1
|
Zhou J, Guo Y, Liu X, Yuan W. Bioinformatics analysis identifies key secretory protein-encoding differentially expressed genes in adipose tissue of metabolic syndrome. Adipocyte 2025; 14:2446243. [PMID: 39819282 DOI: 10.1080/21623945.2024.2446243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 01/19/2025] Open
Abstract
The objective of this study was to identify key secretory protein-encoding differentially expressed genes (SP-DEGs) in adipose tissue in female metabolic syndrome, thus detecting potential targets in treatment. We examined gene expression profiles in 8 women with metabolic syndrome and 7 healthy, normal body weight women. A total of 143 SP-DEGs were screened, including 83 upregulated genes and 60 downregulated genes. GO analyses of these SP-DEGs included proteolysis, angiogenesis, positive regulation of endothelial cell proliferation, immune response, protein processing, positive regulation of neuroblast proliferation, cell adhesion and ER to Golgi vesicle-mediated transport. KEGG pathway analysis of the SP-DEGs were involved in the TGF-beta signalling pathway, cytokine‒cytokine receptor interactions, the hippo signalling pathway, Malaria. Two modules were identified from the PPI network, namely, Module 1 (DNMT1, KDM1A, NCoR1, and E2F1) and Module 2 (IL-7 R, IL-12A, and CSF3). The gene DNMT1 was shared between the network modules and the WGCNA brown module. According to the single-gene GSEA results, DNMT1 was significantly positively correlated with histidine metabolism and phenylalanine metabolism. This study identified 7 key SP-DEGs in adipose tissue. DNMT1 was selected as the central gene in the development of metabolic syndrome and might be a potential therapeutic target.
Collapse
Affiliation(s)
- Jiandong Zhou
- Department of Nephrology, Shanghai General Hospital of Nanjing Medical University, Shanghai, China
| | - Yunshan Guo
- Department of Nephrology, Shanghai General Hospital of Nanjing Medical University, Shanghai, China
| | - Xuan Liu
- Department of Nephrology, Shanghai General Hospital of Nanjing Medical University, Shanghai, China
| | - Weijie Yuan
- Department of Nephrology, Shanghai General Hospital of Nanjing Medical University, Shanghai, China
| |
Collapse
|
2
|
Li X, Jia T, Wu Y, Peng Y, Feng Y, Gong L, Dong S, Tian J, Sun L. Multi-omics integration analysis and association study reveal the potential of ADIPOQ function in gestational diabetes mellitus. Nutr Diabetes 2025; 15:9. [PMID: 40025023 PMCID: PMC11873246 DOI: 10.1038/s41387-025-00356-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 10/27/2024] [Accepted: 01/10/2025] [Indexed: 03/04/2025] Open
Abstract
AIM To investigate the role of ADIPOQ gene in gestational diabetes mellitus (GDM). METHODS We genotyped single nucleotide polymorphisms (SNPs) rs266729 and rs1501299 within the ADIPOQ gene in a cohort of 1157 pregnant women of north Chinese Han ethnicity. This cohort comprised 560 pregnant women diagnosed with GDM and 597 pregnant women who exhibited normal oral glucose tolerance test at 24-28 weeks' gestation. All participants were recruited from the Department of Obstetrics and Gynecology at the Second Affiliated Hospital of Harbin Medical University. Additionally, we used conventional bioinformatics analysis methods to conduct multi-omics analysis (transcriptome, epigenome, and single-cell level) of ADIPOQ-regulated GDM. RESULTS The systolic blood flow velocity/diastolic blood flow velocity (S/D) ratio of the umbilical artery in GDM patients with CC genotype of rs266729 and GG genotype of rs1501299 was higher than control. Single-cell analysis suggested that ADIPOQ was expressed in extravillous trophoblast (EVT), T cell, monocytes, myelocyte, NK cell and syncytiotrophoblast (SCT). Functional enrichment analysis showed ADIPOQ gene was associated with response to nutrient levels, fat cell differentiation. CONCLUSION The findings of our study indicate a correlation between SNPs of ADIPOQ in GDM patients, and ADIPOQ is involved in the transcriptional regulation of GDM.
Collapse
Affiliation(s)
- Xiaoying Li
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), No.158, Shangtang Road, Gongshu District, Hangzhou, Zhejiang, China
| | - Tianshuang Jia
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), No.158, Shangtang Road, Gongshu District, Hangzhou, Zhejiang, China
| | - Yingnan Wu
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), No.158, Shangtang Road, Gongshu District, Hangzhou, Zhejiang, China
| | - Yanqing Peng
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), No.158, Shangtang Road, Gongshu District, Hangzhou, Zhejiang, China
| | - Yanan Feng
- Department of Ultrasound, Second Affiliated Hospital of Harbin Medical University, No.246, Harbin, China
| | - Liping Gong
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), No.158, Shangtang Road, Gongshu District, Hangzhou, Zhejiang, China
| | - Shuang Dong
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), No.158, Shangtang Road, Gongshu District, Hangzhou, Zhejiang, China
| | - Jiawei Tian
- Department of Ultrasound, Second Affiliated Hospital of Harbin Medical University, No.246, Harbin, China
| | - Litao Sun
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), No.158, Shangtang Road, Gongshu District, Hangzhou, Zhejiang, China.
| |
Collapse
|
3
|
Dakal TC, Xiao F, Bhusal CK, Sabapathy PC, Segal R, Chen J, Bai X. Lipids dysregulation in diseases: core concepts, targets and treatment strategies. Lipids Health Dis 2025; 24:61. [PMID: 39984909 PMCID: PMC11843775 DOI: 10.1186/s12944-024-02425-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 12/30/2024] [Indexed: 02/23/2025] Open
Abstract
Lipid metabolism is a well-regulated process essential for maintaining cellular functions and energy homeostasis. Dysregulation of lipid metabolism is associated with various conditions, including cardiovascular diseases, neurodegenerative disorders, and metabolic syndromes. This review explores the mechanisms underlying lipid metabolism, emphasizing the roles of key lipid species such as triglycerides, phospholipids, sphingolipids, and sterols in cellular physiology and pathophysiology. It also examines the genetic and environmental factors contributing to lipid dysregulation and the challenges of diagnosing and managing lipid-related disorders. Recent advancements in lipid-lowering therapies, including PCSK9 inhibitors, ezetimibe, bempedoic acid, and olpasiran, provide promising treatment options. However, these advancements are accompanied by challenges related to cost, accessibility, and patient adherence. The review highlights the need for personalized medicine approaches to address the interplay between genetics and environmental factors in lipid metabolism. As lipidomics and advanced diagnostic tools continue to progress, a deeper understanding of lipid-related disorders could pave the way for more effective therapeutic strategies.
Collapse
Affiliation(s)
- Tikam Chand Dakal
- Genome and Computational Biology Lab, Mohanlal Sukhadia, University, Udaipur, 313001, India
| | - Feng Xiao
- Department of Gastroenterology, The Second Affiliated Hospital of Shandong First Medical University, Taian, 271000, China
| | - Chandra Kanta Bhusal
- Aarupadai Veedu Medical College and Hospital, VMRF-DU, Pondicherry, 607402, India
- Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | | | - Rakesh Segal
- Aarupadai Veedu Medical College and Hospital, VMRF-DU, Pondicherry, 607402, India
- Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Juan Chen
- Department of Gastroenterology, The Second Affiliated Hospital of Shandong First Medical University, Taian, 271000, China.
| | - Xiaodong Bai
- Department of Plastic Surgery, Southern University of Science and Technology Hospital, Shenzhen, 518055, China.
| |
Collapse
|
4
|
Muntean M, Mărginean C, Bernad ES, Bănescu C, Nyulas V, Muntean IE, Săsăran V. Adiponectin C1Q and Collagen Domain Containing rs266729, Cyclin-Dependent Kinase Inhibitor 2A and 2B rs10811661, and Signal Sequence Receptor Subunit 1 rs9505118 Polymorphisms and Their Association with Gestational Diabetes Mellitus: A Case-Control Study in a Romanian Population. Int J Mol Sci 2025; 26:1654. [PMID: 40004118 PMCID: PMC11855124 DOI: 10.3390/ijms26041654] [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: 12/24/2024] [Revised: 02/08/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Gestational diabetes mellitus (GDM) and type 2 diabetes mellitus (T2DM) are public health concerns worldwide. These two diseases share the same pathophysiological and genetic similarities. This study aimed to investigate the T2DM known single nucleotide polymorphisms (SNPs) of the adiponectin C1Q and collagen domain containing (ADIPOQ), cyclin-dependent kinase inhibitor 2A and 2B (CDKN2A/2B), and signal sequence receptor subunit 1 (SSR1) genes in a cohort of Romanian GDM pregnant women and perinatal outcomes. DNA was isolated from the peripheral blood of 213 pregnant women with (n = 71) or without (n = 142) GDM. Afterward, ADIPOQ (rs266729), CDKN2A/2B (rs10811661), and SSR1 (rs9505118) gene polymorphisms were genotyped using TaqMan Real-Time PCR analysis. Women with GDM had a higher pre-pregnancy body mass index (BMI) (p < 0.0001), higher BMI (p < 0.0001), higher insulin resistance homeostatic model assessment (IR-HOMA) (p = 0.0002), higher insulin levels (p = 0.003), and lower adiponectin levels (p = 0.004) at birth compared to pregnant women with normoglycemia. GDM pregnant women had gestational hypertension (GH) more frequently during pregnancy (p < 0.0001), perineal lacerations more frequently during vaginal birth (p = 0.03), and more macrosomic newborns (p < 0.0001) than pregnant women from the control group. We did not find an association under any model (allelic, genotypic, dominant, or recessive) of ADIPOQ rs266729, CDKN2A/2B rs10811661, and SSR1 rs9505118 polymorphisms and GDM. In correlation analysis, we found a weak positive correlation (r = 0.24) between the dominant model GG + CG vs. CC of rs266729 and labor induction failure. In the dominant model TT vs. CC + CT of rs10811661, we found a weak negative correlation between this model and perineal lacerations. Our results suggest that the ADIPOQ rs266729, the CDKN2A/2B rs10811661, and the SSR1 rs9505118 gene polymorphisms are not associated with GDM in a cohort of Romanian pregnant women.
Collapse
Affiliation(s)
- Mihai Muntean
- Department of Obstetrics and Gynecology 2, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania; (M.M.); (V.S.)
| | - Claudiu Mărginean
- Department of Obstetrics and Gynecology 2, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania; (M.M.); (V.S.)
| | - Elena Silvia Bernad
- Department of Obstetrics and Gynecology, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
- Clinic of Obstetrics and Gynecology, “Pius Brinzeu” County Clinical Emergency Hospital, 300723 Timisoara, Romania
- Center for Laparoscopy, Laparoscopic Surgery and In Vitro Fertilization, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Claudia Bănescu
- Genetics Laboratory, Center for Advanced Medical and Pharmaceutical Research, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania;
| | - Victoria Nyulas
- Departament of Informatics and Medical Biostatistics, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania;
| | | | - Vladut Săsăran
- Department of Obstetrics and Gynecology 2, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania; (M.M.); (V.S.)
| |
Collapse
|
5
|
Singh A, Shadangi S, Gupta PK, Rana S. Type 2 Diabetes Mellitus: A Comprehensive Review of Pathophysiology, Comorbidities, and Emerging Therapies. Compr Physiol 2025; 15:e70003. [PMID: 39980164 DOI: 10.1002/cph4.70003] [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: 12/19/2024] [Revised: 02/03/2025] [Accepted: 02/07/2025] [Indexed: 02/22/2025]
Abstract
Humans are perhaps evolutionarily engineered to get deeply addicted to sugar, as it not only provides energy but also helps in storing fats, which helps in survival during starvation. Additionally, sugars (glucose and fructose) stimulate the feel-good factor, as they trigger the secretion of serotonin and dopamine in the brain, associated with the reward sensation, uplifting the mood in general. However, when consumed in excess, it contributes to energy imbalance, weight gain, and obesity, leading to the onset of a complex metabolic disorder, generally referred to as diabetes. Type 2 diabetes mellitus (T2DM) is one of the most prevalent forms of diabetes, nearly affecting all age groups. T2DM is clinically diagnosed with a cardinal sign of chronic hyperglycemia (excessive sugar in the blood). Chronic hyperglycemia, coupled with dysfunctions of pancreatic β-cells, insulin resistance, and immune inflammation, further exacerbate the pathology of T2DM. Uncontrolled T2DM, a major public health concern, also contributes significantly toward the onset and progression of several micro- and macrovascular diseases, such as diabetic retinopathy, nephropathy, neuropathy, atherosclerosis, and cardiovascular diseases, including cancer. The current review discusses the epidemiology, causative factors, pathophysiology, and associated comorbidities, including the existing and emerging therapies related to T2DM. It also provides a future roadmap for alternative drug discovery for the management of T2DM.
Collapse
Affiliation(s)
- Aditi Singh
- Chemical Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Odisha, India
| | - Sucharita Shadangi
- Chemical Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Odisha, India
| | - Pulkit Kr Gupta
- Chemical Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Odisha, India
| | - Soumendra Rana
- Chemical Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Odisha, India
| |
Collapse
|
6
|
Fang L, Kong F, Ou K, Hong L, Wang C, Tong X. Induction of insulin resistance in female mice due to prolonged phenanthrene exposure: Unveiling the low-dose effect and potential mechanisms. ENVIRONMENTAL RESEARCH 2024; 260:119597. [PMID: 39002631 DOI: 10.1016/j.envres.2024.119597] [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: 04/24/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/15/2024]
Abstract
Phenanthrene (Phe) is a commonly occurring polycyclic aromatic hydrocarbon (PAH) found in various food sources and drinking water. Previous studies have shown that long-term exposure to Phe in male mice leads to insulin resistance in a dose-dependent manner. However, the effect of Phe on glucose homeostasis in female mice remains unknown. To address this knowledge gap, female Kunming mice were exposed to Phe through their drinking water at concentrations of 0.05, 0.5, and 5 ng/mL. After 270 d of exposure, we surprisingly discovered a low-dose effect of Phe on insulin resistance in female mice, which differed from the effect observed in male mice and showed sexual dimorphism. Specifically, insulin resistance was only observed in the 0.05 ng/mL treatment, and this low-dose effect was also reflected in the concentration of Phe in white adipose tissue (WAT). Differences in metabolic enzyme activities in the liver may potentially explain this effect. The observed sexual dimorphism in Phe exposure could be attributed to variations in estrogen (E2) level and estrogen receptor beta (ERβ) expression in WAT. These findings highlight the association between environmental factors and the development of insulin resistance, emphasizing the pathogenic effect of even low doses of Phe. Moreover, sex dependent-effect should be given more attention when studying the toxic effects of environmental pollutants.
Collapse
Affiliation(s)
- Lu Fang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, PR China
| | - Feifei Kong
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, PR China
| | - Kunlin Ou
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China
| | - Luning Hong
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, PR China
| | - Chonggang Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China
| | - Xiaomei Tong
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, PR China.
| |
Collapse
|
7
|
Jin Y, Huang Y, Zhu J, Liao D, Zeng S, Jin X. Acupoint catgut embedding regulates community structure of intestinal flora in central obesity during perimenopause. Women Health 2024; 64:857-869. [PMID: 39496462 DOI: 10.1080/03630242.2024.2422876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/19/2024] [Accepted: 10/24/2024] [Indexed: 11/06/2024]
Abstract
Acupoint catgut embedding (ACE) is a safe and effective method for treating obesity. However, how it modulates intestinal flora and adiponectin remains unclear. We employed 16s rRNA sequencing technology to investigate ACE induced changes in intestinal flora and its association with adiponectin in subjects who received real (n = 41) and Sham (n = 41) stimulation. After treatment, the ACE group's body weight, BMI, WC, HC, WHtR, and WHR were significantly lower than those of the Control group (p < .01), there was statistically significant differences in ADPN between the two groups (p < .01). ACE reduces the species abundance and increased the diversity and differences of the gut microbiota in perimenopausal patients with central obesity. Most notably, there was an increase in Kosakonia and Klebsiella after ACE treatment in the patients. Significant negatively correlations were found between body weight/waist circumference and adiponectin. Increases in Klebsiella and Kosakonia were positively correlated with adiponectin, and were negatively correlated with body weight/waist circumference. Our results showed increases in Klebsiella and Kosakonia were correlated with body weight/waist circumference and adiponectin. These findings suggest that ACE-induced weight loss is probably in part associated with increases in adiponectin, Klebsiella and Kosakonia.Trial re-registration: www.chictr.org.cn, ID: ChiCTR2400087718, Registration Date: August 2nd 2024.
Collapse
Affiliation(s)
- Yuanyuan Jin
- Department of Acupuncture and Moxibustion, Zhejiang Hospital, Hangzhou, China
| | - Yifan Huang
- Department of Psychology and Behavioral Sciences, Zhejiang University, Hangzhou, China
| | - Jianfang Zhu
- Department of Acupuncture and Moxibustion, Zhejiang Hospital, Hangzhou, China
| | - Dan Liao
- Department of Nutrition, Zhejiang Hospital, Hangzhou, China
| | - Shumei Zeng
- Department of Gynaecology, Zhejiang Hospital, Hangzhou, China
| | - Xiaoqing Jin
- Department of Acupuncture and Moxibustion, Zhejiang Hospital, Hangzhou, China
| |
Collapse
|
8
|
Jilo DD, Abebe BK, Wang J, Guo J, Li A, Zan L. Long non-coding RNA (LncRNA) and epigenetic factors: their role in regulating the adipocytes in bovine. Front Genet 2024; 15:1405588. [PMID: 39421300 PMCID: PMC11484070 DOI: 10.3389/fgene.2024.1405588] [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: 03/23/2024] [Accepted: 09/02/2024] [Indexed: 10/19/2024] Open
Abstract
Investigating the involvement of long non-coding RNAs (lncRNAs) and epigenetic processes in bovine adipocytes can provide valuable new insights into controlling adipogenesis in livestock. Long non-coding RNAs have been associated with forming chromatin loops that facilitate enhancer-promoter interactions during adipogenesis, as well as regulating important adipogenic transcription factors like C/EBPα and PPARγ. They significantly influence gene expression regulation at the post-transcriptional level and are extensively researched for their diverse roles in cellular functions. Epigenetic modifications such as chromatin reorganization, histone alterations, and DNA methylation subsequently affect the activation of genes related to adipogenesis and the progression of adipocyte differentiation. By investigating how fat deposition is epigenetically regulated in beef cattle, scientists aim to unravel molecular mechanisms, identify key regulatory genes and pathways, and develop targeted strategies for modifying fat deposition to enhance desirable traits such as marbling and meat tenderness. This review paper delves into lncRNAs and epigenetic factors and their role in regulating bovine adipocytes while focusing on their potential as targets for genetic improvement to increase production efficiency. Recent genomics advancements, including molecular markers and genetic variations, can boost animal productivity, meeting global demands for high-quality meat products. This review establishes a foundation for future research on understanding regulatory networks linked to lncRNAs and epigenetic changes, contributing to both scholarly knowledge advancement and practical applications within animal agriculture.
Collapse
Affiliation(s)
- Diba Dedacha Jilo
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
- Department of Animal Science, Bule Hora University, Bule Hora, Ethiopia
| | - Belete Kuraz Abebe
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
- Department of Animal Science, Werabe University, Werabe, Ethiopia
| | - Jianfang Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Juntao Guo
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Anning Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Linsen Zan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
- National Beef Cattle Improvement Center, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
9
|
Luo Y, Ma W, Cheng S, Yuan T, Li J, Hao H, Liu K, Zeng M, Pan Y. Transplantation of Cold-Stimulated Subcutaneous Adipose Tissue Improves Fat Retention and Recipient Metabolism. Aesthet Surg J 2024; 44:NP486-NP500. [PMID: 38518754 DOI: 10.1093/asj/sjae070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/29/2024] [Accepted: 03/12/2024] [Indexed: 03/24/2024] Open
Abstract
BACKGROUND Induction of beige fat for grafting is an emerging transplantation strategy. However, safety concerns associated with pharmaceutical interventions limit its wider application. Moreover, because beige fat is a special type of fat with strong metabolic functions, its effect on the metabolism of recipients after grafting has not been explored in the plastic surgery domain. OBJECTIVES The aim of this study was to explore whether cold-induced inguinal white adipose tissue (iWAT) transplantation has a higher retention rate and beneficial effects on recipient metabolism. METHODS C57/BL6 mice were subjected to cold stimulation for 48 hours to induce the browning of iWAT and harvested immediately. Subsequently, each mouse received a transplant of 0.2 mL cold-induced iWAT or normal iWAT. Fat grafts and recipients' iWAT, epididymal adipose tissue, and brown adipose tissue were harvested at 8 weeks after operation. Immunofluorescence staining, real-time polymerase chain reaction, and western blot were used for histological and molecular analysis. RESULTS Cold-induced iWAT grafting had a higher mean [standard error of the mean] retention rate (67.33% [1.74%] vs 55.83% [2.94%], P < .01) and more satisfactory structural integrity than normal iWAT. Histological changes identified improved adipose tissue homeostasis after cold challenge, including abundant smaller adipocytes, higher levels of adipogenesis, angiogenesis, and proliferation, but lower levels of fibrosis. More importantly, cold-induced iWAT grafting suppressed the inflammation of epididymal adipose tissue caused by conventional fat grafting, and activated the glucose metabolism and thermogenic activity of recipients' adipose tissues. CONCLUSIONS Cold-induced iWAT grafting is an effective nonpharmacological intervention strategy to improve the retention rate and homeostasis of grafts. Furthermore, it improves the adverse effects caused by traditional fat grafting, while also conferring metabolic benefits.
Collapse
|
10
|
Kovácsházi C, Hambalkó S, Sayour NV, Gergely TG, Brenner GB, Pelyhe C, Kapui D, Weber BY, Hültenschmidt AL, Pállinger É, Buzás EI, Zolcsák Á, Kiss B, Bozó T, Csányi C, Kósa N, Kellermayer M, Farkas R, Karvaly GB, Wynne K, Matallanas D, Ferdinandy P, Giricz Z. Effect of hypercholesterolemia on circulating and cardiomyocyte-derived extracellular vesicles. Sci Rep 2024; 14:12016. [PMID: 38797778 PMCID: PMC11128454 DOI: 10.1038/s41598-024-62689-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 05/20/2024] [Indexed: 05/29/2024] Open
Abstract
Hypercholesterolemia (HC) induces, propagates and exacerbates cardiovascular diseases via various mechanisms that are yet not properly understood. Extracellular vesicles (EVs) are involved in the pathomechanism of these diseases. To understand how circulating or cardiac-derived EVs could affect myocardial functions, we analyzed the metabolomic profile of circulating EVs, and we performed an in-depth analysis of cardiomyocyte (CM)-derived EVs in HC. Circulating EVs were isolated with Vezics technology from male Wistar rats fed with high-cholesterol or control chow. AC16 human CMs were treated with Remembrane HC supplement and EVs were isolated from cell culture supernatant. The biophysical properties and the protein composition of CM EVs were analyzed. THP1-ASC-GFP cells were treated with CM EVs, and monocyte activation was measured. HC diet reduced the amount of certain phosphatidylcholines in circulating EVs, independently of their plasma level. HC treatment significantly increased EV secretion of CMs and greatly modified CM EV proteome, enriching several proteins involved in tissue remodeling. Regardless of the treatment, CM EVs did not induce the activation of THP1 monocytes. In conclusion, HC strongly affects the metabolome of circulating EVs and dysregulates CM EVs, which might contribute to HC-induced cardiac derangements.
Collapse
Affiliation(s)
- Csenger Kovácsházi
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Szabolcs Hambalkó
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Nabil V Sayour
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Tamás G Gergely
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Gábor B Brenner
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Csilla Pelyhe
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Dóra Kapui
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Bennet Y Weber
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | | | - Éva Pállinger
- Institute of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Edit I Buzás
- Institute of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
- ELKH-SE Translational Extracellular Vesicle Research Group, Budapest, Hungary
- HCEMM-SU Extracellular Vesicle Research Group, Budapest, Hungary
| | - Ádám Zolcsák
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Bálint Kiss
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
- HUNREN-SE Biophysical Virology Research Group, Budapest, Hungary
| | - Tamás Bozó
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Csilla Csányi
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Nikolett Kósa
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Miklós Kellermayer
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
- HUNREN-SE Biophysical Virology Research Group, Budapest, Hungary
| | - Róbert Farkas
- Department of Laboratory Medicine, Laboratory of Mass Spectrometry and Separation Technology, Semmelweis University, Budapest, Hungary
| | - Gellért B Karvaly
- Department of Laboratory Medicine, Laboratory of Mass Spectrometry and Separation Technology, Semmelweis University, Budapest, Hungary
| | - Kieran Wynne
- Systems Biology Ireland and School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - David Matallanas
- Systems Biology Ireland and School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Zoltán Giricz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.
- Pharmahungary Group, Szeged, Hungary.
| |
Collapse
|
11
|
Taylor BC, Steinthal LH, Dias M, Yalamanchili HK, Ochsner SA, Zapata GE, Mehta NR, McKenna NJ, Young NL, Nuotio-Antar AM. Histone proteoform analysis reveals epigenetic changes in adult mouse brown adipose tissue in response to cold stress. Epigenetics Chromatin 2024; 17:12. [PMID: 38678237 PMCID: PMC11055387 DOI: 10.1186/s13072-024-00536-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 04/09/2024] [Indexed: 04/29/2024] Open
Abstract
BACKGROUND Regulation of the thermogenic response by brown adipose tissue (BAT) is an important component of energy homeostasis with implications for the treatment of obesity and diabetes. Our preliminary analyses of RNA-Seq data uncovered many nodes representing epigenetic modifiers that are altered in BAT in response to chronic thermogenic activation. Thus, we hypothesized that chronic thermogenic activation broadly alters epigenetic modifications of DNA and histones in BAT. RESULTS Motivated to understand how BAT function is regulated epigenetically, we developed a novel method for the first-ever unbiased top-down proteomic quantitation of histone modifications in BAT and validated our results with a multi-omic approach. To test our hypothesis, wildtype male C57BL/6J mice were housed under chronic conditions of thermoneutral temperature (TN, 28°C), mild cold/room temperature (RT, 22°C), or severe cold (SC, 8°C) and BAT was analyzed for DNA methylation and histone modifications. Methylation of promoters and intragenic regions in genomic DNA decrease in response to chronic cold exposure. Integration of DNA methylation and RNA expression datasets suggest a role for epigenetic modification of DNA in regulation of gene expression in response to cold. In response to cold housing, we observe increased bulk acetylation of histones H3.2 and H4, increased histone H3.2 proteoforms with di- and trimethylation of lysine 9 (K9me2 and K9me3), and increased histone H4 proteoforms with acetylation of lysine 16 (K16ac) in BAT. CONCLUSIONS Our results reveal global epigenetically-regulated transcriptional "on" and "off" signals in murine BAT in response to varying degrees of chronic cold stimuli and establish a novel methodology to quantitatively study histones in BAT, allowing for direct comparisons to decipher mechanistic changes during the thermogenic response. Additionally, we make histone PTM and proteoform quantitation, RNA splicing, RRBS, and transcriptional footprint datasets available as a resource for future research.
Collapse
Affiliation(s)
- Bethany C Taylor
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
| | - Loic H Steinthal
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Division of Nutrition, Baylor College of Medicine, Houston, TX, USA
| | - Michelle Dias
- Department of Pediatrics, Division of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Hari Krishna Yalamanchili
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Division of Nutrition, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Division of Neurology, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Scott A Ochsner
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Gladys E Zapata
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Division of Nutrition, Baylor College of Medicine, Houston, TX, USA
| | - Nitesh R Mehta
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Division of Nutrition, Baylor College of Medicine, Houston, TX, USA
| | - Neil J McKenna
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Nicolas L Young
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA.
| | - Alli M Nuotio-Antar
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Division of Nutrition, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
12
|
Zhang Q, Lu C, Lu F, Liao Y, Cai J, Gao J. Challenges and opportunities in obesity: the role of adipocytes during tissue fibrosis. Front Endocrinol (Lausanne) 2024; 15:1365156. [PMID: 38686209 PMCID: PMC11056552 DOI: 10.3389/fendo.2024.1365156] [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: 01/03/2024] [Accepted: 04/01/2024] [Indexed: 05/02/2024] Open
Abstract
Obesity is a chronic disease that affects the energy balance of the whole body. In addition to increasing fat mass, tissue fibrosis occurred in white adipose tissue in obese condition. Fibrosis is the over-activation of fibroblasts leading to excessive accumulation of extracellular matrix, which could be caused by various factors, including the status of adipocytes. The morphology of adipocytes responds rapidly and dynamically to nutrient fluctuations. Adaptive hypertrophy of normal adipocytes protects peripheral organs from damage from lipotoxicity. However, the biological behavior of hypertrophic adipocytes in chronic obesity is abnormally altered. Adipocytes lead to fibrotic remodeling of the extracellular matrix by inducing unresolved chronic inflammation, persistent hypoxia, and increasing myofibroblast numbers. Moreover, adipocyte-induced fibrosis not only restricts the flexible expansion and contraction of adipose tissue but also initiates the development of various diseases through cellular autonomic and paracrine effects. Regarding anti-fibrotic therapy, dysregulated intracellular signaling and epigenetic changes represent potential candidate targets. Thus, modulation of adipocytes may provide potential therapeutic avenues for reversing pathological fibrosis in adipose tissue and achieving the anti-obesity purpose.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Chongxuan Lu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Feng Lu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yunjun Liao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Junrong Cai
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jianhua Gao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
13
|
Wu O, Lu X, Leng J, Zhang X, Liu W, Yang F, Zhang H, Li J, Khederzadeh S, Liu X, Yuan C. Reevaluating Adiponectin's impact on obesity hypertension: a Chinese case-control study. BMC Cardiovasc Disord 2024; 24:208. [PMID: 38615012 PMCID: PMC11015577 DOI: 10.1186/s12872-024-03865-4] [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: 12/03/2023] [Accepted: 03/28/2024] [Indexed: 04/15/2024] Open
Abstract
BACKGROUND Obesity and hypertension are major risk factors for cardiovascular diseases that affect millions of people worldwide. Both conditions are associated with chronic low-grade inflammation, which is mediated by adipokines such as adiponectin. Adiponectin is the most abundant adipokine that has a beneficial impact on metabolic and vascular biology, while high serum concentrations are associated with some syndromes. This "adiponectin paradox" still needs to be clarified in obesity-associated hypertension. The aim of this study was to investigate how adiponectin affects blood pressure, inflammation, and metabolic function in obesity hypertension using a Chinese adult case-control study. METHODS A case-control study that had finished recruiting 153 subjects divided as four characteristic groups. Adiponectin serum levels were tested by ELISA in these subjects among these four characteristic Chinese adult physical examination groups. Waist circumference (WC), body mass index (BMI), systolic blood pressure (SB), diastolic blood pressure (DB), and other clinical laboratory data were collected. Analyzation of correlations between the research index and differences between groups was done by SPSS. RESULTS Serum adiponectin levels in the| normal healthy group (NH group) were significantly higher than those in the newly diagnosed untreated just-obesity group (JO group), and negatively correlated with the visceral adiposity index. With multiple linear egression analysis, it was found that, for serum adiponectin, gender, serum albumin (ALB), alanine aminotransferase (ALT) and high-density lipoprotein cholesterol (HDLC) were the significant independent correlates, and for SB, age and HDLC were the significant independent correlates, and for DB, alkaline phosphatase (ALP) was the significant independent correlate. The other variables did not reach significance in the model. CONCLUSIONS Our study reveals that adiponectin's role in obesity-hypertension is multifaceted and is influenced by the systemic metabolic homeostasis signaling axis. In obesity-related hypertension, compensatory effects, adiponectin resistance, and reduced adiponectin clearance from impaired kidneys and liver all contribute to the "adiponectin paradox".
Collapse
Affiliation(s)
- Ou Wu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, People's Republic of China
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People's Republic of China
| | - Xi Lu
- Hangzhou Vocational and Technical College, Hangzhou, Zhejiang, People's Republic of China
| | - Jianhang Leng
- Department of Central Laboratory/Medical Examination Center of Hangzhou, The Frist People's Hospital of Hangzhou, Hangzhou, Zhejiang, People's Republic of China
| | - Xingyu Zhang
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Wei Liu
- JFIntelligent Healthcare Technology Co., Ltd Building No.5-7, No.699 Tianxiang Avenue, Hi-Tech Zone, Nanchang, Jiangxi Province, People's Republic of China
| | - Fenfang Yang
- Department of Central Laboratory/Medical Examination Center of Hangzhou, The Frist People's Hospital of Hangzhou, Hangzhou, Zhejiang, People's Republic of China
| | - Hu Zhang
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital Affiliated with Medical College of Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Jiajia Li
- Department of Central Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Saber Khederzadeh
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, People's Republic of China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, People's Republic of China
| | - Xiaodong Liu
- Hangzhou Center for Disease Control and Prevention, Hangzhou, Zhejiang, People's Republic of China
| | - Chengda Yuan
- Department of Dermatology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
14
|
Maia RDCA, Lima TC, Barbosa CM, Barbosa MA, de Queiroz KB, Alzamora AC. Intergenerational inheritance induced by a high-fat diet causes hyperphagia and reduced hypothalamic sensitivity to insulin and leptin in the second-generation of rats. Nutrition 2024; 120:112333. [PMID: 38271759 DOI: 10.1016/j.nut.2023.112333] [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: 07/24/2023] [Revised: 11/16/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024]
Abstract
OBJECTIVE The aim was to investigate the intergenerational inheritance induced by a high-fat diet on sensitivity to insulin and leptin in the hypothalamic control of satiety in second-generation offspring, which were fed a control diet. METHODS Progenitor rats were fed a high-fat or a control diet for 59 d until weaning. The first-generation and second-generation offspring were fed the control diet until 90 d of age. Body mass and adiposity index of the progenitors fed the high-fat diet and the second-generation offspring from progenitors fed the high-fat diet were evaluated as were the gene expression of DNA methyltransferase 3a, angiotensin-converting enzyme type 2, angiotensin II type 2 receptor, insulin and leptin signaling pathway (insulin receptor, leptin receptor, insulin receptor substrate 2, protein kinase B, signal transducer and transcriptional activator 3, pro-opiomelanocortin, and neuropeptide Agouti-related protein), superoxide dismutase activity, and the concentration of carbonyl protein and satiety-regulating neuropeptides, pro-opiomelanocortin and neuropeptide Agouti-related protein, in the hypothalamus. RESULTS The progenitor group fed a high-fat diet showed increased insulin resistance and reduced insulin-secreting beta-cell function and reduced food intake, without changes in caloric intake. The second-generation offspring from progenitors fed a high-fat diet, compared with second-generation offspring from progenitors fed a control diet group, had decreased insulin-secreting beta-cell function and increased food and caloric intake, insulin resistance, body mass, and adiposity index. Furthermore, second-generation offspring from progenitors fed a high-fat diet had increased DNA methyltransferase 3a, neuropeptide Agouti-related protein, angiotensin II type 1 receptor, and nicotinamide adenine dinucleotide phosphate oxidase p47phox gene expression, superoxide dismutase activity, and neuropeptide Agouti-related protein concentration in the hypothalamus. In addition, there were reduced in gene expression of the insulin receptor, leptin receptor, insulin receptor substrate 2, pro-opiomelanocortin, angiotensin II type 2 receptor, angiotensin-converting enzyme type 2, and angiotensin-(1-7) receptor and pro-opiomelanocortin concentration in the second-generation offspring from progenitors fed the high-fat diet. CONCLUSIONS Overall, progenitors fed a high-fat diet induced changes in the hypothalamic control of satiety of the second-generation offspring from progenitors fed the high-fat diet through intergenerational inheritance. These changes led to hyperphagia, alterations in the hypothalamic pathways of insulin, and leptin and adiposity index increase, favoring the occurrence of different cardiometabolic disorders in the second-generation offspring from progenitors fed the high-fat diet fed only with the control diet.
Collapse
Affiliation(s)
- Rosana da Conceição Araújo Maia
- Núcleo de Pesquisa em Ciências Biológicas, Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | - Taynara Carolina Lima
- Núcleo de Pesquisa em Ciências Biológicas, Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | - Claudiane Maria Barbosa
- Núcleo de Pesquisa em Ciências Biológicas, Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | - Maria Andréa Barbosa
- Núcleo de Pesquisa em Ciências Biológicas, Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | - Karina Barbosa de Queiroz
- Departamento de Alimentos, Escola de Nutrição, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | - Andréia Carvalho Alzamora
- Núcleo de Pesquisa em Ciências Biológicas, Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Brazil; Departamento de Ciências Biológicas, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Brazil.
| |
Collapse
|
15
|
Lagarde CB, Kavalakatt J, Benz MC, Hawes ML, Arbogast CA, Cullen NM, McConnell EC, Rinderle C, Hebert KL, Khosla M, Belgodere JA, Hoang VT, Collins-Burow BM, Bunnell BA, Burow ME, Alahari SK. Obesity-associated epigenetic alterations and the obesity-breast cancer axis. Oncogene 2024; 43:763-775. [PMID: 38310162 DOI: 10.1038/s41388-024-02954-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 02/05/2024]
Abstract
Both breast cancer and obesity can regulate epigenetic changes or be regulated by epigenetic changes. Due to the well-established link between obesity and an increased risk of developing breast cancer, understanding how obesity-mediated epigenetic changes affect breast cancer pathogenesis is critical. Researchers have described how obesity and breast cancer modulate the epigenome individually and synergistically. In this review, the epigenetic alterations that occur in obesity, including DNA methylation, histone, and chromatin modification, accelerated epigenetic age, carcinogenesis, metastasis, and tumor microenvironment modulation, are discussed. Delineating the relationship between obesity and epigenetic regulation is vital to furthering our understanding of breast cancer pathogenesis.
Collapse
Affiliation(s)
- Courtney B Lagarde
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Joachim Kavalakatt
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Megan C Benz
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Mackenzie L Hawes
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Carter A Arbogast
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Nicole M Cullen
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Emily C McConnell
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Caroline Rinderle
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Katherine L Hebert
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Maninder Khosla
- Department of Biochemistry and Molecular Biology, LSU Health Science Center School of Medicine, New Orleans, LA, 70112, USA
| | - Jorge A Belgodere
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
- Department of Biological and Agricultural Engineering, Louisiana State University and Agricultural Center, Baton Rouge, LA, 70803, USA
| | - Van T Hoang
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Bridgette M Collins-Burow
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Bruce A Bunnell
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Matthew E Burow
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, 70112, USA.
| | - Suresh K Alahari
- Department of Biochemistry and Molecular Biology, LSU Health Science Center School of Medicine, New Orleans, LA, 70112, USA.
- Stanley S. Scott Cancer Center, LSU Health Science Center School of Medicine, New Orleans, LA, 70112, USA.
| |
Collapse
|
16
|
Almohtasib Y, Fancher AJ, Sawalha K. Emerging Trends in Atherosclerosis: Time to Address Atherosclerosis From a Younger Age. Cureus 2024; 16:e56635. [PMID: 38646335 PMCID: PMC11032087 DOI: 10.7759/cureus.56635] [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] [Accepted: 03/21/2024] [Indexed: 04/23/2024] Open
Abstract
Over the past two decades, research efforts into cardiovascular disease (CVD) have uncovered findings that fundamentally challenge our understanding of CVD, particularly atherosclerosis. Atherosclerosis was primarily attributed to the well-described abnormal lipid accumulation theory, involving plaque growth with subsequent plaque hemorrhage resulting in acute vessel thrombosis that may or may not rupture. This perspective has now evolved to encompass more complex pathways, wherein the accumulation of abnormal products of oxidation and inflammation is the most likely factor mediating atherosclerotic plaque growth. Furthermore, atherosclerosis was traditionally thought of as a disease in patients aged 40 and older. However, mounting evidence has demonstrated that significant atherosclerosis and CVD events are more prevalent in younger patients than previously realized and accelerating in incidence. With this alarming trend among younger individuals, our review sought to explore why this trend may be happening and what can be done about this developing problem.
Collapse
Affiliation(s)
- Yazan Almohtasib
- Internal Medicine, University of Missouri Kansas City School of Medicine, Kansas City, USA
| | - Andrew J Fancher
- Internal Medicine, University of Kansas School of Medicine-Wichita, Wichita, USA
| | - Khalid Sawalha
- Cardiometabolic Medicine, University of Missouri Kansas City School of Medicine, Kansas City, USA
| |
Collapse
|
17
|
Greeny A, Nair A, Sadanandan P, Satarker S, Famurewa AC, Nampoothiri M. Epigenetic Alterations in Alzheimer's Disease: Impact on Insulin Signaling and Advanced Drug Delivery Systems. BIOLOGY 2024; 13:157. [PMID: 38534427 DOI: 10.3390/biology13030157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 02/25/2024] [Accepted: 02/26/2024] [Indexed: 03/28/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative condition that predominantly affects the hippocampus and the entorhinal complex, leading to memory lapse and cognitive impairment. This can have a negative impact on an individual's behavior, speech, and ability to navigate their surroundings. AD is one of the principal causes of dementia. One of the most accepted theories in AD, the amyloid β (Aβ) hypothesis, assumes that the buildup of the peptide Aβ is the root cause of AD. Impaired insulin signaling in the periphery and central nervous system has been considered to have an effect on the pathophysiology of AD. Further, researchers have shifted their focus to epigenetic mechanisms that are responsible for dysregulating major biochemical pathways and intracellular signaling processes responsible for directly or indirectly causing AD. The prime epigenetic mechanisms encompass DNA methylation, histone modifications, and non-coding RNA, and are majorly responsible for impairing insulin signaling both centrally and peripherally, thus leading to AD. In this review, we provide insights into the major epigenetic mechanisms involved in causing AD, such as DNA methylation and histone deacetylation. We decipher how the mechanisms alter peripheral insulin signaling and brain insulin signaling, leading to AD pathophysiology. In addition, this review also discusses the need for newer drug delivery systems for the targeted delivery of epigenetic drugs and explores targeted drug delivery systems such as nanoparticles, vesicular systems, networks, and other nano formulations in AD. Further, this review also sheds light on the future approaches used for epigenetic drug delivery.
Collapse
Affiliation(s)
- Alosh Greeny
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Ayushi Nair
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Amrita Health Science Campus, Kochi 682041, India
| | - Prashant Sadanandan
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Amrita Health Science Campus, Kochi 682041, India
| | - Sairaj Satarker
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Ademola C Famurewa
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Medical Sciences, Alex Ekwueme Federal University, Ndufu-Alike, Ikwo 482123, Nigeria
| | - Madhavan Nampoothiri
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| |
Collapse
|
18
|
Cheng C, Yu F, Yuan G, Jia J. Update on N6-methyladenosine methylation in obesity-related diseases. Obesity (Silver Spring) 2024; 32:240-251. [PMID: 37989724 DOI: 10.1002/oby.23932] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 08/28/2023] [Accepted: 08/28/2023] [Indexed: 11/23/2023]
Abstract
Obesity is a chronic metabolic disease that is closely related to type 2 diabetes mellitus, cardiovascular diseases, nonalcoholic fatty liver disease, obstructive sleep apnea, and osteoarthritis. The prevalence of obesity is increasing rapidly every year and is recognized as a global public health problem. In recent years, the role of epigenetics in the development of obesity and related diseases has been recognized and is currently a research hotspot. N6-methyladenosine (m6A) methylation is the most abundant epigenetic modification in the eukaryotic RNA, including mRNA and noncoding RNA. Several studies have shown that the m6A modifications in the target mRNA and the corresponding m6A regulators play a significant role in lipid metabolism and are strongly associated with the pathogenesis of obesity-related diseases. In this review, the latest research findings regarding the role of m6A methylation in obesity and related metabolic diseases are summarized. The authors' aim is to highlight evidence that suggests the clinical utility of m6A modifications and the m6A regulators as novel early prediction biomarkers and precision therapeutics for obesity and obesity-related diseases.
Collapse
Affiliation(s)
- Caiqin Cheng
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Jiangsu University; Institute of Endocrine and Metabolic Diseases, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Fan Yu
- Department of Endocrinology and Metabolism, Jurong Hospital Affiliated to Jiangsu University, Zhenjiang, Jiangsu, China
| | - Guoyue Yuan
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Jiangsu University; Institute of Endocrine and Metabolic Diseases, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Jue Jia
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Jiangsu University; Institute of Endocrine and Metabolic Diseases, Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
19
|
Egashira K, Kajiya H, Tsutsumi T, Taniguchi Y, Kakura K, Ohno J, Kido H. AMPK activation enhances osteoblast differentiation on a titanium disc via autophagy. Int J Implant Dent 2024; 10:2. [PMID: 38286943 PMCID: PMC10825085 DOI: 10.1186/s40729-024-00525-2] [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: 10/05/2023] [Accepted: 01/18/2024] [Indexed: 01/31/2024] Open
Abstract
PURPOSE The acquisition of osseointegration during implant therapy is slower and poorer in patients with diabetes compared with healthy persons. The serum concentration of adiponectin in patients with type II diabetes is lower than that of healthy persons via the suppression of AMP-activated protein kinase (AMPK). Therefore, we hypothesized that the AMPK activation enhances bone formation around implants, resulting in the improved acquisition of osseointegration. The purpose of this study was to evaluate the impact of AMPK activation on osteoblast differentiation and its mechanism of downstream signaling on titanium disc (Ti). METHODS Confluent mouse pre-osteoblasts (MC3T3-E1) cells (1 × 105 cells/well) were cultured with BMP-2 for osteoblast differentiation, in the presence or absence AICAR, an AMPK activator. We examined the effects of AMPK activation on osteoblast differentiation and the underlying mechanism on a Ti using a CCK8 assay, a luciferase assay, quantitative RT-PCR, and western blotting. RESULTS Although the proliferation rate of osteoblasts was not different between a Ti and a tissue culture polystyrene dish, the addition of AICAR, AMPK activator slightly enhanced osteoblast proliferation on the Ti. AICAR enhanced the BMP-2-dependent transcriptional activity on the Ti, leading to upregulation in the expression of osteogenesis-associated molecules. AICAR simultaneously upregulated the expression of autophagy-associated molecules on the Ti, especially LC3-II. AdipoRon, an adiponectin receptor type1/type2 activator activated AMPK, and upregulated osteogenesis-associated molecules on Ti. CONCLUSIONS AMPK activation enhances osteoblast differentiation on a Ti via autophagy, suggesting that it promotes the acquisition of osseointegration during implant therapy.
Collapse
Affiliation(s)
- Kei Egashira
- Section of Oral Implantology, Department of Oral Rehabilitation, Fukuoka Dental College, Fukuoka, Japan
- Oral Medicine Research Center, Fukuoka Dental College, Fukuoka, Japan
| | - Hiroshi Kajiya
- Oral Medicine Research Center, Fukuoka Dental College, Fukuoka, Japan.
- Department of Physiological Science and Molecular Biology, Fukuoka Dental College, Fukuoka, 814-0193, Japan.
| | - Takashi Tsutsumi
- Oral Medicine Research Center, Fukuoka Dental College, Fukuoka, Japan
- Department of General Dentistry, Fukuoka Dental College, Fukuoka, Japan
| | - Yusuke Taniguchi
- Section of Oral Implantology, Department of Oral Rehabilitation, Fukuoka Dental College, Fukuoka, Japan
| | - Kae Kakura
- Section of Oral Implantology, Department of Oral Rehabilitation, Fukuoka Dental College, Fukuoka, Japan
| | - Jun Ohno
- Oral Medicine Research Center, Fukuoka Dental College, Fukuoka, Japan
| | - Hirofumi Kido
- Section of Oral Implantology, Department of Oral Rehabilitation, Fukuoka Dental College, Fukuoka, Japan
| |
Collapse
|
20
|
Taylor BC, Steinthal LH, Dias M, Yalamanchili HK, Ochsner SA, Zapata GE, Mehta NR, McKenna NJ, Young NL, Nuotio-Antar AM. Histone proteoform analysis reveals epigenetic changes in adult mouse brown adipose tissue in response to cold stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.30.551059. [PMID: 38328142 PMCID: PMC10849524 DOI: 10.1101/2023.07.30.551059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Regulation of the thermogenic response by brown adipose tissue (BAT) is an important component of energy homeostasis with implications for the treatment of obesity and diabetes. Our preliminary analyses uncovered many nodes representing epigenetic modifiers that are altered in BAT in response to chronic thermogenic activation. Thus, we hypothesized that chronic thermogenic activation broadly alters epigenetic modifications of DNA and histones in BAT. Motivated to understand how BAT function is regulated epigenetically, we developed a novel method for the first-ever unbiased top-down proteomic quantitation of histone modifications in BAT and validated our results with a multi-omic approach. To test our hypothesis, wildtype male C57BL/6J mice were housed under chronic conditions of thermoneutral temperature (TN, 28.8°C), mild cold/room temperature (RT, 22°C), or severe cold (SC, 8°C) and BAT was analyzed for DNA methylation and histone modifications. Methylation of promoters and intragenic regions in genomic DNA decrease in response to chronic cold exposure. Integration of DNA methylation and RNA expression data suggest a role for epigenetic modification of DNA in gene regulation in response to cold. In response to cold housing, we observe increased bulk acetylation of histones H3.2 and H4, increased histone H3.2 proteoforms with di- and trimethylation of lysine 9 (K9me2 and K9me3), and increased histone H4 proteoforms with acetylation of lysine 16 (K16ac) in BAT. Taken together, our results reveal global epigenetically-regulated transcriptional "on" and "off" signals in murine BAT in response to varying degrees of chronic cold stimuli and establish a novel methodology to quantitatively study histones in BAT, allowing for direct comparisons to decipher mechanistic changes during the thermogenic response. Additionally, we make histone PTM and proteoform quantitation, RNA splicing, RRBS, and transcriptional footprint datasets available as a resource for future research.
Collapse
Affiliation(s)
- Bethany C. Taylor
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX
| | - Loic H. Steinthal
- Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX
| | - Michelle Dias
- Jan and Dan Duncan Neurological Research Institute, Baylor College of Medicine, Houston, TX
| | - Hari K. Yalamanchili
- Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX
- Jan and Dan Duncan Neurological Research Institute, Baylor College of Medicine, Houston, TX
| | - Scott A. Ochsner
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
| | - Gladys E. Zapata
- Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX
| | - Nitesh R. Mehta
- Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX
| | - Neil J. McKenna
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
| | - Nicolas L. Young
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
| | | |
Collapse
|
21
|
Tao Z, Wang Y. The health benefits of dietary short-chain fatty acids in metabolic diseases. Crit Rev Food Sci Nutr 2024; 65:1579-1592. [PMID: 38189336 DOI: 10.1080/10408398.2023.2297811] [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: 01/09/2024]
Abstract
Short-chain fatty acids (SCFAs) are a subset of fatty acids that play crucial roles in maintaining normal physiology and developing metabolic diseases, such as obesity, diabetes, cardiovascular disease, and liver disease. Even though dairy products and vegetable oils are the direct dietary sources of SCFAs, their quantities are highly restricted. SCFAs are produced indirectly through microbial fermentation of fibers. The biological roles of SCFAs in human health and metabolic diseases are mainly due to their receptors, GPR41 and GPR43, FFAR2 and FFAR3. Additionally, it has been demonstrated that SCFAs modulate DNMTs and HDAC activities, inhibit NF-κB-STAT signaling, and regulate G(i/o)βγ-PLC-PKC-PTEN signaling and PPARγ-UCP2-AMPK autophagic signaling, thus mitigating metabolic diseases. Recent studies have uncovered that SCFAs play crucial roles in epigenetic modifications of DNAs, RNAs, and post-translational modifications of proteins, which are critical regulators of metabolic health and diseases. At the same time, dietary recommendations for the purpose of SCFAs have been proposed. The objective of the review is to summarize the most recent research on the role of dietary SCFAs in metabolic diseases, especially the signal transduction of SCFAs in metabolic diseases and their functional efficacy in different backgrounds and models of metabolic diseases, at the same time, to provide dietary and nutritional recommendations for using SCFAs as food ingredients to prevent metabolic diseases.
Collapse
Affiliation(s)
- Zhipeng Tao
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
- Department of Nutrition Sciences, Texas Woman's University, Denton, Texas, USA
| | - Yao Wang
- Diabetes Center, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
22
|
Engin A. Adiponectin Resistance in Obesity: Adiponectin Leptin/Insulin Interaction. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:431-462. [PMID: 39287861 DOI: 10.1007/978-3-031-63657-8_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The adiponectin (APN) levels in obesity are negatively correlated with chronic subclinical inflammation markers. The hypertrophic adipocytes cause obesity-linked insulin resistance and metabolic syndrome. Furthermore, macrophage polarization is a key determinant regulating adiponectin receptor (AdipoR1/R2) expression and differential adiponectin-mediated macrophage inflammatory responses in obese individuals. In addition to decrease in adiponectin concentrations, the decline in AdipoR1/R2 messenger ribonucleic acid (mRNA) expression leads to a decrement in adiponectin binding to cell membrane, and this turns into attenuation in the adiponectin effects. This is defined as APN resistance, and it is linked with insulin resistance in high-fat diet-fed subjects. The insulin-resistant group has a significantly higher leptin-to-APN ratio. The leptin-to-APN ratio is more than twofold higher in obese individuals. An increase in expression of AdipoRs restores insulin sensitivity and β-oxidation of fatty acids via triggering intracellular signal cascades. The ratio of high molecular weight to total APN is defined as the APN sensitivity index (ASI). This index is correlated to insulin sensitivity. Homeostasis model of assessment (HOMA)-APN and HOMA-estimated insulin resistance (HOMA-IR) are the most suitable methods to estimate the metabolic risk in metabolic syndrome. While morbidly obese patients display a significantly higher plasma leptin and soluble (s)E-selectin concentrations, leptin-to-APN ratio, there is a significant negative correlation between leptin-to-APN ratio and sP-selectin in obese patients. When comparing the metabolic dysregulated obese group with the metabolically healthy obese group, postprandial triglyceride clearance, insulin resistance, and leptin resistance are significantly delayed following the oral fat tolerance test in the first group. A neuropeptide, Spexin (SPX), is positively correlated with the quantitative insulin sensitivity check index (QUICKI) and APN. APN resistance together with insulin resistance forms a vicious cycle. Despite normal or high APN levels, an impaired post-receptor signaling due to adaptor protein-containing pleckstrin homology domain, phosphotyrosine-binding domain, and leucine zipper motif 1 (APPL1)/APPL2 may alter APN efficiency and activity. However, APPL2 blocks adiponectin signaling through AdipoR1 and AdipoR2 because of the competitive inhibition of APPL1. APPL1, the intracellular binding partner of AdipoRs, is also an important mediator of adiponectin-dependent insulin sensitization. The elevated adiponectin levels with adiponectin resistance are compensatory responses in the condition of an unusual discordance between insulin resistance and APN unresponsiveness. Hypothalamic recombinant adeno-associated virus (rAAV)-leptin (Lep) gene therapy reduces serum APN levels, and it is a more efficient strategy for long-term weight maintenance.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
23
|
Avtanski D, Stojchevski R. Significance of Adipose Tissue as an Endocrine Organ. CONTEMPORARY ENDOCRINOLOGY 2024:1-46. [DOI: 10.1007/978-3-031-72570-8_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
24
|
Kamiński M, Mierzyński R, Poniedziałek-Czajkowska E, Sadowska A, Sotowski M, Leszczyńska-Gorzelak B. Comparative Evaluation of Adipokine Metrics for the Diagnosis of Gestational Diabetes Mellitus. Int J Mol Sci 2023; 25:175. [PMID: 38203346 PMCID: PMC10778639 DOI: 10.3390/ijms25010175] [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: 11/24/2023] [Revised: 12/19/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
Gestational diabetes mellitus (GDM) is one of the most common medical disorders in pregnancy. Adipokines, predominantly secreted by adipose tissue, are involved in numerous metabolic processes. The exact role of adipokines in the pathogenesis of GDM is still not well known, and numerous adipokines have been analysed throughout pregnancy and proposed as biomarkers of GDM. This study aimed to evaluate serum adiponectin, chemerin, lipocalin and apelin levels in GDM and non-GDM women, to assess them as clinically useful biomarkers of the occurrence of GDM and to demonstrate the correlation between the levels of the above adipokines in the blood serum and the increased risk of the development of GDM. The role of these adipokines in the pathogenesis of GDM was also analysed. The statistically significant differences between the levels of adiponectin (7234.6 vs. 9837.5 ng/mL, p < 0.0001), chemerin (264.0 vs. 206.7 ng/mL, p < 0.0001) and lipocalin (39.5 vs. 19.4 ng/mL, p < 0.0001) were observed between pregnant women with GDM and healthy ones. The diagnostic usefulness of the tested adipokines in detecting GDM was also assessed. The research results confirm the hypothesis on the significance of adiponectin, chemerin, lipocalin and apelin in the pathophysiological mechanisms of GDM. We speculate that these adipokines could potentially be established as novel biomarkers for the prediction and early diagnosis of GDM.
Collapse
Affiliation(s)
| | - Radzisław Mierzyński
- Chair and Department of Obstetrics and Perinatology, Medical University of Lublin, 20-954 Lublin, Poland; (M.K.); (A.S.); (M.S.); (B.L.-G.)
| | - Elżbieta Poniedziałek-Czajkowska
- Chair and Department of Obstetrics and Perinatology, Medical University of Lublin, 20-954 Lublin, Poland; (M.K.); (A.S.); (M.S.); (B.L.-G.)
| | | | | | | |
Collapse
|
25
|
Santos-Pereira M, Pereira SC, Rebelo I, Spadella MA, Oliveira PF, Alves MG. Decoding the Influence of Obesity on Prostate Cancer and Its Transgenerational Impact. Nutrients 2023; 15:4858. [PMID: 38068717 PMCID: PMC10707940 DOI: 10.3390/nu15234858] [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: 10/18/2023] [Revised: 11/12/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
In recent decades, the escalating prevalence of metabolic disorders, notably obesity and being overweight, has emerged as a pressing concern in public health. Projections for the future indicate a continual upward trajectory in obesity rates, primarily attributable to unhealthy dietary patterns and sedentary lifestyles. The ramifications of obesity extend beyond its visible manifestations, intricately weaving a web of hormonal dysregulation, chronic inflammation, and oxidative stress. This nexus of factors holds particular significance in the context of carcinogenesis, notably in the case of prostate cancer (PCa), which is a pervasive malignancy and a leading cause of mortality among men. A compelling hypothesis arises from the perspective of transgenerational inheritance, wherein genetic and epigenetic imprints associated with obesity may wield influence over the development of PCa. This review proposes a comprehensive exploration of the nuanced mechanisms through which obesity disrupts prostate homeostasis and serves as a catalyst for PCa initiation. Additionally, it delves into the intriguing interplay between the transgenerational transmission of both obesity-related traits and the predisposition to PCa. Drawing insights from a spectrum of sources, ranging from in vitro and animal model research to human studies, this review endeavors to discuss the intricate connections between obesity and PCa. However, the landscape remains partially obscured as the current state of knowledge unveils only fragments of the complex mechanisms linking these phenomena. As research advances, unraveling the associated factors and underlying mechanisms promises to unveil novel avenues for understanding and potentially mitigating the nexus between obesity and the development of PCa.
Collapse
Affiliation(s)
- Mariana Santos-Pereira
- iBiMED-Institute of Biomedicine and Department of Medical Science, University of Aveiro, 3810-193 Aveiro, Portugal;
- Endocrine and Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), School of Medicine and Biomedical Sciences (ICBAS), University of Porto, 4050-313 Porto, Portugal;
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, 4099-002 Porto, Portugal
| | - Sara C. Pereira
- Endocrine and Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), School of Medicine and Biomedical Sciences (ICBAS), University of Porto, 4050-313 Porto, Portugal;
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, 4099-002 Porto, Portugal
- LAQV-REQUIMTE and Department of Chemistry, Campus Universitario de Santiago, University of Aveiro, 3810-193 Aveiro, Portugal;
- Department of Pathology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Irene Rebelo
- UCIBIO-REQUIMTE, Laboratory of Biochemistry, Department of Biologic Sciences, Pharmaceutical Faculty, University of Porto, 4050-313 Porto, Portugal;
| | - Maria A. Spadella
- Human Embryology Laboratory, Marília Medical School, Marília 17519-030, SP, Brazil;
| | - Pedro F. Oliveira
- LAQV-REQUIMTE and Department of Chemistry, Campus Universitario de Santiago, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Marco G. Alves
- iBiMED-Institute of Biomedicine and Department of Medical Science, University of Aveiro, 3810-193 Aveiro, Portugal;
| |
Collapse
|
26
|
Guo YC, Cao HD, Lian XF, Wu PX, Zhang F, Zhang H, Lu DH. Molecular mechanisms of noncoding RNA and epigenetic regulation in obesity with consequent diabetes mellitus development. World J Diabetes 2023; 14:1621-1631. [DOI: 10.4239/wjd.v14.i11.1621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/26/2023] [Accepted: 09/27/2023] [Indexed: 11/14/2023] Open
Abstract
Diabetes mellitus (DM) and obesity have become two of the most prevalent and challenging diseases worldwide, with increasing incidence and serious complications. Recent studies have shown that noncoding RNA (ncRNA) and epigenetic regulation play crucial roles in the pathogenesis of DM complicated by obesity. Identification of the involvement of ncRNA and epigenetic regulation in the pathogenesis of diabetes with obesity has opened new avenues of investigation. Targeting these mechanisms with small molecules or RNA-based therapies may provide a more precise and effective approach to diabetes treatment than traditional therapies. In this review, we discuss the molecular mechanisms of ncRNA and epigenetic regulation and their potential therapeutic targets, and the research prospects for DM complicated with obesity.
Collapse
Affiliation(s)
- Yi-Chen Guo
- Department of Endo-crinology, Peking University Shenzhen Hospital, Shenzhen 518036, Guangdong Province, China
- Department of Endocrinology, Zhujiang Hospital of Southern Medical University, Guangzhou 510282, Guangdong Province, China
| | - Hao-Di Cao
- Department of Endocrinology, Zhujiang Hospital of Southern Medical University, Guangzhou 510282, Guangdong Province, China
| | - Xiao-Fen Lian
- Department of Endo-crinology, Peking University Shenzhen Hospital, Shenzhen 518036, Guangdong Province, China
| | - Pei-Xian Wu
- Department of Endo-crinology, Peking University Shenzhen Hospital, Shenzhen 518036, Guangdong Province, China
| | - Fan Zhang
- Department of Endo-crinology, Peking University Shenzhen Hospital, Shenzhen 518036, Guangdong Province, China
| | - Hua Zhang
- Department of Endocrinology, Zhujiang Hospital of Southern Medical University, Guangzhou 510282, Guangdong Province, China
| | - Dong-Hui Lu
- Department of Endo-crinology, Peking University Shenzhen Hospital, Shenzhen 518036, Guangdong Province, China
| |
Collapse
|
27
|
Neto A, Fernandes A, Barateiro A. The complex relationship between obesity and neurodegenerative diseases: an updated review. Front Cell Neurosci 2023; 17:1294420. [PMID: 38026693 PMCID: PMC10665538 DOI: 10.3389/fncel.2023.1294420] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Obesity is a global epidemic, affecting roughly 30% of the world's population and predicted to rise. This disease results from genetic, behavioral, societal, and environmental factors, leading to excessive fat accumulation, due to insufficient energy expenditure. The adipose tissue, once seen as a simple storage depot, is now recognized as a complex organ with various functions, including hormone regulation and modulation of metabolism, inflammation, and homeostasis. Obesity is associated with a low-grade inflammatory state and has been linked to neurodegenerative diseases like multiple sclerosis (MS), Alzheimer's (AD), and Parkinson's (PD). Mechanistically, reduced adipose expandability leads to hypertrophic adipocytes, triggering inflammation, insulin and leptin resistance, blood-brain barrier disruption, altered brain metabolism, neuronal inflammation, brain atrophy, and cognitive decline. Obesity impacts neurodegenerative disorders through shared underlying mechanisms, underscoring its potential as a modifiable risk factor for these diseases. Nevertheless, further research is needed to fully grasp the intricate connections between obesity and neurodegeneration. Collaborative efforts in this field hold promise for innovative strategies to address this complex relationship and develop effective prevention and treatment methods, which also includes specific diets and physical activities, ultimately improving quality of life and health.
Collapse
Affiliation(s)
- Alexandre Neto
- Central Nervous System, Blood and Peripheral Inflammation, Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Adelaide Fernandes
- Central Nervous System, Blood and Peripheral Inflammation, Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Andreia Barateiro
- Central Nervous System, Blood and Peripheral Inflammation, Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
28
|
Jung BC, You D, Lee I, Li D, Schill RL, Ma K, Pi A, Song Z, Mu WC, Wang T, MacDougald OA, Banks AS, Kang S. TET3 plays a critical role in white adipose development and diet-induced remodeling. Cell Rep 2023; 42:113196. [PMID: 37777963 PMCID: PMC10763978 DOI: 10.1016/j.celrep.2023.113196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 07/28/2023] [Accepted: 09/14/2023] [Indexed: 10/03/2023] Open
Abstract
Maintaining healthy adipose tissue is crucial for metabolic health, requiring a deeper understanding of adipocyte development and response to high-calorie diets. This study highlights the importance of TET3 during white adipose tissue (WAT) development and expansion. Selective depletion of Tet3 in adipose precursor cells (APCs) reduces adipogenesis, protects against diet-induced adipose expansion, and enhances whole-body metabolism. Transcriptomic analysis of wild-type and Tet3 knockout (KO) APCs unveiled TET3 target genes, including Pparg and several genes linked to the extracellular matrix, pivotal for adipogenesis and remodeling. DNA methylation profiling and functional studies underscore the importance of DNA demethylation in gene regulation. Remarkably, targeted DNA demethylation at the Pparg promoter restored its transcription. In conclusion, TET3 significantly governs adipogenesis and diet-induced adipose expansion by regulating key target genes in APCs.
Collapse
Affiliation(s)
- Byung Chul Jung
- Nutritional Sciences and Toxicology Department, University of California Berkeley, Berkeley, CA, USA
| | - Dongjoo You
- Nutritional Sciences and Toxicology Department, University of California Berkeley, Berkeley, CA, USA
| | - Ikjun Lee
- Nutritional Sciences and Toxicology Department, University of California Berkeley, Berkeley, CA, USA
| | - Daofeng Li
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA; The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Rebecca L Schill
- Department of Molecular & Integrative Physiology, University of Michigan School of Medicine, Ann Arbor, MO, USA
| | - Katherine Ma
- Nutritional Sciences and Toxicology Department, University of California Berkeley, Berkeley, CA, USA
| | - Anna Pi
- Nutritional Sciences and Toxicology Department, University of California Berkeley, Berkeley, CA, USA
| | - Zehan Song
- Nutritional Sciences and Toxicology Department, University of California Berkeley, Berkeley, CA, USA
| | - Wei-Chieh Mu
- Nutritional Sciences and Toxicology Department, University of California Berkeley, Berkeley, CA, USA
| | - Ting Wang
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA; The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ormond A MacDougald
- Department of Molecular & Integrative Physiology, University of Michigan School of Medicine, Ann Arbor, MO, USA
| | - Alexander S Banks
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Sona Kang
- Nutritional Sciences and Toxicology Department, University of California Berkeley, Berkeley, CA, USA.
| |
Collapse
|
29
|
Abstract
Obesity is a common complex trait that elevates the risk for various diseases, including type 2 diabetes and cardiovascular disease. A combination of environmental and genetic factors influences the pathogenesis of obesity. Advances in genomic technologies have driven the identification of multiple genetic loci associated with this disease, ranging from studying severe onset cases to investigating common multifactorial polygenic forms. Additionally, findings from epigenetic analyses of modifications to the genome that do not involve changes to the underlying DNA sequence have emerged as key signatures in the development of obesity. Such modifications can mediate the effects of environmental factors, including diet and lifestyle, on gene expression and clinical presentation. This review outlines what is known about the genetic and epigenetic contributors to obesity susceptibility, along with the albeit limited therapeutic options currently available. Furthermore, we delineate the potential mechanisms of actions through which epigenetic changes can mediate environmental influences and the related opportunities they present for future interventions in the management of obesity.
Collapse
Affiliation(s)
- Khanh Trang
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104 USA
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104 USA
| | - Struan F.A. Grant
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104 USA
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104 USA
- Division of Diabetes and Endocrinology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104 USA
- Department of Pediatrics, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104 USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104 USA
| |
Collapse
|
30
|
Yang Z, Kubant R, Cho CE, Kranenburg E, Beaudry J, Bottiglieri T, Anderson GH. Micronutrients in High-Fat Diet Modify Insulin Resistance and Its Regulatory Genes in Adult Male Mice. Mol Nutr Food Res 2023; 67:e2300199. [PMID: 37526337 DOI: 10.1002/mnfr.202300199] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/13/2023] [Indexed: 08/02/2023]
Abstract
SCOPE Obesity and insulin resistance (IR) are associated with epigenetic changes of gene expression. However, the relationship between micronutrients, epigenetic regulation of gene expression, and IR during development of diet-induced obesity has yet to be defined. Our objective is to describe the effect of micronutrient addition to diets on IR and its related genes during obesity development. METHODS AND RESULTS Male C57BL/6J mice are fed a high-fat (HFD) or low-fat (LFD) diets with or without a multi-vitamin mineral mix (MVM) addition containing vitamins A, B1, B6, B12, and Zn, and Se for 9 weeks. Compared to LFD mice, HFD mice have higher body weight, IR, fasting glucose, insulin, C-peptide, leptin, and hepatic triglyceride concentrations, and dysregulated gene expression in liver, muscle, pancreas, and fat tissues (p < 0.05). The addition of MVM reduces these HFD-induced effects. HFD downregulates 27 genes associated with insulin regulation and adipose tissue function across all tissues by an average of 47% and upregulates five genes by 230% (p < 0.001). Adding MVM downregulates five genes and upregulates one in HFD-fed mice. Both HFD and MVM alter one-carbon metabolites. CONCLUSION Addition of micronutrients to the HFD decreases IR and modifies associated gene expression in obese and lean mice.
Collapse
Affiliation(s)
- Zeyu Yang
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Ruslan Kubant
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Clara E Cho
- Department of Human Health and Nutritional Sciences, College of Biological Science, University of Guelph, Guelph, Ontario, Canada
| | - Eva Kranenburg
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Jacqueline Beaudry
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Teodoro Bottiglieri
- Institute of Metabolic Disease, Baylor Scott & White Health, Austin, TX, USA
| | - G Harvey Anderson
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
31
|
Oh J, Riek AE, Bauerle KT, Dusso A, McNerney KP, Barve RA, Darwech I, Sprague JE, Moynihan C, Zhang RM, Kutz G, Wang T, Xing X, Li D, Mrad M, Wigge NM, Castelblanco E, Collin A, Bambouskova M, Head RD, Sands MS, Bernal-Mizrachi C. Embryonic vitamin D deficiency programs hematopoietic stem cells to induce type 2 diabetes. Nat Commun 2023; 14:3278. [PMID: 37311757 PMCID: PMC10264405 DOI: 10.1038/s41467-023-38849-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 05/19/2023] [Indexed: 06/15/2023] Open
Abstract
Environmental factors may alter the fetal genome to cause metabolic diseases. It is unknown whether embryonic immune cell programming impacts the risk of type 2 diabetes in later life. We demonstrate that transplantation of fetal hematopoietic stem cells (HSCs) made vitamin D deficient in utero induce diabetes in vitamin D-sufficient mice. Vitamin D deficiency epigenetically suppresses Jarid2 expression and activates the Mef2/PGC1a pathway in HSCs, which persists in recipient bone marrow, resulting in adipose macrophage infiltration. These macrophages secrete miR106-5p, which promotes adipose insulin resistance by repressing PIK3 catalytic and regulatory subunits and down-regulating AKT signaling. Vitamin D-deficient monocytes from human cord blood have comparable Jarid2/Mef2/PGC1a expression changes and secrete miR-106b-5p, causing adipocyte insulin resistance. These findings suggest that vitamin D deficiency during development has epigenetic consequences impacting the systemic metabolic milieu.
Collapse
Affiliation(s)
- Jisu Oh
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Amy E Riek
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Kevin T Bauerle
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Medicine, VA Medical Center, St. Louis, MO, USA
| | - Adriana Dusso
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Kyle P McNerney
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Ruteja A Barve
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Isra Darwech
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Clare Moynihan
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Rong M Zhang
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Greta Kutz
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Ting Wang
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Xiaoyun Xing
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Daofeng Li
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Marguerite Mrad
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Nicholas M Wigge
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Alejandro Collin
- Instituto de Investigaciones en Ciencias de la Salud (INICSA), CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Monika Bambouskova
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Richard D Head
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Mark S Sands
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Carlos Bernal-Mizrachi
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Medicine, VA Medical Center, St. Louis, MO, USA.
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
32
|
Vrânceanu M, Hegheş SC, Cozma-Petruţ A, Banc R, Stroia CM, Raischi V, Miere D, Popa DS, Filip L. Plant-Derived Nutraceuticals Involved in Body Weight Control by Modulating Gene Expression. PLANTS (BASEL, SWITZERLAND) 2023; 12:2273. [PMID: 37375898 DOI: 10.3390/plants12122273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/09/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023]
Abstract
Obesity is the most prevalent health problem in the Western world, with pathological body weight gain associated with numerous co-morbidities that can be the main cause of death. There are several factors that can contribute to the development of obesity, such as diet, sedentary lifestyle, and genetic make-up. Genetic predispositions play an important role in obesity, but genetic variations alone cannot fully explain the explosion of obesity, which is why studies have turned to epigenetics. The latest scientific evidence suggests that both genetics and environmental factors contribute to the rise in obesity. Certain variables, such as diet and exercise, have the ability to alter gene expression without affecting the DNA sequence, a phenomenon known as epigenetics. Epigenetic changes are reversible, and reversibility makes these changes attractive targets for therapeutic interventions. While anti-obesity drugs have been proposed to this end in recent decades, their numerous side effects make them not very attractive. On the other hand, the use of nutraceuticals for weight loss is increasing, and studies have shown that some of these products, such as resveratrol, curcumin, epigallocatechin-3-gallate, ginger, capsaicin, and caffeine, can alter gene expression, restoring the normal epigenetic profile and aiding weight loss.
Collapse
Affiliation(s)
- Maria Vrânceanu
- Department of Toxicology, "Iuliu Haţieganu" University of Medicine and Pharmacy, 6 Pasteur Street, 400349 Cluj-Napoca, Romania
| | - Simona-Codruţa Hegheş
- Department of Drug Analysis, "Iuliu Haţieganu" University of Medicine and Pharmacy, 6 Pasteur Street, 400349 Cluj-Napoca, Romania
| | - Anamaria Cozma-Petruţ
- Department of Bromatology, Hygiene, Nutrition, "Iuliu Haţieganu" University of Medicine and Pharmacy, 6 Pasteur Street, 400349 Cluj-Napoca, Romania
| | - Roxana Banc
- Department of Bromatology, Hygiene, Nutrition, "Iuliu Haţieganu" University of Medicine and Pharmacy, 6 Pasteur Street, 400349 Cluj-Napoca, Romania
| | - Carmina Mariana Stroia
- Department of Pharmacy, Oradea University, 1 Universităţii Street, 410087 Oradea, Romania
| | - Viorica Raischi
- Laboratory of Physiology of Stress, Adaptation and General Sanocreatology, Institute of Physiology and Sanocreatology, 1 Academiei Street, 2028 Chișinău, Moldova
| | - Doina Miere
- Department of Bromatology, Hygiene, Nutrition, "Iuliu Haţieganu" University of Medicine and Pharmacy, 6 Pasteur Street, 400349 Cluj-Napoca, Romania
| | - Daniela-Saveta Popa
- Department of Toxicology, "Iuliu Haţieganu" University of Medicine and Pharmacy, 6 Pasteur Street, 400349 Cluj-Napoca, Romania
| | - Lorena Filip
- Department of Bromatology, Hygiene, Nutrition, "Iuliu Haţieganu" University of Medicine and Pharmacy, 6 Pasteur Street, 400349 Cluj-Napoca, Romania
| |
Collapse
|
33
|
Sawalha K, Norgard N, López-Candales A. Epigenetic Regulation and its Effects on Aging and Cardiovascular Disease. Cureus 2023; 15:e39395. [PMID: 37362531 PMCID: PMC10286850 DOI: 10.7759/cureus.39395] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2023] [Indexed: 06/28/2023] Open
Abstract
Cardiovascular disease (CVD), specifically coronary atherosclerosis, is regulated by an interplay between genetic and lifestyle factors. Most recently, a factor getting much attention is the role epigenetics play in atherosclerosis; particularly the development of coronary artery disease. Furthermore, it is important to understand the intricate interaction between the environment and each individual genetic material and how this interaction affects gene expression and consequently influences the development of atherosclerosis. Our main goal is to discuss epigenetic regulations; particularly, the factors contributing to coronary atherosclerosis and their role in aging and longevity. We reviewed the current literature and provided a simplified yet structured and reasonable appraisal of this topic. This role has also been recently linked to longevity and aging. Epigenetic regulations (modifications) whether through histone modifications or DNA or RNA methylation have been shown to be regulated by environmental factors such as social stress, smoking, chemical contaminants, and diet. These sensitive interactions are further aggravated by racial health disparities that ultimately impact cardiovascular disease outcomes through epigenetic interactions. Certainly, limiting our exposure to such causative events at younger ages seems our "golden opportunity" to tackle the incidence of coronary atherosclerosis and probably the answer to longevity.
Collapse
Affiliation(s)
- Khalid Sawalha
- Cardiometabolic Diseases, Truman Medical Centers - University of Missouri Kansas City, Kansas City, USA
| | - Nicholas Norgard
- Pharmacology and Therapeutics, Truman Medical Centers - University of Missouri Kansas City, Kansas City, USA
| | | |
Collapse
|
34
|
Tang SB, Zhang TT, Yin S, Shen W, Luo SM, Zhao Y, Zhang CL, Klinger FG, Sun QY, Ge ZJ. Inheritance of perturbed methylation and metabolism caused by uterine malnutrition via oocytes. BMC Biol 2023; 21:43. [PMID: 36829148 PMCID: PMC9960220 DOI: 10.1186/s12915-023-01545-x] [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: 08/19/2022] [Accepted: 02/13/2023] [Indexed: 02/26/2023] Open
Abstract
BACKGROUND Undernourishment in utero has deleterious effects on the metabolism of offspring, but the mechanism of the transgenerational transmission of metabolic disorders is not well known. In the present study, we found that undernourishment in utero resulted in metabolic disorders of female F1 and F2 in mouse model. RESULTS Undernutrition in utero induced metabolic disorders of F1 females, which was transmitted to F2 females. The global methylation in oocytes of F1 exposed to undernutrition in utero was decreased compared with the control. KEGG analysis showed that genes with differential methylation regions (DMRs) in promoters were significantly enriched in metabolic pathways. The altered methylation of some DMRs in F1 oocytes located at the promoters of metabolic-related genes were partially observed in F2 tissues, and the expressions of these genes were also changed. Meanwhile, the abnormal DNA methylation of the validated DMRs in F1 oocytes was also observed in F2 oocytes. CONCLUSIONS These results indicate that DNA methylation may mediate the transgenerational inheritance of metabolic disorders induced by undernourishment in utero via female germline.
Collapse
Affiliation(s)
- Shou-Bin Tang
- grid.412608.90000 0000 9526 6338College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109 People’s Republic of China
| | - Ting-Ting Zhang
- grid.412608.90000 0000 9526 6338College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109 People’s Republic of China ,grid.414011.10000 0004 1808 090XReproductive Medicine Center, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou, 450003 People’s Republic of China
| | - Shen Yin
- grid.412608.90000 0000 9526 6338College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109 People’s Republic of China
| | - Wei Shen
- grid.412608.90000 0000 9526 6338College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109 People’s Republic of China
| | - Shi-Ming Luo
- grid.413405.70000 0004 1808 0686Fertility Preservation Lab and Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317 People’s Republic of China
| | - Yong Zhao
- grid.464332.4State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| | - Cui-Lian Zhang
- grid.414011.10000 0004 1808 090XReproductive Medicine Center, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou, 450003 People’s Republic of China
| | - Francesca Gioia Klinger
- grid.512346.7Histology and Embryology, Saint Camillus International University of Health Sciences, Rome, Italy
| | - Qing-Yuan Sun
- Fertility Preservation Lab and Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, People's Republic of China.
| | - Zhao-Jia Ge
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China.
| |
Collapse
|
35
|
Kim YM, Park JS, Choi HJ, Jung KM, Lee KY, Shim JH, Park KJ, Han JY. Efficient production of recombinant human adiponectin in egg white using genome edited chickens. Front Nutr 2023; 9:1068558. [PMID: 36761986 PMCID: PMC9902655 DOI: 10.3389/fnut.2022.1068558] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 12/23/2022] [Indexed: 01/25/2023] Open
Abstract
The prevalence of obesity-related metabolic diseases caused by insulin resistance is rapidly increasing worldwide. Adiponectin (ADPN), a hormone derived from adipose tissue, is a potential therapeutic agent for insulin resistance. Chickens are considered efficient bioreactors for recombinant protein production because they secrete large amounts of high-concentration proteins from the oviduct. Additionally, chickens express high levels of high-molecular-weight (HMW) ADPN, which is considered the active form in the body. Therefore, in this study, a gene-targeted chicken model was produced in which the gene encoding human ADPN was inserted into Ovalbumin (OVA) using the CRISPR/Cas9 system, and the characteristics of the resulting recombinant ADPN protein were evaluated. As a result, human ADPN was expressed in G1 hen oviducts and egg whites of OVA ADPN knock-in (KI) chickens. The concentration of ADPN in egg white ranged from 1.47 to 4.59 mg/mL, of which HMW ADPN accounted for ∼29% (0.24-1.49 mg/mL). Importantly, egg white-derived ADPN promoted expression of genes related to fatty acid oxidation and activated the 5'-AMP-activated protein kinase (AMPK) signaling pathway in muscle cells. In summary, the OVA gene-targeted chicken bioreactor proved to be an advantageous model for production of human ADPN, and the resulting protein was of sufficient quantity and efficacy for industrial use.
Collapse
Affiliation(s)
- Young Min Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea,Avinnogen Co., Ltd., Seoul, Republic of Korea
| | - Jin Se Park
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea,Avinnogen Co., Ltd., Seoul, Republic of Korea
| | - Hee Jung Choi
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Kyung Min Jung
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Kyung Youn Lee
- Bio-MAX/N-Bio, Institute of BioEngineering, Seoul National University, Seoul, Republic of Korea
| | - Ji Hyeon Shim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Kyung Je Park
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jae Yong Han
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea,*Correspondence: Jae Yong Han,
| |
Collapse
|
36
|
Moyce Gruber BL, Dolinsky VW. The Role of Adiponectin during Pregnancy and Gestational Diabetes. Life (Basel) 2023; 13:301. [PMID: 36836658 PMCID: PMC9958871 DOI: 10.3390/life13020301] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/13/2023] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
Pregnancy involves a range of metabolic adaptations to supply adequate energy for fetal growth and development. Gestational diabetes (GDM) is defined as hyperglycemia with first onset during pregnancy. GDM is a recognized risk factor for both pregnancy complications and long-term maternal and offspring risk of cardiometabolic disease development. While pregnancy changes maternal metabolism, GDM can be viewed as a maladaptation by maternal systems to pregnancy, which may include mechanisms such as insufficient insulin secretion, dysregulated hepatic glucose output, mitochondrial dysfunction and lipotoxicity. Adiponectin is an adipose-tissue-derived adipokine that circulates in the body and regulates a diverse range of physiologic mechanisms including energy metabolism and insulin sensitivity. In pregnant women, circulating adiponectin levels decrease correspondingly with insulin sensitivity, and adiponectin levels are low in GDM. In this review, we summarize the current state of knowledge about metabolic adaptations to pregnancy and the role of adiponectin in these processes, with a focus on GDM. Recent studies from rodent model systems have clarified that adiponectin deficiency during pregnancy contributes to GDM development. The upregulation of adiponectin alleviates hyperglycemia in pregnant mice, although much remains to be understood for adiponectin to be utilized clinically for GDM.
Collapse
Affiliation(s)
- Brittany L. Moyce Gruber
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM), Research Theme of the Children’s Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB R3E 0T6, Canada
| | - Vernon W. Dolinsky
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM), Research Theme of the Children’s Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB R3E 0T6, Canada
| |
Collapse
|
37
|
Daneshzad E, Rostami S, Aghamahdi F, Mahdavi-Gorabi A, Qorbani M. Association of cardiometabolic risk factors with insulin resistance in overweight and obese children. BMC Endocr Disord 2022; 22:320. [PMID: 36529727 PMCID: PMC9761952 DOI: 10.1186/s12902-022-01245-7] [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: 09/09/2021] [Accepted: 10/17/2022] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION Regarding the increased prevalence of obesity among children and adolescents, and the impact of obesity on insulin resistance (IR) and other metabolic disorders, this study was performed to determine the association of cardiometabolic risk factors (CMRFs) with IR in overweight and obese children. METHOD In this cross-sectional study 150 overweight and obese children (BMI ≥ 85th and BMI ≥ 95th age-sex specific percentile) and adolescents were selected via convenient sampling method from Endocrinology clinic in Karaj; Iran in 2020. Anthropometric indices, lipid profile, fasting blood glucose (FBG), and Homeostatic Model Assessment for Insulin Resistance (HOMA-IR) were evaluated. IR was defined as HOMA-IR ≥ 2.6. Multivariable linear and logistic regression model was used to assess the association of CMRFs with insulin level and IR respectively. RESULTS The mean age of children was 10.37 (± 2.6) years. Fifty-four percent of the participants were girls. IR was increased through increasing age (P < 0.001). In the multivariate logistic regression model, by increasing each unit increment in waist circumference (OR: 1.03, 95% CI: 1.01-1.06), wrist circumference (OR: 1.47, 95% CI: 1.06-2.02) total cholesterol (OR: 1.01, 95% CI: 1.003-1.03) and FBG (OR: 1.11, 95% CI: 1.05-1.18) the odds of IR increased significantly. Moreover, in the adjusted linear regression model, HOMA-IR was associated significantly with waist to height ratio (β: 2.45), and FBG (β: 0.02). CONCLUSION There was a significant association between some CMRFS with IR in overweight and obese children.
Collapse
Affiliation(s)
- Elnaz Daneshzad
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Sayeh Rostami
- Student Research Committee, Alborz University of Medical Sciences, Karaj, Iran
| | - Fatemeh Aghamahdi
- Department of Pediatric Endocrinology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran.
- Probiotic Research Center, Alborz University of Medical Sciences, Karaj, Iran.
| | | | - Mostafa Qorbani
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran.
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
38
|
Martín MG, Dotti CG. Plasma membrane and brain dysfunction of the old: Do we age from our membranes? Front Cell Dev Biol 2022; 10:1031007. [PMID: 36274849 PMCID: PMC9582647 DOI: 10.3389/fcell.2022.1031007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 09/20/2022] [Indexed: 11/26/2022] Open
Abstract
One of the characteristics of aging is a gradual hypo-responsiveness of cells to extrinsic stimuli, mainly evident in the pathways that are under hormone control, both in the brain and in peripheral tissues. Age-related resistance, i.e., reduced response of receptors to their ligands, has been shown to Insulin and also to leptin, thyroid hormones and glucocorticoids. In addition, lower activity has been reported in aging for ß-adrenergic receptors, adenosine A2B receptor, and several other G-protein-coupled receptors. One of the mechanisms proposed to explain the loss of sensitivity to hormones and neurotransmitters with age is the loss of receptors, which has been observed in several tissues. Another mechanism that is finding more and more experimental support is related to the changes that occur with age in the lipid composition of the neuronal plasma membrane, which are responsible for changes in the receptors’ coupling efficiency to ligands, signal attenuation and pathway desensitization. In fact, recent works have shown that altered membrane composition—as occurs during neuronal aging—underlies reduced response to glutamate, to the neurotrophin BDNF, and to insulin, all these leading to cognition decay and epigenetic alterations in the old. In this review we present evidence that altered functions of membrane receptors due to altered plasma membrane properties may be a triggering factor in physiological decline, decreased brain function, and increased vulnerability to neuropathology in aging.
Collapse
Affiliation(s)
- Mauricio G. Martín
- Cellular and Molecular Neurobiology Department, Instituto Ferreyra (INIMEC)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Córdoba (UNC), Córdoba, Argentina
- *Correspondence: Mauricio G. Martín, ; Carlos G. Dotti,
| | - Carlos G. Dotti
- Molecular Neuropathology Unit, Physiological and Pathological Processes Program, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid (UAM), Madrid, Spain
- *Correspondence: Mauricio G. Martín, ; Carlos G. Dotti,
| |
Collapse
|
39
|
Franzago M, Pilenzi L, Di Rado S, Vitacolonna E, Stuppia L. The epigenetic aging, obesity, and lifestyle. Front Cell Dev Biol 2022; 10:985274. [PMID: 36176280 PMCID: PMC9514048 DOI: 10.3389/fcell.2022.985274] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 08/22/2022] [Indexed: 11/25/2022] Open
Abstract
The prevalence of obesity has dramatically increased worldwide over the past decades. Aging-related chronic conditions, such as type 2 diabetes and cardiovascular disease, are more prevalent in individuals with obesity, thus reducing their lifespan. Epigenetic clocks, the new metrics of biological age based on DNA methylation patterns, could be considered a reflection of the state of one's health. Several environmental exposures and lifestyle factors can induce epigenetic aging accelerations, including obesity, thus leading to an increased risk of age-related diseases. The insight into the complex link between obesity and aging might have significant implications for the promotion of health and the mitigation of future disease risk. The present narrative review takes into account the interaction between epigenetic aging and obesity, suggesting that epigenome may be an intriguing target for age-related physiological changes and that its modification could influence aging and prolong a healthy lifespan. Therefore, we have focused on DNA methylation age as a clinical biomarker, as well as on the potential reversal of epigenetic age using a personalized diet- and lifestyle-based intervention.
Collapse
Affiliation(s)
- Marica Franzago
- Department of Medicine and Aging, School of Medicine and Health Sciences, G. d’Annunzio University, Chieti, Italy
- Center for Advanced Studies and Technology, G. d’Annunzio University, Chieti, Italy
| | - Lucrezia Pilenzi
- Center for Advanced Studies and Technology, G. d’Annunzio University, Chieti, Italy
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, G. d’Annunzio University, Chieti, Italy
| | - Sara Di Rado
- Center for Advanced Studies and Technology, G. d’Annunzio University, Chieti, Italy
| | - Ester Vitacolonna
- Department of Medicine and Aging, School of Medicine and Health Sciences, G. d’Annunzio University, Chieti, Italy
- Center for Advanced Studies and Technology, G. d’Annunzio University, Chieti, Italy
| | - Liborio Stuppia
- Center for Advanced Studies and Technology, G. d’Annunzio University, Chieti, Italy
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, G. d’Annunzio University, Chieti, Italy
| |
Collapse
|
40
|
Zatterale F, Raciti GA, Prevenzano I, Leone A, Campitelli M, De Rosa V, Beguinot F, Parrillo L. Epigenetic Reprogramming of the Inflammatory Response in Obesity and Type 2 Diabetes. Biomolecules 2022; 12:biom12070982. [PMID: 35883538 PMCID: PMC9313117 DOI: 10.3390/biom12070982] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 11/16/2022] Open
Abstract
For the past several decades, the prevalence of obesity and type 2 diabetes (T2D) has continued to rise on a global level. The risk contributing to this pandemic implicates both genetic and environmental factors, which are functionally integrated by epigenetic mechanisms. While these conditions are accompanied by major abnormalities in fuel metabolism, evidence indicates that altered immune cell functions also play an important role in shaping of obesity and T2D phenotypes. Interestingly, these events have been shown to be determined by epigenetic mechanisms. Consistently, recent epigenome-wide association studies have demonstrated that immune cells from obese and T2D individuals feature specific epigenetic profiles when compared to those from healthy subjects. In this work, we have reviewed recent literature reporting epigenetic changes affecting the immune cell phenotype and function in obesity and T2D. We will further discuss therapeutic strategies targeting epigenetic marks for treating obesity and T2D-associated inflammation.
Collapse
Affiliation(s)
- Federica Zatterale
- Department of Translational Medical Science, Federico II University of Naples, 80131 Naples, Italy; (F.Z.); (G.A.R.); (I.P.); (A.L.); (M.C.)
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy;
| | - Gregory Alexander Raciti
- Department of Translational Medical Science, Federico II University of Naples, 80131 Naples, Italy; (F.Z.); (G.A.R.); (I.P.); (A.L.); (M.C.)
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy;
| | - Immacolata Prevenzano
- Department of Translational Medical Science, Federico II University of Naples, 80131 Naples, Italy; (F.Z.); (G.A.R.); (I.P.); (A.L.); (M.C.)
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy;
| | - Alessia Leone
- Department of Translational Medical Science, Federico II University of Naples, 80131 Naples, Italy; (F.Z.); (G.A.R.); (I.P.); (A.L.); (M.C.)
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy;
| | - Michele Campitelli
- Department of Translational Medical Science, Federico II University of Naples, 80131 Naples, Italy; (F.Z.); (G.A.R.); (I.P.); (A.L.); (M.C.)
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy;
| | - Veronica De Rosa
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy;
| | - Francesco Beguinot
- Department of Translational Medical Science, Federico II University of Naples, 80131 Naples, Italy; (F.Z.); (G.A.R.); (I.P.); (A.L.); (M.C.)
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy;
- Correspondence: (F.B.); (L.P.); Tel.: +39-081-746-3248 (F.B.); +39-081-746-3045 (L.P.)
| | - Luca Parrillo
- Department of Translational Medical Science, Federico II University of Naples, 80131 Naples, Italy; (F.Z.); (G.A.R.); (I.P.); (A.L.); (M.C.)
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy;
- Correspondence: (F.B.); (L.P.); Tel.: +39-081-746-3248 (F.B.); +39-081-746-3045 (L.P.)
| |
Collapse
|
41
|
Xue L, Sun J, Liu J, Hu C, Wu D, Nie C, Zhang K, Wang Y, Zhao L, Li X, Lu Y, Zhang L, Zhang D, Fan M, Qian H, Jiang H, Wong J, Li Y, Ying H, Chow BKC, Wang L, Li Y. Maternal secretin ameliorates obesity by promoting white adipose tissue browning in offspring. EMBO Rep 2022; 23:e54132. [PMID: 35652247 PMCID: PMC9253765 DOI: 10.15252/embr.202154132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 04/26/2022] [Accepted: 05/05/2022] [Indexed: 12/09/2023] Open
Abstract
Our knowledge of the coordination of intergenerational inheritance and offspring metabolic reprogramming by gastrointestinal endocrine factors is largely unknown. Here, we showed that secretin (SCT), a brain-gut peptide, is downregulated by overnutrition in pregnant mice and women. More importantly, genetic loss of SCT in the maternal gut results in undesirable phenotypes developed in offspring including enhanced high-fat diet (HFD)-induced obesity and attenuated browning of inguinal white adipose tissue (iWAT). Mechanistically, loss of maternal SCT represses iWAT browning in offspring by a global change in genome methylation pattern through upregulation of DNMT1. SCT functions to facilitate ubiquitination and degradation of DNMT1 by activating AMPKα, which contributes to the observed alteration of DNMT1 in progeny. Lastly, we showed that SCT treatment during pregnancy can reduce the development of obesity and improve glucose tolerance and insulin resistance in offspring of HFD-fed females, suggesting that SCT may serve as a novel biomarker or a strategy for preventing metabolic diseases.
Collapse
Affiliation(s)
- Lamei Xue
- State Key Laboratory of Food Science and TechnologySchool of Food Science and TechnologyJiangnan UniversityWuxiChina
| | - Juan Sun
- State Key Laboratory of Food Science and TechnologySchool of Food Science and TechnologyJiangnan UniversityWuxiChina
| | - Jinxin Liu
- State Key Laboratory of Food Science and TechnologySchool of Food Science and TechnologyJiangnan UniversityWuxiChina
| | - Chaoping Hu
- Department of Neuromuscular DiseaseChildren’s Hospital of Fudan UniversityShanghaiChina
| | - Dandan Wu
- Shanghai Key Laboratory of StomatologyDepartment of Oral & Cranio‐maxillofacial ScienceShanghai 9th People's HospitalCollege of StomatologySchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Chenzhipeng Nie
- State Key Laboratory of Food Science and TechnologySchool of Food Science and TechnologyJiangnan UniversityWuxiChina
| | - Kuiliang Zhang
- State Key Laboratory of Food Science and TechnologySchool of Food Science and TechnologyJiangnan UniversityWuxiChina
| | - Yu Wang
- State Key Laboratory of Food Science and TechnologySchool of Food Science and TechnologyJiangnan UniversityWuxiChina
| | - Lei Zhao
- Department of Neuromuscular DiseaseChildren’s Hospital of Fudan UniversityShanghaiChina
| | - Xihua Li
- Department of Neuromuscular DiseaseChildren’s Hospital of Fudan UniversityShanghaiChina
| | - Yan Lu
- Department of Endocrinology and MetabolismZhongshan HospitalFudan UniversityShanghaiChina
| | - Li Zhang
- Joint International Research Laboratory of CNS RegenerationGuangdong‐Hong Kong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouChina
| | - Duo Zhang
- Clinical and Experimental TherapeuticsCollege of PharmacyUniversity of Georgia and Charlie Norwood VA Medical CenterAugustaGAUSA
| | - Mingcong Fan
- State Key Laboratory of Food Science and TechnologySchool of Food Science and TechnologyJiangnan UniversityWuxiChina
| | - Haifeng Qian
- State Key Laboratory of Food Science and TechnologySchool of Food Science and TechnologyJiangnan UniversityWuxiChina
| | - Haowen Jiang
- State Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
| | - Jiemin Wong
- Shanghai Key Laboratory of Regulatory BiologyFengxian District Central Hospital‐ECNU Joint Center of Translational MedicineInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghaiChina
| | - Yuying Li
- Chinese Academy of Sciences Key Laboratory of Nutrition, Metabolism and Food SafetyShanghai Institutes for Biological SciencesChinese Academy of SciencesShanghaiChina
| | - Hao Ying
- Chinese Academy of Sciences Key Laboratory of Nutrition, Metabolism and Food SafetyShanghai Institutes for Biological SciencesChinese Academy of SciencesShanghaiChina
| | - Billy KC Chow
- School of Biological SciencesUniversity of Hong KongHong KongChina
| | - Li Wang
- State Key Laboratory of Food Science and TechnologySchool of Food Science and TechnologyJiangnan UniversityWuxiChina
| | - Yan Li
- State Key Laboratory of Food Science and TechnologySchool of Food Science and TechnologyJiangnan UniversityWuxiChina
| |
Collapse
|
42
|
Park J, Lee DH, Ham S, Oh J, Noh JR, Lee YK, Park YJ, Lee G, Han SM, Han JS, Kim YY, Jeon YG, Nahmgoong H, Shin KC, Kim SM, Choi SH, Lee CH, Park J, Roh TY, Kim S, Kim JB. Targeted erasure of DNA methylation by TET3 drives adipogenic reprogramming and differentiation. Nat Metab 2022; 4:918-931. [PMID: 35788760 DOI: 10.1038/s42255-022-00597-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 05/24/2022] [Indexed: 01/10/2023]
Abstract
DNA methylation is a crucial epigenetic modification in the establishment of cell-type-specific characteristics. However, how DNA methylation is selectively reprogrammed at adipocyte-specific loci during adipogenesis remains unclear. Here, we show that the transcription factor, C/EBPδ, and the DNA methylation eraser, TET3, cooperatively control adipocyte differentiation. We perform whole-genome bisulfite sequencing to explore the dynamics and regulatory mechanisms of DNA methylation in adipocyte differentiation. During adipogenesis, DNA methylation selectively decreases at adipocyte-specific loci carrying the C/EBP binding motif, which correlates with the activity of adipogenic promoters and enhancers. Mechanistically, we find that C/EBPδ recruits a DNA methylation eraser, TET3, to catalyse DNA demethylation at the C/EBP binding motif and stimulate the expression of key adipogenic genes. Ectopic expression of TET3 potentiates in vitro and in vivo adipocyte differentiation and recovers downregulated adipogenic potential, which is observed in aged mice and humans. Taken together, our study highlights how targeted reprogramming of DNA methylation through cooperative action of the transcription factor C/EBPδ, and the DNA methylation eraser TET3, controls adipocyte differentiation.
Collapse
Affiliation(s)
- Jeu Park
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Do Hoon Lee
- Bioinformatics Institute, Seoul National University, Seoul, South Korea
| | - Seokjin Ham
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, South Korea
| | - Jiyoung Oh
- Department of Biological Sciences, College of Information and Bioengineering, Ulsan National Institute of Science and Technology, Ulsan, South Korea
| | - Jung-Ran Noh
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, University of Science and Technology, Daejeon, South Korea
| | - Yun Kyung Lee
- Internal Medicine, Seoul National University College of Medicine & Seoul National University Bundang Hospital, Seoul, South Korea
| | - Yoon Jeong Park
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Gung Lee
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Sang Mun Han
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Ji Seul Han
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Ye Young Kim
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Yong Geun Jeon
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Han Nahmgoong
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Kyung Cheul Shin
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Sung Min Kim
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Sung Hee Choi
- Internal Medicine, Seoul National University College of Medicine & Seoul National University Bundang Hospital, Seoul, South Korea
| | - Chul-Ho Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, University of Science and Technology, Daejeon, South Korea
| | - Jiyoung Park
- Department of Biological Sciences, College of Information and Bioengineering, Ulsan National Institute of Science and Technology, Ulsan, South Korea
| | - Tae Young Roh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, South Korea
| | - Sun Kim
- Department of Computer Science and Engineering, Institute of Engineering Research, Seoul National University, Seoul, South Korea
| | - Jae Bum Kim
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, South Korea.
| |
Collapse
|
43
|
Zhuo MQ, Chen J, Wu ML, Wang WB. Novel insights for PI3KC3 in mediating lipid accumulation in yellow catfish Pelteobagrus fulvidraco. FISH PHYSIOLOGY AND BIOCHEMISTRY 2022; 48:571-583. [PMID: 35389126 DOI: 10.1007/s10695-022-01071-7] [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: 12/13/2021] [Accepted: 03/25/2022] [Indexed: 06/14/2023]
Abstract
In this study, the transcriptional regulation of PI3KC3 by three transcription factors (PPARγ, PPARα, and STAT3) and the potential role of PI3KC3 in mediating lipid accumulation were determined in yellow catfish Pelteobagrus fulvidraco. The 5'-deletion assay, overexpression assay, site-mutation assay, and electrophoretic mobility shift assay suggested that PPARα, PPARγ, and STAT3 negatively regulated the promoter activity of pi3kc3. Moreover, the transcriptional inactivation of pi3kc3 was directly mediated by PPARα and PPARγ under fatty acid (FA) treatment. Using primary hepatocytes from yellow catfish, FA incubation significantly increased triacylglyceride (TG) content, non-esterified fatty acid (NEFA) content, and lipid drops (LDs) content, the mRNA level of pparα, pparγ, stat3, and dnmt3b, the protein level of PPARα, PPARγ, and STAT3, and the methylation level of pi3kc3, but significantly reduced the mRNA and protein level of PI3KC3. Our findings offer new insights into the mechanisms for transcriptional regulation of PI3KC3 and for PI3KC3-mediated lipid accumulation in fish.
Collapse
Affiliation(s)
- Mei-Qin Zhuo
- School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, 430070, China.
| | - Jun Chen
- School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, 430070, China
| | - Mei-Li Wu
- School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, 430070, China
| | - Wen-Biao Wang
- School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, 430070, China
| |
Collapse
|
44
|
Fu Y, Wang L, Yu B, Xu D, Chu Y. Immunometabolism shapes B cell fate and functions. Immunology 2022; 166:444-457. [PMID: 35569110 DOI: 10.1111/imm.13499] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 03/28/2022] [Indexed: 11/29/2022] Open
Affiliation(s)
- Ying Fu
- Department of Immunology, School of Basic Medical Sciences, and Institutes of Biomedical Sciences Fudan University Shanghai China
| | - Luman Wang
- Department of Immunology, School of Basic Medical Sciences, and Institutes of Biomedical Sciences Fudan University Shanghai China
- Department of Endocrinology and Metabolism, Shanghai Fifth People's Hospital Fudan University Shanghai China
- Biotherapy Research Center Fudan University Shanghai China
| | - Baichao Yu
- Department of Immunology, School of Basic Medical Sciences, and Institutes of Biomedical Sciences Fudan University Shanghai China
| | - Damo Xu
- School of Medicine Shenzhen University Shenzhen China
- Third Affiliated Hospital of Shenzhen University Shenzhen Luohu Hospital Group Shenzhen China
| | - Yiwei Chu
- Department of Immunology, School of Basic Medical Sciences, and Institutes of Biomedical Sciences Fudan University Shanghai China
- Biotherapy Research Center Fudan University Shanghai China
| |
Collapse
|
45
|
Targeting of the Peritumoral Adipose Tissue Microenvironment as an Innovative Antitumor Therapeutic Strategy. Biomolecules 2022; 12:biom12050702. [PMID: 35625629 PMCID: PMC9138344 DOI: 10.3390/biom12050702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/09/2022] [Accepted: 05/11/2022] [Indexed: 12/03/2022] Open
Abstract
The tumor microenvironment (TME) plays a key role in promoting and sustaining cancer growth. Adipose tissue (AT), due to its anatomical distribution, is a prevalent component of TME, and contributes to cancer development and progression. Cancer-associated adipocytes (CAAs), reprogrammed by cancer stem cells (CSCs), drive cancer progression by releasing metabolites and inflammatory adipokines. In this review, we highlight the mechanisms underlying the bidirectional crosstalk among CAAs, CSCs, and stromal cells. Moreover, we focus on the recent advances in the therapeutic targeting of adipocyte-released factors as an innovative strategy to counteract cancer progression.
Collapse
|
46
|
Vander Velden JW, Osborne DM. Prolonged diet-induced obesity modifies DNA methylation and gene expression in the hippocampus. Neurosci Lett 2022; 780:136656. [DOI: 10.1016/j.neulet.2022.136656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 03/04/2022] [Accepted: 04/20/2022] [Indexed: 10/18/2022]
|
47
|
Xie H, Liu X, Zhou Q, Huang T, Zhang L, Gao J, Wang Y, Liu Y, Yan T, Zhang S, Wang CY. DNA Methylation Modulates Aging Process in Adipocytes. Aging Dis 2022; 13:433-446. [PMID: 35371604 PMCID: PMC8947842 DOI: 10.14336/ad.2021.0904] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 09/04/2021] [Indexed: 11/17/2022] Open
Abstract
Aging has been recognized to be a highly complex biological health problem with a high risk of chronic diseases, including type 2 diabetes, atherosclerosis, chronic bronchitis or emphysema, cancer and Alzheimer's disease. Particularly, age-related turnover in adipose tissue is a major contributor to metabolic syndromes and shortened lifespan. Adipocytes undergo senescence in early stage, which results in adipose tissue metabolic dysfunction, redistribution, and inflammation. The well-established association between DNA methylation (DNAm) and aging has been observed in the past few decades. Indeed, age-related alteration in DNAm is highly tissue-specific. This review intends to summarize the advancements how DNAm changes coupled with aging process in adipose tissue, by which DNAm regulates cellular senescence, metabolic function, adipokine secretion and beiging process in adipocytes. Elucidation of the effect of DNAm on adipose aging would have great potential to the development of epigenetic therapeutic strategies against aging-related diseases in clinical settings.
Collapse
Affiliation(s)
- Hao Xie
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xin Liu
- Department of Interventional Radiology, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Qing Zhou
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Teng Huang
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Lu Zhang
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jia Gao
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yuhan Wang
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yanjun Liu
- The Center for Obesity and Metabolic Health, Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Sichuan, China.,The Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People’s Hospital of Chengdu & The affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan, China.
| | - Tong Yan
- The Center for Obesity and Metabolic Health, Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Sichuan, China.
| | - Shu Zhang
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Correspondence should be addressed to: Drs. Cong-Yi Wang () or Shu Zhang (), the Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cong-Yi Wang
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Correspondence should be addressed to: Drs. Cong-Yi Wang () or Shu Zhang (), the Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
48
|
Elghazy AM, Elsaeid AM, Refaat M, Youssef MM. Biochemical studies of adiponectin gene polymorphism in patients with obesity in Egyptians. Arch Physiol Biochem 2022; 128:43-50. [PMID: 31502880 DOI: 10.1080/13813455.2019.1662451] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Adiponectin gene polymorphisms have recently been reported to be associated with obesity. In Egypt, obesity has expanded especially with the changing nourishment propensities and the inexorably inactive ways of life, with almost 70 percent of the Egyptian populations being obese. AIM To assess the relationship of the adiponectin gene (ADIPOQ) polymorphism in patients with Obesity in Egyptians. SUBJECTS AND METHODS This study included 100 patients with obesity and 97 random controls. RESULTS Adiponectin rs1501299 polymorphism showed significant difference cases with different obesity grades where the T/T genotype was relatively higher in higher classes of obesity (Class II and III; (66.7% and 55.6% respectively, p = 000), which determines the susceptibility to obesity. CONCLUSION Adiponectin rs1501299 polymorphism might be a candidate gene, which determines the susceptibility to obesity. Larger studies are necessary to confirm these findings in various populations.
Collapse
Affiliation(s)
- Ahmed M Elghazy
- Biochemistry Division, Chemistry Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Afaf M Elsaeid
- Genetics unit, Department of Pediatrics, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Manar Refaat
- Biochemistry Division, Chemistry Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Magdy M Youssef
- Biochemistry Division, Chemistry Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| |
Collapse
|
49
|
Mahmoud AM. An Overview of Epigenetics in Obesity: The Role of Lifestyle and Therapeutic Interventions. Int J Mol Sci 2022; 23:ijms23031341. [PMID: 35163268 PMCID: PMC8836029 DOI: 10.3390/ijms23031341] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 02/06/2023] Open
Abstract
Obesity has become a global epidemic that has a negative impact on population health and the economy of nations. Genetic predispositions have been demonstrated to have a substantial role in the unbalanced energy metabolism seen in obesity. However, these genetic variations cannot entirely explain the massive growth in obesity over the last few decades. Accumulating evidence suggests that modern lifestyle characteristics such as the intake of energy-dense foods, adopting sedentary behavior, or exposure to environmental factors such as industrial endocrine disruptors all contribute to the rising obesity epidemic. Recent advances in the study of DNA and its alterations have considerably increased our understanding of the function of epigenetics in regulating energy metabolism and expenditure in obesity and metabolic diseases. These epigenetic modifications influence how DNA is transcribed without altering its sequence. They are dynamic, reflecting the interplay between the body and its surroundings. Notably, these epigenetic changes are reversible, making them appealing targets for therapeutic and corrective interventions. In this review, I discuss how these epigenetic modifications contribute to the disordered energy metabolism in obesity and to what degree lifestyle and weight reduction strategies and pharmacological drugs can restore energy balance by restoring normal epigenetic profiles.
Collapse
Affiliation(s)
- Abeer M Mahmoud
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
50
|
Ali MM, Naquiallah D, Qureshi M, Mirza MI, Hassan C, Masrur M, Bianco FM, Frederick P, Cristoforo GP, Gangemi A, Phillips SA, Mahmoud AM. DNA methylation profile of genes involved in inflammation and autoimmunity correlates with vascular function in morbidly obese adults. Epigenetics 2022; 17:93-109. [PMID: 33487124 PMCID: PMC8812729 DOI: 10.1080/15592294.2021.1876285] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 12/12/2020] [Accepted: 01/04/2021] [Indexed: 12/18/2022] Open
Abstract
Obesity is a major risk factor for cardiovascular disease. Blood-detected epigenetic profiles may serve as non-invasive clinically relevant biomarkers. Therefore, we investigated DNA methylation of genes involved in inflammation in peripheral blood of obese subjects and lean controls and their correlation with cardiometabolic measurements. We obtained blood and adipose tissue (AT) samples from bariatric patients (n = 24) and control adults (n = 24). AT-isolated arterioles were tested for flow-induced dilation (FID) and production of nitric oxide (NO) and reactive oxygen species (ROS). Brachial artery flow-mediated dilation (FMD) was measured via doppler ultrasound. Promoter methylation of 94 genes involved in inflammation and autoimmunity were analysed in whole-blood DNA in relation to vascular function and cardiometabolic risk factors. 77 genes had ahigher methylated fraction in the controls compare obese subjects and 28 proinflammatory genes were significantly hypomethylated in the obese individuals; on top of these genes are CXCL1, CXCL12, CXCL6, IGF2BP2, HDAC4, IL12A, and IL17RA. Fifteen of these genes had significantly higher mRNA in obese subjects compared to controls; on top of these genes are CXCL6, TLR5, IL6ST, EGR1, IL15RA, and HDAC4. Methylation % inversely correlated with BMI, total fat %, visceral fat%, blood pressure, fasting plasma insulin, serum IL6 and C-reactive protein, arteriolar ROS, and alcohol consumption and positive correlations with lean %, HDL, plasma folate and vitamin B12, arteriolar FID and NO production, and brachial FMD. Our results suggest that vascular dysfunction in obese adults may be attributed to asystemic hypomethylation and over expression of the immune-related genes.
Collapse
Affiliation(s)
- Mohamed M. Ali
- Department of Physical Therapy, College of Applied Health Sciences, University of Illinois at Chicago, Chicago, IL, USA
- Integrative Physiology Laboratory, College of Applied Health Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Dina Naquiallah
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Maryam Qureshi
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Mohammed Imaduddin Mirza
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Chandra Hassan
- Departments of Surgery, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Mario Masrur
- Departments of Surgery, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Francesco M. Bianco
- Departments of Surgery, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Patrice Frederick
- Departments of Surgery, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | | | - Antonio Gangemi
- Departments of Surgery, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Shane A. Phillips
- Department of Physical Therapy, College of Applied Health Sciences, University of Illinois at Chicago, Chicago, IL, USA
- Integrative Physiology Laboratory, College of Applied Health Sciences, University of Illinois at Chicago, Chicago, IL, USA
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Abeer M. Mahmoud
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|