1
|
Ding S, Xu JL, Tong JY, Cheng YY, Shi LF, Wei W, Zhang LM, Zhang JJ, Meng BY, Peng XY, Xiang L, Li SG, Yue L, Wang ZJ, Xiang GD. Brown adipose tissue alleviates podocyte apoptosis through NRG4 in a male mouse model of diabetic kidney disease. Diabetologia 2025; 68:1057-1075. [PMID: 40014139 DOI: 10.1007/s00125-025-06385-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 12/20/2024] [Indexed: 02/28/2025]
Abstract
AIMS/HYPOTHESIS Brown adipose tissue (BAT) consumes excess energy through heat production by uncoupling protein 1 (UCP1) to regulate the metabolic profile, but the UCP1-independent mechanisms of BAT, such as in endocrine function, are largely unknown. Our previous study showed that BAT-derived neuregulin 4 (NRG4) displays anti-atherosclerotic properties. Thus, we hypothesised that BAT could regulate diabetic nephropathy, a diabetic microvascular complication, via NRG4. METHODS To investigate the influence of NRG4 from BAT on podocyte apoptosis, both loss- and gain-of-function approaches were used in in vivo experiments. Diabetic nephropathy models were created using BAT-specific Nrg4-knockout (BKO) mice, global Nrg4-knockout (KO) mice and wild-type (WT) mice. In in vitro studies, podocytes (MPC5) were exposed to glucose and recombinant NRG4 (rNrg4). Additionally, brown adipocytes were co-cultured with MPC5 podocytes using a transwell system. The expression levels of proteins associated with podocyte apoptosis and signalling pathways were measured. RESULTS BAT-specific NRG4 deficiency aggravated podocyte apoptosis (increased by 47.46%) and increased the urinary albumin/creatinine ratio (increased by 41.71%), decreased nephrin expression and increased desmin expression. As expected, these changes were reversed by NRG4 replenishment using adeno-associated virus-NRG4 interscapular BAT injection and BAT transplantation assays in KO mice. Additionally, co-culture experiments demonstrated that brown adipocytes from WT mice could alleviate high-glucose-induced podocyte apoptosis. In in vitro experiments, recombinant NRG4 inhibited high-glucose-induced podocyte apoptosis. Mechanistically, the Akt-glycogen synthase kinase 3 β (GSK-3β) pathway is crucial for the protection that BAT-derived NRG4 provides to podocytes in diabetic nephropathy. CONCLUSIONS/INTERPRETATION Our data show that BAT had a protective effect on podocyte apoptosis in diabetic nephropathy through BAT-derived NRG4, and the Akt-GSK‑3β signalling pathway may mediate the inhibition of BAT-derived NRG4 on podocyte apoptosis in diabetic nephropathy.
Collapse
Affiliation(s)
- Sheng Ding
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Chronic Disease Management Center, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jin-Ling Xu
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, China
- Department of Endocrinology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia-Yue Tong
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yang-Yang Cheng
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Ling-Feng Shi
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Wei Wei
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of Endocrinology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li-Ming Zhang
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jia-Jia Zhang
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, China
| | - Bi-Ying Meng
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, China
| | - Xiang-Yan Peng
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Lin Xiang
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, China
| | - Shu-Guang Li
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, China
| | - Ling Yue
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, China
| | - Zhong-Jing Wang
- Department of Endocrinology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Guang-da Xiang
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, China.
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China.
| |
Collapse
|
2
|
Wu J, Li W, Tang Y, Wu C, Li W. miR-205-3p inhibits porphyromonas gingivalis lipopolysaccharide-induced human umbilical vein endothelial cells inflammation and apoptosis by targeting PRMT5. Arch Oral Biol 2025; 175:106276. [PMID: 40319839 DOI: 10.1016/j.archoralbio.2025.106276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/12/2025] [Accepted: 04/27/2025] [Indexed: 05/07/2025]
Abstract
OBJECTIVE This study aimed to investigate the regulatory mechanism of miR-205-3p in Porphyromonas gingivalis lipopolysaccharide (P.g-LPS)-induced atherosclerosis. DESIGN In an in vitro setting, human umbilical vein endothelial cells (HUVECs) were exposed to P.g-LPS to simulate the vascular endothelial damage induced by periodontitis. Subsequently, ELISA and flow cytometry were employed to assess the inflammatory response and apoptotic status of these cells.To quantify the expression levels of protein arginine methyltransferase 5 (PRMT5), BCL2-associated X protein (Bax), B-cell lymphoma 2 (Bcl-2), P65 and miR-205-3p within the HUVECs, Western Blot and qPCR were respectively utilized. Moreover, small interfering RNA (siRNA) targeting PRMT5 and miR-205-3p were applied to monitor the changes in PRMT5 expression. Bioinformatics analysis was carried out to predict the potential binding sites between miR-205-3p and PRMT5. Finally, the interaction between miR-205-3p and PRMT5 was validated through the dual-luciferase reporter assay. RESULTS The results indicate that P.g-LPS intervention exacerbates damage to HUVECs and increases the expression of PRMT5. Silencing PRMT5 reduces cell inflammation and apoptosis. After stimulation with P.g-LPS, the level of miR-205-3p decreases, and its overexpression alleviates inflammation and apoptosis in the cells. Bioinformatics analysis and dual luciferase reporter assays confirm that PRMT5 is a target of miR-205-3p, and the overexpression of PRMT5 can reverse the protective effects of miR-205-3p. CONCLUSION miR-205-3p can mitigate vascular endothelial injury by decreasing PRMT5 expression, providing new insights for potential treatments.
Collapse
Affiliation(s)
- Jinsheng Wu
- Stomatology College of Jiamusi University, Jiamusi 154000, China
| | - Weiyi Li
- Stomatology College of Jiamusi University, Jiamusi 154000, China
| | - Ying Tang
- Stomatology College of Jiamusi University, Jiamusi 154000, China
| | - Chang Wu
- Stomatology College of Jiamusi University, Jiamusi 154000, China
| | - Weishan Li
- Stomatology College of Jiamusi University, Jiamusi 154000, China; Department of Periodontal and Mucosal Diseases, Jiamusi University Stomatology Hospital, Jiamusi 154000, China.
| |
Collapse
|
3
|
Gai X, Liu F, Chen Y, Zhang B, Zhang Y, Wu Y, Yang S, Chen L, Deng W, Wang Y, Wang S, Yu C, Du J, Zhang Z, Wang J, Zhang H. GOLM1 Promotes Atherogenesis by Activating Macrophage EGFR-ERK Signaling Cascade. Circ Res 2025; 136:848-861. [PMID: 40026146 DOI: 10.1161/circresaha.124.325880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 02/12/2025] [Accepted: 02/19/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND Atherosclerosis is a chronic inflammatory disease. GOLM1 (Golgi membrane protein 1) is an inflammation-responsive protein and a mediator in some inflammation-associated pathological processes. Because we found a positive correlation between GOLM1 expression and atherosclerosis progression by checking the gene expression data set of human atherosclerotic lesions, we explored the potential significance of GOLM1 in atherosclerosis in this study. METHODS GOLM1 levels in serums and lesions of patients with atherosclerosis and mice with atherosclerosis were examined by immunostaining and ELISA. Gain-of-function and loss-of-function approaches were used to study the impacts of GOLM1 in inflammation and atherogenesis of Apoe-/- mice on a Western diet. The effects of GOLM1 on macrophage behaviors were determined by OxLDL (oxidized low-density lipoprotein) uptake assay, single-cell sequencing analysis, global phosphoproteomics analysis, and molecular biological techniques. The therapeutic potential of GOLM1 neutralization for atherosclerosis was evaluated in Apoe-/- mice. RESULTS GOLM1 was elevated in serums and lesions of patients with atherosclerosis and mice with atherosclerosis. Global deletion of GOLM1 ameliorated mouse inflammation and atherosclerosis, while knock-in of GOLM1 exacerbated these pathological manifestations. Furthermore, hepatic GOLM1 deletion reduced circulating GOLM1 and attenuated atherogenesis. Mechanistically, the expression and secretion of GOLM1 were induced in multiple mouse tissues by atherogenic stimulus, leading to the elevation of extracellular GOLM1. Extracellular GOLM1 then stimulated ERK (extracellular signal-regulated kinase) signaling cascade by binding to its putative receptor EGFR (epidermal growth factor receptor) to promote macrophage uptake of LDL (low-density lipoprotein) and enhance the corresponding macrophage immune response. Moreover, neutralizing GOLM1 by an antibody suppressed mouse inflammation and atherogenesis. CONCLUSIONS GOLM1 is an atherogenic mediator and a promising therapeutic target for the intervention of atherosclerotic diseases.
Collapse
Affiliation(s)
- Xiaochen Gai
- Department of Physiology, State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem (X.G., F.L., B.Z., Y. Wu, S.Y., W.D., H.Z.), Hebei University, Baoding, Hebei, China
| | - Fangming Liu
- Department of Physiology, State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem (X.G., F.L., B.Z., Y. Wu, S.Y., W.D., H.Z.), Hebei University, Baoding, Hebei, China
| | - Yixin Chen
- Department of Cardiovascular Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (Y.C., S.W., C.Y.)
| | - Baohui Zhang
- Department of Physiology, State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem (X.G., F.L., B.Z., Y. Wu, S.Y., W.D., H.Z.), Hebei University, Baoding, Hebei, China
- Department of Physiology, School of Life Science, China Medical University, Shenyang, Liaoning, China (B.Z.)
| | - Yinliang Zhang
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Cellular Homeostasis and Disease, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (Y.Z.)
| | - Yuting Wu
- Department of Physiology, State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem (X.G., F.L., B.Z., Y. Wu, S.Y., W.D., H.Z.), Hebei University, Baoding, Hebei, China
| | - Shuhui Yang
- Department of Physiology, State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem (X.G., F.L., B.Z., Y. Wu, S.Y., W.D., H.Z.), Hebei University, Baoding, Hebei, China
| | | | - Weiwei Deng
- Department of Physiology, State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem (X.G., F.L., B.Z., Y. Wu, S.Y., W.D., H.Z.), Hebei University, Baoding, Hebei, China
| | - Yuan Wang
- Beijing Anzhen Hospital, Beijing, China (Y. Wang, J.D.)
| | - Shuiyun Wang
- Department of Cardiovascular Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (Y.C., S.W., C.Y.)
| | - Cuntao Yu
- Department of Cardiovascular Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (Y.C., S.W., C.Y.)
| | - Jie Du
- Beijing Anzhen Hospital, Beijing, China (Y. Wang, J.D.)
| | - Zhengyi Zhang
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA (Z.Z.)
| | - Jing Wang
- Department of Pathophysiology (J.W.), Hebei University, Baoding, Hebei, China
| | - Hongbing Zhang
- Department of Physiology, State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem (X.G., F.L., B.Z., Y. Wu, S.Y., W.D., H.Z.), Hebei University, Baoding, Hebei, China
- Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China, College of Life Sciences, Hebei University, Baoding, Hebei, China (H.Z.)
| |
Collapse
|
4
|
Ren BX, Zeng ZL, Deng L, Hu JM, Chen MZ, Jiang HW, Zang CZ, Fang ST, Weiss SJ, Liu J, Fu R, Wu ZQ. Genetic and pharmacological targeting of Snail inhibits atherosclerosis by relieving intraplaque endothelium dysfunction and associated inflammation. Acta Pharmacol Sin 2025:10.1038/s41401-025-01519-5. [PMID: 40133628 DOI: 10.1038/s41401-025-01519-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 02/21/2025] [Indexed: 03/27/2025]
Abstract
The intraplaque endothelium dysfunction and associated inflammation contribute to the progression of atherosclerosis. We previously show that zinc-finger transcription factor Snail is predominantly expressed in embryonic vascular endothelial cells (ECs), and deletion of Snail in ECs induces severe defects in vascular development and thus causes embryonic lethality. Snail is essentially absent at postnatal stage, and inducible deletion of Snail in ECs has no impact on physiological angiogenesis in postnatally developing or adult mice. In this study we investigated whether Snail was reactivated in vascular ECs during pathologically angiogenic process (e.g. the formation of atherosclerotic plaque) or could play a functional role in atherosclerosis progression. We showed that the expression levels of Snail were significantly elevated in ECs of human and mouse atherosclerotic plaques, and associated with the disease severity. In the accelerated and canonical mouse models of atherosclerosis, tamoxifen-inducible, EC-specific Snail deletion significantly reduced intraplaque endothelial dysfunction, inflammation and lipid uptake accompanied by enhanced plaque stability. By conducting scRNA-sequencing in ECs of ApoE-/-SnailiΔEC versus ApoE-/-Snailfl/fl arterial vessels, we demonstrated that Snail deletion significantly decreased histone acetylation on Ccl5 and Cxcl10 promoters, thereby decreased CCL5/CXCL10-driven vascular damage and inflammation. Administration with recombinant CXCL10 protein (2 μg/kg, i.v., once per week for three weeks) efficiently restored atherosclerosis in EC-specific Snail-deleted mice. Finally, we developed an orally bioavailable small-molecule Snail inhibitor LFW273 that displayed potent anti-atherosclerotic effects in mice. These results reveal Snail as a promising therapeutic target in atherosclerotic disease.
Collapse
Affiliation(s)
- Bo-Xue Ren
- State Key Laboratory of Natural Medicines, Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Zhao-Lan Zeng
- State Key Laboratory of Natural Medicines, Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
- Department of Organic Chemistry, School of Science, China Pharmaceutical University, Nanjing, 211198, China
| | - Li Deng
- State Key Laboratory of Natural Medicines, Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Jia-Meng Hu
- State Key Laboratory of Natural Medicines, Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Ming-Zhen Chen
- State Key Laboratory of Natural Medicines, Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Hao-Wei Jiang
- State Key Laboratory of Natural Medicines, Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Chen-Zi Zang
- State Key Laboratory of Natural Medicines, Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Shen-Tong Fang
- State Key Laboratory of Natural Medicines, Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Stephen J Weiss
- The Life Sciences Institute, Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jie Liu
- Department of Organic Chemistry, School of Science, China Pharmaceutical University, Nanjing, 211198, China.
| | - Rong Fu
- State Key Laboratory of Natural Medicines, Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| | - Zhao-Qiu Wu
- State Key Laboratory of Natural Medicines, Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
5
|
McLeod K, Datta V, Fuller S. Adipokines as Cardioprotective Factors: BAT Steps Up to the Plate. Biomedicines 2025; 13:710. [PMID: 40149686 PMCID: PMC11940801 DOI: 10.3390/biomedicines13030710] [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: 01/10/2025] [Revised: 03/07/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
Cardiovascular disease is the leading cause of death throughout most of the industrialized world. Metabolic syndrome (MetS) and its associated pathologies are underlying factors in the etiology of cardiovascular disease, as well as a plethora of other maladies which cause excess morbidity and mortality. Adipose tissue (AT) has come to be regarded as a bona fide endocrine organ which secretes specific molecular entities constituting part of a complex web of inter-organ crosstalk that functions as a key determinant of whole-body metabolic phenotype. Brown adipose tissue (BAT) has classically been regarded as a thermogenic tissue exerting its metabolic effects primarily through its capacity to oxidize substrates decoupled from ATP resynthesis, thereby resulting in increased energy expenditure (EE) and heat production. However, in recent years, BAT has begun to receive attention as a secretory organ in its own right. The molecules secreted specifically by BAT have been termed "batokines", and currently available evidence supports the notion that batokines exert favorable metabolic effects on multiple organ systems. While maintenance of healthy body composition by conferring resistance to excessive adiposity is a rather obvious mechanism by which BAT operates via increased EE, effects on critical organs such as the heart remain unclear. This narrative review focuses on four types of batokines (FGF21, neuregulin 4, 12,13-diHOME, and BAT-derived microRNAs) for which evidence of modulation of cardiovascular function exists in the context of pathological states such as hypertension, atherosclerosis, and ischemia/reperfusion injury. Given the overwhelming burden of cardiometabolic disease, further study of the functions of BAT and its secretome is warranted and will intensify in the future.
Collapse
Affiliation(s)
- Keely McLeod
- School of Kinesiology, University of Louisiana at Lafayette, Lafayette, LA 70506, USA; (K.M.); (V.D.)
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA
| | - Victoria Datta
- School of Kinesiology, University of Louisiana at Lafayette, Lafayette, LA 70506, USA; (K.M.); (V.D.)
| | - Scott Fuller
- School of Kinesiology, University of Louisiana at Lafayette, Lafayette, LA 70506, USA; (K.M.); (V.D.)
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA
| |
Collapse
|
6
|
Bonfante ILP, Segantim HDS, Mendonça KNS, de Oliveira MAB, Monfort-Pires M, Duft RG, da Silva Mateus KC, Chacon-Mikahil MPT, Ramos CD, Velloso LA, Cavaglieri CR. Better cardiometabolic/inflammatory profile is associated with differences in the supraclavicular adipose tissue activity of individuals with T2DM. Endocrine 2025; 87:1011-1021. [PMID: 39627400 DOI: 10.1007/s12020-024-04122-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 11/25/2024] [Indexed: 02/22/2025]
Abstract
PURPOSE Brown adipose tissue (BAT), located in the supraclavicular region, has been associated with a better cardiometabolic profile and reduced risk of developing non-communicable chronic diseases (NCD), in addition to being associated with a healthier phenotype in obesity. However, it is unknown whether greater supraclavicular adipose tissue activity could be associated with a healthier metabolic profile in people already diagnosed with type 2 diabetes (T2DM). Thus, the present work evaluated if supraclavicular adipose tissue activity is associated with metabolic and molecular markers in individuals with T2DM. METHODS Based on a cluster study, individuals with T2DM were divided into groups according to high or low-standard uptake value (SUV) evaluated in the supraclavicular adipose tissue area by [18F]-fluorodeoxyglucose and positron emission tomography-computed tomography (18F-FDG-PET/CT) after mild cold exposure). Functional, biochemical, inflammatory, and molecular markers were measured. RESULTS When we evaluated the whole sample, women showed higher SUV, which favored a difference between groups in sex-related markers. On the other hand, volunteers in the high-SUV group showed lower BMI, monocytes count, triglycerides/glucose index (TYG-index) and z score of metabolic syndrome (MS) values, as well as lower triglycerides, and VLDL concentrations. Moreover, they also had enhanced expression of thermogenic genes in subcutaneous fat. When analyzing only women, the differences in markers associated with sex disappear, and a lower count of leukocytes, platelets, along with lower TYG-index, z score of MS values, and triglycerides, VLDL, LDL, and TNFα concentrations were observed in women with the high SUV. In addition, higher expression of thermogenic genes and BECN1 were detected. CONCLUSION Higher supraclavicular adipose tissue SUV in individuals with T2DM is associated with a better cardiometabolic/inflammatory profile and expression of thermogenic genes. CLINICAL TRIAL REGISTRATION UTN: U1111-1202-1476 - 08/20/2020.
Collapse
Affiliation(s)
- Ivan Luiz Padilha Bonfante
- Laboratory of Exercise Physiology, School of Physical Education, University of Campinas, Campinas, SP, Brazil.
- Postdoctoral Researcher Program (PPPD), University of Campinas, Campinas, Brazil.
| | - Higor da Silva Segantim
- Higher Interdisciplinary Training Program (PROFIS), University of Campinas, Campinas, SP, Brazil
| | | | | | - Milena Monfort-Pires
- Laboratory of Cell Signaling, Department of Internal Medicine, University of Campinas, Campinas, SP, Brazil
- Obesity and Comorbidities Research Center, University of Campinas, Campinas, SP, Brazil
- Turku Pet Centre, University of Turku, Turku, Finland
| | - Renata Garbellini Duft
- Laboratory of Exercise Physiology, School of Physical Education, University of Campinas, Campinas, SP, Brazil
- The Rowett Institute of Nutrition and Health, University of Aberdeen, Ashgrove Rd W, Aberdeen, Scotland, UK
| | | | | | - Celso Darío Ramos
- Department of Radiology, University of Campinas, Campinas, SP, Brazil
| | - Licio Augusto Velloso
- Laboratory of Cell Signaling, Department of Internal Medicine, University of Campinas, Campinas, SP, Brazil
- Obesity and Comorbidities Research Center, University of Campinas, Campinas, SP, Brazil
| | - Cláudia Regina Cavaglieri
- Laboratory of Exercise Physiology, School of Physical Education, University of Campinas, Campinas, SP, Brazil
| |
Collapse
|
7
|
Chen M, Li Y, Zhu JY, Mu WJ, Luo HY, Yan LJ, Li S, Li RY, Yin MT, Li X, Chen HM, Guo L. Exercise-induced adipokine Nrg4 alleviates MASLD by disrupting hepatic cGAS-STING signaling. Cell Rep 2025; 44:115251. [PMID: 39891907 DOI: 10.1016/j.celrep.2025.115251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/15/2024] [Accepted: 01/10/2025] [Indexed: 02/03/2025] Open
Abstract
Exercise is an effective non-pharmacological strategy for ameliorating metabolic dysfunction-associated steatotic liver disease (MASLD). Neuregulin-4 (Nrg4) is an adipokine with a potential role in metabolic homeostasis. Previous findings have shown that Nrg4 is upregulated by exercise and that Nrg4 reduces hepatic steatosis, but the underlying mechanism is not fully understood. Here, we show that adipose Nrg4 is transactivated by Pparγ in response to exercise in mice. Adeno-associated virus (AAV)-mediated knockdown of adipose Nrg4 as well as hepatocyte-specific knockout of Erbb4 (Nrg4 receptor) impair exercise-mediated alleviation of MASLD in mice. Conversely, AAV-mediated overexpression of adipose Nrg4 mitigates MASLD in mice in synergy with exercise. Mechanistically, Nrg4/Erbb4/AKT signaling promotes cyclic guanosine monophosphate-AMP synthase (cGAS) phosphorylation to blunt its enzyme activity, thereby inhibiting cGAS-STING pathway-mediated inflammation and steatosis in hepatocytes. Thus, Nrg4 functions as an exercise-induced adipokine that participates in adipose-liver tissue communication to counteract MASLD.
Collapse
Affiliation(s)
- Min Chen
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai 200438, China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China; Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai 200438, China
| | - Yang Li
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai 200438, China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China; Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai 200438, China
| | - Jie-Ying Zhu
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai 200438, China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China; Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai 200438, China
| | - Wang-Jing Mu
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai 200438, China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China; Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai 200438, China
| | - Hong-Yang Luo
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai 200438, China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China; Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai 200438, China
| | - Lin-Jing Yan
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai 200438, China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China; Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai 200438, China
| | - Shan Li
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai 200438, China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China; Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai 200438, China
| | - Ruo-Ying Li
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai 200438, China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China; Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai 200438, China
| | - Meng-Ting Yin
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai 200438, China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China; Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai 200438, China
| | - Xin Li
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai 200438, China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China; Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai 200438, China
| | - Hu-Min Chen
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai 200438, China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China; Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai 200438, China
| | - Liang Guo
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai 200438, China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China; Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai 200438, China.
| |
Collapse
|
8
|
Zhou Q, Wang Y, Si G, Chen X, Mu D, Zhang B. Application of Nanomaterials in Early Imaging and Advanced Treatment of Atherosclerosis. CHEMICAL & BIOMEDICAL IMAGING 2025; 3:51-76. [PMID: 40018650 PMCID: PMC11863161 DOI: 10.1021/cbmi.4c00064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 11/26/2024] [Accepted: 11/26/2024] [Indexed: 03/01/2025]
Abstract
Atherosclerosis (AS) is a serious disease that poses a significant threat to the global population. In this review, we analyze the development of AS from multiple perspectives, aiming to elucidate its molecular mechanisms. We also focus on imaging techniques and therapeutic approaches, highlighting the crucial role of nanomaterials in both imaging and therapy for AS. By leveraging their compatibility and targeting capabilities, nanomaterials can be integrated with traditional medical imaging and therapeutic agents to achieve targeted drug delivery, controlled release, and precise localization and imaging of atherosclerotic plaques.
Collapse
Affiliation(s)
- Qianru Zhou
- Department
of Radiology, Nanjing Drum Tower Hospital Clinical College of Traditional
Chinese and Western Medicine, Nanjing University
of Chinese Medicine, Nanjing 210008, China
| | - Yujie Wang
- Department
of Radiology, Nanjing Drum Tower Hospital
Clinical College of Jiangsu University, Nanjing 210008, China
| | - Guangxiang Si
- Jiangsu
Key Laboratory for Biomaterials and Devices, School of Biological
Science and Medical Engineering, Southeast
University, Nanjing 210000, China
| | - Xingbiao Chen
- Clinical
Science, Philips Healthcare, Shanghai 200233, China
| | - Dan Mu
- Department
of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of
Medical School, Nanjing University, No. 321 Zhongshan Road, Nanjing 210008, China
| | - Bing Zhang
- Department
of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of
Medical School, Nanjing University, No. 321 Zhongshan Road, Nanjing 210008, China
| |
Collapse
|
9
|
Liu H, Wang S, Wang J, Guo X, Song Y, Fu K, Gao Z, Liu D, He W, Yang LL. Energy metabolism in health and diseases. Signal Transduct Target Ther 2025; 10:69. [PMID: 39966374 PMCID: PMC11836267 DOI: 10.1038/s41392-025-02141-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 11/08/2024] [Accepted: 12/25/2024] [Indexed: 02/20/2025] Open
Abstract
Energy metabolism is indispensable for sustaining physiological functions in living organisms and assumes a pivotal role across physiological and pathological conditions. This review provides an extensive overview of advancements in energy metabolism research, elucidating critical pathways such as glycolysis, oxidative phosphorylation, fatty acid metabolism, and amino acid metabolism, along with their intricate regulatory mechanisms. The homeostatic balance of these processes is crucial; however, in pathological states such as neurodegenerative diseases, autoimmune disorders, and cancer, extensive metabolic reprogramming occurs, resulting in impaired glucose metabolism and mitochondrial dysfunction, which accelerate disease progression. Recent investigations into key regulatory pathways, including mechanistic target of rapamycin, sirtuins, and adenosine monophosphate-activated protein kinase, have considerably deepened our understanding of metabolic dysregulation and opened new avenues for therapeutic innovation. Emerging technologies, such as fluorescent probes, nano-biomaterials, and metabolomic analyses, promise substantial improvements in diagnostic precision. This review critically examines recent advancements and ongoing challenges in metabolism research, emphasizing its potential for precision diagnostics and personalized therapeutic interventions. Future studies should prioritize unraveling the regulatory mechanisms of energy metabolism and the dynamics of intercellular energy interactions. Integrating cutting-edge gene-editing technologies and multi-omics approaches, the development of multi-target pharmaceuticals in synergy with existing therapies such as immunotherapy and dietary interventions could enhance therapeutic efficacy. Personalized metabolic analysis is indispensable for crafting tailored treatment protocols, ultimately providing more accurate medical solutions for patients. This review aims to deepen the understanding and improve the application of energy metabolism to drive innovative diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Hui Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuo Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianhua Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin Guo
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yujing Song
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kun Fu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenjie Gao
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Danfeng Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Wei He
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Lei-Lei Yang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
10
|
Feng L, Cui J, Chen W, Zhu L, Li P, Zhou H, Sun Y, Yi W. Nrg4 Secreted by Brown Adipose Tissue Suppresses Ferroptosis of Sepsis-Induced Liver Injury. Inflammation 2025:10.1007/s10753-024-02230-z. [PMID: 39956880 DOI: 10.1007/s10753-024-02230-z] [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: 10/16/2024] [Revised: 12/07/2024] [Accepted: 12/23/2024] [Indexed: 02/18/2025]
Abstract
Sepsis is a leading cause of death, with the liver being particularly vulnerable to sepsis-related injuries. This damage significantly contributes to disease progression, underscoring the need for new treatments. Brown adipose tissue (BAT) secretes various cytokines, including neuregulin 4 (Nrg4), which plays a protective role in hepatic glucose and lipid metabolism. Ferroptosis, a key type of cell death in sepsis-induced liver injury, has recently gained attention. This study aimed to investigate how BAT-secreted cytokines alleviate liver ferroptosis in sepsis. Septic liver injury was induced in the control and BAT group using cecal ligation and puncture (CLP) and lipopolysaccharide injections. BAT removal worsened ferroptosis; in contrast, CL316243 activation reduced it. These findings suggest that Nrg4 secretion following BAT activation protects the liver during sepsis by inhibiting ferroptosis. Future therapies targeting BAT activation and Nrg4 could potentially mitigate sepsis-induced liver damage, offering new insights into treatment strategies.
Collapse
Affiliation(s)
- Linqi Feng
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jun Cui
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Wenlong Chen
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Lei Zhu
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Panpan Li
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Haitao Zhou
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yang Sun
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
| | - Wei Yi
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
11
|
Li K, Feng J, Li M, Han L, Wu Y. Systematic Review of Interleukin-35 in Endothelial Dysfunction: A New Target for Therapeutic Intervention. Mediators Inflamm 2025; 2025:2003124. [PMID: 39974277 PMCID: PMC11839265 DOI: 10.1155/mi/2003124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 01/21/2025] [Indexed: 02/21/2025] Open
Abstract
Endothelial dysfunction is a significant factor in the pathogenesis of various diseases. In pathological states, endothelial cells (ECs) undergo activation, resulting in dysfunction characterized by the stimulation of inflammatory responses, oxidative stress, cell proliferation, blood coagulation, and vascular adhesions. Interleukin-35 (IL-35), a novel member of the IL-12 family, is primarily secreted by regulatory T cells (Tregs) and regulatory B cells (Bregs). The role of IL-35 in immunomodulation, antioxidative stress, resistance to apoptosis, control of EC activation, adhesion, and angiogenesis in ECs remains incompletely understood, as the specific mechanisms of IL-35 action and its regulation have yet to be fully elucidated. Therefore, this systematic review aims to comprehensively investigate the impact of IL-35 on ECs and their physiological roles in a range of conditions, including cardiovascular diseases, tumors, sepsis, and rheumatoid arthritis (RA), with the objective of elucidating the potential of IL-35 as a therapeutic target for these ailments.
Collapse
Affiliation(s)
- Kai Li
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, China
| | - Jie Feng
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, China
| | - Meng Li
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, China
| | - Leilei Han
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, China
| | - Yanqing Wu
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, China
| |
Collapse
|
12
|
Li Y, Li RY, Zhu JY, Chen M, Mu WJ, Luo HY, Li S, Yan LJ, Yin MT, Li X, Chen HM, Guo L. Maternal exercise prevents metabolic disorders in offspring mice through SERPINA3C. Nat Metab 2025; 7:401-420. [PMID: 39891022 DOI: 10.1038/s42255-024-01213-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 12/17/2024] [Indexed: 02/03/2025]
Abstract
Maternal exercise can improve the metabolic health of the offspring. However, the molecular mechanisms underlying the beneficial effects of maternal exercise on the offspring remain unclear. Here, we show that maternal exercise during pregnancy alleviates high-fat diet (HFD)-induced adipose inflammation and glucose intolerance in offspring mice, accompanied by upregulation of the adipokine serine protease inhibitor A3C (SERPINA3C) both in maternal adipose tissues and the fetal circulation. Adipose SERPINA3C knockdown impairs, but its overexpression in dams mimics, maternal exercise-mediated metabolic benefits in HFD-fed offspring. Maternal SERPINA3C is transported into the fetal circulation and promotes Krüppel-like factor 4 (Klf4) gene promoter demethylation in fetal preadipocytes to increase KLF4 expression, which inhibits adipose inflammation in HFD-fed offspring mice. The SERPINA3C-cathepsin G-integrin β1 axis activates phosphatidylinositol 3-kinase signalling in preadipocytes. This promotes nuclear translocation of the p110β subunit to generate phosphatidylinositol 3,4,5-trisphosphate (PIP3) in the nucleus. O-linked β-N-acetylglucosamine (O-GlcNAc) transferase then binds to PIP3 to promote ten-eleven translocation methylcytosine dioxygenase 1 (TET1) O-GlcNAcylation, thereby enhancing TET1 activity to facilitate Klf4 gene promoter demethylation. These results provide mechanistic insights into maternal exercise-mediated improvement of offspring metabolism.
Collapse
Affiliation(s)
- Yang Li
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Ruo-Ying Li
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Jie-Ying Zhu
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Min Chen
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Wang-Jing Mu
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Hong-Yang Luo
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Shan Li
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Lin-Jing Yan
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Meng-Ting Yin
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Xin Li
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Hu-Min Chen
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Liang Guo
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China.
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China.
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China.
| |
Collapse
|
13
|
Deng D, Zhao B, Yang H, Wang S, Geng Z, Zhou J, Yang G, Han L. Investigating the Effect and Potential Mechanism of Rhamnetin 3- O-α-Rhamnoside on Acute Liver Injury In Vivo and In Vitro. Pharmaceuticals (Basel) 2025; 18:116. [PMID: 39861177 PMCID: PMC11769157 DOI: 10.3390/ph18010116] [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: 11/25/2024] [Revised: 01/02/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Rhamnetin 3-O-α-rhamnoside (ARR) is a major flavonoid of the herb Loranthus tanakae Franch. & Sav., which has been used for treating liver diseases in China. However, the protective effect of ARR on the liver has not been reported. Methods: Zebrafish larvae were used as a visual animal model, and liver injury was induced by thioacetamide (TAA) for an acute liver injury (ALI) model. The hepatoprotective activity of ARR was evaluated by assessing liver morphology, liver function indices, oxidative stress, and the mRNA expression levels of inflammation-related genes in the zebrafish model. Additionally, the ROS level, inflammatory factors, and protein expression related to the IKKβ/NF-κB signaling pathway were measured to investigate a potential mechanism of ARR in HepG2 cells. Results: ARR ameliorated TAA-induced growth retardation, reduced liver injury phenotypes, and decreased oxidative stress in the zebrafish. ARR was also able to lower ROS levels in HepG2 cells, effectively inhibit the overactivation of the IKKβ/NF-κB signaling pathway in pathological conditions, inhibit NF-κB p65 translocation from the cytoplasm to the nucleus, and reduce the release of intracellular inflammatory factors. Conclusions: ARR showed significant protective activity against TAA-induced liver injury in in vivo and in vitro models, and its potential mechanism was closely related to the IKKβ/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Dandan Deng
- School of Pharmaceutical Sciences, Shanxi Medical University, No. 56 South Xinjian Road, Taiyuan 030001, China; (D.D.); (B.Z.); (H.Y.); (Z.G.); (J.Z.)
| | - Borong Zhao
- School of Pharmaceutical Sciences, Shanxi Medical University, No. 56 South Xinjian Road, Taiyuan 030001, China; (D.D.); (B.Z.); (H.Y.); (Z.G.); (J.Z.)
| | - Hong Yang
- School of Pharmaceutical Sciences, Shanxi Medical University, No. 56 South Xinjian Road, Taiyuan 030001, China; (D.D.); (B.Z.); (H.Y.); (Z.G.); (J.Z.)
| | - Songsong Wang
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Science, No. 6699 Qingdao Road, Jinan 250117, China;
| | - Ziying Geng
- School of Pharmaceutical Sciences, Shanxi Medical University, No. 56 South Xinjian Road, Taiyuan 030001, China; (D.D.); (B.Z.); (H.Y.); (Z.G.); (J.Z.)
| | - Jiangtao Zhou
- School of Pharmaceutical Sciences, Shanxi Medical University, No. 56 South Xinjian Road, Taiyuan 030001, China; (D.D.); (B.Z.); (H.Y.); (Z.G.); (J.Z.)
| | - Guane Yang
- School of Pharmaceutical Sciences, Shanxi Medical University, No. 56 South Xinjian Road, Taiyuan 030001, China; (D.D.); (B.Z.); (H.Y.); (Z.G.); (J.Z.)
| | - Liwen Han
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Science, No. 6699 Qingdao Road, Jinan 250117, China;
| |
Collapse
|
14
|
Preto AJ, Chanana S, Ence D, Healy MD, Domingo-Fernández D, West KA. Multi-omics data integration identifies novel biomarkers and patient subgroups in inflammatory bowel disease. J Crohns Colitis 2025; 19:jjae197. [PMID: 39756419 PMCID: PMC11792892 DOI: 10.1093/ecco-jcc/jjae197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND Inflammatory bowel disease (IBD), comprising Crohn's disease (CD) and ulcerative colitis (UC), is a complex condition with diverse manifestations; recent advances in multi-omics technologies are helping researchers unravel its molecular characteristics to develop targeted treatments. OBJECTIVES In this work, we explored one of the largest multi-omics cohorts in IBD, the Study of a Prospective Adult Research Cohort (SPARC IBD), with the goal of identifying predictive biomarkers for CD and UC and elucidating patient subtypes. DESIGN We analyzed genomics, transcriptomics (gut biopsy samples), and proteomics (blood plasma) from hundreds of patients from SPARC IBD. We trained a machine learning model that classifies UC versus CD samples. In parallel, we integrated multi-omics data to unveil patient subgroups in each of the 2 indications independently and analyzed the molecular phenotypes of these patient subpopulations. RESULTS The high performance of the model showed that multi-omics signatures are able to discriminate between the 2 indications. The most predictive features of the model, both known and novel omics signatures for IBD, can potentially be used as diagnostic biomarkers. Patient subgroup analysis in each indication uncovered omics features associated with disease severity in UC patients and with tissue inflammation in CD patients. This culminates with the observation of 2 CD subpopulations characterized by distinct inflammation profiles. CONCLUSIONS Our work unveiled potential biomarkers to discriminate between CD and UC and to stratify each population into well-defined subgroups, offering promising avenues for the application of precision medicine strategies.
Collapse
|
15
|
Zhu B, Sun L, Tong J, Ding Y, Shan Y, He M, Tian X, Mei W, Zhao L, Wang Y. Neuregulin 4 attenuates pancreatic β-cell apoptosis induced by lipotoxicity via activating mTOR-mediated autophagy. Islets 2024; 16:2429854. [PMID: 39541216 PMCID: PMC11572226 DOI: 10.1080/19382014.2024.2429854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 10/19/2024] [Accepted: 11/11/2024] [Indexed: 11/16/2024] Open
Abstract
Neuregulin 4 (Nrg4) is a brown fat-enriched endocrine factor that ameliorates lipid metabolism disorders. Autophagy is critical for pancreatic β-cell to counteract lipotoxicity-induced apoptosis. This study aimed at exploring whether Nrg4 attenuates lipotoxicity-induced β-cell apoptosis by regulating autophagy. The mouse pancreatic β-cell line MIN6 was cultured in palmitic acid (PA) with or without Nrg4 administration. Apoptosis rate, together with anti-apoptotic and pro-apoptotic protein levels, was investigated. Autophagic flux and autophagy-related protein levels along with related signaling pathways that regulate autophagy were also evaluated. Results showed that Nrg4 decreased PA-induced MIN6 apoptosis, enhanced anti-apoptotic protein B-cell lymphoma 2 (Bcl-2) expression and reduced pro-apoptotic proteins Bcl-2-associated X protein (Bax) and cleaved-caspase 3 expressions. Autophagy levels in MIN6 also decreased with PA treatment and Nrg4 administration reactivated autophagy. Further, Nrg4 administration activated autophagy via the mammalian target of rapamycin (mTOR) signaling pathway. In addition, when the mTOR pathway was stimulated or autophagy was suppressed, the beneficial effects of Nrg4 administration on MIN6 apoptosis were diminished. These results imply that Nrg4 administration attenuates MIN6 apoptosis by promoting mTOR-dependent autophagy and thus may lead to a new therapeutic method for type 2 diabetes mellitus (T2DM).
Collapse
Affiliation(s)
- Biao Zhu
- Department of Stomatology, Fuxing Hospital, Capital Medical University, Beijing, China
| | - Lei Sun
- Department of Stomatology, The Ninth Medical Center,Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Junyao Tong
- Department of Stomatology, The First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Yan Ding
- Department of Endocrinology, General Hospital of Central Theater Command, Southern Medical University, Wuhan, Hubei Province, China
| | - Yanbo Shan
- Department of Stomatology, The First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Mingjuan He
- Department of Endocrinology, General Hospital of Central Theater Command, Southern Medical University, Wuhan, Hubei Province, China
| | - Xiaoyu Tian
- Department of Stomatology, The First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Wen Mei
- Department of Endocrinology, General Hospital of Central Theater Command, Southern Medical University, Wuhan, Hubei Province, China
| | - Lisheng Zhao
- Department of Stomatology, The First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Ying Wang
- Department of Stomatology, The First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
| |
Collapse
|
16
|
Wei H, Guo X, Yan J, Tian X, Yang W, Cui K, Wang L, Guo B. Neuregulin-4 alleviates isoproterenol (ISO)-induced cardial remodeling by inhibiting inflammation and apoptosis via AMPK/NF-κB pathway. Int Immunopharmacol 2024; 143:113301. [PMID: 39418729 DOI: 10.1016/j.intimp.2024.113301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/19/2024] [Accepted: 09/29/2024] [Indexed: 10/19/2024]
Abstract
Cardiac remodeling refers to the abnormal changes in cardiac structure and function caused by various pathological conditions. It is an inevitable pathological process in the occurrence and development of heart failure and is related to a variety of cardiovascular diseases. Inflammation and apoptosis are critical pathological processes involved in cardiac remodeling. Neuregulin 4 (Nrg 4) is an adipokine produced primarily by brown adipose tissue that may play a protective role in a variety of inflammatory diseases. The aim of this study was to investigate whether Nrg4 can delay the progression of cardiac remodeling by regulating AMPK/NF-κB pathway, inhibiting inflammation and apoptosis. In our study, we established a model of cardiac remodeling in mice after 14 days of isoproterenol (ISO) intervention, and then gave Nrg4 treatment for another 4 weeks. The cardiac function, the degree of myocardial hypertrophy and myocardial fibrosis of the mice were observed. At the same time, the levels of apoptosis-related proteins (Bax,Bcl-2,Caspase-3), IL-6,IL-Iβ and TNF-α, as well as the activation level of AMPK/NF-κB signaling pathway were evaluated.Nrg4 alleviated ISO-induced cardiac dysfunction, cardiac hypertrophy and fibrosis in mice. Nrg4 also attenuated ISO-induced apoptosis and reduces levels of inflammatory factors to protect ISO-induced myocardial damage. At the same time, the effect of Nrg4 on AMPK/NF-κB pathway was measured in vivo and in vitro. The administration of an AMPK inhibitor was found to reverse the anti-hypertrophy, anti-inflammatory, and anti-apoptotic effects of Nrg4. Our findings suggest that Nrg4 may play a protective role in cardiac remodeling by inhibiting inflammation and apoptosis via AMPK/NF-κB pathway.
Collapse
Affiliation(s)
- Huiqing Wei
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China
| | - Xiaohua Guo
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China
| | - Jie Yan
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China
| | - Xiaochao Tian
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China
| | - Wenhui Yang
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China
| | - Kun Cui
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China
| | - Lijie Wang
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China
| | - Bingyan Guo
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China; Hebei Key Laboratory of Laboratory Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China.
| |
Collapse
|
17
|
Wang S, Yin J, Liu Z, Liu X, Tian G, Xin X, Qin Y, Feng X. Metabolic disorders, inter-organ crosstalk, and inflammation in the progression of metabolic dysfunction-associated steatotic liver disease. Life Sci 2024; 359:123211. [PMID: 39491769 DOI: 10.1016/j.lfs.2024.123211] [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: 07/01/2024] [Revised: 08/20/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) represents a global health concern, affecting over 30 % of adults. It is a principal driver in the development of cirrhosis and hepatocellular carcinoma. The complex pathogenesis of MASLD involves an excessive accumulation of lipids, subsequently disrupting lipid metabolism and prompting inflammation within the liver. This review synthesizes the recent research progress in understanding the mechanisms contributing to MASLD progression, with particular emphasis on metabolic disorders and interorgan crosstalk. We highlight the molecular mechanisms linked to these factors and explore their potential as novel targets for pharmacological intervention. The insights gleaned from this article have important implications for both the prevention and therapeutic management of MASLD.
Collapse
Affiliation(s)
- Shendong Wang
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Junhao Yin
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Zhaojun Liu
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Xin Liu
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Ge Tian
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271000, China
| | - Xijian Xin
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Yiming Qin
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Xiujing Feng
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China; Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China.
| |
Collapse
|
18
|
Ziqubu K, Dludla PV, Mthembu SX, Nkambule B, Mazibuko-Mbeje SE. Low circulating levels of neuregulin 4 as a potential biomarker associated with the severity and prognosis of obesity-related metabolic diseases: a systematic review. Adipocyte 2024; 13:2390833. [PMID: 39162358 PMCID: PMC11340757 DOI: 10.1080/21623945.2024.2390833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/03/2024] [Accepted: 08/06/2024] [Indexed: 08/21/2024] Open
Abstract
BACKGROUND Neuregulin 4 (Nrg4) is a brown adipose tissue-derived adipokine that greatly affects systemic metabolism and improves metabolic derangements. Although abnormal circulating levels of Nrg4 are common in obesity, it remains elusive whether low or elevated levels of this batokine are associated with the onset of metabolic diseases. AIM To assess Nrg4 levels and its role as a feasible biomarker to predict the severity of obesity, gestational diabetes mellitus (GDM), type 2 diabetes mellitus (T2DM), non-alcoholic fatty liver disease (NAFLD), and cardiovascular diseases (CVD). METHODS A search for relevant studies was performed systematically using prominent search engines, including PubMed, Google Scholar, and Embase, by following PRISMA guidelines. RESULTS Ample clinical evidence reported low serum/plasma levels of Nrg4 in obesity and these were inversely proportional to the indices of metabolic syndrome, including body mass index, waist circumference, triglycerides, fasting plasma glucose, and homoeostatic model assessment for insulin resistance as well as high-sensitivity C-reactive protein. Low circulating Nrg4 levels may aid in the prediction of morbid obesity, and subsequent GDM, T2DM, NAFLD, and CVD. CONCLUSION Current clinical evidence emphasizes that the circulating levels of Nrg4 are decreased in morbid obesity, and it also highlights that Nrg4 May serve as a potential prognostic biomarker for obesity-related metabolic diseases.
Collapse
Affiliation(s)
- Khanyisani Ziqubu
- Department of Biochemistry, North-West University, Mmabatho, South Africa
| | - Phiwayinkosi V. Dludla
- Cochrane South Africa, South African Medical Research Council, Tygerberg, South Africa
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, South Africa
| | | | - Bongani Nkambule
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | | |
Collapse
|
19
|
Jiang Y, Xing W, Li Z, Zhao D, Xiu B, Xi Y, Bai S, Li X, Zhang Z, Zhang W, Li H. The calcium-sensing receptor alleviates endothelial inflammation in atherosclerosis through regulation of integrin β1-NLRP3 inflammasome. FEBS J 2024. [PMID: 39552549 DOI: 10.1111/febs.17308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 06/21/2024] [Accepted: 10/16/2024] [Indexed: 11/19/2024]
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease of arteries. Endothelial inflammation is key to the initiation and development of AS. The calcium-sensing receptor (CaSR) is expressed in endothelial cells (ECs) but its role in endothelial inflammation during AS remains unclear. This study focused on the involvement of CaSR in regulating endothelial inflammation and its underlying mechanisms, providing novel insights for AS therapy. Here, we observed that CaSR agonist NPS-R568 significantly reduced atherosclerotic lesions and aortic inflammation in high-fat diet (HFD)-fed ApoE-/- mice, while enhancing the expression of CaSR in aortic tissues. In vitro, human umbilical vein endothelial cells (HUVECs) exposed to oxidized low-density lipoprotein (oxLDL) at 20 μg·mL-1 triggered inflammation, as indicated by the upregulation of vascular cell adhesion molecule-1 (VCAM-1), interleukin (IL)-6, and IL-1β expression, along with increased adherence of THP-1 or U937 cells to the HUVECs. Additionally, treatment with 20 μg·mL-1 oxLDL led to downregulation of CaSR expression in HUVECs. The administration of CaSR agonist NPS-R568 or overexpression of CaSR in HUVECs resulted in a significant reversal of inflammation induced by oxLDL. Mechanistically, CaSR was found to mitigate NLRP3 inflammasome activation by downregulating the protein level of integrin β1. In conclusion, our study elucidates the beneficial role of CaSR in reducing endothelial inflammation in AS through the regulation of integrin β1 and the subsequent NLRP3 inflammasome. CaSR emerges as a promising target for potential therapeutic interventions in AS.
Collapse
Affiliation(s)
- Yunge Jiang
- Department of Pathophysiology, Key Laboratory of Cardiovascular Pathophysiology, Harbin Medical University, China
| | - Wenjing Xing
- Department of Immunology, Harbin Medical University, China
| | - Zhong Li
- Department of Nephrology, First Affiliated Hospital of Harbin Medical University, China
| | - Defeng Zhao
- Department of Pathophysiology, Key Laboratory of Cardiovascular Pathophysiology, Harbin Medical University, China
| | - Bingxu Xiu
- Department of Pathophysiology, Key Laboratory of Cardiovascular Pathophysiology, Harbin Medical University, China
| | - Yuhui Xi
- Department of Pathophysiology, Key Laboratory of Cardiovascular Pathophysiology, Harbin Medical University, China
| | - Shuzhi Bai
- Department of Pathophysiology, Key Laboratory of Cardiovascular Pathophysiology, Harbin Medical University, China
| | - Xiaoxue Li
- Department of Pathophysiology, Key Laboratory of Cardiovascular Pathophysiology, Harbin Medical University, China
| | - Zheqi Zhang
- Department of Immunology, Harbin Medical University, China
| | - Weihua Zhang
- Department of Pathophysiology, Key Laboratory of Cardiovascular Pathophysiology, Harbin Medical University, China
| | - Hongxia Li
- Department of Pathophysiology, Key Laboratory of Cardiovascular Pathophysiology, Harbin Medical University, China
| |
Collapse
|
20
|
Dan X, Li K, Xu J, Yan P. The Potential of Neuregulin 4 as a Novel Biomarker and Therapeutic Agent for Vascular Complications in Type 2 Diabetes Mellitus. J Inflamm Res 2024; 17:8543-8554. [PMID: 39539725 PMCID: PMC11559183 DOI: 10.2147/jir.s492115] [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: 08/19/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Neuregulin 4 (Nrg4), a novel adipokine produced primarily by brown adipose tissue (BAT), has been functionally characterized to exert beneficial effects on modulating energy homeostasis and glucolipid metabolism, and is closely associated with the development and progression of obesity and obesity-associated metabolic diseases, such as type 2 diabetes mellitus (T2DM) and cardiovascular diseases. Recently, there has been a growing focus on the relationship between circulating Nrg4 levels and T2DM-related vascular complications. In this review, we discussed the known and potential roles of Nrg4 in various physiological and pathological processes, and its association with vascular complications in T2DM, in the aim of finding a potential biomarker recommended for the clinical diagnosis, prognosis and follow-up of T2DM patients at high risk of developing vascular complications as well as providing new therapeutic approaches.
Collapse
Affiliation(s)
- Xiaofang Dan
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, People’s Republic of China
- Sichuan Clinical Research Center for Diabetes and Metabolism, Luzhou, People’s Republic of China
| | - Ke Li
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, People’s Republic of China
- Sichuan Clinical Research Center for Diabetes and Metabolism, Luzhou, People’s Republic of China
| | - Jiali Xu
- Department of Gastroenterology, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
| | - Pijun Yan
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, People’s Republic of China
- Sichuan Clinical Research Center for Diabetes and Metabolism, Luzhou, People’s Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, People’s Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, People’s Republic of China
| |
Collapse
|
21
|
Hsiao YT, Yoshida Y, Okuda S, Abe M, Mizuno S, Takahashi S, Nakagami H, Morishita R, Kamimura K, Terai S, Aung TM, Li J, Furihata T, Tang JY, Walsh K, Ishigami A, Minamino T, Shimizu I. PCPE-1, a brown adipose tissue-derived cytokine, promotes obesity-induced liver fibrosis. EMBO J 2024; 43:4846-4869. [PMID: 39160276 PMCID: PMC11535236 DOI: 10.1038/s44318-024-00196-0] [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: 10/20/2023] [Revised: 07/10/2024] [Accepted: 07/22/2024] [Indexed: 08/21/2024] Open
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH, previously termed non-alcoholic steatohepatitis (NASH)), is a major complication of obesity that promotes fatty liver disease. MASH is characterized by progressive tissue fibrosis and sterile liver inflammation that can lead to liver cirrhosis, cancer, and death. The molecular mechanisms of fibrosis in MASH and its systemic control remain poorly understood. Here, we identified the secreted-type pro-fibrotic protein, procollagen C-endopeptidase enhancer-1 (PCPE-1), as a brown adipose tissue (BAT)-derived adipokine that promotes liver fibrosis in a murine obesity-induced MASH model. BAT-specific or systemic PCPE-1 depletion in mice ameliorated liver fibrosis, whereas, PCPE-1 gain of function in BAT enhanced hepatic fibrosis. High-calorie diet-induced ER stress increased PCPE-1 production in BAT through the activation of IRE-1/JNK/c-Fos/c-Jun signaling. Circulating PCPE-1 levels are increased in the plasma of MASH patients, suggesting a therapeutic possibility. In sum, our results uncover PCPE-1 as a novel systemic control factor of liver fibrosis.
Collapse
Affiliation(s)
- Yung Ting Hsiao
- Department of Cardiovascular Aging, National Cerebral and Cardiovascular Center Research Institute, Osaka, 564-8565, Japan
| | - Yohko Yoshida
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
- Department of Advanced Senotherapeutics, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Shujiro Okuda
- Division of Bioinformatics, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951-8510, Japan
| | - Manabu Abe
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center in Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Ibaraki, 305-8577, Japan
| | - Satoru Takahashi
- Laboratory Animal Resource Center in Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Ibaraki, 305-8577, Japan
| | - Hironori Nakagami
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan
| | - Ryuichi Morishita
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan
| | - Kenya Kamimura
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 951-8510, Japan
- Department of General Medicine, Niigata University School of Medicine, Niigata, 951-8510, Japan
| | - Shuji Terai
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 951-8510, Japan
| | - Tin May Aung
- Department of Cardiovascular Aging, National Cerebral and Cardiovascular Center Research Institute, Osaka, 564-8565, Japan
| | - Ji Li
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, PR China
| | - Takaaki Furihata
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Jing Yuan Tang
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Kenneth Walsh
- Division of Cardiovascular Medicine, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Akihito Ishigami
- Molecular Regulation of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, 173-0015, Japan
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Ippei Shimizu
- Department of Cardiovascular Aging, National Cerebral and Cardiovascular Center Research Institute, Osaka, 564-8565, Japan.
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, 564-8565, Japan.
| |
Collapse
|
22
|
Xu Z, Li H, Cao G, Li P, Zhou H, Sun Y. The protective role of brown adipose tissue in cardiac cell damage after myocardial infarction and heart failure. Lipids Health Dis 2024; 23:338. [PMID: 39415186 PMCID: PMC11481725 DOI: 10.1186/s12944-024-02326-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 10/06/2024] [Indexed: 10/18/2024] Open
Abstract
Acute myocardial infarction (AMI) and related cardiovascular disease complications are the leading causes of mortality worldwide. Brown adipose tissue (BAT) is thermogenic and characterized by the uncoupling protein expression. Recent studies have found that in cardiovascular diseases, activated BAT can effectively improve the prognosis of AMI and concurrent heart failure through intercellular communication. However, a clear and systematic understanding of the myocardial protective mechanism of BAT after AMI is lacking, especially in the endocrine function of BAT. This review describes the effects of BAT on various cells in the heart after AMI. BAT plays a protective role on cardiac cells and fibroblasts during ischemia/reperfusion (I/R), myocardial remodeling, and myocardial fibrosis. This review also discusses the changes caused by BAT activation in different stages of heart failure. Finally, this review summarizes the treatment methods that target BAT to improve AMI. Further in-depth researches are still needed to clarify the underlying mechanism of the connection between BAT and different cells in cardiac tissue in order to identify potential therapeutic targets.
Collapse
Affiliation(s)
- Zhe Xu
- Department of Geriatric Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Hong Li
- Department of General Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Guojie Cao
- Department of Geriatric Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Panpan Li
- Department of Geriatric Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Haitao Zhou
- Department of Geriatric Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yang Sun
- Department of Geriatric Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
23
|
Jiang T, Wei Y, Xu R, Jin Y, Song T, Wang H, Chen W, Tian H, Xu L, Zhao Y, Fu Y. Renal denervation alleviates vascular remodeling in spontaneously hypertensive rats by regulating perivascular adipose tissue. Hypertens Res 2024; 47:2760-2772. [PMID: 39043918 DOI: 10.1038/s41440-024-01791-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 06/03/2024] [Accepted: 06/15/2024] [Indexed: 07/25/2024]
Abstract
Vascular remodeling is the main pathological process that causes the damage of the target organ of hypertension. Perivascular adipose tissue (PVAT) surrounds blood vessels and plays a key role in the pathogenesis of various cardiovascular diseases. This study aimed to investigate the effects of renal denervation (RDN) on hypertensive vascular remodeling and to elucidate the role of PVAT in this process. Male spontaneously hypertensive rat (SHR) and Wistar-Kyoto (WKY) rat were selected. Aortic vascular remodeling was evaluated using hematoxylin and eosin (H&E) staining and Masson's trichrome staining. Morphological changes in the PVAT were observed through H&E and Oil Red O staining. Dihydroethidium was used to measure oxidative stress levels in PVAT, while western blot analysis was used to determine the expression levels of proteins associated with vascular remodeling. The results showed that the aortic medial thickness, media thickness/lumen diameter, collagen volume fraction, and reactive oxygen species (ROS) level in PVAT were significantly higher in the SHR group than in the WKY group. The indexes mentioned above were lower in the SHR-RDN group than in the SHR group. H&E staining revealed that fat droplets in PVAT in the SHR-RDN group became smaller and browning occurred. Moreover, the protein expression of uncoupling protein-1 (UCP-1) and neuregulin 4 (Nrg4) was significantly increased in the SHR-RDN group. In addition, the expression of adiponectin increased and the expression of leptin decreased in the SHR-RDN group compared to the SHR group. In conclusion, RDN can relieve hypertensive vascular remodeling, which may be associated with PVAT.
Collapse
Affiliation(s)
- Tingting Jiang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yongkang Wei
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Rui Xu
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yuanyuan Jin
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tingting Song
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Huiying Wang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenjia Chen
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hui Tian
- Department of General Practice, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lei Xu
- Department of General Practice, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yong Zhao
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Yu Fu
- Department of General Practice, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
24
|
Tan Y, Huang Z, Jin Y, Wang J, Fan H, Liu Y, Zhang L, Wu Y, Liu P, Li T, Ran J, Tian H, Lam SM, Liu M, Zhou J, Yang Y. Lipid droplets sequester palmitic acid to disrupt endothelial ciliation and exacerbate atherosclerosis in male mice. Nat Commun 2024; 15:8273. [PMID: 39333556 PMCID: PMC11437155 DOI: 10.1038/s41467-024-52621-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 09/17/2024] [Indexed: 09/29/2024] Open
Abstract
Disruption of ciliary homeostasis in vascular endothelial cells has been implicated in the development of atherosclerosis. However, the molecular basis for the regulation of endothelial cilia during atherosclerosis remains poorly understood. Herein, we provide evidence in male mice that the accumulation of lipid droplets in vascular endothelial cells induces ciliary loss and contributes to atherosclerosis. Triglyceride accumulation in vascular endothelial cells differentially affects the abundance of free fatty acid species in the cytosol, leading to stimulated lipid droplet formation and suppressed protein S-palmitoylation. Reduced S-palmitoylation of ciliary proteins, including ADP ribosylation factor like GTPase 13B, results in the loss of cilia. Restoring palmitic acid availability, either through pharmacological inhibition of stearoyl-CoA desaturase 1 or a palmitic acid-enriched diet, significantly restores endothelial cilia and mitigates the progression of atherosclerosis. These findings thus uncover a previously unrecognized role of lipid droplets in regulating ciliary homeostasis and provide a feasible intervention strategy for preventing and treating atherosclerosis.
Collapse
Affiliation(s)
- Yanjie Tan
- Center for Cell Structure and Function, Haihe Laboratory of Cell Ecosystem, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, 250014, Jinan, China
| | - Zhenzhou Huang
- Center for Cell Structure and Function, Haihe Laboratory of Cell Ecosystem, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, 250014, Jinan, China
| | - Yi Jin
- Metabolism and Disease Research Centre, Central Hospital Affiliated to Shandong First Medical University, 250013, Jinan, China
| | - Jiaying Wang
- Center for Cell Structure and Function, Haihe Laboratory of Cell Ecosystem, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, 250014, Jinan, China
| | - Hongjun Fan
- Center for Cell Structure and Function, Haihe Laboratory of Cell Ecosystem, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, 250014, Jinan, China
| | - Yangyang Liu
- Center for Cell Structure and Function, Haihe Laboratory of Cell Ecosystem, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, 250014, Jinan, China
| | - Liang Zhang
- Metabolism and Disease Research Centre, Central Hospital Affiliated to Shandong First Medical University, 250013, Jinan, China
| | - Yue Wu
- Center for Cell Structure and Function, Haihe Laboratory of Cell Ecosystem, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, 250014, Jinan, China
| | - Peiwei Liu
- Center for Cell Structure and Function, Haihe Laboratory of Cell Ecosystem, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, 250014, Jinan, China
| | - Tianliang Li
- Center for Cell Structure and Function, Haihe Laboratory of Cell Ecosystem, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, 250014, Jinan, China
| | - Jie Ran
- Center for Cell Structure and Function, Haihe Laboratory of Cell Ecosystem, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, 250014, Jinan, China
| | - He Tian
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Sin Man Lam
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
- LipidALL Technologies Company Limited, 213022, Changzhou, China
| | - Min Liu
- Laboratory of Tissue Homeostasis, Haihe Laboratory of Cell Ecosystem, 300462, Tianjin, China
| | - Jun Zhou
- Center for Cell Structure and Function, Haihe Laboratory of Cell Ecosystem, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, 250014, Jinan, China.
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, 300071, Tianjin, China.
| | - Yunfan Yang
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 250012, Jinan, China.
| |
Collapse
|
25
|
Hu Y, Huang Y, Jiang Y, Weng L, Cai Z, He B. The Different Shades of Thermogenic Adipose Tissue. Curr Obes Rep 2024; 13:440-460. [PMID: 38607478 DOI: 10.1007/s13679-024-00559-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/12/2024] [Indexed: 04/13/2024]
Abstract
PURPOSE OF REVIEW By providing a concise overview of adipose tissue types, elucidating the regulation of adipose thermogenic capacity in both physiological contexts and chronic wasting diseases (a protracted hypermetabolic state that precipitates sustained catabolism and consequent progressive corporeal atrophy), and most importantly, delving into the ongoing discourse regarding the role of adipose tissue thermogenic activation in chronic wasting diseases, this review aims to provide researchers with a comprehensive understanding of the field. RECENT FINDINGS Adipose tissue, traditionally classified as white, brown, and beige (brite) based on its thermogenic activity and potential, is intricately regulated by complex mechanisms in response to exercise or cold exposure. This regulation is adipose depot-specific and dependent on the duration of exposure. Excessive thermogenic activation of adipose tissue has been observed in chronic wasting diseases and has been considered a pathological factor that accelerates disease progression. However, this conclusion may be confounded by the detrimental effects of excessive lipolysis. Recent research also suggests that such activation may play a beneficial role in the early stages of chronic wasting disease and provide potential therapeutic effects. A more comprehensive understanding of the changes in adipose tissue thermogenesis under physiological and pathological conditions, as well as the underlying regulatory mechanisms, is essential for the development of novel interventions to improve health and prevent disease.
Collapse
Affiliation(s)
- Yunwen Hu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Yijie Huang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Yangjing Jiang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Lvkan Weng
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Zhaohua Cai
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| |
Collapse
|
26
|
Xie H, Jiang Y, Xiang Y, Wu B, Zhao J, Huang R, Wang M, Wang Y, Liu J, Wu D, Tian D, Bian E. Super-enhancer-driven LIF promotes the mesenchymal transition in glioblastoma by activating ITGB2 signaling feedback in microglia. Neuro Oncol 2024; 26:1438-1452. [PMID: 38554116 PMCID: PMC11300025 DOI: 10.1093/neuonc/noae065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Indexed: 04/01/2024] Open
Abstract
BACKGROUND The mesenchymal (MES) subtype of glioblastoma (GBM) is believed to be influenced by both cancer cell-intrinsic alterations and extrinsic cellular interactions, yet the underlying mechanisms remain unexplored. METHODS Identification of microglial heterogeneity by bioinformatics analysis. Transwell migration, invasion assays, and tumor models were used to determine gene function and the role of small molecule inhibitors. RNA sequencing, chromatin immunoprecipitation, and dual-luciferase reporter assays were performed to explore the underlying regulatory mechanisms. RESULTS We identified the inflammatory microglial subtype of tumor-associated microglia (TAM) and found that its specific gene integrin beta 2 (ITGB2) was highly expressed in TAM of MES GBM tissues. Mechanistically, the activation of ITGB2 in microglia promoted the interaction between the SH2 domain of STAT3 and the cytoplasmic domain of ITGB2, thereby stimulating the JAK1/STAT3/IL-6 signaling feedback to promote the MES transition of GBM cells. Additionally, microglia communicated with GBM cells through the interaction between the receptor ITGB2 on microglia and the ligand ICAM-1 on GBM cells, while an increased secretion of ICAM-1 was induced by the proinflammatory cytokine leukemia inhibitory factor (LIF). Further studies demonstrated that inhibition of cyclin-dependent kinase 7 substantially reduced the recruitment of SNW1 to the super-enhancer of LIF, resulting in transcriptional inhibition of LIF. We identified notoginsenoside R1 as a novel LIF inhibitor that exhibited synergistic effects in combination with temozolomide. CONCLUSIONS Our research reveals that the epigenetic-mediated interaction of GBM cells with TAM drives the MES transition of GBM and provides a novel therapeutic avenue for patients with MES GBM.
Collapse
Affiliation(s)
- Han Xie
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Hospital of Anhui Medical University, Hefei, China
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yanyi Jiang
- Institute of Health and Medical Technology, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, China
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
| | - Yufei Xiang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Hospital of Anhui Medical University, Hefei, China
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Baoming Wu
- School of pharmacy, Anhui Medical University, Hefei, China
| | - Jiajia Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Hospital of Anhui Medical University, Hefei, China
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ruixiang Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Hospital of Anhui Medical University, Hefei, China
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Mengting Wang
- School of pharmacy, Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Hospital of Anhui Medical University, Hefei, China
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yunlong Wang
- School of pharmacy, Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Hospital of Anhui Medical University, Hefei, China
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jun Liu
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Hospital of Anhui Medical University, Hefei, China
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Dejun Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Dasheng Tian
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Hospital of Anhui Medical University, Hefei, China
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Erbao Bian
- School of pharmacy, Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Hospital of Anhui Medical University, Hefei, China
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
27
|
Li Y, Chen Z, Xiao Y, Li X. Cross-talks between perivascular adipose tissue and neighbors: multifaceted nature of nereids. Front Pharmacol 2024; 15:1442086. [PMID: 39156105 PMCID: PMC11327032 DOI: 10.3389/fphar.2024.1442086] [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: 06/01/2024] [Accepted: 07/24/2024] [Indexed: 08/20/2024] Open
Abstract
Perivascular adipose tissue (PVAT) is a unique fat depot surrounding blood vessels and plays a vital role in the progression of vascular remodeling and dysfunction. PVAT exhibits remarkable differences in structure, phenotype, origin, and secretome across anatomical locations. The proximity of PVAT to neighboring vascular beds favors a niche for bidirectional communication between adipocytes and vascular smooth muscle cells, endothelial cells, and immune cells. In this review, we update our understanding of PVAT's regional differences and provide a comprehensive exploration of how these differences impact cross-talks between PVAT and the vascular wall. Different PVAT depots show different degrees of vasoprotective function and resilience to pathological changes such as obesity and vasculopathies, shaping multifaceted interactions between PVAT depots and adjacent vasculatures. The depot-specific resilience may lead to innovative strategies to manage cardiometabolic disorders.
Collapse
Affiliation(s)
- Yujuan Li
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau, China
- (R & D Center) Laboratory for Drug Discovery from Natural Resource, Macau University of Science and Technology, Macau, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Zhang Chen
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau, China
- (R & D Center) Laboratory for Drug Discovery from Natural Resource, Macau University of Science and Technology, Macau, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Ying Xiao
- Faculty of Medicine, Macau University of Science and Technology, Macau, China
| | - Xinzhi Li
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau, China
- (R & D Center) Laboratory for Drug Discovery from Natural Resource, Macau University of Science and Technology, Macau, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| |
Collapse
|
28
|
Xu L, Yang Q, Zhou J. Mechanisms of Abnormal Lipid Metabolism in the Pathogenesis of Disease. Int J Mol Sci 2024; 25:8465. [PMID: 39126035 PMCID: PMC11312913 DOI: 10.3390/ijms25158465] [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: 06/25/2024] [Revised: 07/26/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024] Open
Abstract
Lipid metabolism is a critical component in preserving homeostasis and health, and lipids are significant chemicals involved in energy metabolism in living things. With the growing interest in lipid metabolism in recent years, an increasing number of studies have demonstrated the close relationship between abnormalities in lipid metabolism and the development of numerous human diseases, including cancer, cardiovascular, neurological, and endocrine system diseases. Thus, understanding how aberrant lipid metabolism contributes to the development of related diseases and how it works offers a theoretical foundation for treating and preventing related human diseases as well as new avenues for the targeted treatment of related diseases. Therefore, we discuss the processes of aberrant lipid metabolism in various human diseases in this review, including diseases of the cardiovascular system, neurodegenerative diseases, endocrine system diseases (such as obesity and type 2 diabetes mellitus), and other diseases including cancer.
Collapse
Affiliation(s)
| | | | - Jinghua Zhou
- School of Basic Medicine Sciences, Hangzhou Normal University, Hangzhou 311121, China
| |
Collapse
|
29
|
Wang P, Guo X, Wang H, Wang L, Ma M, Guo B. Neuregulin-4 protects cardiomyocytes against high-glucose-induced ferroptosis via the AMPK/NRF2 signalling pathway. Biol Direct 2024; 19:62. [PMID: 39095871 PMCID: PMC11295585 DOI: 10.1186/s13062-024-00505-x] [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: 03/29/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND High glucose levels are key factors and key contributors to several cardiovascular diseases associated with cardiomyocyte injury. Ferroptosis, which was identified in recent years, is a mode of cell death caused by the iron-mediated accumulation of lipid peroxides. Neuregulin-4 (Nrg4) is an adipokine that has protective effects against metabolic disorders and insulin resistance. Our previous study revealed that Nrg4 has a protective effect against diabetic myocardial injury, and the aim of this study was to investigate whether Nrg4 could attenuate the occurrence of high glucose-induced ferroptosis in cardiomyocytes. METHODS We constructed an in vivo diabetic myocardial injury model in which primary cardiomyocytes were cultured in vitro and treated with Nrg4. Changes in ferroptosis-related protein levels and ferroptosis-related indices in cardiomyocytes were observed. In addition, we performed back-validation and explored signalling pathways that regulate ferroptosis in primary cardiomyocytes. RESULTS Nrg4 attenuated cardiomyocyte ferroptosis both in vivo and in vitro. Additionally, the AMPK/NRF2 signalling pathway was activated during this process, and when the AMPK/NRF2 pathway was inhibited, the beneficial effects of Nrg4 were attenuated. CONCLUSION Nrg4 antagonizes high glucose-induced ferroptosis in cardiomyocytes via the AMPK/NRF2 signalling pathway.
Collapse
Affiliation(s)
- Pengfei Wang
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang, 050000, China
| | - Xiaohua Guo
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang, 050000, China
| | - Hongchao Wang
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang, 050000, China
| | - Lijie Wang
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang, 050000, China
| | - Meifang Ma
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang, 050000, China
- Handan Central Hospital, Handan, 056000, China
| | - Bingyan Guo
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang, 050000, China.
- Hebei Key Laboratory of Laboratory Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China.
| |
Collapse
|
30
|
Zheng L, Zhang C, Bu S, Guo W, Li T, Xu Y, Liu Y, Yuan C, Feng C, Zong G, Zhu J, Xing M, Geng X. The Causal Effect of Serum Lipid Levels Mediated by Neuregulin 4 on the Risk of Four Atherosclerosis Subtypes: Evidence from Mendelian Randomization Analysis. Vasc Health Risk Manag 2024; 20:351-357. [PMID: 39104661 PMCID: PMC11299727 DOI: 10.2147/vhrm.s459075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 07/20/2024] [Indexed: 08/07/2024] Open
Abstract
Background Neuregulin 4 (NRG4) was known to be associated with serum lipid levels and atherosclerosis. However, it is unknown whether the role of NRG4 in lipid homeostasis is causal to atherosclerosis and whether the effect is beneficial across different atherosclerosis subtypes. Methods We investigated the causal role of the levels of serum low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol, and triglycerides regulated by NRG4 in subtypes of atherosclerosis through two sample Mendelian randomization. Aggregated genome-wide association study (GWAS) summary data for serum lipid level of 1.32 million individuals with European ancestry were obtained from the Global Lipids Genetics Consortium. GWAS summary data for four atherosclerosis subtypes (peripheral, coronary, cerebral and the other atherosclerosis) were obtained from FinnGen Consortium. Generalized inverse-variance-weighted Mendelian randomization and several sensitivity analyses were used to obtain the causal estimates. Results A 1-SD genetically elevated LDL-C level mediated by NRG4 was validated to be nominally associated with the risk of peripheral atherosclerosis (log (odds ratio)= 4.14, 95% confidence interval 0.11 to 8.17, P = 0.04), and the other associations were not significant or could not be validated by sensitivity analyses. Conclusion LDL-C lowering mediated by NRG4 is likely to prevent peripheral atherosclerosis.
Collapse
Affiliation(s)
- Longyi Zheng
- Department of Endocrinology, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Chengjing Zhang
- Department of Nutrition, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, People’s Republic of China
| | - Shichang Bu
- Department of Endocrinology, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Wencheng Guo
- Department of General Surgery and Vascular Surgery, Huashan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Tongtong Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Ying Xu
- National Center for Liver Cancer, Naval Medical University, Shanghai, People’s Republic of China
| | - Yunan Liu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Caimei Yuan
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Chengwu Feng
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Geng Zong
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Jingwen Zhu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Maoying Xing
- National Center for Liver Cancer, Naval Medical University, Shanghai, People’s Republic of China
| | - Xin Geng
- National Center for Liver Cancer, Naval Medical University, Shanghai, People’s Republic of China
| |
Collapse
|
31
|
Wu G, Liao J, Zhu X, Zhang Y, Lin Y, Zeng Y, Zhao J, Zhang J, Yao T, Shen X, Li H, Hu L, Zhang W. Shexiang Baoxin Pill enriches Lactobacillus to regulate purine metabolism in patients with stable coronary artery disease. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155727. [PMID: 38781732 DOI: 10.1016/j.phymed.2024.155727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 04/29/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND It has been clinically confirmed that the Shexiang Baoxin Pill (SBP) dramatically reduces the frequency of angina in patients with stable coronary artery disease (SCAD). However, potential therapeutic mechanism of SBP has not been fully explored. PURPOSE The study explored the therapeutic mechanism of SBP in the treatment of SCAD patients. METHODS We examined the serum metabolic profiles of patients with SCAD following SBP treatment. A rat model of acute myocardial infarction (AMI) was established, and the potential therapeutic mechanism of SBP was explored using metabolomics, transcriptomics, and 16S rRNA sequencing. RESULTS SBP decreased inosine production and improved purine metabolic disorders in patients with SCAD and in animal models of AMI. Inosine was implicated as a potential biomarker for SBP efficacy. Furthermore, SBP inhibited the expression of genes involved in purine metabolism, which are closely associated with thrombosis, inflammation, and platelet function. The regulation of purine metabolism by SBP was associated with the enrichment of Lactobacillus. Finally, the effects of SBP on inosine production and vascular function could be transmitted through the transplantation of fecal microbiota. CONCLUSION Our study reveals a novel mechanism by which SBP regulates purine metabolism by enriching Lactobacillus to exert cardioprotective effects in patients with SCAD. The data also provide previously undocumented evidence indicating that inosine is a potential biomarker for evaluating the efficacy of SBP in the treatment of SCAD.
Collapse
Affiliation(s)
- Gaosong Wu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jingyu Liao
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiaoyan Zhu
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yuhao Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yuan Lin
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yuanyuan Zeng
- Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Jing Zhao
- Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Jingfang Zhang
- Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Tingting Yao
- Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Xiaoxu Shen
- Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Houkai Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Liang Hu
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Weidong Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Pharmacy, Naval Medical University, Shanghai, 200433, China; Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China.
| |
Collapse
|
32
|
Cai J, Quan Y, Zhu S, Jiayan Lin, Zhang Q, Liu J, Liang Z, Liao Y, Jiang W, He Y, Su T, Lu F. The browning and mobilization of subcutaneous white adipose tissue supports efficient skin repair. Cell Metab 2024; 36:1287-1301.e7. [PMID: 38838641 DOI: 10.1016/j.cmet.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 03/05/2024] [Accepted: 05/08/2024] [Indexed: 06/07/2024]
Abstract
Adipocytes in dermis are considered to be important participants in skin repair and regeneration, but the role of subcutaneous white adipose tissue (sWAT) in skin repair is poorly understood. Here, we revealed the dynamic changes of sWAT during wound healing process. Lineage-tracing mouse studies revealed that sWAT would enter into the large wound bed and participate in the formation of granulation tissue. Moreover, sWAT undergoes beiging after skin injury. Inhibition of sWAT beiging by genetically silencing PRDM16, a key regulator to beiging, hindered wound healing process. The transcriptomics results suggested that beige adipocytes in sWAT abundantly express neuregulin 4 (NRG4), which regulated macrophage polarization and the function of myofibroblasts. In diabetic wounds, the beiging of sWAT was significantly suppressed. Thus, adipocytes from sWAT regulate multiple aspects of repair and may be therapeutic for inflammatory diseases and defective wound healing associated with aging and diabetes.
Collapse
Affiliation(s)
- Junrong Cai
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong 510515, P.R. China
| | - Yuping Quan
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong 510515, P.R. China
| | - Shaowei Zhu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong 510515, P.R. China
| | - Jiayan Lin
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong 510515, P.R. China
| | - Qian Zhang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong 510515, P.R. China
| | - Juzi Liu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong 510515, P.R. China
| | - Zhuokai Liang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong 510515, P.R. China
| | - Yunjun Liao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong 510515, P.R. China
| | - Wenqing Jiang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong 510515, P.R. China
| | - Yufei He
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong 510515, P.R. China
| | - Ting Su
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong 510515, P.R. China.
| | - Feng Lu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong 510515, P.R. China.
| |
Collapse
|
33
|
Bugide S, Reddy DS, Malvi P, Gupta R, Wajapeyee N. ALK inhibitors suppress HCC and synergize with anti-PD-1 therapy and ABT-263 in preclinical models. iScience 2024; 27:109800. [PMID: 38741708 PMCID: PMC11089374 DOI: 10.1016/j.isci.2024.109800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 02/09/2024] [Accepted: 04/18/2024] [Indexed: 05/16/2024] Open
Abstract
Hepatocellular carcinoma (HCC) currently lacks effective therapies, leaving a critical need for new treatment options. A previous study identified the anaplastic lymphoma kinase (ALK) amplification in HCC patients, raising the question of whether ALK inhibitors could be a viable treatment. Here, we showed that both ALK inhibitors and ALK knockout effectively halted HCC growth in cell cultures. Lorlatinib, a potent ALK inhibitor, suppressed HCC tumor growth and metastasis across various mouse models. Additionally, in an advanced immunocompetent humanized mouse model, when combined with an anti-PD-1 antibody, lorlatinib more potently suppressed HCC tumor growth, surpassing individual drug efficacy. Lorlatinib induced apoptosis and senescence in HCC cells, and the senolytic agent ABT-263 enhanced the efficacy of lorlatinib. Additional studies identified that the apoptosis-inducing effect of lorlatinib was mediated via GGN and NRG4. These findings establish ALK inhibitors as promising HCC treatments, either alone or in combination with immunotherapies or senolytic agents.
Collapse
Affiliation(s)
- Suresh Bugide
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Dhana Sekhar Reddy
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Parmanand Malvi
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Romi Gupta
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35233, USA
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Narendra Wajapeyee
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35233, USA
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| |
Collapse
|
34
|
Wang Y, Wang X, Chen Y, Zhang Y, Zhen X, Tao S, Dou J, Li P, Jiang G. Perivascular fat tissue and vascular aging: A sword and a shield. Pharmacol Res 2024; 203:107140. [PMID: 38513826 DOI: 10.1016/j.phrs.2024.107140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 02/16/2024] [Accepted: 03/11/2024] [Indexed: 03/23/2024]
Abstract
The understanding of the function of perivascular adipose tissue (PVAT) in vascular aging has significantly changed due to the increasing amount of information regarding its biology. Adipose tissue surrounding blood vessels is increasingly recognized as a key regulator of vascular disorders. It has significant endocrine and paracrine effects on the vasculature and is mediated by the production of a variety of bioactive chemicals. It also participates in a number of pathological regulatory processes, including oxidative stress, immunological inflammation, lipid metabolism, vasoconstriction, and dilation. Mechanisms of homeostasis and interactions between cells at the local level tightly regulate the function and secretory repertoire of PVAT, which can become dysregulated during vascular aging. The PVAT secretion group changes from being reducing inflammation and lowering cholesterol to increasing inflammation and increasing cholesterol in response to systemic or local inflammation and insulin resistance. In addition, the interaction between the PVAT and the vasculature is reciprocal, and the biological processes of PVAT are directly influenced by the pertinent indicators of vascular aging. The architectural and biological traits of PVAT, the molecular mechanism of crosstalk between PVAT and vascular aging, and the clinical correlation of vascular age-related disorders are all summarized in this review. In addition, this paper aims to elucidate and evaluate the potential benefits of therapeutically targeting PVAT in the context of mitigating vascular aging. Furthermore, it will discuss the latest advancements in technology used for targeting PVAT.
Collapse
Affiliation(s)
- Yan Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xianmin Wang
- Xinjiang Uygur Autonomous Region Hospital of Traditional Chinese Medicine, Xinjiang 830000, China
| | - Yang Chen
- School of Traditional Chinese Medicine, Xinjiang Medical University, Xinjiang 830011, China
| | - Yuelin Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xianjie Zhen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Siyu Tao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jinfang Dou
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Peng Li
- Xinjiang Uygur Autonomous Region Hospital of Traditional Chinese Medicine, Xinjiang 830000, China
| | - Guangjian Jiang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; School of Traditional Chinese Medicine, Xinjiang Medical University, Xinjiang 830011, China.
| |
Collapse
|
35
|
Zhan M, Sun H, Wang Z, Li G, Yang R, Mignani S, Majoral JP, Shen M, Shi X. Nanoparticle-Mediated Multiple Modulation of Bone Microenvironment To Tackle Osteoarthritis. ACS NANO 2024; 18:10625-10641. [PMID: 38563322 DOI: 10.1021/acsnano.4c00909] [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: 04/04/2024]
Abstract
Development of nanomedicines that can collaboratively scavenge reactive oxygen species (ROS) and inhibit inflammatory cytokines, along with osteogenesis promotion, is essential for efficient osteoarthritis (OA) treatment. Herein, we report the design of a ROS-responsive nanomedicine formulation based on fibronectin (FN)-coated polymer nanoparticles (NPs) loaded with azabisdimethylphoaphonate-terminated phosphorus dendrimers (G4-TBP). The constructed G4-TBP NPs-FN with a size of 268 nm are stable under physiological conditions, can be specifically taken up by macrophages through the FN-mediated targeting, and can be dissociated in the oxidative inflammatory microenvironment. The G4-TBP NPs-FN loaded with G4-TBP dendrimer having intrinsic anti-inflammatory property and FN having both anti-inflammatory and antioxidative properties display integrated functions of ROS scavenging, hypoxia attenuation, and macrophage M2 polarization, thus protecting macrophages from apoptosis and creating designed bone immune microenvironment for stem cell osteogenic differentiation. These characteristics of the G4-TBP NPs-FN lead to their effective treatment of an OA model in vivo to reduce pathological changes of joints including synovitis inhibition and cartilage matrix degradation and simultaneously promote osteogenic differentiation for bone repair. The developed nanomedicine formulation combining the advantages of both bioactive phosphorus dendrimers and FN to treat OA may be developed for immunomodulatory therapy of different inflammatory diseases.
Collapse
Affiliation(s)
- Mengsi Zhan
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P. R. China
| | - Huxiao Sun
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P. R. China
| | - Zhiqiang Wang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P. R. China
| | - Gaoming Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P. R. China
| | - Rui Yang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P. R. China
| | - Serge Mignani
- CQM-Centro de Química da Madeira, Universidade da Madeira, Campus Universitário da Penteada, 9020-105 Funchal, Portugal
| | - Jean-Pierre Majoral
- Laboratoire de Chimie de Coordination du CNRS, 205 Route de Narbonne, CEDEX 4, 31077 Toulouse, France
- Université Toulouse, 118 Route de Narbonne, CEDEX 4, 31077 Toulouse, France
| | - Mingwu Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P. R. China
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P. R. China
- CQM-Centro de Química da Madeira, Universidade da Madeira, Campus Universitário da Penteada, 9020-105 Funchal, Portugal
| |
Collapse
|
36
|
Zhang J, Kibret BG, Vatner DE, Vatner SF. The role of brown adipose tissue in mediating healthful longevity. THE JOURNAL OF CARDIOVASCULAR AGING 2024; 4:17. [PMID: 39119146 PMCID: PMC11309368 DOI: 10.20517/jca.2024.01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
There are two major subtypes of adipose tissue, i.e., white adipose tissue (WAT) and brown adipose tissue (BAT). It has been known for a long time that WAT mediates obesity and impairs healthful longevity. More recently, interest has focused on BAT, which, unlike WAT, actually augments healthful aging. The goal of this review is to examine the role of BAT in mediating healthful longevity. A major role for BAT and its related beige adipose tissue is thermogenesis, as a mechanism to maintain body temperature by producing heat through uncoupling protein 1 (UCP1) or through UCP1-independent thermogenic pathways. Our hypothesis is that healthful longevity is, in part, mediated by BAT. BAT protects against the major causes of impaired healthful longevity, i.e., obesity, diabetes, cardiovascular disorders, cancer, Alzheimer's disease, reduced exercise tolerance, and impaired blood flow. Several genetically engineered mouse models have shown that BAT enhances healthful aging and that their BAT is more potent than wild-type (WT) BAT. For example, when BAT, which increases longevity and exercise performance in mice with disruption of the regulator of G protein signaling 14 (RGS14), is transplanted to WT mice, their exercise capacity is enhanced at 3 days after BAT transplantation, whereas BAT transplantation from WT to WT mice also resulted in increased exercise performance, but only at 8 weeks after transplantation. In view of the ability of BAT to mediate healthful longevity, it is likely that a pharmaceutical analog of BAT will become a novel therapeutic modality.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Cell Biology and Molecular Medicine, Rutgers, New Jersey Medical School, Newark, NJ 07103, USA
| | - Berhanu Geresu Kibret
- Department of Cell Biology and Molecular Medicine, Rutgers, New Jersey Medical School, Newark, NJ 07103, USA
| | - Dorothy E. Vatner
- Department of Medicine, Rutgers, New Jersey Medical School, Newark, NJ 07103, USA
| | - Stephen F. Vatner
- Department of Cell Biology and Molecular Medicine, Rutgers, New Jersey Medical School, Newark, NJ 07103, USA
| |
Collapse
|
37
|
Liang D, Li G. Pulling the trigger: Noncoding RNAs in white adipose tissue browning. Rev Endocr Metab Disord 2024; 25:399-420. [PMID: 38157150 DOI: 10.1007/s11154-023-09866-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/11/2023] [Indexed: 01/03/2024]
Abstract
White adipose tissue (WAT) serves as the primary site for energy storage and endocrine regulation in mammals, while brown adipose tissue (BAT) is specialized for thermogenesis and energy expenditure. The conversion of white adipocytes to brown-like fat cells, known as browning, has emerged as a promising therapeutic strategy for reversing obesity and its associated co-morbidities. Noncoding RNAs (ncRNAs) are a class of transcripts that do not encode proteins but exert regulatory functions on gene expression at various levels. Recent studies have shed light on the involvement of ncRNAs in adipose tissue development, differentiation, and function. In this review, we aim to summarize the current understanding of ncRNAs in adipose biology, with a focus on their role and intricate mechanisms in WAT browning. Also, we discuss the potential applications and challenges of ncRNA-based therapies for overweight and its metabolic disorders, so as to combat the obesity epidemic in the future.
Collapse
Affiliation(s)
- Dehuan Liang
- The Key Laboratory of Geriatrics, Institute of Geriatric Medicine, Beijing Institute of Geriatrics, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, People's Republic of China
- Fifth School of Clinical Medicine (Beijing Hospital), Peking University, Beijing, 100730, People's Republic of China
| | - Guoping Li
- The Key Laboratory of Geriatrics, Institute of Geriatric Medicine, Beijing Institute of Geriatrics, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, People's Republic of China.
| |
Collapse
|
38
|
Ghesmati Z, Rashid M, Fayezi S, Gieseler F, Alizadeh E, Darabi M. An update on the secretory functions of brown, white, and beige adipose tissue: Towards therapeutic applications. Rev Endocr Metab Disord 2024; 25:279-308. [PMID: 38051471 PMCID: PMC10942928 DOI: 10.1007/s11154-023-09850-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/30/2023] [Indexed: 12/07/2023]
Abstract
Adipose tissue, including white adipose tissue (WAT), brown adipose tissue (BAT), and beige adipose tissue, is vital in modulating whole-body energy metabolism. While WAT primarily stores energy, BAT dissipates energy as heat for thermoregulation. Beige adipose tissue is a hybrid form of adipose tissue that shares characteristics with WAT and BAT. Dysregulation of adipose tissue metabolism is linked to various disorders, including obesity, type 2 diabetes, cardiovascular diseases, cancer, and infertility. Both brown and beige adipocytes secrete multiple molecules, such as batokines, packaged in extracellular vesicles or as soluble signaling molecules that play autocrine, paracrine, and endocrine roles. A greater understanding of the adipocyte secretome is essential for identifying novel molecular targets in treating metabolic disorders. Additionally, microRNAs show crucial roles in regulating adipose tissue differentiation and function, highlighting their potential as biomarkers for metabolic disorders. The browning of WAT has emerged as a promising therapeutic approach in treating obesity and associated metabolic disorders. Many browning agents have been identified, and nanotechnology-based drug delivery systems have been developed to enhance their efficacy. This review scrutinizes the characteristics of and differences between white, brown, and beige adipose tissues, the molecular mechanisms involved in the development of the adipocytes, the significant roles of batokines, and regulatory microRNAs active in different adipose tissues. Finally, the potential of WAT browning in treating obesity and atherosclerosis, the relationship of BAT with cancer and fertility disorders, and the crosstalk between adipose tissue with circadian system and circadian disorders are also investigated.
Collapse
Affiliation(s)
- Zeinab Ghesmati
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohsen Rashid
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shabnam Fayezi
- Department of Gynecologic Endocrinology and Fertility Disorders, Women's Hospital, Ruprecht-Karls University of Heidelberg, Heidelberg, Germany
| | - Frank Gieseler
- Division of Experimental Oncology, Department of Hematology and Oncology, University Medical Center Schleswig-Holstein, Campus Lübeck, 23538, Lübeck, Germany
| | - Effat Alizadeh
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Masoud Darabi
- Division of Experimental Oncology, Department of Hematology and Oncology, University Medical Center Schleswig-Holstein, Campus Lübeck, 23538, Lübeck, Germany.
| |
Collapse
|
39
|
Liu Y, Qian SW, Tang Y, Tang QQ. The secretory function of adipose tissues in metabolic regulation. LIFE METABOLISM 2024; 3:loae003. [PMID: 39872218 PMCID: PMC11748999 DOI: 10.1093/lifemeta/loae003] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/04/2024] [Accepted: 01/19/2024] [Indexed: 01/30/2025]
Abstract
In addition to their pivotal roles in energy storage and expenditure, adipose tissues play a crucial part in the secretion of bioactive molecules, including peptides, lipids, metabolites, and extracellular vesicles, in response to physiological stimulation and metabolic stress. These secretory factors, through autocrine and paracrine mechanisms, regulate various processes within adipose tissues. These processes include adipogenesis, glucose and lipid metabolism, inflammation, and adaptive thermogenesis, all of which are essential for the maintenance of the balance and functionality of the adipose tissue micro-environment. A subset of these adipose-derived secretory factors can enter the circulation and target the distant tissues to regulate appetite, cognitive function, energy expenditure, insulin secretion and sensitivity, gluconeogenesis, cardiovascular remodeling, and exercise capacity. In this review, we highlight the role of adipose-derived secretory factors and their signaling pathways in modulating metabolic homeostasis. Furthermore, we delve into the alterations in both the content and secretion processes of these factors under various physiological and pathological conditions, shedding light on potential pharmacological treatment strategies for related diseases.
Collapse
Affiliation(s)
- Yang Liu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Shu-Wen Qian
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yan Tang
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qi-Qun Tang
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
40
|
Guo L, Zhou L, Wei P, Li S, He S, Li D. Emerging Roles of UDP-GalNAc Polypeptide N-Acetylgalactosaminyltransferases in Cardiovascular Disease. Aging Dis 2024; 16:AD.2024.0308. [PMID: 38502587 PMCID: PMC11745429 DOI: 10.14336/ad.2024.0308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 03/08/2024] [Indexed: 03/21/2024] Open
Abstract
UDP-GalNAc polypeptide N-acetylgalactosaminyltransferases (GalNAc-Ts) catalyze mucin-type O-glycosylation by transferring α-N-acetylgalactosamine (GalNAc) from UDP-GalNAc to Ser or Thr residues of target proteins. This post-translational modification is common in eukaryotes, yet its biological functions remain unclear. Recent studies have identified specific receptors in the heart and vascular wall cells that can be mucin-type O-glycosylated, and there is now substantial evidence confirming that patients with various cardiovascular diseases (CVDs), such as heart failure, coronary artery disease, myocardial hypertrophy, and vascular calcification, exhibit abnormal changes in GalNAc-Ts. This review aims to highlight recent advances in GalNAc-Ts and their roles in the cardiovascular system, intending to provide evidence for clinical treatment and prevention of CVDs.
Collapse
Affiliation(s)
- Liwei Guo
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China.
- Xinxiang Key Laboratory of Metabolism and Integrative Physiology, Xinxiang, Henan, China.
| | - Lulu Zhou
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China.
| | - Pengcheng Wei
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China.
| | - Shijie Li
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China.
| | - Shanqing He
- Department of Cardiovascular Medicine, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Duan Li
- School of Basic Medicine, Xinxiang Medical University, Xinxiang, Henan, China.
- Henan Key Biological of Biological Psychiatry, Xinxiang Medical University, Xinxiang, Henan, China.
| |
Collapse
|
41
|
Chen J, Zheng H, Wu X, Niu X, Dai Y, Zhou Z, Ye F. Neuregulin 1 as a potential biomarker for disease progression in moyamoya disease: A case-control study in Chinese population. J Stroke Cerebrovasc Dis 2024; 33:107581. [PMID: 38224792 DOI: 10.1016/j.jstrokecerebrovasdis.2024.107581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 01/07/2024] [Accepted: 01/12/2024] [Indexed: 01/17/2024] Open
Abstract
OBJECTIVE Moyamoya disease (MMD) is a rare and progressive stenosis of cerebral arteries characterized by abnormally proliferative vasculopathy. Current studies have demonstrated that Neuregulin 1 (NRG1) plays a key role in angiogenesis-related disorders. Thus, the aim of our study is to investigate the serum NRG1 levels and their clinical correlations in MMD patients. METHODS In this study, thirty adult patients with MMD and age-gender matched healthy controls were enrolled from our hospital between July 2020 and April 2022. Peripheral blood samples were collected at baseline, and clinical data were obtained from the electronic medical record system. Serum NRG1 concentrations were measured by enzyme-linked immunosorbent assay. Sanger sequencing was applied to detect the RNF213 p.R4810K mutation. RESULTS The serum NRG1 levels were significantly higher in MMD patients compared to controls (14.48 ± 10.81 vs.7.54 ± 6.35mmol/L, p < 0.001). No statistical difference in baseline clinical characteristics was found between both groups. Correlation analyses showed that NRG1 levels were positively associated with Suzuki staging (r = 0.4137, p = 0.023) while not related to other clinical features (reduced cerebral blood flow, posterior cerebral artery involvement, bilateral or unilateral steno-occlusive changes). Furthermore, subgroup analysis revealed that MMD patients with the RNF213 p.R4810K mutation presented with significantly higher NRG1 levels than those without the mutation (9.60 ± 0.929 vs. 25.89 ± 4.338 mmol/L, p = 0.001). CONCLUSIONS Our study suggests that increased serum NRG1 levels may constitute a characteristic feature of MMD, indicating a potential positive correlation with disease progression and the presence of the RNF213 mutation. This positions NRG1 as a potentially crucial target for further studies aimed at comprehending the pathogenesis of MMD.
Collapse
Affiliation(s)
- Jie Chen
- Department of Neurology and Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hanyue Zheng
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoxin Wu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xingyang Niu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuanyuan Dai
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhenhua Zhou
- Department of Neurology and Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
| | - Fei Ye
- Department of Neurology and Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Neuroscience and Behavioral Disorders Program, Duke-NUS Medical School, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
42
|
Jin T, Wang H, Liu Y, Wang H. Circular RNAs: Regulators of endothelial cell dysfunction in atherosclerosis. J Mol Med (Berl) 2024; 102:313-335. [PMID: 38265445 DOI: 10.1007/s00109-023-02413-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 12/09/2023] [Accepted: 12/21/2023] [Indexed: 01/25/2024]
Abstract
Endothelial cell (EC) dysfunction is associated with atherosclerosis. Circular RNAs (circRNAs) are covalently closed loops formed by back-splicing, are highly expressed in a tissue-specific or cell-specific manner, and regulate ECs mainly through miRNAs (mircoRNAs) or protein sponges. This review describes the regulatory mechanisms and physiological functions of circRNAs, as well as the differential expression of circRNAs in aberrant ECs. This review focuses on their roles in inflammation, proliferation, migration, angiogenesis, apoptosis, senescence, and autophagy in ECs from the perspective of signaling pathways, such as nuclear factor κB (NF-κB), nucleotide-binding domain, leucine-rich-repeat family, pyrin-domain-containing 3 (NLRP3)/caspase-1, Janus kinase/signal transducer and activator of transcription (JAK/STAT), and phosphoinositide-3 kinase/protein kinase B (PI3K/Akt). Finally, we address the issues and recent advances in circRNAs as well as circRNA-mediated regulation of ECs to improve our understanding of the molecular mechanisms underlying the progression of atherosclerosis and provide a reference for studies on circRNAs that regulate EC dysfunction and thus affect atherosclerosis.
Collapse
Affiliation(s)
- Tengyu Jin
- Hebei Medical University, Shijiazhuang 050011, Hebei, China
- Hebei General Hospital, Affiliated to Hebei Medical University, Shijiazhuang 050051, Hebei, China
| | - Haoyuan Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Yuelin Liu
- Hebei Medical University, Shijiazhuang 050011, Hebei, China
| | - Hebo Wang
- Hebei Medical University, Shijiazhuang 050011, Hebei, China.
- Hebei General Hospital, Affiliated to Hebei Medical University, Shijiazhuang 050051, Hebei, China.
- Hebei Provincial Key Laboratory of Cerebral Networks and Cognitive Disorders, Shijiazhuang 050051, Hebei, China.
| |
Collapse
|
43
|
Chen M, Zhu J, Luo H, Mu W, Guo L. The journey towards physiology and pathology: Tracing the path of neuregulin 4. Genes Dis 2024; 11:687-700. [PMID: 37692526 PMCID: PMC10491916 DOI: 10.1016/j.gendis.2023.03.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 02/11/2023] [Accepted: 03/05/2023] [Indexed: 09/12/2023] Open
Abstract
Neuregulin 4 (Nrg4), an epidermal growth factor (EGF) family member, can bind to and activate the ErbB4 receptor tyrosine kinase. Nrg4 has five different isoforms by alternative splicing and performs a wide variety of functions. Nrg4 is involved in a spectrum of physiological processes including neurobiogenesis, lipid metabolism, glucose metabolism, thermogenesis, and angiogenesis. In pathological processes, Nrg4 inhibits inflammatory factor levels and suppresses apoptosis in inflammatory diseases. In addition, Nrg4 could ameliorate obesity, insulin resistance, and cardiovascular diseases. Furthermore, Nrg4 improves non-alcoholic fatty liver disease (NAFLD) by promoting autophagy, improving lipid metabolism, and inhibiting cell death of hepatocytes. Besides, Nrg4 is closely related to the development of cancer, hyperthyroidism, and some other diseases. Therefore, elucidation of the functional role and mechanisms of Nrg4 will provide a clearer view of the therapeutic potential and possible risks of Nrg4.
Collapse
Affiliation(s)
- Min Chen
- School of Exercise and Health and Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China
| | - Jieying Zhu
- School of Exercise and Health and Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China
| | - Hongyang Luo
- School of Exercise and Health and Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China
| | - Wangjing Mu
- School of Exercise and Health and Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China
| | - Liang Guo
- School of Exercise and Health and Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China
| |
Collapse
|
44
|
Chen Z, Zhang P, Liu T, Qiu X, Li S, Lin JD. Neuregulin 4 mediates the metabolic benefits of mild cold exposure by promoting beige fat thermogenesis. JCI Insight 2024; 9:e172957. [PMID: 38015639 PMCID: PMC10906454 DOI: 10.1172/jci.insight.172957] [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: 06/12/2023] [Accepted: 11/21/2023] [Indexed: 11/30/2023] Open
Abstract
Interorgan crosstalk via secreted hormones and metabolites is a fundamental aspect of mammalian metabolic physiology. Beyond the highly specialized endocrine cells, peripheral tissues are emerging as an important source of metabolic hormones that influence energy and nutrient metabolism and contribute to disease pathogenesis. Neuregulin 4 (Nrg4) is a fat-derived hormone that protects mice from nonalcoholic steatohepatitis (NASH) and NASH-associated liver cancer by shaping hepatic lipid metabolism and the liver immune microenvironment. Despite its enriched expression in brown fat, whether NRG4 plays a role in thermogenic response and mediates the metabolic benefits of cold exposure are areas that remain unexplored. Here we show that Nrg4 expression in inguinal white adipose tissue (iWAT) is highly responsive to chronic cold exposure. Nrg4 deficiency impairs beige fat induction and renders mice more susceptible to diet-induced metabolic disorders under mild cold conditions. Using mice with adipocyte and hepatocyte-specific Nrg4 deletion, we reveal that adipose tissue-derived NRG4, but not hepatic NRG4, is essential for beige fat induction following cold acclimation. Furthermore, treatment with recombinant NRG4-Fc fusion protein promotes beige fat induction in iWAT and improves metabolic health in mice with diet-induced obesity. These findings highlight a critical role of NRG4 in mediating beige fat induction and preserving metabolic health under mild cold conditions.
Collapse
|
45
|
Xu J, Ma H, Shi L, Zhou H, Cheng Y, Tong J, Meng B, Xu X, He K, Ding S, Zhang J, Yue L, Xiang G. Inflammatory Cell-Derived MYDGF Attenuates Endothelial LDL Transcytosis to Protect Against Atherogenesis. Arterioscler Thromb Vasc Biol 2023; 43:e443-e467. [PMID: 37767706 DOI: 10.1161/atvbaha.123.319905] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND Inflammation contributes to the pathogenesis of atherosclerosis. But little is known about the potential benefits of inflammatory cells to atherosclerosis. The aim of this study was to investigate the function of inflammatory cells/endothelium axis and determine whether and how inflammatory cell-derived MYDGF (myeloid-derived growth factor) inhibited endothelial LDL (low-density lipoprotein) transcytosis. METHODS In in vivo experiments, both loss- and gain-of-function strategies were used to evaluate the effect of inflammatory cell-derived MYDGF on LDL transcytosis. We generated monocyte/macrophage-targeted MYDGF-null mice on an Ldlr (LDL receptor)-/- background in the loss-of-function strategy and restored the inflammatory cell-derived MYDGF by bone marrow transplantation and inflammatory cell-specific overexpression of MYDGF mice model in the gain-of-function strategy. In in vitro experiments, coculture experiments between primary mouse aortic endothelial cells and macrophages and mouse aortic endothelial cells supplemented with or without recombinant MYDGF were conducted. RESULTS Inflammatory cell-derived MYDGF deficiency aggravated endothelial LDL transcytosis, drove LDL uptake by artery wall, and thus exacerbated atherosclerosis in vivo. Inflammatory cell-derived MYDGF restoration by bone marrow transplantation and inflammatory cell MYDGF overexpression alleviated LDL transport across the endothelium, prevented LDL accumulation in the subendothelial space, and subsequently ameliorated atherosclerosis in vivo. Furthermore, in the in vitro study, macrophages isolated from MYDGF+/+ mice and recombinant MYDGF attenuated LDL transcytosis and uptake in mouse aortic endothelial cells. Mechanistically, MYDGF inhibited MAP4K4 (mitogen-activated protein kinase kinase kinase kinase isoform 4) phosphorylation, enhanced activation of Akt (protein kinase B)-1, and diminished the FoxO (forkhead box O) 3a signaling cascade to exert protective effects of MYDGF on LDL transcytosis and atherosclerosis. CONCLUSIONS The findings support a role for inflammatory cell-derived MYDGF served as a cross talk factor between inflammatory cells and endothelial cells that inhibits LDL transcytosis across endothelium. MYDGF may become a novel therapeutic drug for atherosclerosis, and the beneficial effects of inflammatory cell in atherosclerosis deserve further attention.
Collapse
Affiliation(s)
- Jinling Xu
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, China (J.X., L.S., Y.C., J.T., B.M., X.X., J.Z., L.Y., G.X.)
- The First School of Clinical Medicine, Southern Medical University, Guangdong, China (J.X., L.S., Y.C., J.T., K.H., S.D., G.X.)
| | - Huaxing Ma
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, China (H.M.)
| | - Lingfeng Shi
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, China (J.X., L.S., Y.C., J.T., B.M., X.X., J.Z., L.Y., G.X.)
- The First School of Clinical Medicine, Southern Medical University, Guangdong, China (J.X., L.S., Y.C., J.T., K.H., S.D., G.X.)
| | - Hui Zhou
- Department of General Surgery, The Third Xiangya Hospital, Central South University, Hunan, China (H.Z.)
| | - Yangyang Cheng
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, China (J.X., L.S., Y.C., J.T., B.M., X.X., J.Z., L.Y., G.X.)
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, China (H.M.)
| | - Jiayue Tong
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, China (J.X., L.S., Y.C., J.T., B.M., X.X., J.Z., L.Y., G.X.)
- The First School of Clinical Medicine, Southern Medical University, Guangdong, China (J.X., L.S., Y.C., J.T., K.H., S.D., G.X.)
| | - Biying Meng
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, China (J.X., L.S., Y.C., J.T., B.M., X.X., J.Z., L.Y., G.X.)
| | - Xiaoli Xu
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, China (J.X., L.S., Y.C., J.T., B.M., X.X., J.Z., L.Y., G.X.)
| | - Kaiyue He
- The First School of Clinical Medicine, Southern Medical University, Guangdong, China (J.X., L.S., Y.C., J.T., K.H., S.D., G.X.)
| | - Sheng Ding
- The First School of Clinical Medicine, Southern Medical University, Guangdong, China (J.X., L.S., Y.C., J.T., K.H., S.D., G.X.)
| | - Jiajia Zhang
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, China (J.X., L.S., Y.C., J.T., B.M., X.X., J.Z., L.Y., G.X.)
| | - Ling Yue
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, China (J.X., L.S., Y.C., J.T., B.M., X.X., J.Z., L.Y., G.X.)
| | - Guangda Xiang
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, China (J.X., L.S., Y.C., J.T., B.M., X.X., J.Z., L.Y., G.X.)
- The First School of Clinical Medicine, Southern Medical University, Guangdong, China (J.X., L.S., Y.C., J.T., K.H., S.D., G.X.)
| |
Collapse
|
46
|
Li C, Cui Z, Deng S, Lei T, Chen P, Yang H. Programmed Cell Death Protein 2-like Promotes Inflammation and Oxidative Stress in Vascular Endothelial Cells. ACS Pharmacol Transl Sci 2023; 6:1453-1470. [PMID: 37854614 PMCID: PMC10580389 DOI: 10.1021/acsptsci.3c00129] [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: 06/25/2023] [Indexed: 10/20/2023]
Abstract
Programmed cell death protein 2-like (PDCD2L) is a shuttle protein of the nucleus and cytoplasm and is related to the ribosome biogenesis. However, there are few reports on the relationship between PDCD2L and inflammation, and the exact relationship between PDCD2L and inflammation has not been determined in vascular endothelial cells yet. Accordingly, we focus on exploring the relationship between PDCD2L and inflammation and its potential mechanisms. Our research findings suggested that PDCD2L is a proinflammatory target. The result showed that, by interfering with the expression of PDCD2L, LPS-induced inflammation of vascular endothelial cells can be reduced, such as IL-6 and IL-1β, as well as the adhesion factor ICAM1. Meanwhile, overexpression of PDCD2L can further increase LPS-induced inflammation levels, ICAM1, and ROS production, reduce CAT, GSH/GSSG levels, and increase SOD levels. Therefore, we determined that PDCD2L has a regulatory effect on inflammation and oxidative stress of vascular endothelial cells, and its regulatory mechanism may be related to inflammatory transcription factors STAT1, NF-κB regulation, transport of inflammatory messenger mRNA, and ribosome biogenesis. Then, we screened that andrographolide (Andro) can bind to PDCD2L, thus inhibiting inflammation and endothelial cell adhesion caused by the overexpression of PDCD2L. This study reveals that PDCD2L is a potential anti-inflammatory therapeutic target, providing new exploration for the development of anti-inflammatory drugs.
Collapse
Affiliation(s)
- Caifeng Li
- Beijing
Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention
and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Zhao Cui
- Institute
of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Shiwen Deng
- Beijing
Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention
and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Tong Lei
- Beijing
Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention
and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Peng Chen
- Experimental
Research Center, China Academy of Chinese
Medical Sciences, Beijing 100700, China
- Robot
Intelligent Laboratory of Traditional Chinese Medicine, Experimental
Research Center & MEGAROBO, China Academy
of Chinese Medical Sciences, Beijing 100700, China
- Hunan
Provincial Key Laboratory of Complex Effects Analysis for Chinese
Patent Medicine, Yongzhou, Hunan Province 425199, China
| | - Hongjun Yang
- Beijing
Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention
and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| |
Collapse
|
47
|
Luo J, He Z, Li Q, Lv M, Cai Y, Ke W, Niu X, Zhang Z. Adipokines in atherosclerosis: unraveling complex roles. Front Cardiovasc Med 2023; 10:1235953. [PMID: 37645520 PMCID: PMC10461402 DOI: 10.3389/fcvm.2023.1235953] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/02/2023] [Indexed: 08/31/2023] Open
Abstract
Adipokines are biologically active factors secreted by adipose tissue that act on local and distant tissues through autocrine, paracrine, and endocrine mechanisms. However, adipokines are believed to be involved in an increased risk of atherosclerosis. Classical adipokines include leptin, adiponectin, and ceramide, while newly identified adipokines include visceral adipose tissue-derived serpin, omentin, and asprosin. New evidence suggests that adipokines can play an essential role in atherosclerosis progression and regression. Here, we summarize the complex roles of various adipokines in atherosclerosis lesions. Representative protective adipokines include adiponectin and neuregulin 4; deteriorating adipokines include leptin, resistin, thrombospondin-1, and C1q/tumor necrosis factor-related protein 5; and adipokines with dual protective and deteriorating effects include C1q/tumor necrosis factor-related protein 1 and C1q/tumor necrosis factor-related protein 3; and adipose tissue-derived bioactive materials include sphingosine-1-phosphate, ceramide, and adipose tissue-derived exosomes. However, the role of a newly discovered adipokine, asprosin, in atherosclerosis remains unclear. This article reviews progress in the research on the effects of adipokines in atherosclerosis and how they may be regulated to halt its progression.
Collapse
Affiliation(s)
- Jiaying Luo
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhiwei He
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qingwen Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Mengna Lv
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuli Cai
- Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Ke
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xuan Niu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhaohui Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
48
|
Adachi Y, Ueda K, Takimoto E. Perivascular adipose tissue in vascular pathologies-a novel therapeutic target for atherosclerotic disease? Front Cardiovasc Med 2023; 10:1151717. [PMID: 37304960 PMCID: PMC10250715 DOI: 10.3389/fcvm.2023.1151717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/28/2023] [Indexed: 06/13/2023] Open
Abstract
Most blood vessels are surrounded by adipose tissues known as perivascular adipose tissue (PVAT). Emerging experimental data have implicated the potential involvement of PVAT in the pathogenesis of cardiovascular disease: PVAT might be a source of inflammatory mediators under pathological conditions such as metabolic disorders, chronic inflammation, and aging, leading to vascular pathologies, while having vasculo-protective roles in a healthy state. PVAT has been also gaining attention in human disease conditions. Recent integrative omics approaches have greatly enhanced our understanding of the molecular mechanisms underlying the diverse functions of PVAT. This review summarizes recent progress in PVAT research and discusses the potential of PVAT as a target for the treatment of atherosclerosis.
Collapse
|
49
|
AlZaim I, Kalucka J. Batokine neuregulin 4 promotes atherosclerotic resolution. Nat Metab 2022; 4:1440-1441. [PMID: 36400934 DOI: 10.1038/s42255-022-00669-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Ibrahim AlZaim
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Joanna Kalucka
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark.
| |
Collapse
|