. Design, synthesis, and biological evaluation of novel 3-naphthylthiophene derivatives as potent SIRT2 inhibitors for the treatment of myocardial fibrosis. Bioorg Chem 2025; 154:108033. [PMID: 39672075 DOI: 10.1016/j.bioorg.2024.108033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 11/26/2024] [Accepted: 12/03/2024] [Indexed: 12/15/2024]
Abstract
SIRT2 (sirtuin2) is a NAD+-dependent deacetylase implicated in fibrosis and inflammation of the liver, kidney, and heart. In this study, we designed and synthesized a series of 3-naphthylthiophene derivatives and evaluated their inhibitory activity against the SIRT2 enzyme. Among them, Z18 demonstrated outstanding SIRT2 inhibitory activity and selectivity. It significantly inhibited both the proliferation of cardiac fibroblasts (CFs) and the activity and expression of SIRT2 in CFs. Moreover, compound Z18 effectively suppressed TGF-β1-induced increases in α-SMA and CoL-1A1 protein expression, as well as hydroxyproline levels. Pharmacological mechanism tests demonstrated that Z18 inhibited SIRT2, thereby suppressing the TGF-β1-induced autocrine production of TGF-β1 and the phosphorylation of Smad2/3 in CFs. In MTT assays, Z18 exhibited a significant inhibitory effect on the proliferation of CFs induced by TGF-β1. In vivo, Z18 effectively ameliorated TAC- and ISO-induced declines in cardiac function, histopathological morphological changes, and collagen deposition. It also inhibited SIRT2 activity and reduced the expression of α-SMA and p-Smad2/3. In hepatorenal toxicity assays, Z18 exhibited an excellent safety profile. These results support the further development of the selective SIRT2 inhibitor Z18 as a potential lead compound for the treatment of myocardial fibrosis.
Collapse
Affiliation(s)
- Zhuo Zhang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, People's Republic of China
| | - Chao Ma
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, People's Republic of China
| | - Xiong Gao
- Department of Pharmacology, School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, People's Republic of China
| | - Chuncheng Wang
- Department of Pharmacology, School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, People's Republic of China
| | - Yanchun Li
- Department of Pharmacology, School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, People's Republic of China
| | - Chen Yang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, People's Republic of China
| | - Enlong Ma
- Department of Pharmacology, School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, People's Republic of China.
| | - Maosheng Cheng
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, People's Republic of China.
| |
Collapse
|
15
|
Hajra D, Rajmani RS, Chaudhary AD, Gupta SK, Chakravortty D. Salmonella-induced SIRT1 and SIRT3 are crucial for maintaining the metabolic switch in bacteria and host for successful pathogenesis. eLife 2024; 13:RP93125. [PMID: 39693143 PMCID: PMC11655064 DOI: 10.7554/elife.93125] [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] [Indexed: 12/19/2024] Open
Abstract
Sirtuins are the major players in host immunometabolic regulation. However, the role of sirtuins in the modulation of the immune metabolism pertaining to salmonellosis is largely unknown. Here, our investigation focussed on the role of two important sirtuins, SIRT1 and SIRT3, shedding light on their impact on intracellular Salmonella's metabolic switch and pathogenesis establishment. Our study indicated the ability of the live Salmonella Typhimurium to differentially regulate the levels of SIRT1 and SIRT3 for maintaining the high glycolytic metabolism and low fatty acid metabolism in Salmonella. Perturbing SIRT1 or SIRT3 through knockdown or inhibition resulted in a remarkable shift in the host metabolism to low fatty acid oxidation and high glycolysis. This switch led to decreased proliferation of Salmonella in the macrophages. Further, Salmonella-induced higher levels of SIRT1 and SIRT3 led to a skewed polarization state of the macrophages from a pro-inflammatory M1 state toward an immunosuppressive M2, making it more conducive for the intracellular life of Salmonella. Alongside, governing immunological functions by modulating p65 NF-κB acetylation, SIRT1, and SIRT3 also skew Salmonella-induced host metabolic switch by regulating the acetylation status of HIF-1α and PDHA1. Interestingly, though knockdown of SIRT1/3 attenuated Salmonella proliferation in macrophages, in in vivo mice model of infection, inhibition or knockdown of SIRT1/3 led to more dissemination and higher organ burden, which can be attributed to enhanced ROS and IL-6 production. Our study hence reports for the first time that Salmonella modulates SIRT1/3 levels to maintain its own metabolism for successful pathogenesis.
Collapse
Grants
- SPM-07/079(0293)/2019-EMR-I (CSIR Shyama Prasad Mukherjee Fellowship) Council of Scientific and Industrial Research, India
- DAE00195 Department of Atomic Energy, Government of India
- DBT-IISC Department of Biotechnology, Ministry of Science & Technology, India
- BT/RLF/re-entry/14/2019 Department of Biotechnology, Ministry of Science and Technology, India
- Department of Biotechnology, Ministry of Science & Technology, India
- Indian Council of Medical Research
- Department of Science & Technology, Ministry of Science and Technology, India
Collapse
Affiliation(s)
- Dipasree Hajra
- Department of Microbiology and Cell Biology, Indian Institute of ScienceBangaloreIndia
| | - Raju S Rajmani
- Centre of Infectious Disease Research, Indian Institute of ScienceBangaloreIndia
| | - Ayushi Devendrasingh Chaudhary
- Pharmacology Division, CSIR-Central Drug Research InstituteLucknowIndia
- Academy of Scientific and Innovative Research (AcSIR)GhaziabadIndia
| | - Shashi Kumar Gupta
- Pharmacology Division, CSIR-Central Drug Research InstituteLucknowIndia
- Academy of Scientific and Innovative Research (AcSIR)GhaziabadIndia
| | - Dipshikha Chakravortty
- Department of Microbiology and Cell Biology, Indian Institute of ScienceBangaloreIndia
- Adjunct Faculty, School of Biology, Indian Institute of Science Education and ResearchThiruvananthapuramIndia
| |
Collapse
|
16
|
Karuga FF, Kaczmarski P, Sochal M, Szmyd B, Białasiewicz P, Gabryelska A. Cross-Sectional Analysis of Hypoxia-Regulated miRNA-181a, miRNA-199a, HIF-1α, and SIRT1 in the Development of Type 2 Diabetes in Patients with Obstructive Sleep Apnea-Preliminary Study. J Clin Med 2024; 13:7644. [PMID: 39768567 PMCID: PMC11728235 DOI: 10.3390/jcm13247644] [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: 10/29/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 01/16/2025] Open
Abstract
Introduction: Obstructive sleep apnea (OSA) is recognized as an independent risk factor for diabetes mellitus type 2 (T2DM) development, which is twice as common in patients with OSA compared to non-OSA patients. Objectives: This study aimed to investigate changes in oxygen metabolism and their role in T2DM development among OSA patients through epigenetic processes via miRNA-181a, miRNA-199a, and enzymatic processes via SIRT1 and HIF-1α. Methods: Based on polysomnography, apnea-hypopnea index and the presence of T2DM patients were divided into three groups: control group (n = 17), OSA group (n = 11), OSA&T2DM (n = 20) group. Total RNA was extracted from the buffy coat. Moreover, HOMA-IR (Homeostatic Model Assessment for Insulin Resistance) was counted. Results: Morning miRNA-181a expression was significantly higher in the OSA&T2DM group than in the control group: 67.618 vs. 32.685 (p = 0.036). Evening miRNA-199a expression was significantly higher in the OSA group than in the control group: 5.043 vs. 2.081 (p = 0.042), while its morning expression was significantly higher in the OSA&T2DM group when compared to the control: 4.065 vs. 1.605 (p = 0.036). MiRNA-181a evening expression revealed a negative correlation with the SIRT1 evening and morning expressions (R = -0.367, p = 0.010 and R = -0.405, p = 0.004, respectively). Moreover, morning miRNA-181a was positively correlated with HOMA-IR (R = 0.321, p = 0.034). MiRNA-199a evening expression presented a moderate positive correlation with the SIRT1 morning expressions (R = 0.48, p < 0.001) and HOMA-IR (R = 0.35, p = 0.02). Conclusions: Patients suffering from OSA and T2DM had an increased expression of miRNA-181a. Moreover, a negative correlation between miRNA-181a and SIRT1 expression was observed, while a correlation between miRNA-181a and insulin resistance was positive. This phenomenon might suggest a possible epigenetic pathway for an increased incidence of T2DM in OSA patients however further research is needed.
Collapse
Affiliation(s)
- Filip Franciszek Karuga
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 90-419 Lodz, Poland; (P.K.); (M.S.); (P.B.)
| | - Piotr Kaczmarski
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 90-419 Lodz, Poland; (P.K.); (M.S.); (P.B.)
| | - Marcin Sochal
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 90-419 Lodz, Poland; (P.K.); (M.S.); (P.B.)
| | - Bartosz Szmyd
- Department of Neurosurgery and Neuro-Oncology, Barlicki University Hospital, Medical University of Lodz, 90-419 Lodz, Poland;
- Department of Pediatrics, Oncology, and Hematology, Medical University of Lodz, 90-419 Lodz, Poland
| | - Piotr Białasiewicz
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 90-419 Lodz, Poland; (P.K.); (M.S.); (P.B.)
| | - Agata Gabryelska
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 90-419 Lodz, Poland; (P.K.); (M.S.); (P.B.)
| |
Collapse
|
17
|
Bo S, Dan M, Li W, Chen C. The regulatory mechanism of natural polysaccharides in type 2 diabetes mellitus treatment. Drug Discov Today 2024; 29:104182. [PMID: 39284523 DOI: 10.1016/j.drudis.2024.104182] [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: 04/11/2024] [Revised: 08/25/2024] [Accepted: 09/11/2024] [Indexed: 09/27/2024]
Abstract
Diabetes is a complex, multifactorial disease that is caused by a pathological combination of insulin resistance and pancreatic islet dysfunction. Polysaccharides are extensively dispersed in nature and have a very complicated structure with various biological properties. Natural polysaccharides have potentially extraordinary beneficial health effects on managing metabolic diseases such as diabetes, obesity and cardiovascular disease. Thus, a systematic review of the latest research into and possible regulatory mechanisms of natural polysaccharides for type 2 diabetes mellitus treatment is of great significance for a better understanding of their pharmaceutical value. We discuss the regulatory mechanisms of natural polysaccharides for the treatment of diabetes, and especially their role in reshaping dysfunctional gut microbiota. Natural polysaccharides could be developed as new and safe antidiabetic drugs, and detailed mechanistic studies could further clarify the molecular targets of polysaccharides in the treatment of diabetes.
Collapse
Affiliation(s)
- Surina Bo
- College of Pharmacy, Inner Mongolia Medical University, Inner Mongolia Jinshan Development Zone, Hohhot, Inner Mongolian Province 010110, China; School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Mu Dan
- College of Pharmacy, Inner Mongolia Medical University, Inner Mongolia Jinshan Development Zone, Hohhot, Inner Mongolian Province 010110, China
| | - Wei Li
- Jilin Agricultural University, Changchun, Jilin Province 130118, China
| | - Chen Chen
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia.
| |
Collapse
|
18
|
Mobasher MA, Shabana MA, Germoush MO, Abuzinadah NY, Abd-Elhameed A, Baioumy SA, ElKot MA, Esawy MM. LncRNA LYPLAL1, miR-204-5p, and SIRT1: novel signatures for risk assessment of diabetic macrovascular complications. Sci Rep 2024; 14:24154. [PMID: 39406930 PMCID: PMC11480381 DOI: 10.1038/s41598-024-75543-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
Long-term, uncontrolled diabetes mellitus can lead to micro- and macrovascular problems. The protective function of lncRNA LYPLAL1 is to reduce endothelium cell inflammation by upregulating sirtuin 1 (SIRT1) and reducing microRNA (miR)-204-5p. This work attempted to examine the lncRNA LYPLAL1/miR-204-5p/SIRT1 molecules as diagnostic biomarkers for diabetic MVC and to assess their clinical correlations. The study enrolled 32 controls, 32 patients with diabetes alone, and 32 patients with diabetic MVC. RT-qPCR, or quantitative real-time PCR, was utilized to determine the expression levels of lncRNA and miR. SIRT1 was measured by ELISA. When comparing cases with MVC to those without MVC, the lncRNA LYPLAL1 and SIRT1 values were significantly lower. Conversely, patients with MVC had significantly higher miR-204-5p levels than those without MVC. The LncRNA LYPLAL1 performed best in terms of detecting MVC. It attained 90.6% specificity and 96.9% sensitivity. A combination of three markers (lncRNA LYPLAL1, miR-204-5p, and SIRT1) yielded the best accuracy at 98.4%. LYPLAL1 expression appeared to be an independent MVC predictor. Adjusted OR for LYPLAL1 expression was 405 (95% CI: 1.4-1200) (p = 0.039). When we compared cases with MVC to those without MVC, the lncRNA LYPLAL1 and SIRT1 values were significantly lower. Patients with MVC had significantly higher miR-204-5p levels than those without MVC. LYPLAL1 LncRNA demonstrated the best performance characteristics. LncRNA LYPLAL1 expression is an independent predictor of MVC.
Collapse
Affiliation(s)
- Maysa A Mobasher
- Department of Pathology, Biochemistry Division, College of Medicine, Jouf University, 72388, Sakaka, Saudi Arabia.
| | - Marwa A Shabana
- Clinical Pathology Department, Faculty of Human Medicine, Zagazig University, Zagazig, Egypt
| | - Mousa O Germoush
- Biology Department, College of Science, Jouf University, Sakakah, Saudi Arabia
| | - Najlaa Yousef Abuzinadah
- Department of biological science, College of Science, University of Jeddah, 23714, Jeddah, Saudi Arabia
| | - Amir Abd-Elhameed
- Internal Medicine Department, Faculty of Human Medicine, Zagazig University, Zagazig, Egypt
| | - Shereen A Baioumy
- Microbiology and Immunology Department, Faculty of Human Medicine, Zagazig University, Zagazig, Egypt
| | - Moataz A ElKot
- Cardiology Department, Faculty of Human Medicine, Zagazig University, Zagazig, Egypt
| | - Marwa M Esawy
- Clinical Pathology Department, Faculty of Human Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
19
|
Li X, Wang J, Zhang M, Li X, Fan Y, Zhou X, Sun Y, Qiu Z. Biological aging mediates the associations of metabolic score for insulin resistance with all-cause and cardiovascular disease mortality among US adults: A nationwide cohort study. Diabetes Obes Metab 2024; 26:3552-3564. [PMID: 38853301 DOI: 10.1111/dom.15694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/11/2024] [Accepted: 05/20/2024] [Indexed: 06/11/2024]
Abstract
AIM To investigate the associations of metabolic score for insulin resistance (METS-IR) with all-cause and cardiovascular disease (CVD)-specific mortality and the potential mediating role of biological ageing. METHODS A cohort of 19 204 participants from the National Health and Nutrition Examination Survey (NHANES) 1999-2018 was recruited for this study. Cox regression models, restricted cubic splines, and Kaplan-Meier survival curves were used to determine the relationships of METS-IR with all-cause and CVD-specific mortality. Mediation analyses were performed to explore the possible intermediary role of biological ageing markers, including phenotypic age (PhenoAge) and biological age (BioAge). RESULTS During a median follow-up of 9.17 years, we observed 2818 deaths, of which 875 were CVD-specific. Multivariable Cox regression showed that the highest METS-IR level (Q4) was associated with increased all-cause (hazard ratio [HR] 1.38, 95% confidence interval [CI] 1.14-1.67) and CVD mortality (HR 1.52, 95% CI 1.10-2.12) compared with the Q1 level. Restricted cubic splines showed a nonlinear relationship between METS-IR and all-cause mortality. Only METS-IR above the threshold (41.02 μg/L) was positively correlated with all-cause death. METS-IR had a linear positive relationship with CVD mortality. In mediation analyses, we found that PhenoAge mediated 51.32% (p < 0.001) and 41.77% (p < 0.001) of the association between METS-IR and all-cause and CVD-specific mortality, respectively. For BioAge, the mediating proportions of PhenoAge were 21.33% (p < 0.001) and 15.88% (p < 0.001), respectively. CONCLUSIONS This study highlights the detrimental effects of insulin resistance, as measured by METS-IR, on all-cause and CVD mortality. Moreover, it underscores the role of biological ageing in mediating these associations, emphasizing the need for interventions targeting both insulin resistance and ageing processes to mitigate mortality risks in metabolic disorders.
Collapse
Affiliation(s)
- Xiaoxuan Li
- Department of Oncology, Key Laboratory of Cancer Molecular and Translational Research, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jia Wang
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Mengqi Zhang
- Department of Oncology, Key Laboratory of Cancer Molecular and Translational Research, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiangjun Li
- Breast Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yuchen Fan
- Department of Medicine, Qingdao University, Qingdao, China
| | - Xinbei Zhou
- Department of Critical Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yuxin Sun
- Department of Oncology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenkang Qiu
- Interventional Medical Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
20
|
Xu Z, Huang J, Wen M, Zhang X, Lyu D, Li S, Xiao H, Li M, Shen C, Huang H. Gentiopicroside ameliorates glucose and lipid metabolism in T2DM via targeting FGFR1. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155780. [PMID: 38885580 DOI: 10.1016/j.phymed.2024.155780] [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: 12/22/2023] [Revised: 05/07/2024] [Accepted: 05/26/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND The suppression of the fibroblast growth factor 21/fibroblast growth factor receptor 1 (FGF21/FGFR1) signaling pathway is considered as a vital factor in the type 2 diabetes mellitus (T2DM) progression. Our previous study showed that gentiopicroside (GPS), the main active compound present in Gentiana macrophylla Pall., has the capacity to control disorders related to glucose and lipid metabolism in individuals with T2DM. Nevertheless, the specific mechanism remains unclear. PURPOSE In light of the fact that the PharmMapper database suggests FGFR1 as the target of GPS, our investigation aims to determine if GPS can enhance glucose and lipid metabolism issues in T2DM by modulating the FGF21/FGFR1 signaling pathway. METHODS In this study, we used palmitic acid (PA)-induced HepG2 cells and db/db mice to investigate the function and mechanism of GPS in the FGF21/FGFR1 signaling pathway. To examine the interaction between GPS and FGFR1, researchers performed Cellular Thermal Shift Assay (CETSA) and Surface Plasmon Resonance (SPR) analysis. RESULTS The results suggest that GPS activates the traditional metabolic pathways, including PI3K/AKT and AMPK, which are the subsequent stages of the FGF21/FGFR1 pathway. This activation leads to the enhancement of glucose and lipid metabolism issues in PA-treated HepG2 cells and db/db mice. Furthermore, the depletion of FGFR1 has been noticed to oppose the stimulation of PI3K/AKT and AMPK pathways by GPS in HepG2 cells subjected to PA. Notability, our research affirms that GPS binds directly to FGFR1, hindering the ubiquitinated degradation of FGFR1 by neural precursor cells expressing developmentally decreased protein 4 (NEDD4) and ultimately promoting FGF21 signal transduction. CONCLUSION This study demonstrates that GPS targeting FGFR1 activates the PI3K/AKT and AMPK pathways, which is an important mechanism for its treatment of T2DM.
Collapse
Affiliation(s)
- Zhanchi Xu
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratoty for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; Guangzhou Hospital of Integrated Traditional and Western Medicine, Guangzhou 510801, China
| | - Jucun Huang
- Hubei NO.3 People's Hospital of Jianghan University, Wuhan 430033, China
| | - Min Wen
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratoty for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Xuting Zhang
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratoty for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Dongxin Lyu
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratoty for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Shanshan Li
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratoty for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Haiming Xiao
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratoty for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Min Li
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratoty for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| | - Cuangpeng Shen
- Department of Endocrinology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| | - Heqing Huang
- Guangzhou Hospital of Integrated Traditional and Western Medicine, Guangzhou 510801, China.
| |
Collapse
|
21
|
Song J, Wang L, Wang L, Guo X, He Q, Cui C, Hu H, Zang N, Yang M, Yan F, Liang K, Wang C, Liu F, Sun Y, Sun Z, Lai H, Hou X, Chen L. Mesenchymal stromal cells ameliorate mitochondrial dysfunction in α cells and hyperglucagonemia in type 2 diabetes via SIRT1/FoxO3a signaling. Stem Cells Transl Med 2024; 13:776-790. [PMID: 38864709 PMCID: PMC11328933 DOI: 10.1093/stcltm/szae038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 04/24/2024] [Indexed: 06/13/2024] Open
Abstract
Dysregulation of α cells results in hyperglycemia and hyperglucagonemia in type 2 diabetes mellitus (T2DM). Mesenchymal stromal cell (MSC)-based therapy increases oxygen consumption of islets and enhances insulin secretion. However, the underlying mechanism for the protective role of MSCs in α-cell mitochondrial dysfunction remains unclear. Here, human umbilical cord MSCs (hucMSCs) were used to treat 2 kinds of T2DM mice and αTC1-6 cells to explore the role of hucMSCs in improving α-cell mitochondrial dysfunction and hyperglucagonemia. Plasma and supernatant glucagon were detected by enzyme-linked immunosorbent assay (ELISA). Mitochondrial function of α cells was assessed by the Seahorse Analyzer. To investigate the underlying mechanisms, Sirtuin 1 (SIRT1), Forkhead box O3a (FoxO3a), glucose transporter type1 (GLUT1), and glucokinase (GCK) were assessed by Western blotting analysis. In vivo, hucMSC infusion improved glucose and insulin tolerance, as well as hyperglycemia and hyperglucagonemia in T2DM mice. Meanwhile, hucMSC intervention rescued the islet structure and decreased α- to β-cell ratio. Glucagon secretion from αTC1-6 cells was consistently inhibited by hucMSCs in vitro. Meanwhile, hucMSC treatment activated intracellular SIRT1/FoxO3a signaling, promoted glucose uptake and activation, alleviated mitochondrial dysfunction, and enhanced ATP production. However, transfection of SIRT1 small interfering RNA (siRNA) or the application of SIRT1 inhibitor EX-527 weakened the therapeutic effects of hucMSCs on mitochondrial function and glucagon secretion. Our observations indicate that hucMSCs mitigate mitochondrial dysfunction and glucagon hypersecretion of α cells in T2DM via SIRT1/FoxO3a signaling, which provides novel evidence demonstrating the potential for hucMSCs in treating T2DM.
Collapse
Affiliation(s)
- Jia Song
- Department of Endocrinology and Metabolism, Qilu Hospital of Shandong University, Jinan 250012, Shandong, People's Republic of China
| | - Lingshu Wang
- Department of Endocrinology and Metabolism, Qilu Hospital of Shandong University, Jinan 250012, Shandong, People's Republic of China
| | - Liming Wang
- Department of Endocrinology and Metabolism, Qilu Hospital of Shandong University, Jinan 250012, Shandong, People's Republic of China
| | - Xinghong Guo
- Department of Endocrinology and Metabolism, Qilu Hospital of Shandong University, Jinan 250012, Shandong, People's Republic of China
| | - Qin He
- Department of Endocrinology and Metabolism, Qilu Hospital of Shandong University, Jinan 250012, Shandong, People's Republic of China
| | - Chen Cui
- Department of Endocrinology and Metabolism, Qilu Hospital of Shandong University, Jinan 250012, Shandong, People's Republic of China
| | - Huiqing Hu
- Department of Endocrinology and Metabolism, Qilu Hospital of Shandong University, Jinan 250012, Shandong, People's Republic of China
| | - Nan Zang
- Department of Endocrinology and Metabolism, Qilu Hospital of Shandong University, Jinan 250012, Shandong, People's Republic of China
| | - Mengmeng Yang
- Department of Endocrinology and Metabolism, Qilu Hospital of Shandong University, Jinan 250012, Shandong, People's Republic of China
| | - Fei Yan
- Department of Endocrinology and Metabolism, Qilu Hospital of Shandong University, Jinan 250012, Shandong, People's Republic of China
| | - Kai Liang
- Department of Endocrinology and Metabolism, Qilu Hospital of Shandong University, Jinan 250012, Shandong, People's Republic of China
| | - Chuan Wang
- Department of Endocrinology and Metabolism, Qilu Hospital of Shandong University, Jinan 250012, Shandong, People's Republic of China
| | - Fuqiang Liu
- Department of Endocrinology and Metabolism, Qilu Hospital of Shandong University, Jinan 250012, Shandong, People's Republic of China
| | - Yujing Sun
- Department of Endocrinology and Metabolism, Qilu Hospital of Shandong University, Jinan 250012, Shandong, People's Republic of China
| | - Zheng Sun
- Department of Endocrinology and Metabolism, Qilu Hospital of Shandong University, Jinan 250012, Shandong, People's Republic of China
| | - Hong Lai
- Department of Endocrinology and Metabolism, Qilu Hospital of Shandong University, Jinan 250012, Shandong, People's Republic of China
- Institute of Endocrine and Metabolic Diseases of Shandong University, Jinan 250012, Shandong, People's Republic of China
- Key Laboratory of Endocrine and Metabolic Diseases, Shandong Province Medicine and Health, Jinan 250012, Shandong, People's Republic of China
- Jinan Clinical Research Center for Endocrine and Metabolic Disease, Jinan 250012, Shandong, People's Republic of China
| | - Xinguo Hou
- Department of Endocrinology and Metabolism, Qilu Hospital of Shandong University, Jinan 250012, Shandong, People's Republic of China
- Institute of Endocrine and Metabolic Diseases of Shandong University, Jinan 250012, Shandong, People's Republic of China
- Key Laboratory of Endocrine and Metabolic Diseases, Shandong Province Medicine and Health, Jinan 250012, Shandong, People's Republic of China
- Jinan Clinical Research Center for Endocrine and Metabolic Disease, Jinan 250012, Shandong, People's Republic of China
| | - Li Chen
- Department of Endocrinology and Metabolism, Qilu Hospital of Shandong University, Jinan 250012, Shandong, People's Republic of China
- Institute of Endocrine and Metabolic Diseases of Shandong University, Jinan 250012, Shandong, People's Republic of China
- Key Laboratory of Endocrine and Metabolic Diseases, Shandong Province Medicine and Health, Jinan 250012, Shandong, People's Republic of China
- Jinan Clinical Research Center for Endocrine and Metabolic Disease, Jinan 250012, Shandong, People's Republic of China
| |
Collapse
|
22
|
Yang L, Liu D, Jiang S, Li H, Chen L, Wu Y, Essien AE, Opoku M, Naranmandakh S, Liu S, Ru Q, Li Y. SIRT1 signaling pathways in sarcopenia: Novel mechanisms and potential therapeutic targets. Biomed Pharmacother 2024; 177:116917. [PMID: 38908209 DOI: 10.1016/j.biopha.2024.116917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/29/2024] [Accepted: 06/09/2024] [Indexed: 06/24/2024] Open
Abstract
Sarcopenia is an aging-related skeletal disease characterized by decreased muscle mass, strength, and physical function, severely affecting the quality of life (QoL) of the elderly population. Sirtuin 1 (SIRT1), as a nicotinamide adenine dinucleotide (NAD+)-dependent histone deacetylases, has been reported to participate in various aging-related signaling pathways and exert protective effect on many human diseases. SIRT1 functioned as an important role in the occurrence and progression of sarcopenia through regulating key pathways related to protein homeostasis, apoptosis, mitochondrial dysfunction, insulin resistance and autophagy in skeletal muscle, including SIRT1/Forkhead Box O (FoxO), AMP-activated protein kinase (AMPK)/SIRT1/nuclear factor κB (NF-κB), SIRT1/p53, AMPK/SIRT1/peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α), and SIRT1/live kinase B1 (LKB1)/AMPK pathways. However, the specific mechanisms of these processes have not been fully illuminated. Currently, several SIRT1-mediated interventions on sarcopenia have been preliminarily developed, such as SIRT1 activator polyphenolic compounds, exercising and calorie restriction. In this review, we summarized the predominant mechanisms of SIRT1 involved in sarcopenia and therapeutic modalities targeting the SIRT1 signaling pathways for the prevention and prognosis of sarcopenia.
Collapse
Affiliation(s)
- Luning Yang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Di Liu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Shide Jiang
- Department of Orthopedics, The Central Hospital of Yongzhou, Yongzhou 425000, China
| | - Hengzhen Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Lin Chen
- Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Yuxiang Wu
- Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Anko Elijah Essien
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Michael Opoku
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Shinen Naranmandakh
- Department of chemistry, School of Arts and Sciences, National University of Mongolia, Ulaanbaatar 14201, Mongolia
| | - ShuGuang Liu
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Qin Ru
- Department of Health and Physical Education, Jianghan University, Wuhan 430056, China.
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| |
Collapse
|
23
|
Wang L, Bai Y, Cao Z, Guo Z, Lian Y, Liu P, Zeng Y, Lyu W, Chen Q. Histone deacetylases and inhibitors in diabetes mellitus and its complications. Biomed Pharmacother 2024; 177:117010. [PMID: 38941890 DOI: 10.1016/j.biopha.2024.117010] [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: 04/11/2024] [Revised: 05/29/2024] [Accepted: 06/17/2024] [Indexed: 06/30/2024] Open
Abstract
Diabetes mellitus (DM) is a metabolic disorder characterized by hyperglycemia, with its prevalence linked to both genetic predisposition and environmental factors. Epigenetic modifications, particularly through histone deacetylases (HDACs), have been recognized for their significant influence on DM pathogenesis. This review focuses on the classification of HDACs, their role in DM and its complications, and the potential therapeutic applications of HDAC inhibitors. HDACs, which modulate gene expression without altering DNA sequences, are categorized into four classes with distinct functions and tissue specificity. HDAC inhibitors (HDACi) have shown efficacy in various diseases, including DM, by targeting these enzymes. The review highlights how HDACs regulate β-cell function, insulin sensitivity, and hepatic gluconeogenesis in DM, as well as their impact on diabetic cardiomyopathy, nephropathy, and retinopathy. Finally, we suggest that targeted histone modification is expected to become a key method for the treatment of diabetes and its complications. The study of HDACi offers insights into new treatment strategies for DM and its associated complications.
Collapse
Affiliation(s)
- Li Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610072, PR China; Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, PR China
| | - Yuning Bai
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, PR China
| | - Zhengmin Cao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, PR China
| | - Ziwei Guo
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, PR China
| | - Yanjie Lian
- Department of Cardiovascular Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, PR China
| | - Pan Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610072, PR China
| | - Yixian Zeng
- Department of Proctology, Beibei Hospital of Traditional Chinese Medicine, Chongqing 400799, PR China
| | - Wenliang Lyu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, PR China.
| | - Qiu Chen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610072, PR China.
| |
Collapse
|
24
|
Zhang M, Hu T, Ma T, Huang W, Wang Y. Epigenetics and environmental health. Front Med 2024; 18:571-596. [PMID: 38806988 DOI: 10.1007/s11684-023-1038-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 10/15/2023] [Indexed: 05/30/2024]
Abstract
Epigenetic modifications including DNA methylation, histone modifications, chromatin remodeling, and RNA modifications complicate gene regulation and heredity and profoundly impact various physiological and pathological processes. In recent years, accumulating evidence indicates that epigenetics is vulnerable to environmental changes and regulates the growth, development, and diseases of individuals by affecting chromatin activity and regulating gene expression. Environmental exposure or induced epigenetic changes can regulate the state of development and lead to developmental disorders, aging, cardiovascular disease, Alzheimer's disease, cancers, and so on. However, epigenetic modifications are reversible. The use of specific epigenetic inhibitors targeting epigenetic changes in response to environmental exposure is useful in disease therapy. Here, we provide an overview of the role of epigenetics in various diseases. Furthermore, we summarize the mechanism of epigenetic alterations induced by different environmental exposures, the influence of different environmental exposures, and the crosstalk between environmental variation epigenetics, and genes that are implicated in the body's health. However, the interaction of multiple factors and epigenetics in regulating the initiation and progression of various diseases complicates clinical treatments. We discuss some commonly used epigenetic drugs targeting epigenetic modifications and methods to prevent or relieve various diseases regulated by environmental exposure and epigenetics through diet.
Collapse
Affiliation(s)
- Min Zhang
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ting Hu
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tianyu Ma
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Wei Huang
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
| | - Yan Wang
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
25
|
de Paula AVL, Dykstra GM, da Rocha RB, Magalhães AT, da Silva BAK, Cardoso VS. The association of diabetic peripheral neuropathy with cardiac autonomic neuropathy in individuals with diabetes mellitus: A systematic review. J Diabetes Complications 2024; 38:108802. [PMID: 38971002 DOI: 10.1016/j.jdiacomp.2024.108802] [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: 03/25/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/08/2024]
Abstract
This systematic review aimed to explore the relationship between diabetic peripheral neuropathy (DPN) and cardiac autonomic neuropathy (CAN) in individuals with type 1 and 2 diabetes mellitus (DM). METHODS The systematic review follow the protocol registered in Prospero (CRD42020182899). Two authors independently searched the PubMed, Scopus, Embase, Cochrane, and Web of Science databases. Discrepancies were resolved by a third author. The review included observational studies investigating the relationship between CAN and DPN in individuals with DM. RESULTS Initially, out of 1165 studies, only 16 were selected, with 42.8 % involving volunteers with one type of diabetes, 14.3 % with both types of diabetes and 14.3 % not specify the type. The total number of volunteers was 2582, mostly with type 2 DM. It was analyzed that there is a relationship between CAN and DPN. It was observed that more severe levels of DPN are associated with worse outcomes in autonomic tests. Some studies suggested that the techniques for evaluating DPN might serve as risk factors for CAN. CONCLUSION The review presents a possible relationship between DPN and CAN, such as in their severity.
Collapse
Affiliation(s)
- Ana Vitoria Lima de Paula
- Programa de Pós-Graduação em Ciências Biomédicas, Universidade Federal do Delta do Parnaíba, Parnaíba, PI, Brazil
| | - Gabrielly Menin Dykstra
- Curso de Bacharelado em Fisioterapia, Universidade Federal do Delta do Parnaíba, Parnaíba, PI, Brazil
| | - Rebeca Barbosa da Rocha
- Programa de Pós Graduação em Biotecnologia, Universidade Federal do Delta do Parnaíba, Parnaíba, PI, Brazil
| | - Alessandra Tanuri Magalhães
- Curso de Bacharelado em Fisioterapia, Universidade Federal do Delta do Parnaíba, Parnaíba, PI, Brazil; Centro Integrado de Especialidades Médicas, Universidade Federal do Piauí (UFPI), Parnaíba, PI, Brazil
| | | | - Vinicius Saura Cardoso
- Programa de Pós-Graduação em Ciências Biomédicas, Universidade Federal do Delta do Parnaíba, Parnaíba, PI, Brazil; Centro Integrado de Especialidades Médicas, Universidade Federal do Piauí (UFPI), Parnaíba, PI, Brazil.
| |
Collapse
|
26
|
Sarma P, Kashyap B, Gurumayum S, Sarma S, Baruah P, Swargiary D, Saikia A, Deka RC, Borah JC. Antihyperglycemic Potential of Quercetin-3-glucoside Isolated from Leucaena leucocephala Seedpods via the SIRT1/AMPK/GLUT4 Signaling Cascade. ACS OMEGA 2024; 9:32429-32443. [PMID: 39100317 PMCID: PMC11292826 DOI: 10.1021/acsomega.3c09672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 05/16/2024] [Accepted: 05/28/2024] [Indexed: 08/06/2024]
Abstract
Leucaena leucocephala. (Lam.) de Wit, a traditional medicinal plant, has been reported among the ethnic communities of Mexico, Indonesia, China, and India for the treatment of diabetes, obesity, and related complications. This study investigates the antihyperglycemic activity of the plant and its isolated active compound quercetin-3-glucoside. Further, bioactivity guided marker assisted development of an enriched bioactive fraction toward enhancing insulin sensitization was carried out. The enriched fraction was also found to contain 397.96 mg/g of quercetin-3-glucoside along with three other marker compounds, which were also isolated and identified. Quercetin-3-glucoside, out of the four isolated marker compounds from the plant, showed the most significant bioactivity when tested in palmitate-induced insulin-resistant C2C12 myotubes. The compound also showed significant upregulation of sirtuin1 (SIRT1) followed by enhancement of insulin-dependent signaling molecules SIRT1/AMPK/PGC1-α and GLUT4 translocation. Molecular dynamics studies showed the compound having stable interactions with the SIRT1 protein. SIRT1 upregulation has been associated with enhanced insulin sensitivity in skeletal muscle, increasing the glucose uptake by muscle cells. The prepared enriched fraction also modulated the SIRT1/AMPK/GLUT4 pathway. The findings of the present study may find future application toward the development of botanical or phytopharmaceutical drugs from the traditionally important plant L. leucocephala against type II diabetes.
Collapse
Affiliation(s)
- Pranamika Sarma
- Chemical
Biology Laboratory 1, Institute of Advanced
Study in Science and Technology (IASST), Paschim Boragaon, Guwahati, Assam 781035, India
- Department
of Chemistry, Gauhati University, Gopinath Bordoloi Nagar, Jalukbari, Guwahati, Assam 781014, India
| | - Bhaswati Kashyap
- Chemical
Biology Laboratory 1, Institute of Advanced
Study in Science and Technology (IASST), Paschim Boragaon, Guwahati, Assam 781035, India
| | - Shalini Gurumayum
- Chemical
Biology Laboratory 1, Institute of Advanced
Study in Science and Technology (IASST), Paschim Boragaon, Guwahati, Assam 781035, India
| | - Srutishree Sarma
- Catalysis
and Molecular Modelling Laboratory, Department of Chemical Sciences, Tezpur University, Tezpur, Assam 784028, India
| | - Paran Baruah
- Chemical
Biology Laboratory 1, Institute of Advanced
Study in Science and Technology (IASST), Paschim Boragaon, Guwahati, Assam 781035, India
| | - Deepsikha Swargiary
- Chemical
Biology Laboratory 1, Institute of Advanced
Study in Science and Technology (IASST), Paschim Boragaon, Guwahati, Assam 781035, India
| | - Abhipsa Saikia
- Chemical
Biology Laboratory 1, Institute of Advanced
Study in Science and Technology (IASST), Paschim Boragaon, Guwahati, Assam 781035, India
| | - Ramesh Ch. Deka
- Catalysis
and Molecular Modelling Laboratory, Department of Chemical Sciences, Tezpur University, Tezpur, Assam 784028, India
| | - Jagat C Borah
- Chemical
Biology Laboratory 1, Institute of Advanced
Study in Science and Technology (IASST), Paschim Boragaon, Guwahati, Assam 781035, India
| |
Collapse
|
27
|
Wu S, Yi B, Dai Y, Liao X, Peng J, Li A. Correlation between polymorphisms of SIRT2 gene and renal injury in patients with type 2 diabetes mellitus. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2024; 49:1005-1014. [PMID: 39788488 PMCID: PMC11495987 DOI: 10.11817/j.issn.1672-7347.2024.240186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Indexed: 01/12/2025]
Abstract
OBJECTIVES Genetic factors play an important role in the pathogenesis of diabetic kidney disease (DKD). Studies have shown that SIRT2 gene polymorphism is associated with the pathogenesis of type 2 diabetes mellitus (T2DM), but its role in DKD remains unclear. This study aims to analyze the distribution of alleles and genotypes of SIRT2 gene in patients with T2DM, and investigate the association between SIRT2 genetic polymorphism and DKD susceptibility in T2DM patients, which may provide new ideas for the pathogenesis of DKD. METHODS A toal of 205 T2DM patients who receiving treatment in the Third Xiangya Hospital of Central South University were divided into a DKD group (n=100) and a DM group (n=105) according to the presence of kidney injury, and 100 healthy volunteers were selected as NC group. Clinical data of the subjects were collected and estimated glomerular filtration rate (eGFR) were calculated. Genomic DNA was extracted and the genotypes of single nucleotide polymorphism (SNP) loci (rs11879029, rs11879010, and rs2241703) were determined using Sanger chain termination method. The genotype/allele frequencies among the 3 groups were compared. Logistic regression was used to analyze the correlation between SNP locus genotype of SIRT2 gene and risk of DKD in T2DM patients. According to the genotypes of rs11879029/rs11879010, T2DM patients were divided into a GG1/GG2 group, a GA1/GA2 group, and an AA1/AA2 group, and the clinical data were compared. Linkage disequilibrium analysis and haplotype analysis were performed. RESULTS The genotype distribution and allele frequencies of the rs11879029 and rs11879010 loci in the DKD group were significantly different in comparison with the NC and DM groups (all P<0.05). For rs2241703, there were no differences in genotype and allele frequencies (all P>0.05). After correcting by age, gender, systolic blood pressure, duration of diabetes, glycosylated hemoglobin, and serum albumin, rs11879029 and rs11879010 genotype were associated with DKD susceptibility in T2DM patients. Carriers of rs11879029 genotype AA were 6.27 times more likely to have DKD than carriers of genotype GG. And carriers of rs11879010 genotype AA were 4.72 times more likely to have DKD than carriers of genotype GG. The eGFR levels in the AA1/AA2 groups were significantly lower than those in the GG1/GG2 groups (both P<0.05). Analysis of linkage disequilibrium showed complete linkage disequilibrium existed between SIRT2 rs11879029 and rs11879010, and the 2 SNPs (rs11879029 and rs11879010) were in strong linkage disequilibrium with rs2241703. Monotype GGG reduced the risk of DKD in T2DM patients (OR=0.53, 95% CI 0.35 to 0.81, P=0.003), while haplotype AAG increased the risk of DKD in patients (OR=1.80, 95% CI 1.16 to 2.80, P=0.008). CONCLUSIONS The genetic polymorphisms rs11879029 and rs11879010 of SIRT2 gene are potential contributors to the susceptibility of DKD in patients with T2DM, and allele A significantly increases the risk of DKD compared with allele G. The AA genotype might be a genetic risk factor for DKD.
Collapse
Affiliation(s)
- Su Wu
- Department of Nephrology, Third Xiangya Hospital, Central South University, Changsha 410013.
- Department of Nephrology, Fourth Hospital of Changsha, Changsha 410017, China.
| | - Bin Yi
- Department of Nephrology, Third Xiangya Hospital, Central South University, Changsha 410013.
| | - Yali Dai
- Department of Nephrology, Third Xiangya Hospital, Central South University, Changsha 410013
| | - Xiangyu Liao
- Department of Nephrology, Third Xiangya Hospital, Central South University, Changsha 410013
| | - Juan Peng
- Department of Nephrology, Third Xiangya Hospital, Central South University, Changsha 410013
| | - Aimei Li
- Department of Nephrology, Third Xiangya Hospital, Central South University, Changsha 410013.
| |
Collapse
|
28
|
Liu Y, Lyons CJ, Ayu C, O’Brien T. Enhancing endothelial colony-forming cells for treating diabetic vascular complications: challenges and clinical prospects. Front Endocrinol (Lausanne) 2024; 15:1396794. [PMID: 39076517 PMCID: PMC11284052 DOI: 10.3389/fendo.2024.1396794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 06/14/2024] [Indexed: 07/31/2024] Open
Abstract
Diabetes mellitus (DM) is a metabolic disease characterized by hyperglycemia, leading to various vascular complications. Accumulating evidence indicates that endothelial colony-forming cells (ECFCs) have attractive prospects for repairing and restoring blood vessels. Thus, ECFCs may be a novel therapeutic option for diabetic patients with vascular complications who require revascularization therapy. However, it has been reported that the function of ECFCs is impaired in DM, which poses challenges for the autologous transplantation of ECFCs. In this review, we summarize the molecular mechanisms that may be responsible for ECFC dysfunction and discuss potential strategies for improving the therapeutic efficacy of ECFCs derived from patients with DM. Finally, we discuss barriers to the use of ECFCs in human studies in light of the fact that there are no published reports using these cells in humans.
Collapse
Affiliation(s)
| | | | | | - Timothy O’Brien
- Regenerative Medicine Institute (REMEDI), Biomedical Sciences Building, University of Galway, Galway, Ireland
| |
Collapse
|
29
|
Młynarska E, Wasiak J, Gajewska A, Steć G, Jasińska J, Rysz J, Franczyk B. Exploring the Significance of Gut Microbiota in Diabetes Pathogenesis and Management-A Narrative Review. Nutrients 2024; 16:1938. [PMID: 38931292 PMCID: PMC11206785 DOI: 10.3390/nu16121938] [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: 05/17/2024] [Revised: 06/13/2024] [Accepted: 06/16/2024] [Indexed: 06/28/2024] Open
Abstract
Type 2 diabetes is a disease with significant health consequences for the individual. Currently, new mechanisms and therapeutic approaches that may affect this disease are being sought. One of them is the association of type 2 diabetes with microbiota. Through the enteric nervous system and the gut-microbiota axis, the microbiota affects the functioning of the body. It has been proven to have a real impact on influencing glucose and lipid metabolism and insulin sensitivity. With dysbiosis, there is increased bacterial translocation through the disrupted intestinal barrier and increased inflammation in the body. In diabetes, the microbiota's composition is altered with, for example, a more abundant class of Betaproteobacteria. The consequences of these disorders are linked to mechanisms involving short-chain fatty acids, branched-chain amino acids, and bacterial lipopolysaccharide, among others. Interventions focusing on the gut microbiota are gaining traction as a promising approach to diabetes management. Studies are currently being conducted on the effects of the supply of probiotics and prebiotics, as well as fecal microbiota transplantation, on the course of diabetes. Further research will allow us to fully develop our knowledge on the subject and possibly best treat and prevent type 2 diabetes.
Collapse
Affiliation(s)
- Ewelina Młynarska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jakub Wasiak
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Agata Gajewska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Greta Steć
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Joanna Jasińska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| |
Collapse
|
30
|
Sun A, Yang H, Li T, Luo J, Zhou L, Chen R, Han L, Lin Y. Molecular mechanisms, targets and clinical potential of berberine in regulating metabolism: a review focussing on databases and molecular docking studies. Front Pharmacol 2024; 15:1368950. [PMID: 38957396 PMCID: PMC11217548 DOI: 10.3389/fphar.2024.1368950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 05/29/2024] [Indexed: 07/04/2024] Open
Abstract
Background: Metabolic imbalance is the common basis of many diseases. As natural isoquinoline alkaloid, berberine (BBR) has shown great promise in regulating glucose and lipids metabolism and treating metabolic disorders. However, the related mechanism still lacks systematic research. Aim: To discuss the role of BBR in the whole body's systemic metabolic regulation and further explore its therapeutic potential and targets. Method: Based on animal and cell experiments, the mechanism of BBR regulating systemic metabolic processes is reviewed. Potential metabolism-related targets were summarized using Therapeutic Target Database (TTD), DrugBank, GeneCards, and cutting-edge literature. Molecular modeling was applied to explore BBR binding to the potential targets. Results: BBR regulates the whole-body metabolic response including digestive, circulatory, immune, endocrine, and motor systems through adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR), sirtuin (SIRT)1/forkhead box O (FOXO)1/sterol regulatory element-binding protein (SREBP)2, nuclear factor erythroid 2-related factor (Nrf) 2/heme oxygenase (HO)-1, and other signaling pathways. Through these reactions, BBR exerts hypoglycemic, lipid-regulating, anti-inflammatory, anti-oxidation, and immune regulation. Molecular docking results showed that BBR could regulate metabolism targeting FOXO3, Nrf2, NAD(P)H quinone oxidoreductase 1 (NQO1), glutathione peroxidase (Gpx) 4 and phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA). Evaluating the target clinical effects, we found that BBR has the therapeutic potential of anti-aging, anti-cancer, relieving kidney disease, regulating the nervous system, and alleviating other chronic diseases. Conclusion: This review elucidates the interaction between potential targets and small molecular metabolites by exploring the mechanism of BBR regulating metabolism. That will help pharmacologists to identify new promising metabolites interacting with these targets.
Collapse
Affiliation(s)
- Aru Sun
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Haoyu Yang
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tao Li
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jinli Luo
- China Traditional Chinese Medicine Holdings Co. Limited, Guangdong e-fong Pharmaceutical Co., Ltd., Foshan, China
- Graduate College, Beijing University of Chinese Medicine, Beijing, China
| | - Ling Zhou
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate College, Beijing University of Chinese Medicine, Beijing, China
| | - Rui Chen
- College of Basic Medical Sciences, Changchun University of Chinese Medicine, Changchun, China
| | - Lin Han
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yiqun Lin
- Department of Endocrinology, Guang’anmen Hospital South Campus, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
31
|
Kumar KK, Aburawi EH, Ljubisavljevic M, Leow MKS, Feng X, Ansari SA, Emerald BS. Exploring histone deacetylases in type 2 diabetes mellitus: pathophysiological insights and therapeutic avenues. Clin Epigenetics 2024; 16:78. [PMID: 38862980 PMCID: PMC11167878 DOI: 10.1186/s13148-024-01692-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 06/04/2024] [Indexed: 06/13/2024] Open
Abstract
Diabetes mellitus is a chronic disease that impairs metabolism, and its prevalence has reached an epidemic proportion globally. Most people affected are with type 2 diabetes mellitus (T2DM), which is caused by a decline in the numbers or functioning of pancreatic endocrine islet cells, specifically the β-cells that release insulin in sufficient quantity to overcome any insulin resistance of the metabolic tissues. Genetic and epigenetic factors have been implicated as the main contributors to the T2DM. Epigenetic modifiers, histone deacetylases (HDACs), are enzymes that remove acetyl groups from histones and play an important role in a variety of molecular processes, including pancreatic cell destiny, insulin release, insulin production, insulin signalling, and glucose metabolism. HDACs also govern other regulatory processes related to diabetes, such as oxidative stress, inflammation, apoptosis, and fibrosis, revealed by network and functional analysis. This review explains the current understanding of the function of HDACs in diabetic pathophysiology, the inhibitory role of various HDAC inhibitors (HDACi), and their functional importance as biomarkers and possible therapeutic targets for T2DM. While their role in T2DM is still emerging, a better understanding of the role of HDACi may be relevant in improving insulin sensitivity, protecting β-cells and reducing T2DM-associated complications, among others.
Collapse
Affiliation(s)
- Kukkala Kiran Kumar
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, PO Box 15551, Al Ain, Abu Dhabi, United Arab Emirates
| | - Elhadi Husein Aburawi
- Department of Pediatrics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Milos Ljubisavljevic
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
- Duke-NUS Medical School, Cardiovascular and Metabolic Disorders Program, Singapore, Singapore
| | - Melvin Khee Shing Leow
- LKC School of Medicine, Nanyang Technological University, Singapore, Singapore
- Dept of Endocrinology, Tan Tock Seng Hospital, Singapore, Singapore
- Duke-NUS Medical School, Cardiovascular and Metabolic Disorders Program, Singapore, Singapore
| | - Xu Feng
- Department of Biochemistry, YLL School of Medicine, National University of Singapore, Singapore, Singapore
| | - Suraiya Anjum Ansari
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Abu Dhabi, United Arab Emirates
- ASPIRE Precision Medicine Research Institute, Abu Dhabi, United Arab Emirates
| | - Bright Starling Emerald
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, PO Box 15551, Al Ain, Abu Dhabi, United Arab Emirates.
- Zayed Center for Health Sciences, United Arab Emirates University, Abu Dhabi, United Arab Emirates.
- ASPIRE Precision Medicine Research Institute, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
32
|
Divya KP, Kanwar N, Anuranjana PV, Kumar G, Beegum F, George KT, Kumar N, Nandakumar K, Kanwal A. SIRT6 in Regulation of Mitochondrial Damage and Associated Cardiac Dysfunctions: A Possible Therapeutic Target for CVDs. Cardiovasc Toxicol 2024; 24:598-621. [PMID: 38689163 DOI: 10.1007/s12012-024-09858-1] [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: 09/01/2023] [Accepted: 04/05/2024] [Indexed: 05/02/2024]
Abstract
Cardiovascular diseases (CVDs) can be described as a global health emergency imploring possible prevention strategies. Although the pathogenesis of CVDs has been extensively studied, the role of mitochondrial dysfunction in CVD development has yet to be investigated. Diabetic cardiomyopathy, ischemic-reperfusion injury, and heart failure are some of the CVDs resulting from mitochondrial dysfunction Recent evidence from the research states that any dysfunction of mitochondria has an impact on metabolic alteration, eventually causes the death of a healthy cell and therefore, progressively directing to the predisposition of disease. Cardiovascular research investigating the targets that both protect and treat mitochondrial damage will help reduce the risk and increase the quality of life of patients suffering from various CVDs. One such target, i.e., nuclear sirtuin SIRT6 is strongly associated with cardiac function. However, the link between mitochondrial dysfunction and SIRT6 concerning cardiovascular pathologies remains poorly understood. Although the Role of SIRT6 in skeletal muscles and cardiomyocytes through mitochondrial regulation has been well understood, its specific role in mitochondrial maintenance in cardiomyocytes is poorly determined. The review aims to explore the domain-specific function of SIRT6 in cardiomyocytes and is an effort to know how SIRT6, mitochondria, and CVDs are related.
Collapse
Affiliation(s)
- K P Divya
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Navjot Kanwar
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab, Technical University, Bathinda, Punjab, 151005, India
| | - P V Anuranjana
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Gautam Kumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
- School of Pharmacy, Sharda University, Greater Noida, Uttar Pradesh, 201310, India
| | - Fathima Beegum
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Krupa Thankam George
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Nitesh Kumar
- Department of Pharmacology, National Institute of Pharmaceutical Educations and Research, Hajipur, Bihar, 844102, India
| | - K Nandakumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India.
| | - Abhinav Kanwal
- Department of Pharmacology, All India Institute of Medical Sciences, Bathinda, Punjab, 151005, India.
| |
Collapse
|
33
|
Pan B, Chen C, Zhao Y, Cai J, Fu S, Liu J. SIRT3: A Potential Target of Different Types of Osteoporosis. Cell Biochem Biophys 2024; 82:489-500. [PMID: 38512537 DOI: 10.1007/s12013-024-01254-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 03/13/2024] [Indexed: 03/23/2024]
Abstract
Osteoporosis (OP) is a common age-related disease. OP is mainly a decrease in bone density and mass caused by the destruction of bone microstructure, which leads to an increase in bone fragility. SIRT3 is a mitochondrial deacetylase that plays critical roles in mitochondrial homeostasis, metabolic regulation, gene transcription, stress response, and gene stability. Studies have shown that the higher expression levels of SIRT3 are associated with decreased levels of oxidative stress in the body and may play important roles in the prevention of age-related diseases. SIRTs can enhance the osteogenic potential and osteoblastic activity of bone marrow mesenchymal stromal cells not only by enhancing PGC-1α, FOXO3, SOD2, and oxidative phosphorylation, but also by anti-aging and reducing mitochondrial autophagy. SIRT3 is able to upregulate antioxidant enzymes to exert an inhibitory effect on osteoclasts, however, it has been shown that the inflammatory cascade response can in turn increase SIRT3 and inhibit osteoclast differentiation through the AMPK-PGC-1β pathway. SIRT3 plays an important role in different types of osteoporosis by affecting osteoblasts, osteoclasts, and bone marrow mesenchymal cells. In this review, we discuss the classification and physiological functions of SIRTs, the effects of SIRT3 on OCs osteoblasts, and BMSCs, and the roles and mechanisms of SIRT3 in different types of OP, such as diabetic OP, glucocorticoid-induced OP, postmenopausal OP, and senile OP.
Collapse
Affiliation(s)
- Binjing Pan
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Chongyang Chen
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Yangting Zhao
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Jing Cai
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Songbo Fu
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
- Department of Endocrinology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Jingfang Liu
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China.
- Department of Endocrinology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.
| |
Collapse
|
34
|
Xie T, Yao L, Li X. Advance in Iron Metabolism, Oxidative Stress and Cellular Dysfunction in Experimental and Human Kidney Diseases. Antioxidants (Basel) 2024; 13:659. [PMID: 38929098 PMCID: PMC11200795 DOI: 10.3390/antiox13060659] [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: 05/06/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
Kidney diseases pose a significant global health issue, frequently resulting in the gradual decline of renal function and eventually leading to end-stage renal failure. Abnormal iron metabolism and oxidative stress-mediated cellular dysfunction facilitates the advancement of kidney diseases. Iron homeostasis is strictly regulated in the body, and disturbance in this regulatory system results in abnormal iron accumulation or deficiency, both of which are associated with the pathogenesis of kidney diseases. Iron overload promotes the production of reactive oxygen species (ROS) through the Fenton reaction, resulting in oxidative damage to cellular molecules and impaired cellular function. Increased oxidative stress can also influence iron metabolism through upregulation of iron regulatory proteins and altering the expression and activity of key iron transport and storage proteins. This creates a harmful cycle in which abnormal iron metabolism and oxidative stress perpetuate each other, ultimately contributing to the advancement of kidney diseases. The crosstalk of iron metabolism and oxidative stress involves multiple signaling pathways, such as hypoxia-inducible factor (HIF) and nuclear factor erythroid 2-related factor 2 (Nrf2) pathways. This review delves into the functions and mechanisms of iron metabolism and oxidative stress, along with the intricate relationship between these two factors in the context of kidney diseases. Understanding the underlying mechanisms should help to identify potential therapeutic targets and develop novel and effective therapeutic strategies to combat the burden of kidney diseases.
Collapse
Affiliation(s)
- Tiancheng Xie
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Li Yao
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110001, China;
| | - Xiaogang Li
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
35
|
Pei MQ, Xu LM, Yang YS, Chen WC, Chen XL, Fang YM, Lin S, He HF. Latest advances and clinical application prospects of resveratrol therapy for neurocognitive disorders. Brain Res 2024; 1830:148821. [PMID: 38401770 DOI: 10.1016/j.brainres.2024.148821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 01/13/2024] [Accepted: 02/21/2024] [Indexed: 02/26/2024]
Abstract
Neurocognitive disorders, such as Alzheimer's disease, vascular dementia, and postoperative cognitive dysfunction, are non-psychiatric brain syndromes in which a significant decline in cognitive function causes great trauma to the mental status of the patient. The lack of effective treatments for neurocognitive disorders imposes a considerable burden on society, including a substantial economic impact. Over the past few decades, the identification of resveratrol, a natural plant compound, has provided researchers with an opportunity to formulate novel strategies for the treatment of neurocognitive disorders. This is because resveratrol effectively protects the brain of those with neurocognitive disorders by targeting some mechanisms such as inflammation and oxidative stress. This article reviews the status of recent research investigating the use of resveratrol for the treatment of different neurocognitive disorders. By examining the possible mechanisms of action of resveratrol and the shared mechanisms of different neurocognitive disorders, treatments for neurocognitive disorders may be further clarified.
Collapse
Affiliation(s)
- Meng-Qin Pei
- Department of Anesthesiology, the Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, Fujian Province, China
| | - Li-Ming Xu
- Department of Anesthesiology, the Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, Fujian Province, China
| | - Yu-Shen Yang
- Department of Anesthesiology, the Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, Fujian Province, China
| | - Wei-Can Chen
- Department of Anesthesiology, the Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, Fujian Province, China
| | - Xin-Li Chen
- Department of Anesthesiology, the Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, Fujian Province, China
| | - Yu-Ming Fang
- Department of Anesthesiology, the Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, Fujian Province, China
| | - Shu Lin
- Center of Neurological and Metabolic Research, the Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, Fujian Province, China; Neuroendocrinology Group, Garvan Institute of Medical Research, 384 Victoria St, Sydney, Australia.
| | - He-Fan He
- Department of Anesthesiology, the Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, Fujian Province, China.
| |
Collapse
|
36
|
Ye W, Han K, Xie M, Li S, Chen G, Wang Y, Li T. Mitochondrial energy metabolism in diabetic cardiomyopathy: Physiological adaption, pathogenesis, and therapeutic targets. Chin Med J (Engl) 2024; 137:936-948. [PMID: 38527931 PMCID: PMC11046025 DOI: 10.1097/cm9.0000000000003075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Indexed: 03/27/2024] Open
Abstract
Diabetic cardiomyopathy is defined as abnormal structure and function of the heart in the setting of diabetes, which could eventually develop heart failure and leads to the death of the patients. Although blood glucose control and medications to heart failure show beneficial effects on this disease, there is currently no specific treatment for diabetic cardiomyopathy. Over the past few decades, the pathophysiology of diabetic cardiomyopathy has been extensively studied, and an increasing number of studies pinpoint that impaired mitochondrial energy metabolism is a key mediator as well as a therapeutic target. In this review, we summarize the latest research in the field of diabetic cardiomyopathy, focusing on mitochondrial damage and adaptation, altered energy substrates, and potential therapeutic targets. A better understanding of the mitochondrial energy metabolism in diabetic cardiomyopathy may help to gain more mechanistic insights and generate more precise mitochondria-oriented therapies to treat this disease.
Collapse
Affiliation(s)
- Wanlin Ye
- Department of Anesthesiology, Laboratory of Mitochondria and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Kun Han
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan 610041, China
| | - Maodi Xie
- Department of Anesthesiology, Laboratory of Mitochondria and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Sheyu Li
- Department of Endocrinology and Metabolism, Division of Guideline and Rapid Recommendation, Cochrane China Center, MAGIC China Center, Chinese Evidence-Based Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Guo Chen
- Department of Anesthesiology, Laboratory of Mitochondria and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yanyan Wang
- Nursing Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Tao Li
- Department of Anesthesiology, Laboratory of Mitochondria and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
37
|
Wang T, Chen Y, Liu Z, Zhou J, Li N, Shan Y, He Y. Long noncoding RNA Glis2 regulates podocyte mitochondrial dysfunction and apoptosis in diabetic nephropathy via sponging miR-328-5p. J Cell Mol Med 2024; 28:e18204. [PMID: 38506068 PMCID: PMC10951868 DOI: 10.1111/jcmm.18204] [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: 08/12/2023] [Revised: 01/13/2024] [Accepted: 02/20/2024] [Indexed: 03/21/2024] Open
Abstract
Podocyte apoptosis exerts a crucial role in the pathogenesis of DN. Recently, long noncoding RNAs (lncRNAs) have been gradually identified to be functional in a variety of different mechanisms associated with podocyte apoptosis. This study aimed to investigate whether lncRNA Glis2 could regulate podocyte apoptosis in DN and uncover the underlying mechanism. The apoptosis rate was detected by flow cytometry. Mitochondrial membrane potential (ΔΨM) was measured using JC-1 staining. Mitochondrial morphology was detected by MitoTracker Deep Red staining. Then, the histopathological and ultrastructure changes of renal tissues in diabetic mice were observed using periodic acid-Schiff (PAS) staining and transmission electron microscopy. We found that lncRNA Glis2 was significantly downregulated in high-glucose cultured podocytes and renal tissues of db/db mice. LncRNA Glis2 overexpression was found to alleviate podocyte mitochondrial dysfunction and apoptosis. The direct interaction between lncRNA Glis2 and miR-328-5p was confirmed by dual luciferase reporter assay. Furthermore, lncRNA Glis2 overexpression alleviated podocyte apoptosis in diabetic mice. Taken together, this study demonstrated that lncRNA Glis2, acting as a competing endogenous RNA (ceRNA) of miRNA-328-5p, regulated Sirt1-mediated mitochondrial dysfunction and podocyte apoptosis in DN.
Collapse
Affiliation(s)
- Ting Wang
- Department of EndocrinologySecond Hospital of Hebei Medical UniversityShijiazhuangHebeiP.R. China
| | - Yanxia Chen
- Department of EndocrinologySecond Hospital of Hebei Medical UniversityShijiazhuangHebeiP.R. China
| | - Zhihong Liu
- Department of EndocrinologySecond Hospital of Hebei Medical UniversityShijiazhuangHebeiP.R. China
| | - Jing Zhou
- Department of EndocrinologySecond Hospital of Hebei Medical UniversityShijiazhuangHebeiP.R. China
| | - Na Li
- Department of EndocrinologySecond Hospital of Hebei Medical UniversityShijiazhuangHebeiP.R. China
| | - Yue Shan
- Department of EndocrinologySecond Hospital of Hebei Medical UniversityShijiazhuangHebeiP.R. China
| | - Yinxi He
- Department of Orthopaedic TraumaThe Third Hospital of ShijiazhuangShijiazhuangHebeiP.R. China
| |
Collapse
|
38
|
Kong W, Chen J, Ruan X, Xu X, Li X, Bao M, Shao Y, Bian X, Li R, Jiang Q, Zhang Y, Li Z, Yan F, Ye J. Cardiac injury activates STING signaling via upregulating SIRT6 in macrophages after myocardial infarction. Life Sci 2024; 341:122474. [PMID: 38296191 DOI: 10.1016/j.lfs.2024.122474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 02/19/2024]
Abstract
AIMS This work sought to investigate the mechanism underlying the STING signaling pathway during myocardial infarction (MI), and explore the involvement and the role of SIRT6 in the process. MAIN METHODS Mice underwent the surgery of permanent left anterior descending (LAD) artery constriction. Primary cardiomyocytes (CMs) and fibroblasts were subjected to hypoxia to mimic MI in vitro. STING expression was assessed in the infarct heart, and the effect of STING inhibition on cardiac fibrosis was explored. This study also evaluated the regulatory effect of STING by SIRT6 in macrophages. KEY FINDINGS STING protein was increased in the infarct heart tissue, highlighting its involvement in the post-MI inflammatory response. Hypoxia-induced death of CMs and fibroblasts contributed to the upregulation of STING in macrophages, establishing the involvement of STING in the intercellular signaling during MI. Inhibition of STING resulted in a significant reduction of cardiac fibrosis at day 14 after MI. Additionally, this study identified SIRT6 as a key regulator of STING via influencing its acetylation and ubiquitination in macrophages, providing novel insights into the posttranscriptional modification and expression of STING at the acute phase after myocardial infarction. SIGNIFICANCE This work shows the key role of SIRT6/STING signaling in the pathogenesis of cardiac injury after MI, suggesting that targeting this regulatory pathway could be a promising strategy to attenuate cardiac fibrosis after MI.
Collapse
Affiliation(s)
- Weixian Kong
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Jiawen Chen
- State Key Laboratory of Natural Medicines, Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China
| | - Xinjia Ruan
- State Key Laboratory of Natural Medicines, Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China
| | - Xiaozhi Xu
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Xie Li
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Mengmeng Bao
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Yuru Shao
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Xiaohong Bian
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Ruiyan Li
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Qizhou Jiang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Yubin Zhang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Zhe Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular research Institute, Wuhan University, Wuhan 430060, China; Hubei key Laboratory of Cardiology, Wuhan 430060, China.
| | - Fangrong Yan
- State Key Laboratory of Natural Medicines, Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China.
| | - Junmei Ye
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China.
| |
Collapse
|
39
|
Fedorczak A, Lewiński A, Stawerska R. Sirtuin 1 serum concentration in healthy children - dependence on sex, age, stage of puberty, body weight and diet. Front Endocrinol (Lausanne) 2024; 15:1356612. [PMID: 38529393 PMCID: PMC10961438 DOI: 10.3389/fendo.2024.1356612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 02/21/2024] [Indexed: 03/27/2024] Open
Abstract
Introduction Sirtuin 1 (SIRT1) is known to be involved in sensing cellular energy levels and regulating energy metabolism. This study aimed to evaluate fasting serum SIRT1 levels in healthy children, and to analyse the influence of age, sex, puberty, body weight, height, and diet on its concentration. Methods 47 healthy children aged 4-14 with weight and height within normal range and no chronic disease were included into the study. Fasting serum SIRT1 concentrations were estimated by Enzyme Linked Immunosorbent Assay (ELISA). Results Results showed that serum SIRT1 concentrations in healthy children did not differ with respect to sex, age, height, weight and puberty. Whereas, it appeared that a higher frequency of fruits, vegetables and dairy products consumption was associated with an increase in serum SIRT1 levels. Discussion Studying SIRT1 in the context of children's health may have implications for a broader understanding of growth processes, pubertal development, metabolic disorders and nutrition.
Collapse
Affiliation(s)
- Anna Fedorczak
- Department of Endocrinology and Metabolic Diseases, Polish Mother’s Memorial Hospital – Research Institute, Lodz, Poland
| | - Andrzej Lewiński
- Department of Endocrinology and Metabolic Diseases, Polish Mother’s Memorial Hospital – Research Institute, Lodz, Poland
- Department of Paediatric Endocrinology, Medical University of Lodz, Lodz, Poland
| | - Renata Stawerska
- Department of Endocrinology and Metabolic Diseases, Polish Mother’s Memorial Hospital – Research Institute, Lodz, Poland
- Department of Paediatric Endocrinology, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
40
|
Chen H, Guo J, Cai Y, Zhang C, Wei F, Sun H, Cheng C, Liu W, He Z. Elucidation of the anti-β-cell dedifferentiation mechanism of a modified Da Chaihu Decoction by an integrative approach of network pharmacology and experimental verification. JOURNAL OF ETHNOPHARMACOLOGY 2024; 321:117481. [PMID: 38007164 DOI: 10.1016/j.jep.2023.117481] [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: 06/09/2023] [Revised: 11/17/2023] [Accepted: 11/17/2023] [Indexed: 11/27/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Modified Da Chaihu decoction (MDCH) is a traditional Chinese herbal prescription that has been used in the clinic to treat type 2 diabetes (T2D). Previous studies have confirmed that MDCH improves glycemic and lipid metabolism, enhances pancreatic function, and alleviates insulin resistance in patients with T2D and diabetic rats. Evidence has demonstrated that MDCH protects pancreatic β cells via regulating the gene expression of sirtuin 1 (SIRT1) and forkhead box protein O1 (FOXO1). However, the detailed mechanism remains unclear. AIM OF THE STUDY Dedifferentiation of pancreatic β cells mediated by FOXO1 has been recognized as the main pathogenesis of T2D. This study aims to investigate the therapeutic effects of MDCH on T2D in vitro and in vivo to elucidate the potential molecular mechanisms. MATERIALS AND METHODS To predict the key targets of MDCH in treating T2D, network pharmacology methods were used. A T2D model was induced in diet-induced obese (DIO) C57BL/6 mice with a single intraperitoneal injection of streptozotocin. Glucose metabolism indicators (oral glucose tolerance test, insulin tolerance test), lipid metabolism indicators (total cholesterol, triglyceride, low-density lipoprotein cholesterol, high-density lipoprotein cholesterol), inflammatory factors (C-reactive protein, interleukin 6, tumor necrosis factor alpha), oxidative stress indicators (total antioxidant capacity, superoxide dismutase, malondialdehyde), and hematoxylin and eosin staining were analyzed to evaluate the therapeutic effect of MDCH on T2D. Immunofluorescence staining and quantification of FOXO1, pancreatic and duodenal homeobox 1 (PDX1), NK6 homeobox 1 (NKX6.1), octamer-binding protein 4 (OCT4), neurogenin 3 (Ngn3), insulin, and SIRT1, and Western blot analysis of insulin, SIRT1, and FOXO1 were performed to investigate the mechanism by which MDCH inhibited pancreatic β-cell dedifferentiation. RESULTS The chemical ingredients identified in MDCH were predicted to be important for signaling pathways related to lipid metabolism and insulin resistance, including lipids in atherosclerosis, the advanced glycation end product receptor of the advanced glycation end product signaling pathway, and the FOXO signaling pathway. Experimental studies showed that MDCH improved glucose and lipid metabolism in T2D mice, alleviated inflammation and oxidative stress damage, and reduced pancreatic pathological damage. Furthermore, MDCH upregulated the expression levels of SIRT1, FOXO1, PDX1, and NKX6.1, while downregulating the expression levels of OCT4 and Ngn3, which indicated that MDCH inhibited pancreatic dedifferentiation of β cells. CONCLUSIONS MDCH has therapeutic effects on T2D, through regulating the SIRT1/FOXO1 signaling pathway to inhibit pancreatic β-cell dedifferentiation, which has not been reported previously.
Collapse
Affiliation(s)
- Hongdong Chen
- Department of Endocrinology, Beijng Hepingli Hospital, NO.18th Hepingli North Street, Beijing, 100013, China; Renal Research Institution of Beijing University of Chinese Medicine, and Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Jing Guo
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China; Renal Research Institution of Beijing University of Chinese Medicine, and Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Yuzi Cai
- Renal Research Institution of Beijing University of Chinese Medicine, and Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Chao Zhang
- Renal Research Institution of Beijing University of Chinese Medicine, and Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Fudong Wei
- Renal Research Institution of Beijing University of Chinese Medicine, and Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Hao Sun
- Renal Research Institution of Beijing University of Chinese Medicine, and Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Cheng Cheng
- Department of Endocrinology, Beijng Hepingli Hospital, NO.18th Hepingli North Street, Beijing, 100013, China
| | - Weijing Liu
- Renal Research Institution of Beijing University of Chinese Medicine, and Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Zhongchen He
- Department of Endocrinology, Beijng Hepingli Hospital, NO.18th Hepingli North Street, Beijing, 100013, China.
| |
Collapse
|
41
|
Aftab R, Akbar F, Afroz A, Asif A, Khan MR, Rehman N, Zeeshan N. Mentha piperita silver nanoparticle-loaded hydrocolloid film for enhanced diabetic wound healing in rats. J Wound Care 2024; 33:xlviii-lx. [PMID: 38457268 DOI: 10.12968/jowc.2024.33.sup3a.xlviii] [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] [Indexed: 03/10/2024]
Abstract
OBJECTIVE To investigate the role of Mentha piperita silver nanoparticle-loaded carbopol gel for enhanced wound healing in a diabetic rat model. This research further aims to explore bioactive compounds derived from Mentha piperita obtained from high altitude. METHOD Methanolic extracts of Mentha piperita (MP), Mentha spicata (MS) and Mentha longifolia (ML) were used to synthesise silver nanoparticles (AgNP). AgNP synthesis was confirmed by ultraviolet-visible (UV-Vis) spectroscopy, X-ray diffraction (XRD), Fourier transform infrared spectroscopy (FTIR) and scanning electron microscopy (SEM). The antioxidant activity was assessed by 2, 2-diphenyl-1-picrylhydrazyl (DDPH) assay. Antiglycation potential was determined by measuring the fluorescent advanced glycation end products. The bioactive compound identified in the Mentha piperita methanolic (MPM) fraction through electrospray ionisation tandem mass spectrometric analysis (ESI-MS) was responsible for the highest antiglycation. The effects of MPM and MPM.AgNP-loaded Carbopol (Sanare Lab, India) on wound healing were compared in male, alloxan-induced, diabetic albino rats (200-250g), divided into control and treated groups. Effects on wound healing were assessed via histopathology. RESULTS UV-Vis and FTIR confirmed NP synthesis with peaks for flavonoids and polyphenols. SEM and XRD explored the cubical, 30-63nm crystalline NP. The maximum antioxidant and antiglycation potential was observed in order of; MP.AgNP>MS.AgNP>ML.AgNP. The highest antioxidant activity was observed by methanolic and aqueous MP.AgNPs (88.55% and 83.63%, respectively) at 2mg.ml-1, and (75.16% and 69.73%, respectively) at 1mg.ml-1, compared to ascorbic acid (acting as a positive control, 90.01%). MPM.AgNPs demonstrated the best antiglycation potential of 75.2% and 83.3% at 1mg.ml-1 and 2mg.ml-1, respectively, comparable to positive control (rutin: 88.1%) at 14 days post-incubation. A similar trend was observed for antimicrobial activity against Bacillus subtilis, Micrococcus luteus and Escherichia coli with an inhibition zone of 21mm, 21.6mm and 24.6mm. Rosmarinic acid was the active compound present in Mentha piperita, as identified by ESI-MS. MPM.AgNP-loaded Carbopol resulted in 100% wound closure compared with control at 20 days post-wounding. In the treatment group, re-epithelialisation was achieved by day 18, compared with 25 days for the positive control group. CONCLUSION MPM.AgNP-loaded Carbopol demonstrated safer and more effective biological properties, hence accelerating the diabetic excision wound healing process in alloxan-induced diabetic rats.
Collapse
Affiliation(s)
- Reema Aftab
- Department of Biochemistry and Biotechnology, University of Gujrat, Hafiz Hayat Campus Gujrat, Punjab, Pakistan
| | - Fatima Akbar
- Department of Biochemistry and Biotechnology, University of Gujrat, Hafiz Hayat Campus Gujrat, Punjab, Pakistan
| | - Amber Afroz
- Department of Biochemistry and Biotechnology, University of Gujrat, Hafiz Hayat Campus Gujrat, Punjab, Pakistan
| | - Awais Asif
- Nawaz Sharif Medical College, University of Gujrat, Hafiz Hayat Campus Gujrat, Punjab, Pakistan
| | - Muhammad Ramzan Khan
- National Institute for Genomics and Advanced Biotechnology, National Agricultural Research Centre, Park Road, Islamabad 45500, Pakistan
| | - Nazia Rehman
- National Institute for Genomics and Advanced Biotechnology, National Agricultural Research Centre, Park Road, Islamabad 45500, Pakistan
| | - Nadia Zeeshan
- Department of Biochemistry and Biotechnology, University of Gujrat, Hafiz Hayat Campus Gujrat, Punjab, Pakistan
| |
Collapse
|
42
|
Zhang H, Wang C, Sun H, Zhou T, Ma C, Han X, Zhang T, Xia J. Glutamine supplementation alleviated aortic atherosclerosis in mice model and in vitro. Proteomics 2024; 24:e2300179. [PMID: 37679095 DOI: 10.1002/pmic.202300179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 08/23/2023] [Accepted: 08/29/2023] [Indexed: 09/09/2023]
Abstract
This study aimed to clarify the role of glutamine in atherosclerosis and its participating mechanism. Forty C57BL/6J mice were divided into wild control (wild Con), ApoE- /- control (ApoE- /- Con), glutamine + ApoE- /- control (Glut + ApoE- /- Con), ApoE- /- high fat diet (ApoE- /- HFD), and glutamine + ApoE- /- HFD (Glut + ApoE- /- HFD) groups. The degree of atherosclerosis, western blotting, and multiomics were detected at 18 weeks. An in vitro study was also performed. Glutamine treatment significantly decreased the degree of aortic atherosclerosis (p = 0.03). O-GlcNAcylation (O-GlcNAc), IL-1β, IL-1α, and pyruvate kinase M2 (PKM2) in the ApoE- /- HFD group were significantly higher than those in the ApoE- /- Con group (p < 0.05). These differences were attenuated by glutamine treatment (p < 0.05), and aggravated by O-GlcNA transferase (OGT) overexpression in the in vitro study (p < 0.05). Multiomics showed that the ApoE- /- HFD group had higher levels of oxidative stress regulatory molecules (guanine deaminase [GUAD], xanthine dehydrogenase [XDH]), proinflammatory regulatory molecules (myristic acid and myristoleic acid), and stress granules regulatory molecules (caprin-1 and deoxyribose-phosphate aldolase [DERA]) (p < 0.05). These differences were attenuated by glutamine treatment (p < 0.05). We conclude that glutamine supplementation might alleviate atherosclerosis through downregulation of O-GlcNAc, glycolysis, oxidative stress, and proinflammatory pathway.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Cardiology, National Clinical Research Centre for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Chunxiu Wang
- Department of Evidence-Based Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Haichen Sun
- Surgical Laboratory, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Tian Zhou
- Department of Cardiology, National Clinical Research Centre for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Chang Ma
- Department of Cardiology, National Clinical Research Centre for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xuexue Han
- Department of Cardiology, National Clinical Research Centre for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Tianxing Zhang
- Department of Cardiology, National Clinical Research Centre for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jinggang Xia
- Department of Cardiology, National Clinical Research Centre for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
43
|
Liu X, Zhao Z, Chen D, Zhang Z, Lin X, Shen Z, Lin Q, Fan K, Wang Q, Zhang W, Ou Q. SIRT1 and miR-34a-5p Expression in PBMCs as Potential Biomarkers for Patients With Type 2 Diabetes With Cognitive Impairments. J Clin Endocrinol Metab 2024; 109:815-826. [PMID: 37758217 DOI: 10.1210/clinem/dgad562] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 09/18/2023] [Accepted: 09/21/2023] [Indexed: 10/03/2023]
Abstract
CONTEXT Patients with type 2 diabetes mellitus (T2DM) are at significantly increased risk of Alzheimer disease (AD). However, no biomarkers are available for early identification of patients with T2DM with cognitive impairment (T2DM-CI). Mitochondrial dysfunction is linked to AD. Silent Information Regulator 1 (SIRT1), which is responsible for regulating mitochondrial biogenesis, and its related miRNAs were also altered in AD. OBJECTIVE This study aimed to determine whether mitochondrial function in peripheral blood mononuclear cells (PBMCs) of patients with T2DM-CI was altered and if these alterations could be used as biomarkers. METHODS A total of 374 subjects were enrolled, including AD, T2DM-CI, T2DM-nCI (T2DM without cognitive impairment), and healthy controls. The mitochondrial function was determined using a commercial assay kit. The mitochondrial DNA (mtDNA) content, the expression of SIRT1, and selected miRNAs in PBMCs were measured by quantitative polymerase chain reaction. The correlations and diagnostic accuracy were assessed using the Spearman correlation coefficient or receiver operating characteristics analysis, respectively. RESULTS We found significant changes in mitochondrial function in PBMCs of patients with AD compared with controls (all P < .05), which were not found in T2DM-CI. However, mtDNA content and SIRT1 mRNA expression were lower in PBMCs of patients with T2DM-CI, while miR-34a-5p expression was higher than in patients with T2DM-nCI (all P < .05). A combination of SIRT1 and miR-34a-5p demonstrated excellent discrimination between T2DM-CI and T2DM-nCI (area under the curve = 0.793; sensitivity: 80.01%; specificity: 78.46%). Furthermore, correlation analysis revealed a link between miR-34a-5p expression and hyperglycemia in T2DM-CI. CONCLUSION Our findings revealed that there was an alteration of mitochondria at the peripheral level in patients with T2DM-CI. SIRT1 combined with miR-34a-5p in PBMCs performed well in identifying patients with T2DM-CI and may be a promising biomarker.
Collapse
Affiliation(s)
- Xiaofeng Liu
- Department of Laboratory Medicine, Gene Diagnosis Research Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Department of Laboratory Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
- Fujian Key Laboratory of Laboratory Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Fujian Clinical Research Center for Clinical Immunology Laboratory Test, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Zhipei Zhao
- Department of Laboratory Medicine, Gene Diagnosis Research Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Dengbin Chen
- Department of Laboratory Medicine, Gene Diagnosis Research Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Department of Laboratory Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Zeqin Zhang
- Department of Laboratory Medicine, Gene Diagnosis Research Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Department of Laboratory Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Xiaozhen Lin
- Department of Geriatrics, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Zhanbo Shen
- Department of Laboratory Medicine, Gene Diagnosis Research Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Qingwen Lin
- Department of Laboratory Medicine, Gene Diagnosis Research Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Department of Laboratory Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Kengna Fan
- Department of Laboratory Medicine, Gene Diagnosis Research Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Department of Laboratory Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Qi Wang
- Department of Laboratory Medicine, Gene Diagnosis Research Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Department of Laboratory Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Weiqing Zhang
- Department of Laboratory Medicine, Gene Diagnosis Research Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Department of Laboratory Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Qishui Ou
- Department of Laboratory Medicine, Gene Diagnosis Research Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Department of Laboratory Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
- Fujian Key Laboratory of Laboratory Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Fujian Clinical Research Center for Clinical Immunology Laboratory Test, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| |
Collapse
|
44
|
Lombardo GE, Russo C, Maugeri A, Navarra M. Sirtuins as Players in the Signal Transduction of Citrus Flavonoids. Int J Mol Sci 2024; 25:1956. [PMID: 38396635 PMCID: PMC10889095 DOI: 10.3390/ijms25041956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/02/2024] [Accepted: 02/03/2024] [Indexed: 02/25/2024] Open
Abstract
Sirtuins (SIRTs) belong to the family of nicotine adenine dinucleotide (NAD+)-dependent class III histone deacetylases, which come into play in the regulation of epigenetic processes through the deacetylation of histones and other substrates. The human genome encodes for seven homologs (SIRT1-7), which are localized into the nucleus, cytoplasm, and mitochondria, with different enzymatic activities and regulatory mechanisms. Indeed, SIRTs are involved in different physio-pathological processes responsible for the onset of several human illnesses, such as cardiovascular and neurodegenerative diseases, obesity and diabetes, age-related disorders, and cancer. Nowadays, it is well-known that Citrus fruits, typical of the Mediterranean diet, are an important source of bioactive compounds, such as polyphenols. Among these, flavonoids are recognized as potential agents endowed with a wide range of beneficial properties, including antioxidant, anti-inflammatory, hypolipidemic, and antitumoral ones. On these bases, we offer a comprehensive overview on biological effects exerted by Citrus flavonoids via targeting SIRTs, which acted as modulator of several signaling pathways. According to the reported studies, Citrus flavonoids appear to be promising SIRT modulators in many different pathologies, a role which might be potentially evaluated in future therapies, along with encouraging the study of those SIRT members which still lack proper evidence on their support.
Collapse
Affiliation(s)
- Giovanni Enrico Lombardo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (G.E.L.); (C.R.); (M.N.)
| | - Caterina Russo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (G.E.L.); (C.R.); (M.N.)
| | - Alessandro Maugeri
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Michele Navarra
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (G.E.L.); (C.R.); (M.N.)
| |
Collapse
|
45
|
Rani N, Sahu M, Ambasta RK, Kumar P. Triaging between post-translational modification of cell cycle regulators and their therapeutics in neurodegenerative diseases. Ageing Res Rev 2024; 94:102174. [PMID: 38135008 DOI: 10.1016/j.arr.2023.102174] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/18/2023] [Accepted: 12/18/2023] [Indexed: 12/24/2023]
Abstract
Neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, and Huntington's disease, present challenges in healthcare because of their complicated etiologies and absence of healing remedies. Lately, the emerging role of post-translational modifications (PTMs), in the context of cell cycle regulators, has garnered big interest as a potential avenue for therapeutic intervention. The review explores the problematic panorama of PTMs on cell cycle regulators and their implications in neurodegenerative diseases. We delve into the dynamic phosphorylation, acetylation, ubiquitination, SUMOylation, Glycation, and Neddylation that modulate the key cell cycle regulators, consisting of cyclins, cyclin-dependent kinases (CDKs), and their inhibitors. The dysregulation of these PTMs is related to aberrant cell cycle in neurons, which is one of the factors involved in neurodegenerative pathologies. Moreover, the effect of exogenous activation of CDKs and CDK inhibitors through PTMs on the signaling cascade was studied in postmitotic conditions of NDDs. Furthermore, the therapeutic implications of CDK inhibitors and associated alteration in PTMs were discussed. Lastly, we explored the putative mechanism of PTMs to restore normal neuronal function that might reverse NDDs.
Collapse
Affiliation(s)
- Neetu Rani
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042
| | - Mehar Sahu
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042; Department of Biotechnology and Microbiology, SRM University, Sonepat, Haryana, India.
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042.
| |
Collapse
|
46
|
Wei W, Li T, Chen J, Fan Z, Gao F, Yu Z, Jiang Y. SIRT3/6: an amazing challenge and opportunity in the fight against fibrosis and aging. Cell Mol Life Sci 2024; 81:69. [PMID: 38294557 PMCID: PMC10830597 DOI: 10.1007/s00018-023-05093-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/29/2023] [Accepted: 12/09/2023] [Indexed: 02/01/2024]
Abstract
Fibrosis is a typical aging-related pathological process involving almost all organs, including the heart, kidney, liver, lung, and skin. Fibrogenesis is a highly orchestrated process defined by sequences of cellular response and molecular signals mechanisms underlying the disease. In pathophysiologic conditions associated with organ fibrosis, a variety of injurious stimuli such as metabolic disorders, epigenetic changes, and aging may induce the progression of fibrosis. Sirtuins protein is a kind of deacetylase which can regulate cell metabolism and participate in a variety of cell physiological functions. In this review, we outline our current understanding of common principles of fibrogenic mechanisms and the functional role of SIRT3/6 in aging-related fibrosis. In addition, sequences of novel protective strategies have been identified directly or indirectly according to these mechanisms. Here, we highlight the role and biological function of SIRT3/6 focus on aging fibrosis, as well as their inhibitors and activators as novel preventative or therapeutic interventions for aging-related tissue fibrosis.
Collapse
Affiliation(s)
- Wenxin Wei
- School of Queen Mary, Nanchang University, Nanchang, 330031, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| | - Jinlong Chen
- School of Chemistry and Chemical Engineering, Nangchang University, 999 Xuefu Rd, Nanchang, 330031, China
| | - Zhen Fan
- The Hospital Affiliated to Shanxi University of Chinese Medicine, Xianyang, 712000, China.
| | - Feng Gao
- Shanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Zhibiao Yu
- School of Chemistry and Chemical Engineering, Nangchang University, 999 Xuefu Rd, Nanchang, 330031, China
| | - Yihao Jiang
- School of Chemistry and Chemical Engineering, Nangchang University, 999 Xuefu Rd, Nanchang, 330031, China.
| |
Collapse
|
47
|
Casagrande V, Menini S, Internò C, Pugliese G, Federici M, Menghini R. TIMP3 overexpression in myeloid lineage alleviates pancreatic damage and confers resistance to the development of type 1 diabetes in the MLDS -induced model. Front Endocrinol (Lausanne) 2024; 14:1297847. [PMID: 38313841 PMCID: PMC10835381 DOI: 10.3389/fendo.2023.1297847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/27/2023] [Indexed: 02/06/2024] Open
Abstract
Introduction Type 1 diabetes mellitus (T1DM) development involves a complex interplay of genetic, environmental, and immunological factors. By modulating the activity of proteases and receptors, the protein tissue inhibitor of metalloproteinase 3 (TIMP3) plays a role in limiting the expression and function of pro-inflammatory cytokines, which have been implicated in the advancement of T1DM. This study was aimed at examining the effect of TIMP3 overexpression in myeloid cells on the development of T1DM. Methods and results Twelve weeks after multiple low doses of streptozotocin (MLDS) treatment, diabetic mice overexpressing TIMP3 specifically in myeloid cells under the CD68 promoter (MacT3 mice) showed improved insulin secretion, islet morphology and vascularization, antioxidant defense system, and regulatory factors of mitochondrial biosynthesis and function. To get mechanistic insights into the origin of this protection, the severity of insulitis and inflammatory parameters were evaluated in pancreatic tissues 11 days after MLSD treatment, showing significantly reduced insulitis and levels of the pro-inflammatory cytokine tumor necrosis factor-α, interleukin -1β, and interferon -γ in MacT3 mice. Discussion The results indicate that TIMP3 is involved in maintaining islet architecture and functions, at least in part, through modulation of pro-inflammatory cytokine production associated with insulitis and may represent a novel therapeutic strategy for T1DM.
Collapse
Affiliation(s)
- Viviana Casagrande
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Stefano Menini
- Department of Clinical and Molecular Medicine, Sapienza University, Rome, Italy
| | - Chiara Internò
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Giuseppe Pugliese
- Department of Clinical and Molecular Medicine, Sapienza University, Rome, Italy
| | - Massimo Federici
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Center for Atherosclerosis, Department of Medical Sciences, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Rossella Menghini
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
48
|
Bettiol A, Urban ML, Emmi G, Galora S, Argento FR, Fini E, Borghi S, Bagni G, Mattioli I, Prisco D, Fiorillo C, Becatti M. SIRT1 and thrombosis. Front Mol Biosci 2024; 10:1325002. [PMID: 38304233 PMCID: PMC10833004 DOI: 10.3389/fmolb.2023.1325002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 12/29/2023] [Indexed: 02/03/2024] Open
Abstract
Thrombosis is a major cause of morbidity and mortality worldwide, with a complex and multifactorial pathogenesis. Recent studies have shown that SIRT1, a member of the sirtuin family of NAD + -dependent deacetylases, plays a crucial role in regulating thrombosis, modulating key pathways including endothelial activation, platelet aggregation, and coagulation. Furthermore, SIRT1 displays anti-inflammatory activity both in vitro, in vivo and in clinical studies, particularly via the reduction of oxidative stress. On these bases, several studies have investigated the therapeutic potential of targeting SIRT1 for the prevention of thrombosis. This review provides a comprehensive and critical overview of the main preclinical and clinical studies and of the current understanding of the role of SIRT1 in thrombosis.
Collapse
Affiliation(s)
- Alessandra Bettiol
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze, Italy
| | - Maria Letizia Urban
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze, Italy
| | - Giacomo Emmi
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze, Italy
| | - Silvia Galora
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Firenze, Firenze, Italy
| | - Flavia Rita Argento
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Firenze, Firenze, Italy
| | - Eleonora Fini
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Firenze, Firenze, Italy
| | - Serena Borghi
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Firenze, Firenze, Italy
| | - Giacomo Bagni
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze, Italy
| | - Irene Mattioli
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze, Italy
| | - Domenico Prisco
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze, Italy
| | - Claudia Fiorillo
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Firenze, Firenze, Italy
| | - Matteo Becatti
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Firenze, Firenze, Italy
| |
Collapse
|
49
|
Abdalla MMI. Therapeutic potential of adiponectin in prediabetes: strategies, challenges, and future directions. Ther Adv Endocrinol Metab 2024; 15:20420188231222371. [PMID: 38250316 PMCID: PMC10798122 DOI: 10.1177/20420188231222371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/28/2023] [Indexed: 01/23/2024] Open
Abstract
Adiponectin, an adipose-derived hormone, plays a pivotal role in glucose regulation and lipid metabolism, with a decrease in circulating adiponectin levels being linked to insulin resistance and prediabetes. This review examines the therapeutic potential of adiponectin in managing prediabetes, elucidating on multiple aspects including its role in glucose and lipid metabolism, influence on insulin sensitivity, and anti-inflammatory properties. Moreover, the paper highlights the latest strategies to augment adiponectin levels, such as gene therapy, pharmacological interventions, dietary modifications, and lifestyle changes. It also addresses the challenges encountered in translating preclinical findings into clinical practice, primarily related to drug delivery, safety, and efficacy. Lastly, the review proposes future directions, underlining the need for large-scale human trials, novel adiponectin analogs, and personalized treatment strategies to harness adiponectin's full therapeutic potential in preventing the transition from prediabetes to diabetes.
Collapse
Affiliation(s)
- Mona Mohamed Ibrahim Abdalla
- Human Biology Department, School of Medicine, International Medical University, 126, Jln Jalil Perkasa 19, Bukit Jalil, Kuala Lumpur, Federal Territory of Kuala Lumpur 57000, Malaysia
| |
Collapse
|
50
|
Petrariu OA, Barbu IC, Niculescu AG, Constantin M, Grigore GA, Cristian RE, Mihaescu G, Vrancianu CO. Role of probiotics in managing various human diseases, from oral pathology to cancer and gastrointestinal diseases. Front Microbiol 2024; 14:1296447. [PMID: 38249451 PMCID: PMC10797027 DOI: 10.3389/fmicb.2023.1296447] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/18/2023] [Indexed: 01/23/2024] Open
Abstract
The imbalance of microbial composition and diversity in favor of pathogenic microorganisms combined with a loss of beneficial gut microbiota taxa results from factors such as age, diet, antimicrobial administration for different infections, other underlying medical conditions, etc. Probiotics are known for their capacity to improve health by stimulating the indigenous gut microbiota, enhancing host immunity resistance to infection, helping digestion, and carrying out various other functions. Concurrently, the metabolites produced by these microorganisms, termed postbiotics, which include compounds like bacteriocins, lactic acid, and hydrogen peroxide, contribute to inhibiting a wide range of pathogenic bacteria. This review presents an update on using probiotics in managing and treating various human diseases, including complications that may emerge during or after a COVID-19 infection.
Collapse
Affiliation(s)
- Oana-Alina Petrariu
- Microbiology-Immunology Department, Faculty of Biology, University of Bucharest, Bucharest, Romania
- The Research Institute of the University of Bucharest, Bucharest, Romania
| | - Ilda Czobor Barbu
- Microbiology-Immunology Department, Faculty of Biology, University of Bucharest, Bucharest, Romania
- The Research Institute of the University of Bucharest, Bucharest, Romania
- Academy of Romanian Scientists, Bucharest, Romania
| | - Adelina-Gabriela Niculescu
- The Research Institute of the University of Bucharest, Bucharest, Romania
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Politehnica University of Bucharest, Bucharest, Romania
| | - Marian Constantin
- The Research Institute of the University of Bucharest, Bucharest, Romania
- Institute of Biology of Romanian Academy, Bucharest, Romania
| | - Georgiana Alexandra Grigore
- Microbiology-Immunology Department, Faculty of Biology, University of Bucharest, Bucharest, Romania
- The Research Institute of the University of Bucharest, Bucharest, Romania
- Academy of Romanian Scientists, Bucharest, Romania
- National Institute of Research and Development for Biological Sciences, Bucharest, Romania
| | - Roxana-Elena Cristian
- The Research Institute of the University of Bucharest, Bucharest, Romania
- National Institute of Research and Development for Biological Sciences, Bucharest, Romania
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Grigore Mihaescu
- Microbiology-Immunology Department, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Corneliu Ovidiu Vrancianu
- Microbiology-Immunology Department, Faculty of Biology, University of Bucharest, Bucharest, Romania
- The Research Institute of the University of Bucharest, Bucharest, Romania
- National Institute of Research and Development for Biological Sciences, Bucharest, Romania
| |
Collapse
|