1
|
Sun Y, Shan X, Li M, Niu Y, Sun Z, Ma X, Wang T, Zhang J, Niu D. Autoimmune mechanisms and inflammation in obesity-associated type 2 diabetes, atherosclerosis, and non-alcoholic fatty liver disease. Funct Integr Genomics 2025; 25:84. [PMID: 40205260 DOI: 10.1007/s10142-025-01587-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 04/11/2025]
Abstract
Obesity, characterized by the excessive accumulation of white adipose tissue, is a significant global health burden and a major risk factor for a range of diseases, including malignancies and metabolic disorders. Individuals with high visceral fat content are particularly susceptible to severe complications such as type 2 diabetes, cardiovascular diseases, and liver disorders. However, the pathogenesis of obesity-related metabolic diseases extends beyond simple adiposity. Chronic obesity triggers a prolonged inflammatory response, which leads to tissue fibrosis and sustained organ damage, contributing to multi-organ dysfunction. This review explores the autoimmune mechanisms and inflammatory pathways underlying obesity-induced type 2 diabetes, atherosclerosis, and non-alcoholic fatty liver disease, with an emphasis on their interrelated pathophysiology and the potential for therapeutic interventions.
Collapse
Grants
- LZ22C010003 Key Project of Zhejiang Provincial Natural Science Foundation of China
- LZ22C010003 Key Project of Zhejiang Provincial Natural Science Foundation of China
- LZ22C010003 Key Project of Zhejiang Provincial Natural Science Foundation of China
- LZ22C010003 Key Project of Zhejiang Provincial Natural Science Foundation of China
- LZ22C010003 Key Project of Zhejiang Provincial Natural Science Foundation of China
- 2021R52043 Scientific and Technological Innovation Leading Talents Project of Zhejiang Provincial "High-level Talents Special Support Plan"
- 2021R52043 Scientific and Technological Innovation Leading Talents Project of Zhejiang Provincial "High-level Talents Special Support Plan"
- 2021R52043 Scientific and Technological Innovation Leading Talents Project of Zhejiang Provincial "High-level Talents Special Support Plan"
- 2021R52043 Scientific and Technological Innovation Leading Talents Project of Zhejiang Provincial "High-level Talents Special Support Plan"
- 2021R52043 Scientific and Technological Innovation Leading Talents Project of Zhejiang Provincial "High-level Talents Special Support Plan"
- 32202656, 32402753 National Natural Science Foundation of China
- 32202656, 32402753 National Natural Science Foundation of China
- 32202656, 32402753 National Natural Science Foundation of China
- 32202656, 32402753 National Natural Science Foundation of China
- 32202656, 32402753 National Natural Science Foundation of China
- LQ23C170003, LQ23C180003 & LQ24C170001 Zhejiang Provincial Natural Science Foundation of China
- LQ23C170003, LQ23C180003 & LQ24C170001 Zhejiang Provincial Natural Science Foundation of China
- LQ23C170003, LQ23C180003 & LQ24C170001 Zhejiang Provincial Natural Science Foundation of China
- LQ23C170003, LQ23C180003 & LQ24C170001 Zhejiang Provincial Natural Science Foundation of China
- LQ23C170003, LQ23C180003 & LQ24C170001 Zhejiang Provincial Natural Science Foundation of China
- 2021C02068-4 Zhejiang Science and Technology Major Program on Agricultural New Variety Breeding
- 2021C02068-4 Zhejiang Science and Technology Major Program on Agricultural New Variety Breeding
Collapse
Affiliation(s)
- Yuanyuan Sun
- College of Animal Science and Technology & College of Veterinary Medicine, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Xueting Shan
- College of Animal Science and Technology & College of Veterinary Medicine, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Mingyang Li
- College of Animal Science and Technology & College of Veterinary Medicine, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Yifan Niu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Zhongxin Sun
- Department of Plastic, Reconstructive & Hand Microsurgery, Ningbo NO.6 Hospital, Ningbo, 315000, Zhejiang, China
| | - Xiang Ma
- College of Animal Science and Technology & College of Veterinary Medicine, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Tao Wang
- Nanjing Kgene Genetic Engineering Co., Ltd, Nanjing, 211300, Jiangsu, China.
| | - Jufang Zhang
- Department of Plastic and Aesthetic Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, Zhejiang, China.
| | - Dong Niu
- College of Animal Science and Technology & College of Veterinary Medicine, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China.
| |
Collapse
|
2
|
Zhou H, Gizlenci M, Xiao Y, Martin F, Nakamori K, Zicari EM, Sato Y, Tullius SG. Obesity-associated Inflammation and Alloimmunity. Transplantation 2025; 109:588-596. [PMID: 39192462 PMCID: PMC11868468 DOI: 10.1097/tp.0000000000005183] [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] [Indexed: 08/29/2024]
Abstract
Obesity is a worldwide health problem with a rapidly rising incidence. In organ transplantation, increasing numbers of patients with obesity accumulate on waiting lists and undergo surgery. Obesity is in general conceptualized as a chronic inflammatory disease, potentially impacting alloimmune response and graft function. Here, we summarize our current understanding of cellular and molecular mechanisms that control obesity-associated adipose tissue inflammation and provide insights into mechanisms affecting transplant outcomes, emphasizing on the beneficial effects of weight loss on alloimmune responses.
Collapse
Affiliation(s)
- Hao Zhou
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
| | - Merih Gizlenci
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
- Department of General, Visceral, Cancer and Transplant Surgery, University Hospital of Cologne, Cologne, Germany
| | - Yao Xiao
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
| | - Friederike Martin
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
- Department of Surgery, CVK/CCM, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Keita Nakamori
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan
| | - Elizabeth M. Zicari
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
- Faculté de Pharmacie, Université Paris Cité, Paris, France
| | - Yuko Sato
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
| | - Stefan G. Tullius
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
| |
Collapse
|
3
|
Ho PY, Chou YC, Koh YC, Lin WS, Chen WJ, Tseng AL, Gung CL, Wei YS, Pan MH. Lactobacillus rhamnosus 069 and Lactobacillus brevis 031: Unraveling Strain-Specific Pathways for Modulating Lipid Metabolism and Attenuating High-Fat-Diet-Induced Obesity in Mice. ACS OMEGA 2024; 9:28520-28533. [PMID: 38973907 PMCID: PMC11223209 DOI: 10.1021/acsomega.4c02514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/29/2024] [Accepted: 05/09/2024] [Indexed: 07/09/2024]
Abstract
Obesity is a global health crisis, marked by excessive fat in tissues that function as immune organs, linked to microbiota dysregulation and adipose inflammation. Investigating the effects of Lactobacillus rhamnosus SG069 (LR069) and Lactobacillus brevis SG031 (LB031) on obesity and lipid metabolism, this research highlights adipose tissue's critical immune-metabolic role and the probiotics' potential against diet-induced obesity. Mice fed a high-fat diet were treated with either LR069 or LB031 for 12 weeks. Administration of LB031 boosted lipid metabolism, indicated by higher AMP-activated protein kinase (AMPK) and acetyl-CoA carboxylase (ACC) phosphorylation, and increased the M2/M1 macrophage ratio, indicating LB031's anti-inflammatory effect. Meanwhile, LR069 administration not only led to significant weight loss by enhancing lipolysis which evidenced by increased phosphorylation of hormone-sensitive lipase (HSL) and adipose triglyceride lipase (ATGL) but also elevated Akkermansia and fecal acetic acid levels, showing the gut microbiota's pivotal role in its antiobesity effects. LR069 and LB031 exhibit distinct effects on lipid metabolism and obesity, underscoring their potential for precise interventions. This research elucidates the unique impacts of these strains on metabolic health and highlights the intricate relationship between gut microbiota and obesity, advancing our knowledge of probiotics' therapeutic potential.
Collapse
Affiliation(s)
- Pin-Yu Ho
- Institute
of Food Science and Technology, National
Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 10617, Taiwan, ROC
| | - Ya-Chun Chou
- Institute
of Food Science and Technology, National
Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 10617, Taiwan, ROC
| | - Yen-Chun Koh
- Institute
of Food Science and Technology, National
Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 10617, Taiwan, ROC
| | - Wei-Sheng Lin
- Institute
of Food Science and Technology, National
Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 10617, Taiwan, ROC
- Department
of Food Science, National Quemoy University, Quemoy County 89250, Taiwan, ROC
| | - Wei-Jen Chen
- Syngen
Biotech Co., Ltd., Building
A, No. 154, Kaiyuan Rd., Sinying, Tainan 73055, Taiwan
| | - Ai-Lun Tseng
- Syngen
Biotech Co., Ltd., Building
A, No. 154, Kaiyuan Rd., Sinying, Tainan 73055, Taiwan
| | - Chiau-Ling Gung
- Syngen
Biotech Co., Ltd., Building
A, No. 154, Kaiyuan Rd., Sinying, Tainan 73055, Taiwan
| | - Yu-Shan Wei
- Syngen
Biotech Co., Ltd., Building
A, No. 154, Kaiyuan Rd., Sinying, Tainan 73055, Taiwan
| | - Min-Hsiung Pan
- Institute
of Food Science and Technology, National
Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 10617, Taiwan, ROC
- Department
of Public Health, China Medical University, 91, Hsueh-Shih Road, Taichung 40402, Taiwan, ROC
- Department
of Food Nutrition and Health Biotechnology, Asia University, 500,
Lioufeng Rd., Wufeng, Taichung 41354, Taiwan, ROC
| |
Collapse
|
4
|
Qian Q, Li M, Zhang Z, Davis SW, Rahmouni K, Norris AW, Cao H, Ding WX, Hotamisligil GS, Yang L. Obesity disrupts the pituitary-hepatic UPR communication leading to NAFLD progression. Cell Metab 2024; 36:1550-1565.e9. [PMID: 38718793 PMCID: PMC11222033 DOI: 10.1016/j.cmet.2024.04.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 03/05/2024] [Accepted: 04/17/2024] [Indexed: 07/05/2024]
Abstract
Obesity alters levels of pituitary hormones that govern hepatic immune-metabolic homeostasis, dysregulation of which leads to nonalcoholic fatty liver disease (NAFLD). However, the impact of obesity on intra-pituitary homeostasis is largely unknown. Here, we uncovered a blunted unfolded protein response (UPR) but elevated inflammatory signatures in pituitary glands of obese mice and humans. Furthermore, we found that obesity inflames the pituitary gland, leading to impaired pituitary inositol-requiring enzyme 1α (IRE1α)-X-box-binding protein 1 (XBP1) UPR branch, which is essential for protecting against pituitary endocrine defects and NAFLD progression. Intriguingly, pituitary IRE1-deletion resulted in hypothyroidism and suppressed the thyroid hormone receptor B (THRB)-mediated activation of Xbp1 in the liver. Conversely, activation of the hepatic THRB-XBP1 axis improved NAFLD in mice with pituitary UPR defect. Our study provides the first evidence and mechanism of obesity-induced intra-pituitary cellular defects and the pathophysiological role of pituitary-liver UPR communication in NAFLD progression.
Collapse
Affiliation(s)
- Qingwen Qian
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Mark Li
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Zeyuan Zhang
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Shannon W Davis
- Department of Biological Sciences, College of Arts and Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Kamal Rahmouni
- Department of Neuroscience and Pharmacology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Andrew W Norris
- Division of Endocrinology and Diabetes, Department of Pediatrics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Huojun Cao
- Iowa Institute for Oral Health Research, Division of Biostatistics and Computational Biology, Department of Endodontics, University of Iowa College of Dentistry, Iowa City, IA 52242, USA
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Gökhan S Hotamisligil
- Sabri Ülker Center for Metabolic Research, Department of Molecular Metabolism, Harvard T.H. School of Public Health, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02141, USA
| | - Ling Yang
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA.
| |
Collapse
|
5
|
Chirivi M, Contreras GA. Endotoxin-induced alterations of adipose tissue function: a pathway to bovine metabolic stress. J Anim Sci Biotechnol 2024; 15:53. [PMID: 38581064 PMCID: PMC10998405 DOI: 10.1186/s40104-024-01013-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 02/14/2024] [Indexed: 04/07/2024] Open
Abstract
During the periparturient period, dairy cows exhibit negative energy balance due to limited appetite and increased energy requirements for lactogenesis. The delicate equilibrium between energy availability and expenditure puts cows in a state of metabolic stress characterized by excessive lipolysis in white adipose tissues (AT), increased production of reactive oxygen species, and immune cell dysfunction. Metabolic stress, especially in AT, increases the risk for metabolic and inflammatory diseases. Around parturition, cows are also susceptible to endotoxemia. Bacterial-derived toxins cause endotoxemia by promoting inflammatory processes and immune cell infiltration in different organs and systems while impacting metabolic function by altering lipolysis, mitochondrial activity, and insulin sensitivity. In dairy cows, endotoxins enter the bloodstream after overcoming the defense mechanisms of the epithelial barriers, particularly during common periparturient conditions such as mastitis, metritis, and pneumonia, or after abrupt changes in the gut microbiome. In the bovine AT, endotoxins induce a pro-inflammatory response and stimulate lipolysis in AT, leading to the release of free fatty acids into the bloodstream. When excessive and protracted, endotoxin-induced lipolysis can impair adipocyte's insulin signaling pathways and lipid synthesis. Endotoxin exposure can also induce oxidative stress in AT through the production of reactive oxygen species by inflammatory cells and other cellular components. This review provides insights into endotoxins' impact on AT function, highlighting the gaps in our knowledge of the mechanisms underlying AT dysfunction, its connection with periparturient cows' disease risk, and the need to develop effective interventions to prevent and treat endotoxemia-related inflammatory conditions in dairy cattle.
Collapse
Affiliation(s)
- Miguel Chirivi
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA
| | - G Andres Contreras
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
6
|
Lee J, Rogers HM, Springer DA, Noguchi CT. Neuronal nitric oxide synthase required for erythropoietin modulation of heart function in mice. Front Physiol 2024; 15:1338476. [PMID: 38628440 PMCID: PMC11019009 DOI: 10.3389/fphys.2024.1338476] [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: 11/14/2023] [Accepted: 03/04/2024] [Indexed: 04/19/2024] Open
Abstract
Introduction: Erythropoietin (EPO) acts primarily in regulating red blood cell production mediated by high EPO receptor (EPOR) expression in erythroid progenitor cells. EPO activity in non-erythroid tissue is evident in mice with EPOR restricted to erythroid tissues (ΔEPORE) that become obese, glucose-intolerant, and insulin-resistant. In animal models, nitric oxide synthase (NOS) contributes to EPO activities including erythropoiesis, neuroprotection, and cardioprotection against ischemia-reperfusion injury. However, we found that extended EPO treatment to increase hematocrit compromised heart function, while the loss of neuronal NOS (nNOS) was protective against the deleterious activity of EPO to promote heart failure. Methods: Wild-type (WT) mice, ΔEPORE mice, and nNOS-knockout mice (nNOS-/-) were placed on a high-fat diet to match the ΔEPORE obese phenotype and were treated with EPO for 3 weeks. Hematocrit and metabolic response to EPO treatment were monitored. Cardiac function was assessed by echocardiography and ultrasonography. Results: ΔEPORE mice showed a decrease in the left ventricular outflow tract (LVOT) peak velocity, ejection fraction, and fractional shortening, showing that endogenous non-erythroid EPO response is protective for heart function. EPO treatment increased hematocrit in all mice and decreased fat mass in male WT, demonstrating that EPO regulation of fat mass requires non-erythroid EPOR. EPO treatment also compromised heart function in WT mice, and decreased the pulmonary artery peak velocity (PA peak velocity), LVOT peak velocity, ejection fraction, and fractional shortening, but it had minimal effect in further reducing the heart function in ΔEPORE mice, indicating that the adverse effect of EPO on heart function is not related to EPO-stimulated erythropoiesis. ΔEPORE mice had increased expression of heart failure-associated genes, hypertrophic cardiomyopathy-related genes, and sarcomeric genes that were also elevated with EPO treatment in WT mice. Male and female nNOS-/- mice were protected against diet-induced obesity. EPO treatment in nNOS-/- mice increased the hematocrit that tended to be lower than WT mice and decreased the PA peak velocity but did not affect the LVOT peak velocity, ejection fraction, and fractional shortening, suggesting that nNOS is required for the adverse effect of EPO treatment on heart function in WT mice. EPO treatment did not change expression of heart failure-associated gene expression in nNOS-/- mice. Discussion: Endogenous EPO has a protective effect on heart function. With EPO administration, in contrast to the protective effect to the cardiac injury of acute EPO treatment, extended EPO treatment to increase hematocrit in WT mice adversely affected the heart function with a corresponding increase in expression of heart failure-associated genes. This EPO activity was independent of EPO-stimulated erythropoiesis and required EPOR in non-erythroid tissue and nNOS activity, while nNOS-/- mice were protected from the EPO-associated adverse effect on heart function. These data provide evidence that nNOS contributes to the negative impact on the heart function of high-dose EPO treatment for anemia.
Collapse
Affiliation(s)
- Jeeyoung Lee
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Heather M. Rogers
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Danielle A. Springer
- Murine Phenotyping Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Constance T. Noguchi
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
7
|
Bahadoran Z, Mirmiran P, Ghasemi A. Adipose organ dysfunction and type 2 diabetes: Role of nitric oxide. Biochem Pharmacol 2024; 221:116043. [PMID: 38325496 DOI: 10.1016/j.bcp.2024.116043] [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: 10/30/2023] [Revised: 01/07/2024] [Accepted: 02/01/2024] [Indexed: 02/09/2024]
Abstract
Adipose organ, historically known as specialized lipid-handling tissue serving as the long-term fat depot, is now appreciated as the largest endocrine organ composed of two main compartments, i.e., subcutaneous and visceral adipose tissue (AT), madding up white and beige/brown adipocytes. Adipose organ dysfunction manifested as maldistribution of the compartments, hypertrophic, hypoxic, inflamed, and insulin-resistant AT, contributes to the development of type 2 diabetes (T2D). Here, we highlight the role of nitric oxide (NO·) in AT (dys)function in relation to developing T2D. The key aspects determining lipid and glucose homeostasis in AT depend on the physiological levels of the NO· produced via endothelial NO· synthases (eNOS). In addition to decreased NO· bioavailability (via decreased expression/activity of eNOS or scavenging NO·), excessive NO· produced by inducible NOS (iNOS) in response to hypoxia and AT inflammation may be a critical interfering factor diverting NO· signaling to the formation of reactive oxygen and nitrogen species, resulting in AT and whole-body metabolic dysfunction. Pharmacological approaches boosting AT-NO· availability at physiological levels (by increasing NO· production and its stability), as well as suppression of iNOS-NO· synthesis, are potential candidates for developing NO·-based therapeutics in T2D.
Collapse
Affiliation(s)
- Zahra Bahadoran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parvin Mirmiran
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Engin A. Endothelial Dysfunction in Obesity and Therapeutic Targets. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:489-538. [PMID: 39287863 DOI: 10.1007/978-3-031-63657-8_17] [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
Parallel to the increasing prevalence of obesity in the world, the mortality from cardiovascular disease has also increased. Low-grade chronic inflammation in obesity disrupts vascular homeostasis, and the dysregulation of adipocyte-derived endocrine and paracrine effects contributes to endothelial dysfunction. Besides the adipose tissue inflammation, decreased nitric oxide (NO)-bioavailability, insulin resistance (IR), and oxidized low-density lipoproteins (oxLDLs) are the main factors contributing to endothelial dysfunction in obesity and the development of cardiorenal metabolic syndrome. While normal healthy perivascular adipose tissue (PVAT) ensures the dilation of blood vessels, obesity-associated PVAT leads to a change in the profile of the released adipo-cytokines, resulting in a decreased vasorelaxing effect. Higher stiffness parameter β, increased oxidative stress, upregulation of pro-inflammatory cytokines, and nicotinamide adenine dinucleotide phosphate (NADP) oxidase in PVAT turn the macrophages into pro-atherogenic phenotypes by oxLDL-induced adipocyte-derived exosome-macrophage crosstalk and contribute to the endothelial dysfunction. In clinical practice, carotid ultrasound, higher leptin levels correlate with irisin over-secretion by human visceral and subcutaneous adipose tissues, and remnant cholesterol (RC) levels predict atherosclerotic disease in obesity. As a novel therapeutic strategy for cardiovascular protection, liraglutide improves vascular dysfunction by modulating a cyclic adenosine monophosphate (cAMP)-independent protein kinase A (PKA)-AMP-activated protein kinase (AMPK) pathway in PVAT in obese individuals. Because the renin-angiotensin-aldosterone system (RAAS) activity, hyperinsulinemia, and the resultant IR play key roles in the progression of cardiovascular disease in obesity, RAAS-targeted therapies contribute to improving endothelial dysfunction. By contrast, arginase reciprocally inhibits NO formation and promotes oxidative stress. Thus, targeting arginase activity as a key mediator in endothelial dysfunction has therapeutic potential in obesity-related vascular comorbidities. Obesity-related endothelial dysfunction plays a pivotal role in the progression of type 2 diabetes (T2D). The peroxisome proliferator-activated receptor gamma (PPARγ) agonist, rosiglitazone (thiazolidinedione), is a popular drug for treating diabetes; however, it leads to increased cardiovascular risk. Selective sodium-glucose co-transporter-2 (SGLT-2) inhibitor empagliflozin (EMPA) significantly improves endothelial dysfunction and mortality occurring through redox-dependent mechanisms. Although endothelial dysfunction and oxidative stress are alleviated by either metformin or EMPA, currently used drugs to treat obesity-related diabetes neither possess the same anti-inflammatory potential nor simultaneously target endothelial cell dysfunction and obesity equally. While therapeutic interventions with glucagon-like peptide-1 (GLP-1) receptor agonist liraglutide or bariatric surgery reverse regenerative cell exhaustion, support vascular repair mechanisms, and improve cardiometabolic risk in individuals with T2D and obesity, the GLP-1 analog exendin-4 attenuates endothelial endoplasmic reticulum stress.
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
|
9
|
Zhou HL, Grimmett ZW, Venetos NM, Stomberski CT, Qian Z, McLaughlin PJ, Bansal PK, Zhang R, Reynolds JD, Premont RT, Stamler JS. An enzyme that selectively S-nitrosylates proteins to regulate insulin signaling. Cell 2023; 186:5812-5825.e21. [PMID: 38056462 PMCID: PMC10794992 DOI: 10.1016/j.cell.2023.11.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 09/01/2023] [Accepted: 11/03/2023] [Indexed: 12/08/2023]
Abstract
Acyl-coenzyme A (acyl-CoA) species are cofactors for numerous enzymes that acylate thousands of proteins. Here, we describe an enzyme that uses S-nitroso-CoA (SNO-CoA) as its cofactor to S-nitrosylate multiple proteins (SNO-CoA-assisted nitrosylase, SCAN). Separate domains in SCAN mediate SNO-CoA and substrate binding, allowing SCAN to selectively catalyze SNO transfer from SNO-CoA to SCAN to multiple protein targets, including the insulin receptor (INSR) and insulin receptor substrate 1 (IRS1). Insulin-stimulated S-nitrosylation of INSR/IRS1 by SCAN reduces insulin signaling physiologically, whereas increased SCAN activity in obesity causes INSR/IRS1 hypernitrosylation and insulin resistance. SCAN-deficient mice are thus protected from diabetes. In human skeletal muscle and adipose tissue, SCAN expression increases with body mass index and correlates with INSR S-nitrosylation. S-nitrosylation by SCAN/SNO-CoA thus defines a new enzyme class, a unique mode of receptor tyrosine kinase regulation, and a revised paradigm for NO function in physiology and disease.
Collapse
Affiliation(s)
- Hua-Lin Zhou
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Zachary W Grimmett
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Nicholas M Venetos
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Colin T Stomberski
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Zhaoxia Qian
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Precious J McLaughlin
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Puneet K Bansal
- Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Rongli Zhang
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - James D Reynolds
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Anesthesiology and Perioperative Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Richard T Premont
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Jonathan S Stamler
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA.
| |
Collapse
|
10
|
Sirmakesyan S, Hajj A, Hamouda A, Cammisotto P, Campeau L. Synthesis and secretion of Nerve Growth Factor is regulated by Nitric Oxide in bladder cells in vitro under a hyperglycemic environment. Nitric Oxide 2023; 140-141:30-40. [PMID: 37699453 DOI: 10.1016/j.niox.2023.09.002] [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: 07/14/2023] [Revised: 08/23/2023] [Accepted: 09/09/2023] [Indexed: 09/14/2023]
Abstract
Urine samples of female patients with overactive bladder (OAB) are characterized by low levels of nerve growth factor (NGF) and elevated concentrations of nitric oxide (NO) compared to healthy controls. We therefore examined how NO might regulate NGF synthesis using rat bladder smooth muscle (SMCs) and urothelial (UROs) cells in culture. In UROs, incubation in hyperglycemic conditions to mimic insulin insensitivity present in the OAB cohort increased secretion of NO and concomitantly decreased NGF, except when the NO synthase inhibitor, l-NAME (1 mM) was present. Sodium nitroprusside (SNP) (300 μM, 24 h), a NO generator, decreased NGF levels and decreased cyclic GMP (cGMP) content, a process validated by the cGMP synthase inhibitor ODQ (100 μM). Alternatively, SNP increased mRNA of both NGF and matrix metalloproteinase-9 (MMP-9). MMP-9 knockout of UROs by Crispr-Cas9 potently decreased the effect of SNP on NGF, implying a dependent role of NO on MMP-9. On the other hand, matrix metalloproteinase-7 (MMP-7) activity was increased by SNP, which taken together with increase in NGF mRNA, suggests a compensatory mechanism. In SMCs, hyperglycemic conditions had the same effect on extracellular content of NO and NGF than in UROs. SNP also decreased NGF secretion but increased cGMP content. Stable permeable analogs of cGMP 8-(4-Chlorophenylthio)-cGMP (1 mM) and N2,2'-O-Dibutyryl-cGMP (3 mM) inhibited NGF release. NGF and MMP-9 mRNA expression was unchanged by SNP. Deletion of MMP-9 in SMCs by Crispr-Cas9 did not alter the effect of SNP. Finally, SNP decreased MMP-7 activity, diminishing the conversion of proNGF to NGF. These results demonstrate that enhanced NO secretion triggered by high glucose decreases NGF secretion through pathways unique for each cell type that involve cGMP and proteases MMP-7 and MMP-9. These results might help to explain our observations from the urine from patients with OAB associated with metabolic syndrome.
Collapse
Affiliation(s)
| | - Aya Hajj
- Lady Davis Institute for Medical Research, Montreal, Quebec, Canada
| | - Aalya Hamouda
- Lady Davis Institute for Medical Research, Montreal, Quebec, Canada
| | | | - Lysanne Campeau
- Lady Davis Institute for Medical Research, Montreal, Quebec, Canada; Urology Department, Jewish General Hospital, Montreal, Quebec, Canada.
| |
Collapse
|
11
|
Jun JS, You YL, Byun HJ, Han KH, Kim J, Jung JB, Choi HS, Han SH. Enzyme Activity and Lipogenesis Inhibition by Fermented Grain Using Natural Enzymes. Molecules 2023; 28:7285. [PMID: 37959705 PMCID: PMC10647246 DOI: 10.3390/molecules28217285] [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: 09/15/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
This study aims to compare the effects of three enzyme-rich foods, including one fermented (grain enzyme) and two non-fermented foods (enzyme foods 1 and 2), by investigating their antioxidant, anti-inflammatory, and anti-adipogenic properties. Grain enzyme exhibited the highest radical scavenging activity and was rich in antioxidant components, including total polyphenol and total flavonoid contents. Grain enzyme and enzyme foods 1 and 2 inhibited nitric oxide production by 27, 34, and 17%, respectively, at a concentration of 200 μg/mL in LPS-stimulated macrophages. Among the tested enzymes, grain enzyme demonstrated the strongest inhibition on the expression of inducible nitric oxide synthase (INOS), cyclooxygenase-2 (COX-2), and interleukin (IL)-1β, while Enzyme Food 2 exhibited the most significant suppression of IL-6 mRNA levels. Furthermore, Grain Enzyme demonstrated a stronger inhibitory effect compared to Enzyme Food 1 and 2. Grain Enzyme decreased the mRNA expression of peroxisome proliferator-activated receptor (PPAR)γ, CCAAT/enhancer-binding protein (C/EBP)α, and fatty acid-binding protein (FABP)4 by 28, 21, and 30%, respectively, at a concentration of 400 μg/mL. In summary, fermented grain enzymes outperformed non-fermented enzymes in suppressing inflammation and adipogenesis. This study highlights the anti-inflammatory and anti-adipogenic effects of grain enzyme, suggesting its potential as a valuable dietary supplement for managing metabolic disorders.
Collapse
Affiliation(s)
- Jin-Sung Jun
- Naraentech Co., Ltd., Sanhangni-gil, Janggun-myeon, Sejong-si 30054, Republic of Korea;
| | - Ye-Lim You
- Department of Food Nutrition, Sangmyung University, Hongjimun 2-Gil 20, Jongno-gu, Seoul 03016, Republic of Korea; (Y.-L.Y.); (H.-J.B.); (H.-S.C.)
| | - Ha-Jun Byun
- Department of Food Nutrition, Sangmyung University, Hongjimun 2-Gil 20, Jongno-gu, Seoul 03016, Republic of Korea; (Y.-L.Y.); (H.-J.B.); (H.-S.C.)
| | - Kyung-Hoon Han
- Institute of Human Behavior & Genetics, Korea University College of Medicine, Seoul 02841, Republic of Korea; (K.-H.H.); (J.K.)
| | - Jay Kim
- Institute of Human Behavior & Genetics, Korea University College of Medicine, Seoul 02841, Republic of Korea; (K.-H.H.); (J.K.)
| | - Jea-Bum Jung
- Wisedom Science Lab, Korea University, Seoul 02841, Republic of Korea;
| | - Hyeon-Son Choi
- Department of Food Nutrition, Sangmyung University, Hongjimun 2-Gil 20, Jongno-gu, Seoul 03016, Republic of Korea; (Y.-L.Y.); (H.-J.B.); (H.-S.C.)
| | - Sung-Hee Han
- Institute of Human Behavior & Genetics, Korea University College of Medicine, Seoul 02841, Republic of Korea; (K.-H.H.); (J.K.)
| |
Collapse
|
12
|
La Russa D, Barberio L, Marrone A, Perri A, Pellegrino D. Caloric Restriction Mitigates Kidney Fibrosis in an Aged and Obese Rat Model. Antioxidants (Basel) 2023; 12:1778. [PMID: 37760081 PMCID: PMC10525959 DOI: 10.3390/antiox12091778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/31/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Caloric restriction is an effective intervention to protract healthspan and lifespan in several animal models from yeast to primates, including humans. Caloric restriction has been found to induce cardiometabolic adaptations associated with improved health and to delay the onset and progression of kidney disease in different species, particularly in rodent models. In both aging and obesity, fibrosis is a hallmark of kidney disease, and epithelial-mesenchymal transition is a key process that leads to fibrosis and renal dysfunction during aging. In this study, we used an aged and obese rat model to evaluate the effect of long-term (6 months) caloric restriction (-40%) on renal damage both from a structural and functional point of view. Renal interstitial fibrosis was analyzed by histological techniques, whereas effects on mesenchymal (N-cadherin, Vimentin, Desmin and α-SMA), antioxidant (SOD1, SOD2, Catalase and GSTP1) inflammatory (YM1 and iNOS) markers and apoptotic/cell cycle (BAX, BCL2, pJNK, Caspase 3 and p27) pathways were investigated using Western blot analysis. Our results clearly showed that caloric restriction promotes cell cycle division and reduces apoptotic injury and fibrosis phenotype through inflammation attenuation and leukocyte infiltration. In conclusion, we highlight the beneficial effects of caloric restriction to preserve elderly kidney function.
Collapse
Affiliation(s)
- Daniele La Russa
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy; (L.B.); (A.M.); (D.P.)
- LARSO (Analysis and Research on Oxidative Stress Laboratory), University of Calabria, 87036 Rende, Italy
| | - Laura Barberio
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy; (L.B.); (A.M.); (D.P.)
- LARSO (Analysis and Research on Oxidative Stress Laboratory), University of Calabria, 87036 Rende, Italy
| | - Alessandro Marrone
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy; (L.B.); (A.M.); (D.P.)
| | - Anna Perri
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy;
| | - Daniela Pellegrino
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy; (L.B.); (A.M.); (D.P.)
- LARSO (Analysis and Research on Oxidative Stress Laboratory), University of Calabria, 87036 Rende, Italy
| |
Collapse
|
13
|
Daehn IS, Ekperikpe US, Stadler K. Redox regulation in diabetic kidney disease. Am J Physiol Renal Physiol 2023; 325:F135-F149. [PMID: 37262088 PMCID: PMC10393330 DOI: 10.1152/ajprenal.00047.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/08/2023] [Accepted: 05/20/2023] [Indexed: 06/03/2023] Open
Abstract
Diabetic kidney disease (DKD) is one of the most devastating complications of diabetes mellitus, where currently there is no cure available. Several important mechanisms contribute to the pathogenesis of this complication, with oxidative stress being one of the key factors. The past decades have seen a large number of publications with various aspects of this topic; however, the specific details of redox regulation in DKD are still unclear. This is partly because redox biology is very complex, coupled with a complex and heterogeneous organ with numerous cell types. Furthermore, often times terms such as "oxidative stress" or reactive oxygen species are used as a general term to cover a wide and rich variety of reactive species and their differing reactions. However, no reactive species are the same, and not all of them are capable of biologically relevant reactions or "redox signaling." The goal of this review is to provide a biochemical background for an array of specific reactive oxygen species types with varying reactivity and specificity in the kidney as well as highlight some of the advances in redox biology that are paving the way to a better understanding of DKD development and risk.
Collapse
Affiliation(s)
- Ilse S Daehn
- Division of Nephrology, Department of Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Ubong S Ekperikpe
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Krisztian Stadler
- Oxidative Stress and Disease Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, United States
| |
Collapse
|
14
|
Mauchauffée E, Leroy J, Chamcham J, Ejjoummany A, Maurel M, Nauton L, Ramassamy B, Mezghenna K, Boucher JL, Lajoix AD, Hernandez JF. S-Ethyl-Isothiocitrullin-Based Dipeptides and 1,2,4-Oxadiazole Pseudo-Dipeptides: Solid Phase Synthesis and Evaluation as NO Synthase Inhibitors. Molecules 2023; 28:5085. [PMID: 37446746 DOI: 10.3390/molecules28135085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/23/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
We previously reported dipeptidomimetic compounds as inhibitors of neuronal and/or inducible NO synthases (n/iNOS) with significant selectivity against endothelial NOS (eNOS). They were composed of an S-ethylisothiocitrullin-like moiety linked to an extension through a peptide bond or a 1,2,4-oxadiazole link. Here, we developed two further series where the extension size was increased to establish more favorable interactions in the NOS substrate access channel. The extension was introduced on the solid phase by the reductive alkylation of an amino-piperidine moiety or an aminoethyl segment in the case of dipeptide-like and 1,2,4-oxadiazole compounds, respectively, with various benzaldehydes. Compared to the previous series, more potent inhibitors were identified with IC50 in the micromolar to the submicromolar range, with significant selectivity toward nNOS. As expected, most compounds did not inhibit eNOS, and molecular modeling was carried out to characterize the reasons for the selectivity toward nNOS over eNOS. Spectral studies showed that compounds were interacting at the heme active site. Finally, selected inhibitors were found to inhibit intra-cellular iNOS and nNOS expressed in RAW264.7 and INS-1 cells, respectively.
Collapse
Affiliation(s)
- Elodie Mauchauffée
- Institut des Biomolécules Max Mousseron, CNRS, Univ. Montpellier, ENSCM, Pôle Chimie Balard, 34293 Montpellier, France
| | - Jérémy Leroy
- Centre Biocommunication en Cardio-Métabolique, Univ. Montpellier, UFR Pharmacie, 34093 Montpellier, France
| | - Jihanne Chamcham
- Institut des Biomolécules Max Mousseron, CNRS, Univ. Montpellier, ENSCM, Pôle Chimie Balard, 34293 Montpellier, France
| | - Abdelaziz Ejjoummany
- Institut des Biomolécules Max Mousseron, CNRS, Univ. Montpellier, ENSCM, Pôle Chimie Balard, 34293 Montpellier, France
| | - Manon Maurel
- Institut des Biomolécules Max Mousseron, CNRS, Univ. Montpellier, ENSCM, Pôle Chimie Balard, 34293 Montpellier, France
| | - Lionel Nauton
- Institut de Chimie de Clermont-Ferrand, Université Clermont-Auvergne, CNRS, 63178 Aubière, France
| | - Booma Ramassamy
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, UMR 8601, CNRS, Université Paris Descartes, CEDEX 06, 75270 Paris, France
| | - Karima Mezghenna
- Centre Biocommunication en Cardio-Métabolique, Univ. Montpellier, UFR Pharmacie, 34093 Montpellier, France
| | - Jean-Luc Boucher
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, UMR 8601, CNRS, Université Paris Descartes, CEDEX 06, 75270 Paris, France
| | - Anne-Dominique Lajoix
- Centre Biocommunication en Cardio-Métabolique, Univ. Montpellier, UFR Pharmacie, 34093 Montpellier, France
| | - Jean-François Hernandez
- Institut des Biomolécules Max Mousseron, CNRS, Univ. Montpellier, ENSCM, Pôle Chimie Balard, 34293 Montpellier, France
| |
Collapse
|
15
|
Abo-Zaid OA, Moawed FS, Ismail ES, Farrag MA. β-sitosterol attenuates high- fat diet-induced hepatic steatosis in rats by modulating lipid metabolism, inflammation and ER stress pathway. BMC Pharmacol Toxicol 2023; 24:31. [PMID: 37173727 PMCID: PMC10182633 DOI: 10.1186/s40360-023-00671-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most prevalent chronic hepatic disorder. The naturally occurring phytosterol; β-sitosterol has antiobesogenic and anti-diabetic properties. The purpose of this study was to explore the role of β-sitosterol in preventing hepatic steatosis induced by a high-fat diet (HFD) in rats. In the current study, to induce NAFLD in the female Wister rats, an HFD was administered to them for 8 weeks. The pathogenic severity of steatosis in rats receiving an HFD diet was dramatically decreased by oral administration of β-sitosterol. After administering β-sitosterol to HFD-induced steatosis for three weeks, several oxidative stress-related markers were then assessed. We showed that β-sitosterol reduced steatosis and the serum levels of triglycerides, transaminases (ALT and AST) and inflammatory markers (IL-1β and iNOS) compared to HFD-fed rats. Additionally, β-sitosterol reduced endoplasmic reticulum stress by preventing the overexpression of inositol-requiring enzyme-1 (IRE-1α), X-box binding protein 1(sXBP1) and C/EBP homologous protein (CHOP) genes which, showing a function in the homeostatic regulation of protein folding. Also, it was found that the expression of the lipogenic factors; peroxisome proliferator-activated receptor (PPAR-α), sterol regulatory element binding protein (SREBP-1c) and carnitine palmitoyltransferase-1(CPT-1), which are involved in the regulation of the fatty acid oxidation process, may be regulated by β-sitosterol. It can be concluded that β-sitosterol may prevent NAFLD by reducing oxidative stress, endoplasmic reticulum stress and inflammatory responses, which supports the possibility of using β-sitosterol as an alternative therapy for NAFLD. Together, β-sitosterol may be an option for NAFLD prevention.
Collapse
Affiliation(s)
- Omayma Ar Abo-Zaid
- Molecular Biology Department, Faculty of Vet. Med, Benha University, Banha, Egypt
| | - Fatma Sm Moawed
- Health Radiation Research, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt.
| | - Effet Soliman Ismail
- Molecular Biology Department, Faculty of Vet. Med, Benha University, Banha, Egypt
| | - Mostafa A Farrag
- Radiation Biology, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|
16
|
Abstract
Insulin action is impaired in type 2 diabetes. The functions of the hormone are an integrated product of insulin secretion from pancreatic β-cells and insulin clearance by receptor-mediated endocytosis and degradation, mostly in liver (hepatocytes) and, to a lower extent, in extrahepatic peripheral tissues. Substantial evidence indicates that genetic or acquired abnormalities of insulin secretion or action predispose to type 2 diabetes. In recent years, along with the discovery of the molecular foundation of receptor-mediated insulin clearance, such as through the membrane glycoprotein CEACAM1, a consensus has begun to emerge that reduction of insulin clearance contributes to the disease process. In this review, we consider the evidence suggesting a pathogenic role for reduced insulin clearance in insulin resistance, obesity, hepatic steatosis, and type 2 diabetes.
Collapse
Affiliation(s)
- Sonia M Najjar
- Department of Biomedical Sciences and the Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA;
| | - Sonia Caprio
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Amalia Gastaldelli
- Cardiometabolic Risk Unit, Institute of Clinical Physiology-National Research Council, Pisa, Italy
| |
Collapse
|
17
|
Fraga CG, Trostchansky A, Rocha BS, Laranjinha J, Rubbo H, Galleano M. (Poly)phenols and nitrolipids: Relevant participants in nitric oxide metabolism. Mol Aspects Med 2023; 89:101158. [PMID: 36517273 DOI: 10.1016/j.mam.2022.101158] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 12/14/2022]
Abstract
Nitric oxide (•NO) is an essential molecule able to control and regulate many biological functions. Additionally, •NO bears a potential toxicity or damaging effects under conditions of uncontrolled production, and because of its participation in redox-sensitive pathways and oxidizing reactions. Several plant (poly)phenols present in the diet are able to regulate the enzymes producing •NO (NOSs). In addition, (poly)phenols are implicated in defining •NO bioavailability, especially by regulating NADPH oxidases (NOXs), and the subsequent generation of superoxide and •NO depletion. Nitrolipids are compounds that are present in animal tissues because of dietary consumption, e.g. of olive oil, and/or as result of endogenous production. This endogenous production of nitrolipids is dependent on the nitrate/nitrite presence in the diet. Select nitrolipids, e.g. the nitroalkenes, are able to exert •NO-like signaling actions, and act as •NO reservoirs, becoming relevant for systemic •NO bioavailability. Furthermore, the presence of (poly)phenols in the stomach reduces dietary nitrite to •NO favoring nitrolipids formation. In this review we focus on the capacity of molecules representing these two groups of bioactives, i.e. (poly)phenols and nitrolipids, as relevant participants in •NO metabolism and bioavailability. This participation acquires especial relevance when human homeostasis is lost, for example under inflammatory conditions, in which the protective actions of (poly)phenols and/or nitrolipids have been associated with local and systemic •NO bioavailability.
Collapse
Affiliation(s)
- César G Fraga
- Physical Chemistry, School of Pharmacy and Biochemistry, Universidad de Buenos Aires, Buenos Aires, Argentina; Instituto de Bioquímica y Medicina Molecular-Dr. Alberto Boveris (IBIMOL), Universidad de Buenos Aires-CONICET, Buenos Aires, Argentina; Department of Nutrition, University of California, Davis, CA, USA
| | - Andrés Trostchansky
- Departamento de Bioquímica, Facultad de Medicina, Centro de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo, Uruguay
| | - Barbara S Rocha
- Faculty of Pharmacy and Center for Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - João Laranjinha
- Faculty of Pharmacy and Center for Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Homero Rubbo
- Departamento de Bioquímica, Facultad de Medicina, Centro de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo, Uruguay
| | - Monica Galleano
- Physical Chemistry, School of Pharmacy and Biochemistry, Universidad de Buenos Aires, Buenos Aires, Argentina; Instituto de Bioquímica y Medicina Molecular-Dr. Alberto Boveris (IBIMOL), Universidad de Buenos Aires-CONICET, Buenos Aires, Argentina.
| |
Collapse
|
18
|
Amer OE, Sabico S, Khattak MNK, Al-Daghri NM. Circulating Nitric Oxide and Metabolic Syndrome in Arab Children and Adolescents: A Case-Control Study. CHILDREN (BASEL, SWITZERLAND) 2023; 10:children10020210. [PMID: 36832339 PMCID: PMC9954847 DOI: 10.3390/children10020210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/12/2023] [Accepted: 01/21/2023] [Indexed: 01/27/2023]
Abstract
BACKGROUND Metabolic syndrome (MetS) represents a cluster of known cardiometabolic risk factors, which elevates the risk of type 2 diabetes mellitus (T2DM), atherosclerotic cardiovascular disease (CVD) and chronic kidney disease (CKD) in adults and, only recently, even in children and adolescents. Circulating nitric oxide (NOx) has been observed to influence MetS risk factors in adults, but this has been scarcely investigated in children. The aim of the present study was to determine whether circulating NOx levels correlate with known components of MetS in Arab children and adolescents. METHODS Anthropometrics, serum NOx, lipid profile and fasting glucose levels were measured in 740 Saudi Arabs aged 10-17 years (68.8% girls). The presence of MetS was screened using the criteria of de Ferranti et al. Results: Overall, serum NOx levels were significantly higher in MetS participants compared to non-MetS (25.7 µmol/L (10.1-46.7) versus 11.9 µmol/L (5.5-22.9), p < 0.001) even after adjustments for age, BMI and sex. With the exception of elevated blood pressure, higher circulating NOx significantly increased the odds for MetS and its components. Lastly, receiver operating characteristics (ROC) showed that NOx, as a diagnostic marker for MetS, had good sensitivity and was higher in boys than girls (all MetS participants: area under the curve (AUC) = 0.68, p < 0.001), (girls with MetS: AUC = 0.62, p = 0.002), (boys with MetS: AUC = 0.83, p < 0.001)). CONCLUSIONS MetS and most of its components were significantly associated with circulating NOx levels in Arab adolescents and may be a promising diagnostic biomarker for MetS.
Collapse
|
19
|
Exploratory Longitudinal Analysis of the Circulating CHIT1 Activity in Pediatric Patients with Obesity. CHILDREN (BASEL, SWITZERLAND) 2023; 10:children10010124. [PMID: 36670674 PMCID: PMC9857224 DOI: 10.3390/children10010124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/20/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023]
Abstract
Macrophage activation and cytokine release play a pivotal role in inflammation-mediated metabolic disturbances in obesity. The proinflammatory macrophage secretes human chitotriosidase (CHIT1). The expression of the CHIT1 in visceral adipose tissue is associated with cytokine production. Our study aimed to assess whether the CHIT1 circulating activity, as a macrophage activation indicator, reflects the change of the adiposity level and the insulin resistance (IR) in children with obesity. We longitudinally (median follow-up period of 7 months; IQR [5 to 8.5] and {2 to 13} months) evaluated the CHIT1 circulating activity, the adiposity level (waist circumference (WC), waist-to-hip ratio (WHR), waist-to-height ratio (WtHR), and body mass index (BMI)-for-age z score), and two surrogate markers of IR (Homeostatic Model Assessment for Insulin Resistance, HOMA-IR and the triglycerides-to-high density lipoprotein cholesterol ratio, TG/HDLc) in 29 pediatric patients (16 girls and 13 boys) with obesity. We found a significant reduction in CHIT1 circulating activity (Wilcoxon test, p = 0.015) and a decrease in TG/HDLc at the follow-up evaluation (Wilcoxon test, p < 0.001). Indicators of adiposity were positively correlated with HOMA-IR at baseline, among which WC was the sole indicator associated with HOMA-IR (Spearman’s rank correlation coefficients, p < 0.05) at follow-up. Human chitotriosidase has the potential to be a valuable measure of the progression of subclinical inflammation in children with obesity. Subclinical inflammation, as expressed by the circulating CHIT1 activity, progresses independently of the abdominal adiposity, as measured by the clinical indicators, and is associated with a change in insulin resistance.
Collapse
|
20
|
|
21
|
Khan I, Khan I, Usman M, Jianye Z, Wei ZX, Ping X, Zhiqiang L, Lizhe A. Analysis of the blood bacterial composition of patients with acute coronary syndrome and chronic coronary syndrome. Front Cell Infect Microbiol 2022; 12:943808. [PMID: 36268223 PMCID: PMC9577097 DOI: 10.3389/fcimb.2022.943808] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 09/13/2022] [Indexed: 11/30/2022] Open
Abstract
Emerging evidence revealed that the blood microbiota plays a role in several non-communicable diseases, including cardiovascular disease. However, the role of circulating microbes in atherosclerosis remains understudied. To test this hypothesis, we performed this study to investigate the microbial profile in the blood of Chines atherosclerosis volunteers. A total of seventy Acute Coronary Syndrome patients, seventy Chronic Coronary Syndrome patients, and seventy healthy individuals were examined using high-throughput Illumina Novaseq targeting the V3-V4 regions of the 16S rRNA gene. The relationship between atherosclerosis and blood microbiome, clinical variables, and their functional pathways were also investigated. Our study observed significantly higher alpha diversity indices (Chao1, p = 0.001, and Shannon, p = 0.004) in the acute coronary syndrome group compared with chronic coronary syndrome and healthy group, although a significantly lower alpha diversity was observed in the chronic coronary syndrome compared to acute coronary syndrome and healthy group. Beta diversity based on principal coordinate analysis demonstrated a major separation among the three groups. In addition, using linear discriminant analysis, a significant distinct taxon such as Actinobacteria _ phylum, and Staphylococcus_ genus in the healthy group; Firmicutes_ phylum, and Lactobacillus_ genus in the chronic coronary syndrome group, and Proteobacteria and Acidobacteriota _ phyla in acute coronary syndrome group were observed among three groups. Clusters of Orthologous Genes grouped and Kyoto Encyclopedia of Genes and Genomes pathways suggested a significant variation among all groups (p < 0.05). The blood microbiota analysis provides potential biomarkers for the detection of coronary syndromes in this population.
Collapse
Affiliation(s)
- Ikram Khan
- Department of Microbiology, School of Life Sciences, Lanzhou University, Lanzhou, China
- School of Stomatology, Northwest Minzu University, Lanzhou, China
| | - Imran Khan
- Department of Microbiology, Khyber Medical University Peshawar, Peshawar, Pakistan
| | - Muhammad Usman
- State Key Laboratory of Grassland Agro-ecosystem, Key Laboratory of Grassland Livestock Industry Innovation, Ministry of Agriculture and Ruler Affairs, College of Pastoral Agriculture Sciences and Technology, Lanzhou University, Lanzhou, China
| | - Zhou Jianye
- School of Stomatology, Northwest Minzu University, Lanzhou, China
| | - Zhang Xiao Wei
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou, China
| | - Xie Ping
- Department of Cardiology, Gansu Provincial Hospital, Lanzhou, China
| | - Li Zhiqiang
- School of Stomatology, Northwest Minzu University, Lanzhou, China
- *Correspondence: Li Zhiqiang, ; An Lizhe,
| | - An Lizhe
- Department of Microbiology, School of Life Sciences, Lanzhou University, Lanzhou, China
- *Correspondence: Li Zhiqiang, ; An Lizhe,
| |
Collapse
|
22
|
Liu X, Lu F, Chen X. Examination of the role of necroptotic damage-associated molecular patterns in tissue fibrosis. Front Immunol 2022; 13:886374. [PMID: 36110858 PMCID: PMC9468929 DOI: 10.3389/fimmu.2022.886374] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 08/08/2022] [Indexed: 11/29/2022] Open
Abstract
Fibrosis is defined as the abnormal and excessive deposition of extracellular matrix (ECM) components, which leads to tissue or organ dysfunction and failure. However, the pathological mechanisms underlying fibrosis remain unclear. The inflammatory response induced by tissue injury is closely associated with tissue fibrosis. Recently, an increasing number of studies have linked necroptosis to inflammation and fibrosis. Necroptosis is a type of preprogrammed death caused by death receptors, interferons, Toll-like receptors, intracellular RNA and DNA sensors, and other mediators. These activate receptor-interacting protein kinase (RIPK) 1, which recruits and phosphorylates RIPK3. RIPK3 then phosphorylates a mixed lineage kinase domain-like protein and causes its oligomerization, leading to rapid plasma membrane permeabilization, the release of cellular contents, and exposure of damage-associated molecular patterns (DAMPs). DAMPs, as inflammatory mediators, are involved in the loss of balance between extensive inflammation and tissue regeneration, leading to remodeling, the hallmark of fibrosis. In this review, we discuss the role of necroptotic DAMPs in tissue fibrosis and highlight the inflammatory responses induced by DAMPs in tissue ECM remodeling. By summarizing the existing literature on this topic, we underscore the gaps in the current research, providing a framework for future investigations into the relationship among necroptosis, DAMPs, and fibrosis, as well as a reference for later transformation into clinical treatment.
Collapse
Affiliation(s)
| | - Feng Lu
- *Correspondence: Feng Lu, ; Xihang Chen,
| | | |
Collapse
|
23
|
Najjar SM, Abdolahipour R, Ghadieh HE, Jahromi MS, Najjar JA, Abuamreh BAM, Zaidi S, Kumarasamy S, Muturi HT. Regulation of Insulin Clearance by Non-Esterified Fatty Acids. Biomedicines 2022; 10:biomedicines10081899. [PMID: 36009446 PMCID: PMC9405499 DOI: 10.3390/biomedicines10081899] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/27/2022] [Accepted: 08/03/2022] [Indexed: 11/16/2022] Open
Abstract
Insulin stores lipid in adipocytes and prevents lipolysis and the release of non-esterified fatty acids (NEFA). Excessive release of NEFA during sustained energy supply and increase in abdominal adiposity trigger systemic insulin resistance, including in the liver, a major site of insulin clearance. This causes a reduction in insulin clearance as a compensatory mechanism to insulin resistance in obesity. On the other hand, reduced insulin clearance in the liver can cause chronic hyperinsulinemia, followed by downregulation of insulin receptor and insulin resistance. Delineating the cause–effect relationship between reduced insulin clearance and insulin resistance has been complicated by the fact that insulin action and clearance are mechanistically linked to insulin binding to its receptors. This review discusses how NEFA mobilization contributes to the reciprocal relationship between insulin resistance and reduced hepatic insulin clearance, and how this may be implicated in the pathogenesis of non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Sonia M. Najjar
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- Correspondence: ; Tel.: +1-740-593-2376; Fax: +1-740-593-2320
| | - Raziyeh Abdolahipour
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Hilda E. Ghadieh
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Balamand P.O. Box 100, Lebanon
| | - Marziyeh Salehi Jahromi
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - John A. Najjar
- Department of Internal Medicine, College of Medicine, University of Toledo, Toledo, OH 43606, USA
| | - Basil A. M. Abuamreh
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Sobia Zaidi
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Sivarajan Kumarasamy
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Harrison T. Muturi
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| |
Collapse
|
24
|
Lundberg JO, Weitzberg E. Nitric oxide signaling in health and disease. Cell 2022; 185:2853-2878. [DOI: 10.1016/j.cell.2022.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/01/2022] [Accepted: 06/06/2022] [Indexed: 10/16/2022]
|
25
|
Chakarov S, Blériot C, Ginhoux F. Role of adipose tissue macrophages in obesity-related disorders. J Exp Med 2022; 219:213212. [PMID: 35543703 PMCID: PMC9098652 DOI: 10.1084/jem.20211948] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 04/17/2022] [Accepted: 04/18/2022] [Indexed: 11/04/2022] Open
Abstract
The obesity epidemic has led researchers and clinicians to reconsider the etiology of this disease and precisely decipher its molecular mechanisms. The excessive accumulation of fat by cells, most notably adipocytes, which play a key role in this process, has many repercussions in tissue physiology. Herein, we focus on how macrophages, immune cells well known for their tissue gatekeeping functions, assume fundamental, yet ill-defined, roles in the genesis and development of obesity-related metabolic disorders. We first discuss the determinants of the biology of these cells before introducing the specifics of the adipose tissue environment, while highlighting its heterogeneity. Finally, we detail how obesity transforms both adipose tissue and local macrophage populations. Understanding macrophage diversity and their cross talk with the diverse cell types constituting the adipose tissue environment will allow us to frame the therapeutic potential of adipose tissue macrophages in obesity.
Collapse
Affiliation(s)
- Svetoslav Chakarov
- Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Camille Blériot
- Institut Gustave Roussy, Batiment de Médecine Moléculaire, Villejuif, France
| | - Florent Ginhoux
- Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai, China.,Institut Gustave Roussy, Batiment de Médecine Moléculaire, Villejuif, France.,Singapore Immunology Network, Agency for Science, Technology, and Research, Singapore, Singapore.,Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| |
Collapse
|
26
|
Kociszewska D, Vlajkovic S. Age-Related Hearing Loss: The Link between Inflammaging, Immunosenescence, and Gut Dysbiosis. Int J Mol Sci 2022; 23:7348. [PMID: 35806352 PMCID: PMC9266910 DOI: 10.3390/ijms23137348] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/30/2022] [Accepted: 06/30/2022] [Indexed: 12/12/2022] Open
Abstract
This article provides a theoretical overview of the association between age-related hearing loss (ARHL), immune system ageing (immunosenescence), and chronic inflammation. ARHL, or presbyacusis, is the most common sensory disability that significantly reduces the quality of life and has a high economic impact. This disorder is linked to genetic risk factors but is also influenced by a lifelong cumulative effect of environmental stressors, such as noise, otological diseases, or ototoxic drugs. Age-related hearing loss and other age-related disorders share common mechanisms which often converge on low-grade chronic inflammation known as "inflammaging". Various stimuli can sustain inflammaging, including pathogens, cell debris, nutrients, and gut microbiota. As a result of ageing, the immune system can become defective, leading to the accumulation of unresolved inflammatory processes in the body. Gut microbiota plays a central role in inflammaging because it can release inflammatory mediators and crosstalk with other organ systems. A proinflammatory gut environment associated with ageing could result in a leaky gut and the translocation of bacterial metabolites and inflammatory mediators to distant organs via the systemic circulation. Here, we postulate that inflammaging, as a result of immunosenescence and gut dysbiosis, accelerates age-related cochlear degeneration, contributing to the development of ARHL. Age-dependent gut dysbiosis was included as a hypothetical link that should receive more attention in future studies.
Collapse
Affiliation(s)
| | - Srdjan Vlajkovic
- Department of Physiology and The Eisdell Moore Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, Auckland 1142, New Zealand;
| |
Collapse
|
27
|
Dubé L, Spahis S, Lachaîne K, Lemieux A, Monhem H, Poulin SM, Randoll C, Travaillaud E, Ould-Chikh NEH, Marcil V, Delvin E, Levy E. Specialized Pro-Resolving Mediators Derived from N-3 Polyunsaturated Fatty Acids: Role in Metabolic Syndrome and Related Complications. Antioxid Redox Signal 2022; 37:54-83. [PMID: 35072542 DOI: 10.1089/ars.2021.0156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Significance: Metabolic syndrome (MetS) prevalence continues to grow and represents a serious public health issue worldwide. This multifactorial condition carries the risk of hastening the development of type 2 diabetes (T2D), non-alcoholic fatty liver disease (NAFLD), and cardiovascular diseases (CVD). Another troubling aspect of MetS is the requirement of poly-pharmacological therapy not devoid of side effects. Therefore, there is an urgent need for prospecting alternative nutraceuticals as effective therapeutic agents for MetS. Recent Advances: Currently, there is an increased interest in understanding the regulation of metabolic derangements by specialized pro-resolving lipid mediators (SPMs), especially those derived from the long chain n-3 polyunsaturated fatty acids. Critical Issues: The SPMs are recognized as efficient modulators that are capable of inhibiting the production of pro-inflammatory cytokines, blocking neutrophil activation/recruitment, and inducing non-phlogistic (anti-inflammatory) activation of macrophage engulfment and removal of apoptotic inflammatory cells and debris. The aim of the present review is precisely to first underline key concepts relative to SPM functions before focusing on their status and actions on MetS components (e.g., obesity, glucose dysmetabolism, hyperlipidemia, hypertension) and complications such as T2D, NAFLD, and CVD. Future Directions: Valuable data from preclinical and clinical investigations have emphasized the SPM functions and influence on oxidative stress- and inflammation-related MetS. Despite these promising findings obtained without compromising host defense, additional efforts are needed to evaluate their potential therapeutic applications and further develop practical tools to monitor their bioavailability to cope with cardiometabolic disorders. Antioxid. Redox Signal. 37, 54-83.
Collapse
Affiliation(s)
- Laurent Dubé
- Research Centre, Sainte-Justine Hospital, Université de Montréal, Montreal, Canada.,Department of Nutrition, Université de Montréal, Montreal, Canada
| | - Schohraya Spahis
- Research Centre, Sainte-Justine Hospital, Université de Montréal, Montreal, Canada.,Department of Nutrition, Université de Montréal, Montreal, Canada.,Institute of Nutrition and Functional Foods, Laval University, Quebec City, Canada
| | - Karelle Lachaîne
- Department of Nutrition, Université de Montréal, Montreal, Canada
| | | | - Hanine Monhem
- Department of Nutrition, Université de Montréal, Montreal, Canada
| | | | - Carolane Randoll
- Department of Nutrition, Université de Montréal, Montreal, Canada
| | - Eva Travaillaud
- Department of Nutrition, Université de Montréal, Montreal, Canada
| | | | - Valérie Marcil
- Research Centre, Sainte-Justine Hospital, Université de Montréal, Montreal, Canada.,Department of Nutrition, Université de Montréal, Montreal, Canada.,Institute of Nutrition and Functional Foods, Laval University, Quebec City, Canada
| | - Edgard Delvin
- Research Centre, Sainte-Justine Hospital, Université de Montréal, Montreal, Canada.,Department of Biochemistry, Université de Montréal, Montreal, Canada
| | - Emile Levy
- Research Centre, Sainte-Justine Hospital, Université de Montréal, Montreal, Canada.,Department of Nutrition, Université de Montréal, Montreal, Canada.,Institute of Nutrition and Functional Foods, Laval University, Quebec City, Canada.,Department of Pediatrics, Gastroenterology & Hepatology Unit, Université de Montréal, Montreal, Canada
| |
Collapse
|
28
|
Mendes NF, Velloso LA. Perivascular macrophages in high-fat diet-induced hypothalamic inflammation. J Neuroinflammation 2022; 19:136. [PMID: 35681242 PMCID: PMC9185933 DOI: 10.1186/s12974-022-02519-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/01/2022] [Indexed: 02/06/2023] Open
Abstract
Brain macrophages and microglia are centrally involved in immune surveillance of the central nervous system. Upon inflammatory stimuli, they become reactive and release key molecules to prevent further damage to the neuronal network. In the hypothalamic area, perivascular macrophages (PVMs) are the first line of host defence against pathogenic organisms, particles and/or substances from the blood. They are distributed throughout the circumventricular organ median eminence, wrapping endothelial cells from fenestrated portal capillaries and in the hypothalamic vascular network, where they are localised in the perivascular space of the blood-brain barrier (BBB). Some studies have indicated that PVMs from the hypothalamus increase the expression of inducible nitric oxide synthase and vascular endothelial growth factor upon feeding for a long time on a high-fat diet. This adaptive response contributes to the impairment of glucose uptake, facilitates BBB leakage and leads to increased lipid and inflammatory cell influx towards the hypothalamic parenchyma. Despite these early findings, there is still a lack of studies exploring the mechanisms by which PVMs contribute to the development of obesity-related hypothalamic dysfunction, particularly at the early stages when there is chemotaxis of peripheral myeloid cells into the mediobasal hypothalamus. Here, we reviewed the studies involving the ontogeny, hallmarks and main features of brain PVMs in vascular homeostasis, inflammation and neuroendocrine control. This review provides a framework for understanding the potential involvement of PVMs in diet-induced hypothalamic inflammation.
Collapse
Affiliation(s)
- Natalia F Mendes
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of Campinas, Rua Carl Von Linnaeus s/n, Instituto de Biologia - Bloco Z. Campus Universitário Zeferino Vaz - Barão Geraldo, Campinas, SP, 13083-864, Brazil.
| | - Licio A Velloso
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of Campinas, Rua Carl Von Linnaeus s/n, Instituto de Biologia - Bloco Z. Campus Universitário Zeferino Vaz - Barão Geraldo, Campinas, SP, 13083-864, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation, Rio de Janeiro, Brazil
| |
Collapse
|
29
|
Zhou HL, Premont RT, Stamler JS. The manifold roles of protein S-nitrosylation in the life of insulin. Nat Rev Endocrinol 2022; 18:111-128. [PMID: 34789923 PMCID: PMC8889587 DOI: 10.1038/s41574-021-00583-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/08/2021] [Indexed: 02/04/2023]
Abstract
Insulin, which is released by pancreatic islet β-cells in response to elevated levels of glucose in the blood, is a critical regulator of metabolism. Insulin triggers the uptake of glucose and fatty acids into the liver, adipose tissue and muscle, and promotes the storage of these nutrients in the form of glycogen and lipids. Dysregulation of insulin synthesis, secretion, transport, degradation or signal transduction all cause failure to take up and store nutrients, resulting in type 1 diabetes mellitus, type 2 diabetes mellitus and metabolic dysfunction. In this Review, we make the case that insulin signalling is intimately coupled to protein S-nitrosylation, in which nitric oxide groups are conjugated to cysteine thiols to form S-nitrosothiols, within effectors of insulin action. We discuss the role of S-nitrosylation in the life cycle of insulin, from its synthesis and secretion in pancreatic β-cells, to its signalling and degradation in target tissues. Finally, we consider how aberrant S-nitrosylation contributes to metabolic diseases, including the roles of human genetic mutations and cellular events that alter S-nitrosylation of insulin-regulating proteins. Given the growing influence of S-nitrosylation in cellular metabolism, the field of metabolic signalling could benefit from renewed focus on S-nitrosylation in type 2 diabetes mellitus and insulin-related disorders.
Collapse
Affiliation(s)
- Hua-Lin Zhou
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Richard T Premont
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Jonathan S Stamler
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA.
| |
Collapse
|
30
|
Aggarwal H, Pathak P, Singh V, Kumar Y, Shankar M, Das B, Jagavelu K, Dikshit M. Vancomycin-Induced Modulation of Gram-Positive Gut Bacteria and Metabolites Remediates Insulin Resistance in iNOS Knockout Mice. Front Cell Infect Microbiol 2022; 11:795333. [PMID: 35127558 PMCID: PMC8807491 DOI: 10.3389/fcimb.2021.795333] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/23/2021] [Indexed: 12/27/2022] Open
Abstract
The role of oxidative and nitrosative stress has been implied in both physiology and pathophysiology of metabolic disorders. Inducible nitric oxide synthase (iNOS) has emerged as a crucial regulator of host metabolism and gut microbiota activity. The present study examines the role of the gut microbiome in determining host metabolic functions in the absence of iNOS. Insulin-resistant and dyslipidemic iNOS-/- mice displayed reduced microbial diversity, with a higher relative abundance of Allobaculum and Bifidobacterium, gram-positive bacteria, and altered serum metabolites along with metabolic dysregulation. Vancomycin, which largely depletes gram-positive bacteria, reversed the insulin resistance (IR), dyslipidemia, and related metabolic anomalies in iNOS-/- mice. Such improvements in metabolic markers were accompanied by alterations in the expression of genes involved in fatty acid synthesis in the liver and adipose tissue, lipid uptake in adipose tissue, and lipid efflux in the liver and intestine tissue. The rescue of IR in vancomycin-treated iNOS-/- mice was accompanied with the changes in select serum metabolites such as 10-hydroxydecanoate, indole-3-ethanol, allantoin, hippurate, sebacic acid, aminoadipate, and ophthalmate, along with improvement in phosphatidylethanolamine to phosphatidylcholine (PE/PC) ratio. In the present study, we demonstrate that vancomycin-mediated depletion of gram-positive bacteria in iNOS-/- mice reversed the metabolic perturbations, dyslipidemia, and insulin resistance.
Collapse
Affiliation(s)
- Hobby Aggarwal
- Pharmacology Division, Council of Scientific and Industrial Research (CSIR)-Central Drug Research Institute, Lucknow, India
| | - Priya Pathak
- Pharmacology Division, Council of Scientific and Industrial Research (CSIR)-Central Drug Research Institute, Lucknow, India
| | - Vishal Singh
- Department of Nutritional Sciences, The Pennsylvania State University, State College, PA, United States
| | - Yashwant Kumar
- Non-Communicable Diseases Division, Translational Health Science and Technology Institute, Faridabad, India
| | - Manoharan Shankar
- Microbial Physiology Laboratory, Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, India
| | - Bhabatosh Das
- Molecular Genetics Laboratory, Infection and Immunology Division, Translational Health Science and Technology Institute, Faridabad, India
| | - Kumaravelu Jagavelu
- Pharmacology Division, Council of Scientific and Industrial Research (CSIR)-Central Drug Research Institute, Lucknow, India
| | - Madhu Dikshit
- Pharmacology Division, Council of Scientific and Industrial Research (CSIR)-Central Drug Research Institute, Lucknow, India
- Non-Communicable Diseases Division, Translational Health Science and Technology Institute, Faridabad, India
| |
Collapse
|
31
|
Vilela VR, Samson N, Nachbar R, Perazza LR, Lachance G, Rokatoarivelo V, Centano-Baez C, Zancan P, Sola-Penna M, Bellmann K, Di Marzo V, Laplante M, Marette A. Adipocyte-specific Nos2 deletion improves insulin resistance and dyslipidemia through brown fat activation in diet-induced obese mice. Mol Metab 2022; 57:101437. [PMID: 35033724 PMCID: PMC8802131 DOI: 10.1016/j.molmet.2022.101437] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 12/22/2021] [Accepted: 01/03/2022] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE Inducible nitric oxide (NO) synthase (NOS2) is a well documented inflammatory mediator of insulin resistance in obesity. NOS2 expression is induced in both adipocytes and macrophages within adipose tissue during high-fat (HF)-induced obesity. METHODS Eight week old male mice with adipocyte selective deletion of the Nos2 gene (Nos2AD-KO) and their wildtype littermates (Nos2fl/fl) were subjected to chow or high-fat high-sucrose (HFHS) diet for 10 weeks followed by metabolic phenotyping and determination of brown adipose tissue (BAT) thermogenesis. The direct impact of NO on BAT mitochondrial respiration was also assessed in brown adipocytes. RESULTS Here, we show that HFHS-fed Nos2AD-KO mice had improved insulin sensitivity as compared to Nos2fl/fl littermates. Nos2AD-KO mice were also protected from HF-induced dyslipidemia and exhibited increased energy expenditure compared to Nos2fl/fl mice. This was linked to activation of BAT in HFHS-fed Nos2AD-KO mice as shown by increased Ucp1 and Ucp2 gene expression and augmented respiratory capacity of BAT mitochondria. Furthermore, mitochondrial respiration was inhibited by NO, or upon cytokine-induced NOS2 activation, but improved by NOS2 inhibition in brown adipocytes. CONCLUSIONS These results demonstrate a key role for adipocyte NOS2 in the development of obesity-linked insulin resistance and dyslipidemia, partly through NO dependent inhibition of BAT mitochondrial bioenergetics.
Collapse
Affiliation(s)
| | - Nolwenn Samson
- Quebec Heart & Lung Institute, Université Laval, 2725 Ch Ste-Foy, Québec, QC, G1V 4G5, Canada
| | - Renato Nachbar
- Quebec Heart & Lung Institute, Université Laval, 2725 Ch Ste-Foy, Québec, QC, G1V 4G5, Canada
| | - Lia Rossi Perazza
- Quebec Heart & Lung Institute, Université Laval, 2725 Ch Ste-Foy, Québec, QC, G1V 4G5, Canada
| | - Gabriel Lachance
- Quebec Heart & Lung Institute, Université Laval, 2725 Ch Ste-Foy, Québec, QC, G1V 4G5, Canada
| | - Volatiana Rokatoarivelo
- Quebec Heart & Lung Institute, Université Laval, 2725 Ch Ste-Foy, Québec, QC, G1V 4G5, Canada
| | - Carolina Centano-Baez
- Quebec Heart & Lung Institute, Université Laval, 2725 Ch Ste-Foy, Québec, QC, G1V 4G5, Canada
| | - Patricia Zancan
- Quebec Heart & Lung Institute, Université Laval, 2725 Ch Ste-Foy, Québec, QC, G1V 4G5, Canada
| | - Mauro Sola-Penna
- Quebec Heart & Lung Institute, Université Laval, 2725 Ch Ste-Foy, Québec, QC, G1V 4G5, Canada
| | - Kerstin Bellmann
- Quebec Heart & Lung Institute, Université Laval, 2725 Ch Ste-Foy, Québec, QC, G1V 4G5, Canada
| | - Vincenzo Di Marzo
- Quebec Heart & Lung Institute, Université Laval, 2725 Ch Ste-Foy, Québec, QC, G1V 4G5, Canada; Institute of Nutrition and Functional Foods, Centre NUTRISS, Université Laval, 2440 Boulevard Hochelaga Suite 1710, Québec, QC, G1V 0A6, Canada; Canada Excellence Research Chair Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND)
| | - Mathieu Laplante
- Quebec Heart & Lung Institute, Université Laval, 2725 Ch Ste-Foy, Québec, QC, G1V 4G5, Canada
| | - André Marette
- Quebec Heart & Lung Institute, Université Laval, 2725 Ch Ste-Foy, Québec, QC, G1V 4G5, Canada; Institute of Nutrition and Functional Foods, Centre NUTRISS, Université Laval, 2440 Boulevard Hochelaga Suite 1710, Québec, QC, G1V 0A6, Canada.
| |
Collapse
|
32
|
Luk C, Haywood NJ, Bridge KI, Kearney MT. Paracrine Role of the Endothelium in Metabolic Homeostasis in Health and Nutrient Excess. Front Cardiovasc Med 2022; 9:882923. [PMID: 35557517 PMCID: PMC9086712 DOI: 10.3389/fcvm.2022.882923] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 04/04/2022] [Indexed: 02/02/2023] Open
Abstract
The vascular endothelium traditionally viewed as a simple physical barrier between the circulation and tissue is now well-established as a key organ mediating whole organism homeostasis by release of a portfolio of anti-inflammatory and pro-inflammatory vasoactive molecules. Healthy endothelium releases anti-inflammatory signaling molecules such as nitric oxide and prostacyclin; in contrast, diseased endothelium secretes pro-inflammatory signals such as reactive oxygen species, endothelin-1 and tumor necrosis factor-alpha (TNFα). Endothelial dysfunction, which has now been identified as a hallmark of different components of the cardiometabolic syndrome including obesity, type 2 diabetes and hypertension, initiates and drives the progression of tissue damage in these disorders. Recently it has become apparent that, in addition to vasoactive molecules, the vascular endothelium has the potential to secrete a diverse range of small molecules and proteins mediating metabolic processes in adipose tissue (AT), liver, skeletal muscle and the pancreas. AT plays a pivotal role in orchestrating whole-body energy homeostasis and AT dysfunction, characterized by local and systemic inflammation, is central to the metabolic complications of obesity. Thus, understanding and targeting the crosstalk between the endothelium and AT may generate novel therapeutic opportunities for the cardiometabolic syndrome. Here, we provide an overview of the role of the endothelial secretome in controlling the function of AT. The endothelial-derived metabolic regulatory factors are grouped and discussed based on their physical properties and their downstream signaling effects. In addition, we focus on the therapeutic potential of these regulatory factors in treating cardiometabolic syndrome, and discuss areas of future study of potential translatable and clinical significance. The vascular endothelium is emerging as an important paracrine/endocrine organ that secretes regulatory factors in response to nutritional and environmental cues. Endothelial dysfunction may result in imbalanced secretion of these regulatory factors and contribute to the progression of AT and whole body metabolic dysfunction. As the vascular endothelium is the first responder to local nutritional changes and adipocyte-derived signals, future work elucidating the changes in the endothelial secretome is crucial to improve our understanding of the pathophysiology of cardiometabolic disease, and in aiding our development of new therapeutic strategies to treat and prevent cardiometabolic syndrome.
Collapse
Affiliation(s)
- Cheukyau Luk
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom
| | - Natalie J Haywood
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom
| | - Katherine I Bridge
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom
| | - Mark T Kearney
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
33
|
Sebag SC, Zhang Z, Qian Q, Li M, Zhu Z, Harata M, Li W, Zingman LV, Liu L, Lira VA, Potthoff MJ, Bartelt A, Yang L. ADH5-mediated NO bioactivity maintains metabolic homeostasis in brown adipose tissue. Cell Rep 2021; 37:110003. [PMID: 34788615 PMCID: PMC8640996 DOI: 10.1016/j.celrep.2021.110003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 08/23/2021] [Accepted: 10/22/2021] [Indexed: 01/21/2023] Open
Abstract
Brown adipose tissue (BAT) thermogenic activity is tightly regulated by cellular redox status, but the underlying molecular mechanisms are incompletely understood. Protein S-nitrosylation, the nitric-oxide-mediated cysteine thiol protein modification, plays important roles in cellular redox regulation. Here we show that diet-induced obesity (DIO) and acute cold exposure elevate BAT protein S-nitrosylation, including UCP1. This thermogenic-induced nitric oxide bioactivity is regulated by S-nitrosoglutathione reductase (GSNOR; alcohol dehydrogenase 5 [ADH5]), a denitrosylase that balances the intracellular nitroso-redox status. Loss of ADH5 in BAT impairs cold-induced UCP1-dependent thermogenesis and worsens obesity-associated metabolic dysfunction. Mechanistically, we demonstrate that Adh5 expression is induced by the transcription factor heat shock factor 1 (HSF1), and administration of an HSF1 activator to BAT of DIO mice increases Adh5 expression and significantly improves UCP1-mediated respiration. Together, these data indicate that ADH5 controls BAT nitroso-redox homeostasis to regulate adipose thermogenesis, which may be therapeutically targeted to improve metabolic health.
Collapse
Affiliation(s)
- Sara C. Sebag
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA,These authors contributed equally
| | - Zeyuan Zhang
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA,These authors contributed equally
| | - Qingwen Qian
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Mark Li
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Zhiyong Zhu
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Mikako Harata
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Wenxian Li
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Leonid V. Zingman
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Limin Liu
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Vitor A. Lira
- Department of Health and Human Physiology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA,College of Liberal Arts and Sciences, University of Iowa, Iowa City, IA 52242, USA
| | - Matthew J. Potthoff
- Department of Neuroscience and Pharmacology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Alexander Bartelt
- Institute for Cardiovascular Prevention, Ludwig Maximilians University Munich Pettenkoferstr. 9, 80336 Munich, Germany,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Technische Universität München, Biedersteiner Str. 29, 80802 München, Germany,Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Ingolstädter Landstr. 1, 85764 Neuherberg, Germany,Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA 02115, USA
| | - Ling Yang
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA,Lead contact,Correspondence:
| |
Collapse
|
34
|
Abstract
Obesity is a major risk factor for the development of comorbidities such as type 2 diabetes, neurodegenerative disorders, osteoarthritis, cancer, cardiovascular and renal diseases. The onset of obesity is linked to an increase of senescent cells within adipose tissue and other organs. Cellular senescence is a stress response that has been shown to be causally linked to aging and development of various age-related diseases such as obesity. The senescence-associated-secretory phenotype of senescent cells creates a chronic inflammatory milieu that leads to local and systemic dysfunction. The elimination of senescent cells using pharmacological approaches (i.e., senolytics) has been shown to delay, prevent, or alleviate obesity-related organ dysfunction.
Collapse
Affiliation(s)
- Selim Chaib
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Tamara Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
35
|
Khatchadourian C, Sisliyan C, Nguyen K, Poladian N, Tian Q, Tamjidi F, Luong B, Singh M, Robison J, Venketaraman V. Hyperlipidemia and Obesity's Role in Immune Dysregulation Underlying the Severity of COVID-19 Infection. Clin Pract 2021; 11:694-707. [PMID: 34698139 PMCID: PMC8544571 DOI: 10.3390/clinpract11040085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/17/2021] [Accepted: 09/17/2021] [Indexed: 12/23/2022] Open
Abstract
Obesity and hyperlipidemia are known to be risk factors for various pathological disorders, including various forms of infectious respiratory disease, including the current Coronavirus outbreak termed Coronavirus Disease 19 (COVID-19). This review studies the effects of hyperlipidemia and obesity on enhancing the inflammatory response seen in COVID-19 and potential therapeutic pathways related to these processes. In order to better understand the underlying processes of cytokine and chemokine-induced inflammation, we must further investigate the immunomodulatory effects of agents such as Vitamin D and the reduced form of glutathione as adjunctive therapies for COVID-19 disease.
Collapse
Affiliation(s)
- Christopher Khatchadourian
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (C.K.); (C.S.); (K.N.); (N.P.); (Q.T.); (F.T.); (B.L.)
| | - Christina Sisliyan
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (C.K.); (C.S.); (K.N.); (N.P.); (Q.T.); (F.T.); (B.L.)
| | - Kevin Nguyen
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (C.K.); (C.S.); (K.N.); (N.P.); (Q.T.); (F.T.); (B.L.)
| | - Nicole Poladian
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (C.K.); (C.S.); (K.N.); (N.P.); (Q.T.); (F.T.); (B.L.)
| | - Qi Tian
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (C.K.); (C.S.); (K.N.); (N.P.); (Q.T.); (F.T.); (B.L.)
| | - Faraaz Tamjidi
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (C.K.); (C.S.); (K.N.); (N.P.); (Q.T.); (F.T.); (B.L.)
| | - Bao Luong
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (C.K.); (C.S.); (K.N.); (N.P.); (Q.T.); (F.T.); (B.L.)
| | - Manpreet Singh
- Department of Emergency Medicine, St. Barnabas Hospital Health System, Bronx, NY 10457, USA; (M.S.); (J.R.)
| | - Jeremiah Robison
- Department of Emergency Medicine, St. Barnabas Hospital Health System, Bronx, NY 10457, USA; (M.S.); (J.R.)
| | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (C.K.); (C.S.); (K.N.); (N.P.); (Q.T.); (F.T.); (B.L.)
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 309 E Second Street, Pomona, CA 91766, USA
| |
Collapse
|
36
|
Koufakis T, Dimitriadis G, Metallidis S, Zebekakis P, Kotsa K. The role of autoimmunity in the pathophysiology of type 2 diabetes: Looking at the other side of the moon. Obes Rev 2021; 22:e13231. [PMID: 33682984 DOI: 10.1111/obr.13231] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 12/12/2022]
Abstract
Efforts to unravel the pathophysiological mechanisms of type 2 diabetes (T2D) have been traditionally trapped into a metabolic perspective. However, T2D is a phenotypically and pathophysiologically heterogenous disorder, and the need for a tailored approach in its management is becoming increasingly evident. There is emerging evidence that irregular immune responses contribute to the development of hyperglycemia in T2D and, inversely, that insulin resistance is a component of the pathogenesis of autoimmune diabetes. Nevertheless, it has not yet been fully elucidated to what extent the presence of conventional autoimmune markers, such as autoantibodies, in subjects with T2D might affect the natural history of the disease and particularly each response to various treatments. The challenge for future research in the field is the discovery of novel genetic, molecular, or phenotypical indicators that would enable the characterization of specific subpopulations of people with T2D who would benefit most from the addition of immunomodulatory therapies to standard glucose-lowering treatment. This narrative review aims to discuss the plausible mechanisms through which the immune system might be implicated in the development of metabolic disturbances in T2D and obesity and explore a potential role of immunotherapy in the future management of the disorder and its complications.
Collapse
Affiliation(s)
- Theocharis Koufakis
- Division of Endocrinology and Metabolism and Diabetes Center, First Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| | - George Dimitriadis
- Athens University Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Symeon Metallidis
- Infectious Diseases Division, First Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| | - Pantelis Zebekakis
- Division of Endocrinology and Metabolism and Diabetes Center, First Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece.,Infectious Diseases Division, First Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| | - Kalliopi Kotsa
- Division of Endocrinology and Metabolism and Diabetes Center, First Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| |
Collapse
|
37
|
Mohammad S, Al Zoubi S, Collotta D, Krieg N, Wissuwa B, Ferreira Alves G, Purvis GSD, Norata GD, Baragetti A, Catapano AL, Solito E, Zechendorf E, Schürholz T, Correa-Vargas W, Brandenburg K, Coldewey SM, Collino M, Yaqoob MM, Martin L, Thiemermann C. A Synthetic Peptide Designed to Neutralize Lipopolysaccharides Attenuates Metaflammation and Diet-Induced Metabolic Derangements in Mice. Front Immunol 2021; 12:701275. [PMID: 34349763 PMCID: PMC8328475 DOI: 10.3389/fimmu.2021.701275] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 06/29/2021] [Indexed: 12/17/2022] Open
Abstract
Metabolic endotoxemia has been suggested to play a role in the pathophysiology of metaflammation, insulin-resistance and ultimately type-2 diabetes mellitus (T2DM). The role of endogenous antimicrobial peptides (AMPs), such as the cathelicidin LL-37, in T2DM is unknown. We report here for the first time that patients with T2DM compared to healthy volunteers have elevated plasma levels of LL-37. In a reverse-translational approach, we have investigated the effects of the AMP, peptide 19-2.5, in a murine model of high-fat diet (HFD)-induced insulin-resistance, steatohepatitis and T2DM. HFD-fed mice for 12 weeks caused obesity, an impairment in glycemic regulations, hypercholesterolemia, microalbuminuria and steatohepatitis, all of which were attenuated by Peptide 19-2.5. The liver steatosis caused by feeding mice a HFD resulted in the activation of nuclear factor kappa light chain enhancer of activated B cells (NF-ĸB) (phosphorylation of inhibitor of kappa beta kinase (IKK)α/β, IκBα, translocation of p65 to the nucleus), expression of NF-ĸB-dependent protein inducible nitric oxide synthase (iNOS) and activation of the NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome, all of which were reduced by Peptide 19-2.5. Feeding mice, a HFD also resulted in an enhanced expression of the lipid scavenger receptor cluster of differentiation 36 (CD36) secondary to activation of extracellular signal-regulated kinases (ERK)1/2, both of which were abolished by Peptide 19-2.5. Taken together, these results demonstrate that the AMP, Peptide 19-2.5 reduces insulin-resistance, steatohepatitis and proteinuria. These effects are, at least in part, due to prevention of the expression of CD36 and may provide further evidence for a role of metabolic endotoxemia in the pathogenesis of metaflammation and ultimately T2DM. The observed increase in the levels of the endogenous AMP LL-37 in patients with T2DM may serve to limit the severity of the disease.
Collapse
Affiliation(s)
- Shireen Mohammad
- William Harvey Research Institute, Bart’s and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Sura Al Zoubi
- William Harvey Research Institute, Bart’s and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Department of Basic Medical Sciences, School of Medicine, Al-Balqa Applied University, As-Salt, Jordan
| | - Debora Collotta
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Nadine Krieg
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Bianka Wissuwa
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | | | - Gareth S. D. Purvis
- William Harvey Research Institute, Bart’s and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Sir William Dunn School Pathology, University of Oxford, Oxford, United Kingdom
| | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
- IRCCS Multimedica, Sesto San Giovanni, Milan, Italy
- Società Italiana per lo Studio della Aterosclerosi (S.I.S.A.) Centre for the Study of Atherosclerosis, Bassini Hospital, Milan, Italy
| | - Andrea Baragetti
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
- IRCCS Multimedica, Sesto San Giovanni, Milan, Italy
| | - Alberico Luigi Catapano
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
- IRCCS Multimedica, Sesto San Giovanni, Milan, Italy
- Società Italiana per lo Studio della Aterosclerosi (S.I.S.A.) Centre for the Study of Atherosclerosis, Bassini Hospital, Milan, Italy
| | - Egle Solito
- William Harvey Research Institute, Bart’s and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Universitá degli Studi di Napoli “Federico II”, Napoli, Italy
| | - Elisabeth Zechendorf
- Department of Intensive Care and Intermediate Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Tobias Schürholz
- Department of Intensive Care and Intermediate Care, University Hospital RWTH Aachen, Aachen, Germany
| | | | - Klaus Brandenburg
- Brandenburg Antiinfektiva GmbH, c/o Forschungszentrum Borstel, Borstel, Germany
| | - Sina M. Coldewey
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Massimo Collino
- Department of Neurosciences “Rita Levi Montalcini”, University of Turin, Turin, Italy
| | - Muhammad M. Yaqoob
- William Harvey Research Institute, Bart’s and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Lukas Martin
- William Harvey Research Institute, Bart’s and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Department of Intensive Care and Intermediate Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Christoph Thiemermann
- William Harvey Research Institute, Bart’s and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
38
|
Ozbayer C, Kebapci MN, Kurt H, Colak E, Gunes HV, Degirmenci I. Potential associations between variants of genes encoding regulators of inflammation, and mediators of inflammation in type 2 diabetes and insulin resistance. J Clin Pharm Ther 2021; 46:1395-1403. [PMID: 34145611 DOI: 10.1111/jcpt.13471] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/01/2021] [Accepted: 06/05/2021] [Indexed: 02/06/2023]
Abstract
WHAT IS KNOWN AND OBJECTIVE Type 2 diabetes (T2DM) is a multigenic disease that develops with impaired β-cell function and insulin sensitivity and has a high prevalence worldwide. A cause often postulated for type 2 diabetes is chronic inflammation. It has been suggested that inflammatory regulators can inhibit insulin signal transduction and that inflammation is involved in insulin resistance (IR) and the pathogenesis of type 2 diabetes. In this direction, we aimed to investigate the gene variants of MyD88 (rs1319438, rs199396), IRAK4 (rs1461567, rs4251513, rs4251559) and TRAF6 (rs331455, rs331457) and serum levels of COX-2, NF-κB, iNOS in T2DM and IR. METHODS The MyD88, IRAK4 and TRAF6 variations were genotyped in 100 newly diagnosed T2DM patients and 100 non-diabetic individuals using The MassARRAY® Iplex GOLD SNP genotyping method. The COX-2, iNOS and NF-κB levels were measured in serum samples with the sandwich-ELISA method. Results were analysed using SPSS Statistics software and the online FINNETI program. RESULTS AND DISCUSSION In our study, a total of the 7 variants in the MyD88, IRAK4 and TRAF6 genes were genotyped, and as a result, no relationship was found between most of these variants and the risk of type 2 diabetes and insulin resistance (p > 0.05). Only, the rs1461567 variant of the IRAK4 gene was significant in the heterozygous model (CC vs. CT), and the CT genotype was most frequent in diabetic individuals compared with the non-diabetics (p = 0.033). Additionally, COX-2 and iNOS levels were found to be associated with diabetes and insulin resistance (p < 0.05). WHAT IS NEW AND CONCLUSION Our results show that high COX-2 and iNOS levels are associated with T2DM, besides MyD88, IRAK4 and TRAF6 gene variations may not be closely related to type 2 diabetes and insulin resistance. Nevertheless, studies in this pathway with a different population and a large number of patients are important.
Collapse
Affiliation(s)
- Cansu Ozbayer
- Medical Faculty, Department of Medical Biology, Kutahya Health Sciences University, Kutahya, Turkey
| | - Medine Nur Kebapci
- Medical Faculty, Department of Endocrinology, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Hulyam Kurt
- Medical Faculty, Department of Medical Biology, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Ertugrul Colak
- Medical Faculty, Department of Biostatistics, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Hasan Veysi Gunes
- Medical Faculty, Department of Medical Biology, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Irfan Degirmenci
- Medical Faculty, Department of Medical Biology, Kutahya Health Sciences University, Kutahya, Turkey
| |
Collapse
|
39
|
McDowell SAC, Luo RBE, Arabzadeh A, Doré S, Bennett NC, Breton V, Karimi E, Rezanejad M, Yang RR, Lach KD, Issac MSM, Samborska B, Perus LJM, Moldoveanu D, Wei Y, Fiset B, Rayes RF, Watson IR, Kazak L, Guiot MC, Fiset PO, Spicer JD, Dannenberg AJ, Walsh LA, Quail DF. Neutrophil oxidative stress mediates obesity-associated vascular dysfunction and metastatic transmigration. ACTA ACUST UNITED AC 2021; 2:545-562. [DOI: 10.1038/s43018-021-00194-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 03/10/2021] [Indexed: 12/22/2022]
|
40
|
Zhang L, Wang L, Yuan X, Zhong M, Chen H, Zhang D, Han X, Xie S, He L, Li Y, Chen F, Liu Y, Tan W. Decoding the Complex Free Radical Cascade by Using a DNA Framework-Based Artificial DNA Encoder. Angew Chem Int Ed Engl 2021; 60:10745-10755. [PMID: 33555644 DOI: 10.1002/anie.202014088] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/21/2020] [Indexed: 02/06/2023]
Abstract
DNA-based molecular communications (DMC) are critical for regulating biological networks to maintain stable organismic functions. However, the complicated, time-consuming information transmission process involved in genome-coded DMC and the limited, vulnerable decoding activity generally lead to communication impairment or failure, in response to external stimuli. Herein, we present a conceptually innovative DMC strategy mediated by the DNA framework-based artificial DNA encoder. With the free-radical cascade as a proof-of-concept study, the artificial DNA encoder shows active sensing and real-time actuation, in situ and broad free radical-decoding efficacy, as well as robust resistance to environmental noise. It can also block undesirable short-to-medium-range communications between free radicals and inflammatory networks, leading to a synergistic anti-obesity effect. The artificial DNA encoder-based DMC may be generalized to other communication systems for a variety of applications.
Collapse
Affiliation(s)
- Lili Zhang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Linlin Wang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Xi Yuan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Minjuan Zhong
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Hong Chen
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Dailiang Zhang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Xiaoyan Han
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Sitao Xie
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Lei He
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Yazhou Li
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Fengming Chen
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Yanlan Liu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China.,The Cancer Hospital of the University of, Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China.,Foundation for Applied Molecular Evolution, 13709 Progress Boulevard, Alachua, FL, 32615, USA
| |
Collapse
|
41
|
Huffman AM, Rezq S, Basnet J, Yanes Cardozo LL, Romero DG. SARS-CoV-2 Viral Entry Proteins in Hyperandrogenemic Female Mice: Implications for Women with PCOS and COVID-19. Int J Mol Sci 2021; 22:4472. [PMID: 33922918 PMCID: PMC8123333 DOI: 10.3390/ijms22094472] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/14/2021] [Accepted: 04/20/2021] [Indexed: 02/06/2023] Open
Abstract
SARS-CoV-2, the causative agent of COVID-19, infects host cells using the angiotensin I converting enzyme 2 (ACE2) as its receptor after priming by host proteases, including TMPRSS2. COVID-19 affects multiple organ systems, and male patients suffer increased severity and mortality. Polycystic Ovary Syndrome (PCOS) is the most common endocrine disorder in reproductive-age women and is characterized by hyperandrogenism, ovulatory dysfunction, and polycystic ovarian morphology. PCOS is associated with obesity and cardiometabolic comorbidities, both being risk factors associated with severe COVID-19 pathology. We hypothesize that elevated androgens in PCOS regulate SARS-CoV-2 entry proteins in multiple tissues increasing the risk for this population. Female mice were treated with dihydrotestosterone (DHT) for 90 days. Body composition was measured by EchoMRI. Fasting glucose was determined by an enzymatic method. mRNA and protein levels of ACE2, Tmprss2, Cathepsin L, Furin, Tmprss4, and Adam17 were quantified by RT-qPCR, Western-blot, or ELISA in tissues, serum, and urine. DHT treatment increased body weight, fat and lean mass, and fasting glucose. Ace2 mRNA was upregulated in the lung, cecum, heart, and kidney, while downregulated in the brain by DHT. ACE2 protein was upregulated by DHT in the small intestine, heart, and kidney. The SARS-CoV-2 priming proteases Tmprss2, Cathepsin L, and Furin mRNA were upregulated by DHT in the kidney. ACE2 sheddase Adam17 mRNA was upregulated by DHT in the kidney, which corresponded with increased urinary ACE2 in DHT treated mice. Our results highlight the potential for increased cardiac, renal, and gastrointestinal dysfunction in PCOS women with COVID-19.
Collapse
Affiliation(s)
- Alexandra M. Huffman
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (A.M.H.); (S.R.); (J.B.); (L.L.Y.C.)
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Cardio Renal Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Samar Rezq
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (A.M.H.); (S.R.); (J.B.); (L.L.Y.C.)
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Cardio Renal Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Jelina Basnet
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (A.M.H.); (S.R.); (J.B.); (L.L.Y.C.)
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Cardio Renal Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Licy L. Yanes Cardozo
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (A.M.H.); (S.R.); (J.B.); (L.L.Y.C.)
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Cardio Renal Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Damian G. Romero
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (A.M.H.); (S.R.); (J.B.); (L.L.Y.C.)
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Cardio Renal Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
42
|
Zhang L, Wang L, Yuan X, Zhong M, Chen H, Zhang D, Han X, Xie S, He L, Li Y, Chen F, Liu Y, Tan W. Decoding the Complex Free Radical Cascade by Using a DNA Framework‐Based Artificial DNA Encoder. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202014088] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Lili Zhang
- Molecular Science and Biomedicine Laboratory (MBL) State Key Laboratory of Chemo/Bio-Sensing and Chemometrics College of Chemistry and Chemical Engineering Aptamer Engineering Center of Hunan Province Hunan University Changsha Hunan 410082 China
| | - Linlin Wang
- Molecular Science and Biomedicine Laboratory (MBL) State Key Laboratory of Chemo/Bio-Sensing and Chemometrics College of Chemistry and Chemical Engineering Aptamer Engineering Center of Hunan Province Hunan University Changsha Hunan 410082 China
| | - Xi Yuan
- Molecular Science and Biomedicine Laboratory (MBL) State Key Laboratory of Chemo/Bio-Sensing and Chemometrics College of Chemistry and Chemical Engineering Aptamer Engineering Center of Hunan Province Hunan University Changsha Hunan 410082 China
| | - Minjuan Zhong
- Molecular Science and Biomedicine Laboratory (MBL) State Key Laboratory of Chemo/Bio-Sensing and Chemometrics College of Chemistry and Chemical Engineering Aptamer Engineering Center of Hunan Province Hunan University Changsha Hunan 410082 China
| | - Hong Chen
- Molecular Science and Biomedicine Laboratory (MBL) State Key Laboratory of Chemo/Bio-Sensing and Chemometrics College of Chemistry and Chemical Engineering Aptamer Engineering Center of Hunan Province Hunan University Changsha Hunan 410082 China
| | - Dailiang Zhang
- Molecular Science and Biomedicine Laboratory (MBL) State Key Laboratory of Chemo/Bio-Sensing and Chemometrics College of Chemistry and Chemical Engineering Aptamer Engineering Center of Hunan Province Hunan University Changsha Hunan 410082 China
| | - Xiaoyan Han
- Molecular Science and Biomedicine Laboratory (MBL) State Key Laboratory of Chemo/Bio-Sensing and Chemometrics College of Chemistry and Chemical Engineering Aptamer Engineering Center of Hunan Province Hunan University Changsha Hunan 410082 China
| | - Sitao Xie
- Molecular Science and Biomedicine Laboratory (MBL) State Key Laboratory of Chemo/Bio-Sensing and Chemometrics College of Chemistry and Chemical Engineering Aptamer Engineering Center of Hunan Province Hunan University Changsha Hunan 410082 China
| | - Lei He
- Molecular Science and Biomedicine Laboratory (MBL) State Key Laboratory of Chemo/Bio-Sensing and Chemometrics College of Chemistry and Chemical Engineering Aptamer Engineering Center of Hunan Province Hunan University Changsha Hunan 410082 China
| | - Yazhou Li
- Molecular Science and Biomedicine Laboratory (MBL) State Key Laboratory of Chemo/Bio-Sensing and Chemometrics College of Chemistry and Chemical Engineering Aptamer Engineering Center of Hunan Province Hunan University Changsha Hunan 410082 China
| | - Fengming Chen
- Molecular Science and Biomedicine Laboratory (MBL) State Key Laboratory of Chemo/Bio-Sensing and Chemometrics College of Chemistry and Chemical Engineering Aptamer Engineering Center of Hunan Province Hunan University Changsha Hunan 410082 China
| | - Yanlan Liu
- Molecular Science and Biomedicine Laboratory (MBL) State Key Laboratory of Chemo/Bio-Sensing and Chemometrics College of Chemistry and Chemical Engineering Aptamer Engineering Center of Hunan Province Hunan University Changsha Hunan 410082 China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory (MBL) State Key Laboratory of Chemo/Bio-Sensing and Chemometrics College of Chemistry and Chemical Engineering Aptamer Engineering Center of Hunan Province Hunan University Changsha Hunan 410082 China
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital) Institute of Basic Medicine and Cancer (IBMC) Chinese Academy of Sciences Hangzhou Zhejiang 310022 China
- Foundation for Applied Molecular Evolution 13709 Progress Boulevard Alachua FL 32615 USA
| |
Collapse
|
43
|
Gómez-Zorita S, Queralt M, Vicente MA, González M, Portillo MP. Metabolically healthy obesity and metabolically obese normal weight: a review. J Physiol Biochem 2021; 77:175-189. [PMID: 33704694 DOI: 10.1007/s13105-020-00781-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 12/23/2020] [Indexed: 02/07/2023]
Abstract
Despite the general relationship between obesity and its co-morbidities, there are both obese individuals who scarcely present the associated pathologies (metabolically healthy obese; MHO) and individuals who present obesity alterations despite having normal weight (metabolically obese normal weight; MONW). It is still difficult to define metabolically MHO and MONW individuals because different classifications have been used in the studies reported. Indeed, different inclusion criteria have been used to discriminate between metabolically healthy and metabolically unhealthy subjects. Due to this and other reasons, such as differences in ethnicity, genetics, and lifestyle of the populations, data concerning the prevalence of MHO and MONW are very variable. The main determinants of MHO are type of growth (hypertrophy or hyperplasia), anatomical location, inflammation of adipose tissue, ectopic fat accumulation, genetic factors, and lifestyles factors. In the case of MONW, the main determinants are genetic background and lifestyle factors. With regard to treatment, it is not clear whether MHO subjects would benefit from traditional lifestyle interventions, based on diet energy restriction and increased physical activity. For MONW subjects, there is still no specialized treatment, and the therapies are the same as those used in obese subjects.
Collapse
Affiliation(s)
- Saioa Gómez-Zorita
- Nutrition and Obesity Group. Department of Nutrition and Food Science, University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Institute, Vitoria, Spain. .,BIOARABA Health Research Institute, Vitoria, Spain. .,CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Vitoria, Spain.
| | - Maite Queralt
- Nutrition and Obesity Group. Department of Nutrition and Food Science, University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Institute, Vitoria, Spain
| | - Maria Angeles Vicente
- BIOARABA Health Research Institute, Vitoria, Spain.,Alava University Hospital (Osakidetza), Vitoria, Spain
| | - Marcela González
- Nutrition and Food Science Department, Faculty of Biochemistry and Biological Sciences, National University of Litoral and National Scientific and Technical Research Council (CONICET), 3000, Santa Fe, Argentina
| | - María P Portillo
- Nutrition and Obesity Group. Department of Nutrition and Food Science, University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Institute, Vitoria, Spain.,BIOARABA Health Research Institute, Vitoria, Spain.,CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Vitoria, Spain
| |
Collapse
|
44
|
Kowalska K, Dembczyński R, Gołąbek A, Olkowicz M, Olejnik A. ROS Modulating Effects of Lingonberry ( Vaccinium vitis-idaea L.) Polyphenols on Obese Adipocyte Hypertrophy and Vascular Endothelial Dysfunction. Nutrients 2021; 13:nu13030885. [PMID: 33803343 PMCID: PMC7999824 DOI: 10.3390/nu13030885] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/28/2021] [Accepted: 03/05/2021] [Indexed: 02/06/2023] Open
Abstract
Oxidative stress and dysregulated adipocytokine secretion accompanying hypertrophied adipose tissue induce chronic inflammation, which leads to vascular endothelial dysfunction. The present study investigated the ability of anthocyanin (ACN) and non-anthocyanin polyphenol (PP) fractions from lingonberry fruit to mitigate adipose tissue hypertrophy and endothelial dysfunction using 3T3-L1 adipocytes and human umbilical vein endothelial cells (HUVECs). This study showed that the PP fraction decreased intracellular ROS generation in hypertrophied adipocytes by enhancing antioxidant enzyme expression (SOD2) and inhibiting oxidant enzyme expression (NOX4, iNOS). Moreover, PP and ACN fractions reduced triglyceride content in adipocytes accompanied by downregulation of the expression of lipogenic genes such as aP2, FAS, and DAGT1. Treatment with both fractions modulated the mRNA expression and protein secretion of key adipokines in hypertrophied adipocytes. Expression and secretion of leptin and adiponectin were, respectively, down- and upregulated. Furthermore, PP and ACN fractions alleviated the inflammatory response in TNF-α-induced HUVECs by inhibiting the expression of pro-inflammatory genes (IL-6, IL-1β) and adhesion molecules (VCAM-1, ICAM-1, SELE). The obtained results suggest that consuming polyphenol-rich lingonberry fruit may help prevent and treat obesity and endothelial dysfunction due to their antioxidant and anti-inflammatory actions.
Collapse
Affiliation(s)
- Katarzyna Kowalska
- Department of Biotechnology and Food Microbiology, Poznan University of Life Sciences, 48 Wojska Polskiego St., 60-627 Poznan, Poland; (K.K.); (R.D.); (A.G.)
| | - Radosław Dembczyński
- Department of Biotechnology and Food Microbiology, Poznan University of Life Sciences, 48 Wojska Polskiego St., 60-627 Poznan, Poland; (K.K.); (R.D.); (A.G.)
| | - Agata Gołąbek
- Department of Biotechnology and Food Microbiology, Poznan University of Life Sciences, 48 Wojska Polskiego St., 60-627 Poznan, Poland; (K.K.); (R.D.); (A.G.)
| | - Mariola Olkowicz
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, 14 Bobrzynskiego St., 30-348 Krakow, Poland;
| | - Anna Olejnik
- Department of Biotechnology and Food Microbiology, Poznan University of Life Sciences, 48 Wojska Polskiego St., 60-627 Poznan, Poland; (K.K.); (R.D.); (A.G.)
- Correspondence:
| |
Collapse
|
45
|
Proinflammatory cytokine polarization in type 2 diabetes. Cent Eur J Immunol 2021; 45:170-175. [PMID: 33456327 PMCID: PMC7792447 DOI: 10.5114/ceji.2020.97904] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 08/16/2017] [Indexed: 11/17/2022] Open
Abstract
Subclinical inflammatory reaction is associated with non-insulin dependent diabetes. Therefore, the aim of the present study is to describe the effect of the three cytokines: interferon γ (IFN-γ), interleukin (IL)-4 and IL-5 on the development of type 2 diabetes (T2D). Forty-five volunteers (after their permission) were participated in this work; according to their clinical examination and laboratory investigations (fasting blood sugar, 2 hours postprandial, HbA1c and lipid profile), they were divided into thirteen control (non-diabetic) (five females and eight males) and thirty-two diabetic patients (twenty-one females and eleven males). Thereafter, their sera were evaluated for C-reactive protein (CRP), IFN-γ, IL-4 and IL-5. The results revealed an increasing trend of CRP and a significant increase of IFN-γ in diabetic patients with no sex difference. A positive correlation between IFN-γ and both IL-4 and IL-5 in control, and a positive correlation between IL-4 and IL-5 in diabetic patients had been visualized. These results denoted that there may be an association of the pro-inflammatory cytokines in the etiology of diabetes mellitus type 2.
Collapse
|
46
|
Patel B, New LE, Griffiths JC, Deuchars J, Filippi BM. Inhibition of mitochondrial fission and iNOS in the dorsal vagal complex protects from overeating and weight gain. Mol Metab 2020; 43:101123. [PMID: 33227495 PMCID: PMC7753200 DOI: 10.1016/j.molmet.2020.101123] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/17/2020] [Accepted: 11/17/2020] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES The dorsal vagal complex (DVC) senses insulin and controls glucose homeostasis, feeding behaviour and body weight. Three-days of high-fat diet (HFD) in rats are sufficient to induce insulin resistance in the DVC and impair its ability to regulate feeding behaviour. HFD-feeding is associated with increased dynamin-related protein 1 (Drp1)-dependent mitochondrial fission in the DVC. We investigated the effects that altered Drp1 activity in the DVC has on feeding behaviour. Additionally, we aimed to uncover the molecular events and the neuronal cell populations associated with DVC insulin sensing and resistance. METHODS Eight-week-old male Sprague Dawley rats received DVC stereotactic surgery for brain infusion to facilitate the localised administration of insulin or viruses to express mutated forms of Drp1 or to knockdown inducible nitric oxide synthase (iNOS) in the NTS of the DVC. High-Fat diet feeding was used to cause insulin resistance and obesity. RESULTS We showed that Drp1 activation in the DVC increases weight gain in rats and Drp1 inhibition in HFD-fed rats reduced food intake, weight gain and adipose tissue. Rats expressing active Drp1 in the DVC had higher levels of iNOS and knockdown of DVC iNOS in HFD-fed rats led to a reduction of food intake, weight gain and adipose tissue. Finally, inhibiting mitochondrial fission in DVC astrocytes was sufficient to protect rats from HFD-dependent insulin resistance, hyperphagia, weight gain and fat deposition. CONCLUSION We uncovered new molecular and cellular targets for brain regulation of whole-body metabolism, which could inform new strategies to combat obesity and diabetes.
Collapse
Affiliation(s)
- Bianca Patel
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, United Kingdom
| | - Lauryn E New
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, United Kingdom
| | - Joanne C Griffiths
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, United Kingdom
| | - Jim Deuchars
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, United Kingdom
| | - Beatrice M Filippi
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, United Kingdom.
| |
Collapse
|
47
|
Durand R, Ouellette A, Houde VP, Guénard F, Varin TV, Marcotte B, Pilon G, Fraboulet E, Vohl MC, Marette A, Bazinet L. Animal and Cellular Studies Demonstrate Some of the Beneficial Impacts of Herring Milt Hydrolysates on Obesity-Induced Glucose Intolerance and Inflammation. Nutrients 2020; 12:nu12113235. [PMID: 33105775 PMCID: PMC7690616 DOI: 10.3390/nu12113235] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 10/13/2020] [Accepted: 10/16/2020] [Indexed: 12/14/2022] Open
Abstract
The search for bioactive compounds from enzymatic hydrolysates has increased in the last few decades. Fish by-products have been shown to be rich in these valuable molecules; for instance, herring milt is a complex matrix composed of lipids, nucleotides, minerals, and proteins. However, limited information is available on the potential health benefits of this by-product. In this context, three industrial products containing herring milt hydrolysate (HMH) were tested in both animal and cellular models to measure their effects on obesity-related metabolic disorders. Male C57Bl/6J mice were fed either a control chow diet or a high-fat high-sucrose (HFHS) diet for 8 weeks and received either the vehicle (water) or one of the three HMH products (HMH1, HMH2, and HMH3) at a dose of 208.8 mg/kg (representing 1 g/day for a human) by daily oral gavage. The impact of HMH treatments on insulin and glucose tolerance, lipid homeostasis, liver gene expression, and the gut microbiota profile was studied. In parallel, the effects of HMH on glucose uptake and inflammation were studied in L6 myocytes and J774 macrophages, respectively. In vivo, daily treatment with HMH2 and HMH3 improved early time point glycemia during the oral glucose tolerance test (OGTT) induced by the HFHS diet, without changes in weight gain and insulin secretion. Interestingly, we also observed that HMH2 consumption partially prevented a lower abundance of Lactobacillus species in the gut microbiota of HFHS diet-fed animals. In addition to this, modulations of gene expression in the liver, such as the upregulation of sucrose nonfermenting AMPK-related kinase (SNARK), were reported for the first time in mice treated with HMH products. While HMH2 and HMH3 inhibited inducible nitric oxide synthase (iNOS) induction in J774 macrophages, glucose uptake was not modified in L6 muscle cells. These results indicate that milt herring hydrolysates reduce some metabolic and inflammatory alterations in cellular and animal models, suggesting a possible novel marine ingredient to help fight against obesity-related immunometabolic disorders.
Collapse
Affiliation(s)
- Rachel Durand
- Department of food Sciences and Laboratory of Food Processing and Electromembrane Process (LTAPEM), Université Laval, Québec, QC G1V 0A6, Canada;
- Institute of Nutrition and Functional Foods (INAF), Université Laval, Québec, QC G1V 0A6, Canada; (A.O.); (V.P.H.); (F.G.); (T.V.V.); (B.M.); (G.P.); (M.-C.V.); (A.M.)
| | - Adia Ouellette
- Institute of Nutrition and Functional Foods (INAF), Université Laval, Québec, QC G1V 0A6, Canada; (A.O.); (V.P.H.); (F.G.); (T.V.V.); (B.M.); (G.P.); (M.-C.V.); (A.M.)
- Québec Heart and Lung Institute, Department of medicine, Université Laval, QC G1V 4G5 Québec, Canada
| | - Vanessa P. Houde
- Institute of Nutrition and Functional Foods (INAF), Université Laval, Québec, QC G1V 0A6, Canada; (A.O.); (V.P.H.); (F.G.); (T.V.V.); (B.M.); (G.P.); (M.-C.V.); (A.M.)
- Québec Heart and Lung Institute, Department of medicine, Université Laval, QC G1V 4G5 Québec, Canada
| | - Frédéric Guénard
- Institute of Nutrition and Functional Foods (INAF), Université Laval, Québec, QC G1V 0A6, Canada; (A.O.); (V.P.H.); (F.G.); (T.V.V.); (B.M.); (G.P.); (M.-C.V.); (A.M.)
- School of Nutrition, Université Laval, Québec, QC G1V 0A6, Canada
| | - Thibaut V. Varin
- Institute of Nutrition and Functional Foods (INAF), Université Laval, Québec, QC G1V 0A6, Canada; (A.O.); (V.P.H.); (F.G.); (T.V.V.); (B.M.); (G.P.); (M.-C.V.); (A.M.)
- Québec Heart and Lung Institute, Department of medicine, Université Laval, QC G1V 4G5 Québec, Canada
| | - Bruno Marcotte
- Institute of Nutrition and Functional Foods (INAF), Université Laval, Québec, QC G1V 0A6, Canada; (A.O.); (V.P.H.); (F.G.); (T.V.V.); (B.M.); (G.P.); (M.-C.V.); (A.M.)
- Québec Heart and Lung Institute, Department of medicine, Université Laval, QC G1V 4G5 Québec, Canada
| | - Geneviève Pilon
- Institute of Nutrition and Functional Foods (INAF), Université Laval, Québec, QC G1V 0A6, Canada; (A.O.); (V.P.H.); (F.G.); (T.V.V.); (B.M.); (G.P.); (M.-C.V.); (A.M.)
- Québec Heart and Lung Institute, Department of medicine, Université Laval, QC G1V 4G5 Québec, Canada
| | | | - Marie-Claude Vohl
- Institute of Nutrition and Functional Foods (INAF), Université Laval, Québec, QC G1V 0A6, Canada; (A.O.); (V.P.H.); (F.G.); (T.V.V.); (B.M.); (G.P.); (M.-C.V.); (A.M.)
- School of Nutrition, Université Laval, Québec, QC G1V 0A6, Canada
| | - André Marette
- Institute of Nutrition and Functional Foods (INAF), Université Laval, Québec, QC G1V 0A6, Canada; (A.O.); (V.P.H.); (F.G.); (T.V.V.); (B.M.); (G.P.); (M.-C.V.); (A.M.)
- Québec Heart and Lung Institute, Department of medicine, Université Laval, QC G1V 4G5 Québec, Canada
| | - Laurent Bazinet
- Department of food Sciences and Laboratory of Food Processing and Electromembrane Process (LTAPEM), Université Laval, Québec, QC G1V 0A6, Canada;
- Institute of Nutrition and Functional Foods (INAF), Université Laval, Québec, QC G1V 0A6, Canada; (A.O.); (V.P.H.); (F.G.); (T.V.V.); (B.M.); (G.P.); (M.-C.V.); (A.M.)
- Correspondence: ; Tel.: +418-656-2131-7445; Fax: +418-656-3353
| |
Collapse
|
48
|
Posa DK, Baba SP. Intracellular pH Regulation of Skeletal Muscle in the Milieu of Insulin Signaling. Nutrients 2020; 12:nu12102910. [PMID: 32977552 PMCID: PMC7598285 DOI: 10.3390/nu12102910] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/24/2020] [Accepted: 08/31/2020] [Indexed: 12/18/2022] Open
Abstract
Type 2 diabetes (T2D), along with obesity, is one of the leading health problems in the world which causes other systemic diseases, such as cardiovascular diseases and kidney failure. Impairments in glycemic control and insulin resistance plays a pivotal role in the development of diabetes and its complications. Since skeletal muscle constitutes a significant tissue mass of the body, insulin resistance within the muscle is considered to initiate the onset of diet-induced metabolic syndrome. Insulin resistance is associated with impaired glucose uptake, resulting from defective post-receptor insulin responses, decreased glucose transport, impaired glucose phosphorylation, oxidation and glycogen synthesis in the muscle. Although defects in the insulin signaling pathway have been widely studied, the effects of cellular mechanisms activated during metabolic syndrome that cross-talk with insulin responses are not fully elucidated. Numerous reports suggest that pathways such as inflammation, lipid peroxidation products, acidosis and autophagy could cross-talk with insulin-signaling pathway and contribute to diminished insulin responses. Here, we review and discuss the literature about the defects in glycolytic pathway, shift in glucose utilization toward anaerobic glycolysis and change in intracellular pH [pH]i within the skeletal muscle and their contribution towards insulin resistance. We will discuss whether the derangements in pathways, which maintain [pH]i within the skeletal muscle, such as transporters (monocarboxylate transporters 1 and 4) and depletion of intracellular buffers, such as histidyl dipeptides, could lead to decrease in [pH]i and the onset of insulin resistance. Further we will discuss, whether the changes in [pH]i within the skeletal muscle of patients with T2D, could enhance the formation of protein aggregates and activate autophagy. Understanding the mechanisms by which changes in the glycolytic pathway and [pH]i within the muscle, contribute to insulin resistance might help explain the onset of obesity-linked metabolic syndrome. Finally, we will conclude whether correcting the pathways which maintain [pH]i within the skeletal muscle could, in turn, be effective to maintain or restore insulin responses during metabolic syndrome.
Collapse
Affiliation(s)
- Dheeraj Kumar Posa
- Diabetes and Obesity Center, University of Louisville, Louisville, KY 40202, USA
- Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY 40202, USA
| | - Shahid P Baba
- Diabetes and Obesity Center, University of Louisville, Louisville, KY 40202, USA
- Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
49
|
Mancusi C, Izzo R, di Gioia G, Losi MA, Barbato E, Morisco C. Insulin Resistance the Hinge Between Hypertension and Type 2 Diabetes. High Blood Press Cardiovasc Prev 2020; 27:515-526. [PMID: 32964344 PMCID: PMC7661395 DOI: 10.1007/s40292-020-00408-8] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 09/05/2020] [Indexed: 12/19/2022] Open
Abstract
Epidemiological studies have documented a high incidence of diabetes in hypertensive patients.Insulin resistance is defined as a less than expected biologic response to a given concentration of the hormone and plays a pivotal role in the pathogenesis of diabetes. However, over the last decades, it became evident that insulin resistance is not merely a metabolic abnormality, but is a complex and multifaceted syndrome that can also affect blood pressure homeostasis. The dysregulation of neuro-humoral and neuro-immune systems is involved in the pathophysiology of both insulin resistance and hypertension. These mechanisms induce a chronic low grade of inflammation that interferes with insulin signalling transduction. Molecular abnormalities associated with insulin resistance include the defects of insulin receptor structure, number, binding affinity, and/or signalling capacity. For instance, hyperglycaemia impairs insulin signalling through the generation of reactive oxygen species, which abrogate insulin-induced tyrosine autophosphorylation of the insulin receptor. Additional mechanisms have been described as responsible for the inhibition of insulin signalling, including proteasome-mediated degradation of insulin receptor substrate 1/2, phosphatase-mediated dephosphorylation and kinase-mediated serine/threonine phosphorylation of both insulin receptor and insulin receptor substrates. Insulin resistance plays a key role also in the pathogenesis and progression of hypertension-induced target organ damage, like left ventricular hypertrophy, atherosclerosis and chronic kidney disease. Altogether these abnormalities significantly contribute to the increase the risk of developing type 2 diabetes.
Collapse
Affiliation(s)
- Costantino Mancusi
- Department of Advanced Biomedical Sciences, Federico II University of Naples, Via S. Pansini n. 5, 80131, Naples, Italy
| | - Raffaele Izzo
- Department of Advanced Biomedical Sciences, Federico II University of Naples, Via S. Pansini n. 5, 80131, Naples, Italy
| | - Giuseppe di Gioia
- Department of Advanced Biomedical Sciences, Federico II University of Naples, Via S. Pansini n. 5, 80131, Naples, Italy
| | - Maria Angela Losi
- Department of Advanced Biomedical Sciences, Federico II University of Naples, Via S. Pansini n. 5, 80131, Naples, Italy
| | - Emanuele Barbato
- Department of Advanced Biomedical Sciences, Federico II University of Naples, Via S. Pansini n. 5, 80131, Naples, Italy
| | - Carmine Morisco
- Department of Advanced Biomedical Sciences, Federico II University of Naples, Via S. Pansini n. 5, 80131, Naples, Italy.
| |
Collapse
|
50
|
Regulation of lymphatic function and injury by nitrosative stress in obese mice. Mol Metab 2020; 42:101081. [PMID: 32941994 PMCID: PMC7536739 DOI: 10.1016/j.molmet.2020.101081] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 09/02/2020] [Accepted: 09/10/2020] [Indexed: 12/11/2022] Open
Abstract
Objective Obesity results in lymphatic dysfunction, but the cellular mechanisms that mediate this effect remain largely unknown. Previous studies in obese mice have shown that inducible nitric oxide synthase-expressing (iNOS+) inflammatory cells accumulate around lymphatic vessels. In the current study, we therefore tested the hypothesis that increased expression of iNOS results in nitrosative stress and injury to the lymphatic endothelial cells (LECs). In addition, we tested the hypothesis that lymphatic injury, independent of obesity, can modulate glucose and lipid metabolism. Methods We compared the metabolic changes and lymphatic function of wild-type and iNOS knockout mice fed a normal chow or high-fat diet for 16 weeks. To corroborate our in vivo findings, we analyzed the effects of reactive nitrogen species on isolated LECs. Finally, using a genetically engineered mouse model that allows partial ablation of the lymphatic system, we studied the effects of acute lymphatic injury on glucose and lipid metabolism in lean mice. Results The mesenteric lymphatic vessels of obese wild-type animals were dilated, leaky, and surrounded by iNOS+ inflammatory cells with resulting increased accumulation of reactive nitrogen species when compared with lean wild-type or obese iNOS knockout animals. These changes in obese wild-type mice were associated with systemic glucose and lipid abnormalities, as well as decreased mesenteric LEC expression of lymphatic-specific genes, including vascular endothelial growth factor receptor 3 (VEGFR-3) and antioxidant genes as compared with lean wild-type or obese iNOS knockout animals. In vitro experiments demonstrated that isolated LECs were more sensitive to reactive nitrogen species than blood endothelial cells, and that this sensitivity was ameliorated by antioxidant therapies. Finally, using mice in which the lymphatics were specifically ablated using diphtheria toxin, we found that the interaction between metabolic abnormalities caused by obesity and lymphatic dysfunction is bidirectional. Targeted partial ablation of mesenteric lymphatic channels of lean mice resulted in increased accumulation of iNOS+ inflammatory cells and increased reactive nitrogen species. Lymphatic ablation also caused marked abnormalities in insulin sensitivity, serum glucose and insulin concentrations, expression of insulin-sensitive genes, lipid metabolism, and significantly increased systemic and mesenteric white adipose tissue (M-WAT) inflammatory responses. Conclusions Our studies suggest that increased iNOS production in obese animals plays a key role in regulating lymphatic injury by increasing nitrosative stress. In addition, our studies suggest that obesity-induced lymphatic injury may amplify metabolic abnormalities by increasing systemic and local inflammatory responses and regulating insulin sensitivity. These findings suggest that manipulation of the lymphatic system may represent a novel means of treating metabolic abnormalities associated with obesity. Increased iNOS+ cells around mesenteric lymphatics of high fat diet-induced obese mice. iNOS knockout mice are protected from obesity-induced lymphatic dysfunction. Lymphatic endothelial cells are highly sensitive to nitrosative stress. Nitrosative stress causes lymphatic gene regulation. Lymphatic injury alone enhances iNOS+ cells and causes insulin resistance and dyslipidemia.
Collapse
|