1
|
Chen XH, Li L, Ma SY, Ling CQ, Lai JQ, Yao HJ, Guo WB, Yang Y, Zhang Y, Lin WJ, He FZ. Arginase 1 genetic variation is associated with the risk of vascular complications in type 2 diabetes. Biomark Med 2025; 19:63-72. [PMID: 39885775 DOI: 10.1080/17520363.2025.2455924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/16/2025] [Indexed: 02/01/2025] Open
Abstract
OBJECTIVE This study aims to explore the association between arginase 1 (ARG1) genetic variation and susceptibility to type 2 diabetes (T2DM) vascular complications, a primary cause of morbidity and mortality in diabetics. METHODS ARG1, a risk gene for cardiovascular disease, was identified from GEO datasets GSE22255 and GSE58294. The ENCODE database identified four candidate single-nucleotide polymorphism (SNP) loci. Nine hundred ninety-two T2DM patients underwent SNP genotyping, and relevant biochemical markers were tested. Logistic regression analysis calculated the odds ratio (OR) and 95% confidence interval (CI) between ARG1 SNP and diabetic vascular complications. RESULTS Out of 985 patients, 250 had CHD, with the TTTG group accounting for 64/250. This group showed a significant reduction in CHD risk (non-TTTG factor-adjusted OR = 1.61, 95% CI: 1.14-2.29, p = 0.008). The combination of the TTTG group, age, central obesity, and hypertension better predicted CHD risk (Area under the curve = 0.72, p < 0.001). CONCLUSIONS ARG1 polymorphisms significantly impact vascular complications in T2DM patients, implying that ARG1 genetic variation may be a potential prevention and treatment target. TRIAL REGISTRATION ChiCTR1800015661.
Collapse
Affiliation(s)
- X H Chen
- College of Pharmacy, Jinan University, Guangzhou, China
- Department of Pharmacy, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| | - L Li
- Department of Pharmacy, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - S Y Ma
- Department of Pharmacy, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - C Q Ling
- Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| | - J Q Lai
- College of Pharmacy, Jinan University, Guangzhou, China
- Department of Pharmacy, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| | - H J Yao
- College of Pharmacy, Jinan University, Guangzhou, China
- Department of Pharmacy, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| | - W B Guo
- The 3rd People's Hospital of Yuxi City, Yuxi, China
| | - Y Yang
- Department of Pharmacy, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| | - Y Zhang
- Department of Pharmacy, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| | - W J Lin
- Department of Pharmacy, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| | - F Z He
- Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| |
Collapse
|
2
|
Molaro MC, Battisegola C, Schiano ME, Failla M, Rimoli MG, Lazzarato L, Chegaev K, Sodano F. Synthesis of Arginase Inhibitors: An Overview. Pharmaceutics 2025; 17:117. [PMID: 39861764 DOI: 10.3390/pharmaceutics17010117] [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: 12/17/2024] [Revised: 01/12/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Arginase (ARG) is a binuclear manganese-containing metalloenzyme that can convert L-arginine to L-ornithine and urea and plays a key role in the urea cycle. It also mediates different cellular functions and processes such as proliferation, senescence, apoptosis, autophagy, and inflammatory responses in various cell types. In mammals, there are two isoenzymes, ARG-1 and ARG-2; they are functionally similar, but their coding genes, tissue distribution, subcellular localization, and molecular regulation are distinct. In recent decades, the abnormal expression of ARG-1 or ARG-2 has been reported to be increasingly linked to a variety of diseases, including cardiovascular disease, inflammatory bowel disease, Alzheimer's disease, and cancer. Therefore, considering the current relevance of this topic and the need to address the growing demand for new and more potent ARG inhibitors in the context of various diseases, this review was conceived. We will provide an overview of all classes of ARG inhibitors developed so far including compounds of synthetic, natural, and semisynthetic origin. For the first time, the synthesis protocol and optimized reaction conditions of each molecule, including those reported in patent applications, will be described. For each molecule, its inhibitory activity in terms of IC50 towards ARG-1 and ARG-2 will be reported specifying the type of assay conducted.
Collapse
Affiliation(s)
| | - Chiara Battisegola
- Department of Pharmacy, "Federico II" University of Naples, 80131 Naples, Italy
| | | | - Mariacristina Failla
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | - Maria Grazia Rimoli
- Department of Pharmacy, "Federico II" University of Naples, 80131 Naples, Italy
| | - Loretta Lazzarato
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | - Konstantin Chegaev
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | - Federica Sodano
- Department of Pharmacy, "Federico II" University of Naples, 80131 Naples, Italy
| |
Collapse
|
3
|
Lin L, Zhong S, Zhou Y, Xia J, Deng S, Jiang T, Jiang A, Huang Z, Wang J. Dapagliflozin improves the dysfunction of human umbilical vein endothelial cells (HUVECs) by downregulating high glucose/high fat-induced autophagy through inhibiting SGLT-2. J Diabetes Complications 2025; 39:108907. [PMID: 39580877 DOI: 10.1016/j.jdiacomp.2024.108907] [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: 06/01/2024] [Revised: 11/10/2024] [Accepted: 11/10/2024] [Indexed: 11/26/2024]
Abstract
OBJECTIVE To investigate the effect of Dapagliflozin (Da) on the disorders of human umbilical vein endothelial cells (HUVECs) induced by high glucose and high fat (HG/HF). METHODS Immunohistochemistry and immunofluorescence were used to detect the SGLT-2 expression in thoracic aortic tissues. After transfected with overexpressed plasmid SLC5A2, autophagy and cell functions of HUVECs were detected with the treatment of autophagy inhibitor 3-MA (5 mM). HUVECs were exposed to mannitol (MAN), glucose/palmitate (Hg/PA), and Hg/PA/Da for 24 h, and the proliferation of HUVECs was detected by CCK-8. The protein expression levels, endothelial cell functions (cell proliferation, migration, tubular formation, apoptosis, and autophagy) in endothelial cells were evaluated. RESULTS The SGLT-2 expression was found in atherosclerotic human thoracic aorta tissues and HG/PA induced HUVECs (P < 0.05). After the overexpression of SGLT-2 in HUVECs, the proliferation, migration and tubule formation ability of HUVECs were inhibited, and autophagy and apoptosis were increased, which were reversed by 3-MA (P < 0.05). After the addition of Sodium-glucose co-transporters 2 inhibitor, Dapagliflozin, the proliferation of HUVECs, the tubule formation, autophagy, apoptosis and migration ability of cells inhibited by HG/PA were significantly improved (P < 0.05). Moreover, the increased protein expression levels of autophagy and apoptosis in HG/PA induced HUVECs were also decreased by the treatment of Dapagliflozin (P < 0.05). CONCLUSIONS Dapagliflozin can improve the dysfunction of high glucose/high fat-induced human umbilical vein endothelial cells by downregulate autophagy through inhibiting SGLT-2.
Collapse
Affiliation(s)
- Lijiahui Lin
- Department of Endocrinology The Second Affiliated Hospital of the University of South China, Hengyang, Hunan 421001, China; Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde, Hunan 415000, China
| | - Siyu Zhong
- Department of Endocrinology The Second Affiliated Hospital of the University of South China, Hengyang, Hunan 421001, China
| | - Ying Zhou
- Department of Endocrinology The Second Affiliated Hospital of the University of South China, Hengyang, Hunan 421001, China
| | - Jie Xia
- Department of Endocrinology The Second Affiliated Hospital of the University of South China, Hengyang, Hunan 421001, China
| | - Shanshan Deng
- Department of Endocrinology The Second Affiliated Hospital of the University of South China, Hengyang, Hunan 421001, China
| | - Tao Jiang
- Department of Endocrinology The Second Affiliated Hospital of the University of South China, Hengyang, Hunan 421001, China
| | - Aihua Jiang
- Department of Endocrinology The Second Affiliated Hospital of the University of South China, Hengyang, Hunan 421001, China.
| | - Zhimei Huang
- Department of Endocrinology The Second Affiliated Hospital of the University of South China, Hengyang, Hunan 421001, China
| | - Jianping Wang
- Department of Endocrinology The Second Affiliated Hospital of the University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
4
|
Failla M, Molaro MC, Schiano ME, Serafini M, Tiburtini GA, Gianquinto E, Scoccia R, Battisegola C, Rimoli MG, Chegaev K, Ercolano G, Lazzarato L, Spyrakis F, Sodano F. Opportunities and Challenges of Arginase Inhibitors in Cancer: A Medicinal Chemistry Perspective. J Med Chem 2024; 67:19988-20021. [PMID: 39558532 DOI: 10.1021/acs.jmedchem.4c01429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2024]
Abstract
The overexpression of two arginase (ARG) isoforms, ARG1 and ARG2, contributes to the onset of numerous disorders, including cardiovascular and immune-mediated diseases, as well as tumors. To elucidate the specific roles of ARG1 and ARG2 without interfering with their physiological functions, it is crucial to develop effective ARG inhibitors that target only one isoform, while maintaining low toxicity and an adequate pharmacokinetic profile. In this context, we present a comprehensive overview of the different generations of ARG inhibitors. Given the general lack of selectivity in most existing inhibitors, we analyzed the structural features and plasticity of the ARG1 and ARG2 binding sites to explore the potential for designing inhibitors with novel binding patterns. We also review ongoing preclinical and clinical studies on selected inhibitors, highlighting both progress and challenges in developing potent, selective ARG inhibitors. Furthermore, we discuss medicinal chemistry strategies that may accelerate the discovery of selective ARG inhibitors.
Collapse
Affiliation(s)
- Mariacristina Failla
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | | | | | - Marta Serafini
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | | | - Eleonora Gianquinto
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | - Riccardo Scoccia
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | - Chiara Battisegola
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Maria Grazia Rimoli
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Konstantin Chegaev
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | - Giuseppe Ercolano
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Loretta Lazzarato
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | - Francesca Spyrakis
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | - Federica Sodano
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
5
|
Jiang H, Zhang C, Lin M, Yin Y, Deng S, Liu W, Zhuo B, Tian G, Du Y, Meng Z. Deciphering the mechanistic impact of acupuncture on the neurovascular unit in acute ischemic stroke: Insights from basic research in a narrative review. Ageing Res Rev 2024; 101:102536. [PMID: 39384155 DOI: 10.1016/j.arr.2024.102536] [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: 08/18/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/11/2024]
Abstract
Ischemic stroke(IS), a severe acute cerebrovascular disease, not only imposes a heavy economic burden on society but also presents numerous challenges in treatment. During the acute phase, while thrombolysis and thrombectomy serve as primary treatments, these approaches are restricted by a narrow therapeutic window. During rehabilitation, commonly used neuroprotective agents struggle with their low drug delivery efficiency and inadequate preclinical testing, and the long-term pharmacological and toxicity effects of nanomedicines remain undefined. Meanwhile, acupuncture as a therapeutic approach is widely acknowledged for its effectiveness in treating IS and has been recommended by the World Health Organization (WHO) as an alternative and complementary therapy, even though its exact mechanisms remain unclear. This review aims to summarize the known mechanisms of acupuncture and electroacupuncture (EA) in the treatment of IS. Research shows that acupuncture treatment mainly protects the neurovascular unit through five mechanisms: 1) reducing neuronal apoptosis and promoting neuronal repair and proliferation; 2) maintaining the integrity of the blood-brain barrier (BBB); 3) inhibiting the overactivation and polarization imbalance of microglia; 4) regulating the movement of vascular smooth muscle (VSM) cells; 5) promoting the proliferation of oligodendrocyte precursors. Through an in-depth analysis, this review reveals the multi-level, multi-dimensional impact of acupuncture treatment on the neurovascular unit (NVU) following IS, providing stronger evidence and a theoretical basis for its clinical application.
Collapse
Affiliation(s)
- Hailun Jiang
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Chao Zhang
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Mengxuan Lin
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Yu Yin
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shizhe Deng
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Wei Liu
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Bifang Zhuo
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Guang Tian
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Yuzheng Du
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| | - Zhihong Meng
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| |
Collapse
|
6
|
Heuser SK, Li J, Pudewell S, LoBue A, Li Z, Cortese-Krott MM. Biochemistry, pharmacology and in vivo function of arginases. Pharmacol Rev 2024; 77:PHARMREV-AR-2024-001271. [PMID: 39406506 DOI: 10.1124/pharmrev.124.001271] [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: 06/03/2024] [Revised: 09/24/2024] [Accepted: 10/07/2024] [Indexed: 01/22/2025] Open
Abstract
Arginase catalyzes the hydrolysis of L-arginine into L-ornithine and urea. The two existing isoforms Arg1 and Arg2 show different cellular localizations and metabolic functions. Arginase activity is crucial for nitrogen detoxification in the urea cycle, synthesis of polyamines, and control of l-arginine bioavailability and nitric oxide production. Despite significant progress in the understanding of the biochemistry and function of arginases, several open questions remain. Recent studies have revealed that the regulation and function of Arg1 and Arg2 are cell-type-specific, species-specific, and profoundly different in mice and humans. The main differences were found in the distribution and function of Arg1 and Arg2 in immune and erythroid cells. Contrary to what was previously thought, Arg1 activity appears to be only partially related to vascular NO signaling under homeostatic conditions in the vascular wall, but its expression is increased under disease conditions and may be targeted by treatment with arginase inhibitors. Arg2 appears to be mainly a catabolic enzyme involved in the synthesis of L-ornithine, polyamine, and proline but may play a putative role in blood pressure control, at least in mice. The immunosuppressive role of arginase-mediated arginine depletion is a promising target for cancer treatment. This review critically revises and discusses the biochemistry, pharmacology, and in vivo function of arginase, focusing on the insights gained from the analysis of cell-specific Arg1 and Arg2 knockout mice and human studies using arginase inhibitors or pegylated recombinant arginase. Significance Statement The review emphasizes the need for further research to deepen our understanding of the regulation of Arg1 and Arg 2 in different cell types under consideration of their localization, species-specificity, and multiple biochemical and physiological roles. This could lead to better pharmacological strategies to target arginase activity in liver, cardiovascular, hematological, immune/infection diseases and cancer.
Collapse
Affiliation(s)
- Sophia K Heuser
- Department of Cardiology, Pulmonology, and Angiology, University of Duesseldorf, Germany
| | - Junjie Li
- Department of Cardiology, Pulmonology, and Angiology, University of Duesseldorf, Germany
| | - Silke Pudewell
- Department of Biochemistry and Molecular Biology II, University of Duesseldorf, Germany
| | - Anthea LoBue
- Department of Cardiology, Pulmonology, and Angiology, University of Duesseldorf, Germany
| | - Zhixin Li
- Department of Cardiology, Pulmonology, and Angiology, University of Duesseldorf, Germany
| | - Miriam M Cortese-Krott
- Department of Cardiology, Pulmonology, and Angiology, University of Duesseldorf, Germany
| |
Collapse
|
7
|
Marzęta-Assas P, Jacenik D, Zasłona Z. Pathophysiology of Arginases in Cancer and Efforts in Their Pharmacological Inhibition. Int J Mol Sci 2024; 25:9782. [PMID: 39337272 PMCID: PMC11431790 DOI: 10.3390/ijms25189782] [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: 08/05/2024] [Revised: 09/02/2024] [Accepted: 09/05/2024] [Indexed: 09/30/2024] Open
Abstract
Arginases are key enzymes that hydrolyze L-arginine to urea and L-ornithine in the urea cycle. The two arginase isoforms, arginase 1 (ARG1) and arginase 2 (ARG2), regulate the proliferation of cancer cells, migration, and apoptosis; affect immunosuppression; and promote the synthesis of polyamines, leading to the development of cancer. Arginases also compete with nitric oxide synthase (NOS) for L-arginine, and their participation has also been confirmed in cardiovascular diseases, stroke, and inflammation. Due to the fact that arginases play a crucial role in the development of various types of diseases, finding an appropriate candidate to inhibit the activity of these enzymes would be beneficial for the therapy of many human diseases. In this review, based on numerous experimental, preclinical, and clinical studies, we provide a comprehensive overview of the biological and physiological functions of ARG1 and ARG2, their molecular mechanisms of action, and affected metabolic pathways. We summarize the recent clinical trials' advances in targeting arginases and describe potential future drugs.
Collapse
Affiliation(s)
| | - Damian Jacenik
- Molecure S.A., 101 Żwirki i Wigury St., 02-089 Warsaw, Poland
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, 141/143 Pomorska St., 90-236 Lodz, Poland
| | | |
Collapse
|
8
|
Liu B, Zeng H, Su H, Williams QA, Besanson J, Chen Y, Chen JX. Endothelial Cell-Specific Prolyl Hydroxylase-2 Deficiency Augments Angiotensin II-Induced Arterial Stiffness and Cardiac Pericyte Recruitment in Mice. J Am Heart Assoc 2024; 13:e035769. [PMID: 39056332 DOI: 10.1161/jaha.124.035769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 06/18/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND Endothelial prolyl hydroxylase-2 (PHD2) is essential for pulmonary remodeling and hypertension. In the present study, we investigated the role of endothelial PHD2 in angiotensin II-mediated arterial stiffness, pericyte recruitment, and cardiac fibrosis. METHODS AND RESULTS Chondroitin sulfate proteoglycan 4 tracing reporter chondroitin sulfate proteoglycan 4- red fluorescent protein (DsRed) transgenic mice were crossed with PHD2flox/flox (PHD2f/f) mice and endothelial-specific knockout of PHD2 (PHD2ECKO) mice. Transgenic PHD2f/f (TgPHD2f/f) mice and TgPHD2ECKO mice were infused with angiotensin II for 4 weeks. Arterial thickness, stiffness, and histological and immunofluorescence of pericytes and fibrosis were measured. Infusion of TgPHD2f/f mice with angiotensin II resulted in a time-dependent increase in pulse-wave velocity. Angiotensin II-induced pulse-wave velocity was further elevated in the TgPHD2ECKO mice. TgPHD2ECKO also reduced coronary flow reserve compared with TgPHD2f/f mice infused with angiotensin II. Mechanistically, knockout of endothelial PHD2 promoted aortic arginase activity and angiotensin II-induced aortic thickness together with increased transforming growth factor-β1 and ICAM-1/VCAM-1 expression in coronary arteries. TgPHD2f/f mice infused with angiotensin II for 4 weeks exhibited a significant increase in cardiac fibrosis and hypertrophy, which was further developed in the TgPHD2ECKO mice. Chondroitin sulfate proteoglycan 4 pericyte was traced by DsRed+ staining and angiotensin II infusion displayed a significant increase of DsRed+ pericytes in the heart, as well as a deficiency of endothelial PHD2, which further promoted angiotensin II-induced pericyte increase. DsRed+ pericytes were costained with fibroblast-specific protein 1 and α-smooth muscle actin for measuring pericyte-myofibroblast cell transition. The knockout of endothelial PHD2 increased the amount of DsRed+/fibroblast-specific protein 1+ and DsRed+/α-smooth muscle actin+ cells induced by angiotensin II infusion. CONCLUSIONS Knockout of endothelial PHD2 enhanced angiotensin II-induced cardiac fibrosis by mechanisms involving increasing arterial stiffness and pericyte-myofibroblast cell transitions.
Collapse
Affiliation(s)
- Bo Liu
- Department of Pharmacology and Toxicology University of Mississippi Medical Center, School of Medicine Jackson MS
| | - Heng Zeng
- Department of Pharmacology and Toxicology University of Mississippi Medical Center, School of Medicine Jackson MS
| | - Han Su
- Department of Pharmacology and Toxicology University of Mississippi Medical Center, School of Medicine Jackson MS
| | - Quinesha A Williams
- Department of Pharmacology and Toxicology University of Mississippi Medical Center, School of Medicine Jackson MS
| | - Jessie Besanson
- Department of Pharmacology and Toxicology University of Mississippi Medical Center, School of Medicine Jackson MS
| | - Yingjie Chen
- Department of Physiology and Biophysics University of Mississippi Medical Center, School of Medicine Jackson MS
| | - Jian-Xiong Chen
- Department of Pharmacology and Toxicology University of Mississippi Medical Center, School of Medicine Jackson MS
| |
Collapse
|
9
|
Tengbom J, Kontidou E, Collado A, Yang J, Alvarsson M, Brinck J, Rössner S, Zhou Z, Pernow J, Mahdi A. Differences in endothelial function between patients with Type 1 and Type 2 diabetes: effects of red blood cells and arginase. Clin Sci (Lond) 2024; 138:975-985. [PMID: 39037711 DOI: 10.1042/cs20240447] [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: 03/12/2024] [Revised: 06/20/2024] [Accepted: 07/22/2024] [Indexed: 07/23/2024]
Abstract
The mechanisms underlying endothelial dysfunction in Type 1 and Type 2 diabetes (T1DM and T2DM) are unresolved. The red blood cells (RBCs) with increased arginase activity induce endothelial dysfunction in T2DM, but the implications of RBCs and the role of arginase inhibition in T1DM are unexplored. We aimed to investigate the differences in endothelial function in patients with T1DM and T2DM, with focus on RBCs and arginase. Thirteen patients with T1DM and twenty-six patients with T2DM, matched for HbA1c and sex were included. In vivo endothelium-dependent and -independent vasodilation (EDV and EIDV) were assessed by venous occlusion plethysmography before and after administration of an arginase inhibitor. RBCs were co-incubated with rat aortic segments for 18h followed by evaluation of endothelium-dependent (EDR) and -independent relaxation (EIDR) in isolated organ chambers. In vivo EDV, but not EIDV, was significantly impaired in patients with T2DM compared with patients with T1DM. Arginase inhibition resulted in improved EDV only in T2DM. RBCs from patients with T2DM induced impaired EDR but not EIDR in isolated aortic segments, whereas RBCs from patients with T1DM did not affect EDR nor EIDR. The present study demonstrates markedly impaired EDV in patients with T2DM in comparison with T1DM. In addition, it highlights the divergent roles of RBCs and arginase in mediating endothelial dysfunction in T1DM and T2DM. While endothelial dysfunction is mediated via RBCs and arginase in T2DM, these phenomena are not prominent in T1DM thereby indicating distinct differences in underlying mechanisms.
Collapse
Affiliation(s)
- John Tengbom
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Eftychia Kontidou
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Aida Collado
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Jiangning Yang
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Michael Alvarsson
- Division of Endocrinology and Diabetology, Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Jonas Brinck
- Division of Endocrinology, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Sophia Rössner
- Division of Endocrinology, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Zhichao Zhou
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - John Pernow
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Ali Mahdi
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
10
|
Zhu Z, Wang M, Lu S, Dai S, Liu J. Role of macrophage polarization in heart failure and traditional Chinese medicine treatment. Front Pharmacol 2024; 15:1434654. [PMID: 39104386 PMCID: PMC11298811 DOI: 10.3389/fphar.2024.1434654] [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: 05/21/2024] [Accepted: 07/01/2024] [Indexed: 08/07/2024] Open
Abstract
Heart failure (HF) has a severe impact on public health development due to high morbidity and mortality and is associated with imbalances in cardiac immunoregulation. Macrophages, a major cell population involved in cardiac immune response and inflammation, are highly heterogeneous and polarized into M1 and M2 types depending on the microenvironment. M1 macrophage releases inflammatory factors and chemokines to activate the immune response and remove harmful substances, while M2 macrophage releases anti-inflammatory factors to inhibit the overactive immune response and promote tissue repair. M1 and M2 restrict each other to maintain cardiac homeostasis. The dynamic balance of M1 and M2 is closely related to the Traditional Chinese Medicine (TCM) yin-yang theory, and the imbalance of yin and yang will result in a pathological state of the organism. Studies have confirmed that TCM produces positive effects on HF by regulating macrophage polarization. This review describes the critical role of macrophage polarization in inflammation, fibrosis, angiogenesis and electrophysiology in the course of HF, as well as the potential mechanism of TCM regulation of macrophage polarization in preventing and treating HF, thereby providing new ideas for clinical treatment and scientific research design of HF.
Collapse
Affiliation(s)
- Zheqin Zhu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Min Wang
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Shenghua Lu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Sisi Dai
- Hunan University of Chinese Medicine, Changsha, China
| | - Jianhe Liu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
11
|
Engin A. Endothelial Dysfunction in Obesity and Therapeutic Targets. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:489-538. [PMID: 39287863 DOI: 10.1007/978-3-031-63657-8_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Parallel to the increasing prevalence of obesity in the world, the mortality from cardiovascular disease has also increased. Low-grade chronic inflammation in obesity disrupts vascular homeostasis, and the dysregulation of adipocyte-derived endocrine and paracrine effects contributes to endothelial dysfunction. Besides the adipose tissue inflammation, decreased nitric oxide (NO)-bioavailability, insulin resistance (IR), and oxidized low-density lipoproteins (oxLDLs) are the main factors contributing to endothelial dysfunction in obesity and the development of cardiorenal metabolic syndrome. While normal healthy perivascular adipose tissue (PVAT) ensures the dilation of blood vessels, obesity-associated PVAT leads to a change in the profile of the released adipo-cytokines, resulting in a decreased vasorelaxing effect. Higher stiffness parameter β, increased oxidative stress, upregulation of pro-inflammatory cytokines, and nicotinamide adenine dinucleotide phosphate (NADP) oxidase in PVAT turn the macrophages into pro-atherogenic phenotypes by oxLDL-induced adipocyte-derived exosome-macrophage crosstalk and contribute to the endothelial dysfunction. In clinical practice, carotid ultrasound, higher leptin levels correlate with irisin over-secretion by human visceral and subcutaneous adipose tissues, and remnant cholesterol (RC) levels predict atherosclerotic disease in obesity. As a novel therapeutic strategy for cardiovascular protection, liraglutide improves vascular dysfunction by modulating a cyclic adenosine monophosphate (cAMP)-independent protein kinase A (PKA)-AMP-activated protein kinase (AMPK) pathway in PVAT in obese individuals. Because the renin-angiotensin-aldosterone system (RAAS) activity, hyperinsulinemia, and the resultant IR play key roles in the progression of cardiovascular disease in obesity, RAAS-targeted therapies contribute to improving endothelial dysfunction. By contrast, arginase reciprocally inhibits NO formation and promotes oxidative stress. Thus, targeting arginase activity as a key mediator in endothelial dysfunction has therapeutic potential in obesity-related vascular comorbidities. Obesity-related endothelial dysfunction plays a pivotal role in the progression of type 2 diabetes (T2D). The peroxisome proliferator-activated receptor gamma (PPARγ) agonist, rosiglitazone (thiazolidinedione), is a popular drug for treating diabetes; however, it leads to increased cardiovascular risk. Selective sodium-glucose co-transporter-2 (SGLT-2) inhibitor empagliflozin (EMPA) significantly improves endothelial dysfunction and mortality occurring through redox-dependent mechanisms. Although endothelial dysfunction and oxidative stress are alleviated by either metformin or EMPA, currently used drugs to treat obesity-related diabetes neither possess the same anti-inflammatory potential nor simultaneously target endothelial cell dysfunction and obesity equally. While therapeutic interventions with glucagon-like peptide-1 (GLP-1) receptor agonist liraglutide or bariatric surgery reverse regenerative cell exhaustion, support vascular repair mechanisms, and improve cardiometabolic risk in individuals with T2D and obesity, the GLP-1 analog exendin-4 attenuates endothelial endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
12
|
Lee DH, Kain V, Wang DZ, Rokosh DG, Prabhu SD, Halade GV. Genetic deletion of 12/15 lipoxygenase delays vascular remodeling and limits cardiorenal dysfunction after pressure overload. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2023; 5:100046. [PMID: 39802176 PMCID: PMC11708313 DOI: 10.1016/j.jmccpl.2023.100046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/16/2023] [Accepted: 08/16/2023] [Indexed: 01/16/2025]
Abstract
The lipid metabolizing enzyme 12/15 lipoxygenase (12/15LOX) induces proinflammatory responses that may increase cardiovascular and renal complications after cardiac insult. To define the role of 12/15LOX, 8-12-week-old male C57BL/6J wild-type (WT; n = 49) and 12/15LOX-/- mice (n = 50) were subject to transverse aortic constriction (TAC) and monitored for 7, 28, and 56 days (d) post-TAC. Compared with WT, 12/15LOX-/- mice experienced less left ventricle (LV) dysfunction with limited LV hypertrophy and lung edema post-TAC. 12/15LOX deletion decreased TAC-induced proinflammatory mediators 12-HETE and prostaglandins with modulation in mir-7a-5p, mir 26a-5p, miR-21e-5p, and miR-107-3p during chronic remodeling period (after d28). At d7 post-TAC, 12/15LOX-/- mice showed increased cardiac gene expression of Arg-1 and the prostanoid receptors EP2 and EP4. The EP4 receptor expression was consistently elevated from d7 till d56 in 12/15LOX-/- mice post-TAC compared with WT controls. Post-TAC, wheat germ agglutinin staining revealed less cardiomyocyte hypertrophy at d28 and d56 in 12/15LOX-/- mice compared with WT. TAC-induced vascular remodeling was marked by disruption in the endothelium, evident by irregular CD31 staining and increased alpha-smooth muscle actin (α-SMA) in WT mice at d28 and d56. Compared to WT, 12/15LOX-/- mice exhibited a diminished expression of NGAL in the kidney, suggesting that 12/15LOX-/- reduced cardiorenal dysfunction post-TAC. In WT-TAC mice, structural analyses of the kidney revealed glomerular swelling during the maladaptive phase of heart failure, with decreases in the capsula glomeruli space and glomerular sclerosis compared to 12/15LOX-/- mice. Overall, vascular and kidney inflammation markers were higher in WT than in 12/15LOX-/- post-TAC. Thus, deletion of 12/15LOX limits LV hypertrophy associated with perivascular inflammation and cardiorenal remodeling after pressure overload. Deficiency of 12/15 LOX serves a dual role in delaying an early adaptive interstitial remodeling with long-term protective effects on cardiac hypertrophy and cardiac fibrosis and detrimental adverse vascular remodeling during later maladaptive remodeling after pressure overload.
Collapse
Affiliation(s)
- Dae Hyun Lee
- Division of Cardiovascular Disease, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Florida 33602, United States
| | - Vasundhara Kain
- Division of Cardiovascular Disease, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Florida 33602, United States
| | - Da-Zhi Wang
- Division of Cardiovascular Disease, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Florida 33602, United States
| | - Donald G. Rokosh
- Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine, MO 63110, United States
| | - Sumanth D. Prabhu
- Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine, MO 63110, United States
| | - Ganesh V. Halade
- Division of Cardiovascular Disease, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Florida 33602, United States
| |
Collapse
|
13
|
Chen M, Cheng H, Chen X, Gu J, Su W, Cai G, Yan Y, Wang C, Xia X, Zhang K, Zhang M, Jiang H, Chen Y, Yao L. The activation of histone deacetylases 4 prevented endothelial dysfunction: A crucial mechanism of HuangqiGuizhiWuwu Decoction in improving microcirculation dysfunction in diabetes. JOURNAL OF ETHNOPHARMACOLOGY 2023; 307:116240. [PMID: 36764560 DOI: 10.1016/j.jep.2023.116240] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 01/18/2023] [Accepted: 02/02/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The regulation of epigenetic factors is considered a crucial target for solving complex chronic diseases such as cardio-cerebrovascular diseases. HuangqiGuizhiWuwu Decoction (HGWWD), a classic Chinese prescription, is mainly used to treat various vascular diseases. Although our previous studies reported that HGWWD could effectively prevent vascular dysfunction in diabetic rodent models, the precise mechanism is still elusive. AIM OF THE STUDY In this study, we investigated the epigenetic mechanisms of modulating the damage of vascular endothelial cells in diabetes by HGWWD. METHODS We first analyzed common active components of HGWWD by using HPLC-Q-TOF-MS/MS analysis, and predicted the isoforms of histone deacetylase (HDAC) that can potentially combine the above active components by systems pharmacology. Next, we screened the involvement of specific HDAC isoforms in the protective effect of HGWWD on vascular injury by using pharmacological blockade combined with the evaluation of vascular function in vivo and in vitro. RESULTS Firstly, HDAC1, HDAC2, HDAC3, HDAC4, HDAC6, HDAC7, SIRT2, and SIRT3 have been implicated with the possibility of binding to the thirty-one common active components in HGWWD. Furthermore, the protective effect of HGWWD is reversed by both TSA (HDAC inhibitor) and MC1568 (class II HDAC inhibitor) on vascular impairment accompanied by reduced aortic HDAC activity in STZ mice. Finally, inhibition of HDAC4 blocked the protective effect of HGWWD on microvascular and endothelial dysfunction in diabetic mice. CONCLUSIONS These results prove the key role of HDAC4 in diabetes-induced microvascular dysfunction and underlying epigenetic mechanisms for the protective effect of HGWWD in diabetes.
Collapse
Affiliation(s)
- Meijiang Chen
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China.
| | - Hong Cheng
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China.
| | - Xinyi Chen
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China.
| | - Jiangyong Gu
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China.
| | - Weiwei Su
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-evaluation of Post-marketed TCM, Guangdong Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, PR China.
| | - Gaize Cai
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China.
| | - Yue Yan
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China.
| | - Chen Wang
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China.
| | - Xiaoye Xia
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China.
| | - Kaitong Zhang
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China.
| | - Meng Zhang
- Research Institute of Acupuncture and Moxibustion, Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China.
| | - Haiqiang Jiang
- Research Institute of Acupuncture and Moxibustion, Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China.
| | - Yongjun Chen
- Research Institute of Acupuncture and Moxibustion, Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China.
| | - Lin Yao
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China; Research Institute of Acupuncture and Moxibustion, Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China.
| |
Collapse
|
14
|
Janaszak-Jasiecka A, Płoska A, Wierońska JM, Dobrucki LW, Kalinowski L. Endothelial dysfunction due to eNOS uncoupling: molecular mechanisms as potential therapeutic targets. Cell Mol Biol Lett 2023; 28:21. [PMID: 36890458 PMCID: PMC9996905 DOI: 10.1186/s11658-023-00423-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 01/19/2023] [Indexed: 03/10/2023] Open
Abstract
Nitric oxide (NO) is one of the most important molecules released by endothelial cells, and its antiatherogenic properties support cardiovascular homeostasis. Diminished NO bioavailability is a common hallmark of endothelial dysfunction underlying the pathogenesis of the cardiovascular disease. Vascular NO is synthesized by endothelial nitric oxide synthase (eNOS) from the substrate L-arginine (L-Arg), with tetrahydrobiopterin (BH4) as an essential cofactor. Cardiovascular risk factors such as diabetes, dyslipidemia, hypertension, aging, or smoking increase vascular oxidative stress that strongly affects eNOS activity and leads to eNOS uncoupling. Uncoupled eNOS produces superoxide anion (O2-) instead of NO, thus becoming a source of harmful free radicals exacerbating the oxidative stress further. eNOS uncoupling is thought to be one of the major underlying causes of endothelial dysfunction observed in the pathogenesis of vascular diseases. Here, we discuss the main mechanisms of eNOS uncoupling, including oxidative depletion of the critical eNOS cofactor BH4, deficiency of eNOS substrate L-Arg, or accumulation of its analog asymmetrical dimethylarginine (ADMA), and eNOS S-glutathionylation. Moreover, potential therapeutic approaches that prevent eNOS uncoupling by improving cofactor availability, restoration of L-Arg/ADMA ratio, or modulation of eNOS S-glutathionylation are briefly outlined.
Collapse
Affiliation(s)
- Anna Janaszak-Jasiecka
- Department of Medical Laboratory Diagnostics - Fahrenheit Biobank BBMRI.Pl, Medical University of Gdansk, 7 Debinki Street, 80-211, Gdansk, Poland
| | - Agata Płoska
- Department of Medical Laboratory Diagnostics - Fahrenheit Biobank BBMRI.Pl, Medical University of Gdansk, 7 Debinki Street, 80-211, Gdansk, Poland
| | - Joanna M Wierońska
- Department of Neurobiology, Polish Academy of Sciences, Maj Institute of Pharmacology, 12 Smętna Street, 31-343, Kraków, Poland
| | - Lawrence W Dobrucki
- Department of Medical Laboratory Diagnostics - Fahrenheit Biobank BBMRI.Pl, Medical University of Gdansk, 7 Debinki Street, 80-211, Gdansk, Poland.,Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Beckman Institute for Advanced Science and Technology, 405 N Mathews Ave, MC-251, Urbana, IL, 61801, USA.,Department of Biomedical and Translational Sciences, Carle-Illinois College of Medicine, Urbana, IL, USA
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics - Fahrenheit Biobank BBMRI.Pl, Medical University of Gdansk, 7 Debinki Street, 80-211, Gdansk, Poland. .,BioTechMed Centre, Department of Mechanics of Materials and Structures, Gdansk University of Technology, 11/12 Gabriela Narutowicza Street, 80-233, Gdansk, Poland.
| |
Collapse
|
15
|
McFarlin BK, Tanner EA, Hill DW, Vingren JL. Prebiotic/probiotic supplementation resulted in reduced visceral fat and mRNA expression associated with adipose tissue inflammation, systemic inflammation, and chronic disease risk. GENES & NUTRITION 2022; 17:15. [PMID: 36437471 PMCID: PMC9703693 DOI: 10.1186/s12263-022-00718-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 11/14/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Prebiotic/probiotic supplementation represents a viable option for addressing elevated systemic inflammation and chronic disease risk in overweight individuals. The purpose of this study was to determine if 90 days of prebiotic/probiotic supplementation could alter mRNA responsible for inflammation and chronic disease risk in weight-stable overweight adults. Nanostring mRNA analysis (574 plex) was used to survey targets associated with adipose tissue inflammation, systemic inflammation, and chronic disease risk. All protocols were approved by the University IRB, and participants gave written informed consent. Participants were randomly assigned to either placebo (N = 7; rice flour) or combined (N = 8) prebiotic (PreticX® Xylooligosaccharide; 0.8 g/day; ADIP) and probiotic (MegaDuo® Bacillus subtilis HU58 and Bacillus coagulans SC-208; billion CFU/day) supplementation. Participants were diverse population of healthy individuals with the exception of excess body weight. Measurements were made at baseline, 30, 60, and 90 days. Whole-body DXA scans (GE iDXA®; body composition) and blood 574-plex mRNA analysis (Nanostring®) were used to generate primary outcomes. Significance was set to p < 0.05 and adjusted for multiple comparisons where necessary. RESULTS Compared to placebo, prebiotic/probiotic supplementation was associated with a 35% reduction in visceral adipose tissue (VAT; p = 0.002) but no change in body weight or overall percent body fat. Prebiotic/probiotic supplementation resulted in significant (p < 0.05), differential expression of 15 mRNA associated with adipose tissue inflammation (GATA3, TNFAIP6, ST2, CMKLR1, and CD9), systemic inflammation (LTF, SOCS1, and SERPING1), and/or chronic disease risk (ARG1, IL-18, CCL4, CEACAM6, ATM, CD80, and LAMP3). We also found 6 additional mRNA that had no obvious relationship to three previous biological functions (CSF1, SRC, ICAM4CD24, CD274, and CLEC6A). CONCLUSION The key findings support that 90-day prebiotic/probiotic supplementation may be associated with reduced adipose tissue inflammation, reduced systemic inflammation, and reduced chronic disease risk. Combined with the unexpected finding of reduced VAT, this intervention may have resulted in improved overall health and reduced chronic disease risk.
Collapse
Affiliation(s)
- Brian K McFarlin
- Applied Physiology Laboratory, University of North Texas, Denton, TX, 76203, USA. .,Department of Biological Sciences, University of North Texas, Denton, TX, 76203, USA.
| | - Elizabeth A Tanner
- Applied Physiology Laboratory, University of North Texas, Denton, TX, 76203, USA.,Department of Biological Sciences, University of North Texas, Denton, TX, 76203, USA
| | - David W Hill
- Applied Physiology Laboratory, University of North Texas, Denton, TX, 76203, USA
| | - Jakob L Vingren
- Applied Physiology Laboratory, University of North Texas, Denton, TX, 76203, USA.,Department of Biological Sciences, University of North Texas, Denton, TX, 76203, USA
| |
Collapse
|
16
|
Suvorava T, Metry S, Pick S, Kojda G. Alterations in endothelial nitric oxide synthase activity and their relevance to blood pressure. Biochem Pharmacol 2022; 205:115256. [DOI: 10.1016/j.bcp.2022.115256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 12/15/2022]
|
17
|
Larios-Cárdenas M, González-Radillo OI, Trujillo-Quirós J, Cardona-Müller D, Barocio-Pantoja M, Cardona-Muñoz EG, Grover-Páez F. Tadalafil Improves Haemodynamics and Arterial Stiffness but Not Flow- Mediated Dilation in Grade 1 Obesity. A Single-dose, Placebo-controlled Clinical Trial. Curr Vasc Pharmacol 2022; 20:527-533. [PMID: 36043781 DOI: 10.2174/1570161120666220827154417] [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: 02/14/2022] [Revised: 06/21/2022] [Accepted: 07/04/2022] [Indexed: 01/25/2023]
Abstract
OBJECTIVE Obesity, a major health issue worldwide, is associated with increased cardiovascular risk, endothelial dysfunction, and arterial stiffness. Tadalafil has been demonstrated to improve vascular parameters. AIM To evaluate the effect of a single 20 mg dose of tadalafil on flow-mediated dilation and hemodynamic and arterial stiffness markers. METHODS A randomized, double-blind, placebo-controlled study was conducted on 80 participants (41 assigned to placebo and 39 to tadalafil) with grade 1 obesity, to evaluate the acute effect of a single dose of 20 mg of tadalafil on flow-mediated dilation and hemodynamic and arterial stiffness markers. RESULTS Tadalafil did not modify flow-mediated dilation. However, it significantly lowered systolic blood pressure (SBP) (130.6±17.1 vs. 125.0±12.7 mmHg, p=0.011), diastolic blood pressure (82.7±18.2 vs. 76.5±11.8 mmHg, p≤0.001), central systolic blood pressure (116.33±19.16 vs. 109.90±15.05 mmHg, p=0.001), the augmentation index (69.1±17.1 vs. 65.7±14.4, p=0.012), and brachial-ankle pulse wave velocity (1229.7±218.4 vs. 1164.0±181.7, p=0.001). CONCLUSION A single dose of tadalafil did not modify flow-mediated dilation in patients with grade 1 obesity but improved blood pressure and brachial-ankle pulse wave velocity.
Collapse
Affiliation(s)
- Mariana Larios-Cárdenas
- Vascular Mechanics Laboratory, Experimental Therapeutic and Clinic Institute, Health Sciences University Centre, University of Guadalajara, Guadalajara, Mexico.,Department of Physiology, Pharmacology, Health Sciences University Centre, University of Guadalajara, Guadalajara, Mexico
| | - Oscar I González-Radillo
- Vascular Mechanics Laboratory, Experimental Therapeutic and Clinic Institute, Health Sciences University Centre, University of Guadalajara, Guadalajara, Mexico.,Department of Physiology, Pharmacology, Health Sciences University Centre, University of Guadalajara, Guadalajara, Mexico
| | - Jhonatan Trujillo-Quirós
- Vascular Mechanics Laboratory, Experimental Therapeutic and Clinic Institute, Health Sciences University Centre, University of Guadalajara, Guadalajara, Mexico.,Department of Physiology, Pharmacology, Health Sciences University Centre, University of Guadalajara, Guadalajara, Mexico
| | - David Cardona-Müller
- Vascular Mechanics Laboratory, Experimental Therapeutic and Clinic Institute, Health Sciences University Centre, University of Guadalajara, Guadalajara, Mexico.,Department of Physiology, Pharmacology, Health Sciences University Centre, University of Guadalajara, Guadalajara, Mexico
| | - Marycruz Barocio-Pantoja
- Vascular Mechanics Laboratory, Experimental Therapeutic and Clinic Institute, Health Sciences University Centre, University of Guadalajara, Guadalajara, Mexico.,Department of Physiology, Pharmacology, Health Sciences University Centre, University of Guadalajara, Guadalajara, Mexico
| | - Ernesto G Cardona-Muñoz
- Vascular Mechanics Laboratory, Experimental Therapeutic and Clinic Institute, Health Sciences University Centre, University of Guadalajara, Guadalajara, Mexico.,Department of Physiology, Pharmacology, Health Sciences University Centre, University of Guadalajara, Guadalajara, Mexico
| | - Fernando Grover-Páez
- Vascular Mechanics Laboratory, Experimental Therapeutic and Clinic Institute, Health Sciences University Centre, University of Guadalajara, Guadalajara, Mexico.,Department of Physiology, Pharmacology, Health Sciences University Centre, University of Guadalajara, Guadalajara, Mexico
| |
Collapse
|
18
|
Foratori-Junior GA, Guennec AL, Fidalgo TKDS, Cleaver L, Buzalaf MAR, Carpenter GH, Sales-Peres SHDC. Metabolomic Profiles Associated with Obesity and Periodontitis during Pregnancy: Cross-Sectional Study with Proton Nuclear Magnetic Resonance ( 1H-NMR)-Based Analysis. Metabolites 2022; 12:metabo12111029. [PMID: 36355112 PMCID: PMC9694155 DOI: 10.3390/metabo12111029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/18/2022] [Accepted: 10/24/2022] [Indexed: 12/27/2022] Open
Abstract
This study aimed to elucidate the metabolomic signature associated with obesity and periodontitis during pregnancy in plasma and saliva biofluids. Ninety-eight pregnant women were divided into: with obesity and periodontitis (OP = 20), with obesity but without periodontitis (OWP = 27), with normal BMI but with periodontitis (NP = 21), with normal BMI and without periodontitis (NWP = 30). Saliva and plasma were analyzed by 1H-NMR for metabolites identification. Partial Least Squares-Discriminant Analysis (PLS-DA), Sparse PLS-DA (sPLS-DA), and Variable Importance of Projection (VIP) were performed. ANOVA and Pearson’s correlation were applied (p < 0.05). Plasmatic analysis indicated the levels of glucose (p = 0.041) and phenylalanine (p = 0.015) were positively correlated with periodontal parameters and BMI, respectively. In saliva, periodontitis was mainly associated with high levels of acetic acid (p = 0.024), isovaleric acid, butyric acid, leucine, valine, isoleucine, and propionic acid (p < 0.001). High salivary concentrations of glycine (p = 0.015), succinic acid (p = 0.015), and lactate (p = 0.026) were associated with obesity. Saliva demonstrated a more elucidative difference than plasma, indicating the glucose-alanine cycle, alanine metabolism, valine, leucine and isoleucine degradation, glutamate metabolism, and Warburg effect as the main metabolic pathways.
Collapse
Affiliation(s)
- Gerson Aparecido Foratori-Junior
- Department of Pediatric Dentistry, Orthodontics and Public Health, Bauru School of Dentistry, University of São Paulo, Bauru 17012-901, Brazil
- Centre for Host-Microbiome Interactions, Faculty of Dental, Oral & Craniofacial Sciences, Guy’s Campus, King’s College London, London SE1 1UL, UK
- Correspondence: (G.A.F.-J.); (S.H.d.C.S.-P.)
| | - Adrien Le Guennec
- Nuclear Magnetic Resonance Facility, Guy’s Campus, King’s College London, London SE1 1UL, UK
| | - Tatiana Kelly da Silva Fidalgo
- Department of Preventive and Community Dentistry, School of Dentistry, Rio de Janeiro State University, Rio de Janeiro 20551-030, Brazil
| | - Leanne Cleaver
- Centre for Host-Microbiome Interactions, Faculty of Dental, Oral & Craniofacial Sciences, Guy’s Campus, King’s College London, London SE1 1UL, UK
| | | | - Guy Howard Carpenter
- Centre for Host-Microbiome Interactions, Faculty of Dental, Oral & Craniofacial Sciences, Guy’s Campus, King’s College London, London SE1 1UL, UK
| | - Silvia Helena de Carvalho Sales-Peres
- Department of Pediatric Dentistry, Orthodontics and Public Health, Bauru School of Dentistry, University of São Paulo, Bauru 17012-901, Brazil
- Correspondence: (G.A.F.-J.); (S.H.d.C.S.-P.)
| |
Collapse
|
19
|
Ren Y, Li Z, Li W, Fan X, Han F, Huang Y, Yu Y, Qian L, Xiong Y. Arginase: Biological and Therapeutic Implications in Diabetes Mellitus and Its Complications. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2419412. [PMID: 36338341 PMCID: PMC9629921 DOI: 10.1155/2022/2419412] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 10/18/2022] [Indexed: 09/21/2023]
Abstract
Arginase is a ubiquitous enzyme in the urea cycle (UC) that hydrolyzes L-arginine to urea and L-ornithine. Two mammalian arginase isoforms, arginase1 (ARG1) and arginase2 (ARG2), play a vital role in the regulation of β-cell functions, insulin resistance (IR), and vascular complications via modulating L-arginine metabolism, nitric oxide (NO) production, and inflammatory responses as well as oxidative stress. Basic and clinical studies reveal that abnormal alterations of arginase expression and activity are strongly associated with the onset and development of diabetes mellitus (DM) and its complications. As a result, targeting arginase may be a novel and promising approach for DM treatment. An increasing number of arginase inhibitors, including chemical and natural inhibitors, have been developed and shown to protect against the development of DM and its complications. In this review, we discuss the fundamental features of arginase. Next, the regulatory roles and underlying mechanisms of arginase in the pathogenesis and progression of DM and its complications are explored. Furthermore, we review the development and discuss the challenges of arginase inhibitors in treating DM and its related pathologies.
Collapse
Affiliation(s)
- Yuanyuan Ren
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Zhuozhuo Li
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Wenqing Li
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Xiaobin Fan
- Department of Obstetrics and Gynecology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, Shaanxi, China
| | - Feifei Han
- Department of Endocrinology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, Shaanxi, China
| | - Yaoyao Huang
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Yi Yu
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Lu Qian
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China
- Department of Obstetrics and Gynecology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, Shaanxi, China
| | - Yuyan Xiong
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, China
| |
Collapse
|
20
|
Heuser SK, LoBue A, Li J, Zhuge Z, Leo F, Suvorava T, Olsson A, Schneckmann R, Guimaraes Braga DD, Srivrastava T, Montero L, Schmitz OJ, Schmitt JP, Grandoch M, Weitzberg E, Lundberg JO, Pernow J, Kelm M, Carlström M, Cortese-Krott MM. Downregulation of eNOS and preserved endothelial function in endothelial-specific arginase 1-deficient mice. Nitric Oxide 2022; 125-126:69-77. [PMID: 35752264 DOI: 10.1016/j.niox.2022.06.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 06/08/2022] [Accepted: 06/10/2022] [Indexed: 12/20/2022]
Abstract
Arginase 1 (Arg1) is a ubiquitous enzyme belonging to the urea cycle that catalyzes the conversion of l-arginine into l-ornithine and urea. In endothelial cells (ECs), Arg1 was proposed to limit the availability of l-arginine for the endothelial nitric oxide synthase (eNOS) and thereby reduce nitric oxide (NO) production, thus promoting endothelial dysfunction and vascular disease. The role of EC Arg1 under homeostatic conditions is in vivo less understood. The aim of this study was to investigate the role of EC Arg1 on the regulation of eNOS, vascular tone, and endothelial function under normal homeostatic conditions in vivo and ex vivo. By using a tamoxifen-inducible EC-specific gene-targeting approach, we generated EC Arg1 KO mice. Efficiency and specificity of the gene targeting strategy was demonstrated by DNA recombination and loss of Arg1 expression measured after tamoxifen treatment in EC only. In EC Arg1 KO mice we found a significant decrease in Arg1 expression in heart and lung ECs and in the aorta, however, vascular enzymatic activity was preserved likely due to the presence of high levels of Arg1 in smooth muscle cells. Moreover, we found a downregulation of eNOS expression in the aorta, and a fully preserved systemic l-arginine and NO bioavailability, as demonstrated by the levels of l-arginine, l-ornithine, and l-citrulline as well as nitrite, nitrate, and nitroso-species. Lung and liver tissues from EC Arg1 KO mice showed respectively increase or decrease in nitrosyl-heme species, indicating that the lack of endothelial Arg1 affects NO bioavailability in these organs. In addition, EC Arg1 KO mice showed fully preserved acetylcholine-mediated vascular relaxation in both conductance and resistant vessels but increased phenylephrine-induced vasoconstriction. Systolic, diastolic, and mean arterial pressure and cardiac performance in EC Arg1 KO mice were not different from the wild-type littermate controls. In conclusion, under normal homeostatic conditions, lack of EC Arg1 expression is associated with a down-regulation of eNOS expression but a preserved NO bioavailability and vascular endothelial function. These results suggest that a cross-talk exists between Arg1 and eNOS to control NO production in ECs, which depends on both L-Arg availability and EC Arg1-dependent eNOS expression.
Collapse
Affiliation(s)
- Sophia K Heuser
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Anthea LoBue
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Junjie Li
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Zhengbing Zhuge
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Francesca Leo
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Tatsiana Suvorava
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany; Department of Pharmacology and Clinical Pharmacology, Medical Faculty, Heinrich-Heine-University, Germany
| | - Annika Olsson
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Rebekka Schneckmann
- Department of Pharmacology and Clinical Pharmacology, Medical Faculty, Heinrich-Heine-University, Germany
| | | | - Tanu Srivrastava
- Department of Pharmacology and Clinical Pharmacology, Medical Faculty, Heinrich-Heine-University, Germany
| | - Lidia Montero
- Applied Analytical Chemistry, Faculty of Chemistry, University of Duisburg-Essen, Germany
| | - Oliver J Schmitz
- Applied Analytical Chemistry, Faculty of Chemistry, University of Duisburg-Essen, Germany
| | - Joachim P Schmitt
- Department of Pharmacology and Clinical Pharmacology, Medical Faculty, Heinrich-Heine-University, Germany
| | - Maria Grandoch
- Department of Pharmacology and Clinical Pharmacology, Medical Faculty, Heinrich-Heine-University, Germany
| | - Eddie Weitzberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jon O Lundberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - John Pernow
- Department of Cardiology, Karolinska Institute, Stockholm, Sweden
| | - Malte Kelm
- Cardiovascular Research Laboratory, Department of Cardiology Pneumology and Angiology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany; CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Mattias Carlström
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Miriam M Cortese-Krott
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
21
|
Niu F, Yu Y, Li Z, Ren Y, Li Z, Ye Q, Liu P, Ji C, Qian L, Xiong Y. Arginase: An emerging and promising therapeutic target for cancer treatment. Biomed Pharmacother 2022; 149:112840. [PMID: 35316752 DOI: 10.1016/j.biopha.2022.112840] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/03/2022] [Accepted: 03/16/2022] [Indexed: 11/19/2022] Open
Abstract
Arginase is a key hydrolase in the urea cycle that hydrolyses L-arginine to urea and L-ornithine. Increasing number of studies in recent years demonstrate that two mammalian arginase isoforms, arginase 1 (ARG1) and arginase 2 (ARG2), were aberrantly upregulated in various types of cancers, and played crucial roles in the regulation of tumor growth and metastasis through various mechanisms such as regulating L-arginine metabolism, influencing tumor immune microenvironment, etc. Thus, arginase receives increasing focus as an attractive target for cancer therapy. In this review, we provide a comprehensive overview of the physiological and biological roles of arginase in a variety of cancers, and shed light on the underlying mechanisms of arginase mediating cancer cells growth and development, as well as summarize the recent clinical research advances of targeting arginase for cancer therapy.
Collapse
Affiliation(s)
- Fanglin Niu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an 710069, Shaanxi, China
| | - Yi Yu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an 710069, Shaanxi, China
| | - Zhuozhuo Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an 710069, Shaanxi, China
| | - Yuanyuan Ren
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an 710069, Shaanxi, China
| | - Zi Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an 710069, Shaanxi, China
| | - Qiang Ye
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an 710069, Shaanxi, China
| | - Ping Liu
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, China; Department of Endocrinology, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an 710018, Shaanxi, China
| | - Chenshuang Ji
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an 710069, Shaanxi, China
| | - Lu Qian
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, China; Department of Endocrinology, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an 710018, Shaanxi, China.
| | - Yuyan Xiong
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an 710069, Shaanxi, China; Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, China.
| |
Collapse
|
22
|
Sabe SA, Feng J, Sellke FW, Abid MR. Mechanisms and clinical implications of endothelium-dependent vasomotor dysfunction in coronary microvasculature. Am J Physiol Heart Circ Physiol 2022; 322:H819-H841. [PMID: 35333122 PMCID: PMC9018047 DOI: 10.1152/ajpheart.00603.2021] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 03/21/2022] [Accepted: 03/21/2022] [Indexed: 12/16/2022]
Abstract
Coronary microvascular disease (CMD), which affects the arterioles and capillary endothelium that regulate myocardial perfusion, is an increasingly recognized source of morbidity and mortality, particularly in the setting of metabolic syndrome. The coronary endothelium plays a pivotal role in maintaining homeostasis, though factors such as diabetes, hypertension, hyperlipidemia, and obesity can contribute to endothelial injury and consequently arteriolar vasomotor dysfunction. These disturbances in the coronary microvasculature clinically manifest as diminished coronary flow reserve, which is a known independent risk factor for cardiac death, even in the absence of macrovascular atherosclerotic disease. Therefore, a growing body of literature has examined the molecular mechanisms by which coronary microvascular injury occurs at the level of the endothelium and the consequences on arteriolar vasomotor responses. This review will begin with an overview of normal coronary microvascular physiology, modalities of measuring coronary microvascular function, and clinical implications of CMD. These introductory topics will be followed by a discussion of recent advances in the understanding of the mechanisms by which inflammation, oxidative stress, insulin resistance, hyperlipidemia, hypertension, shear stress, endothelial cell senescence, and tissue ischemia dysregulate coronary endothelial homeostasis and arteriolar vasomotor function.
Collapse
Affiliation(s)
- Sharif A Sabe
- Cardiovascular Research Center, Rhode Island Hospital, Providence, Rhode Island
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island
| | - Jun Feng
- Cardiovascular Research Center, Rhode Island Hospital, Providence, Rhode Island
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island
| | - Frank W Sellke
- Cardiovascular Research Center, Rhode Island Hospital, Providence, Rhode Island
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island
| | - M Ruhul Abid
- Cardiovascular Research Center, Rhode Island Hospital, Providence, Rhode Island
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island
| |
Collapse
|
23
|
Targeting Arginine in COVID-19-Induced Immunopathology and Vasculopathy. Metabolites 2022; 12:metabo12030240. [PMID: 35323682 PMCID: PMC8953281 DOI: 10.3390/metabo12030240] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/06/2022] [Accepted: 03/09/2022] [Indexed: 01/27/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) represents a major public health crisis that has caused the death of nearly six million people worldwide. Emerging data have identified a deficiency of circulating arginine in patients with COVID-19. Arginine is a semi-essential amino acid that serves as key regulator of immune and vascular cell function. Arginine is metabolized by nitric oxide (NO) synthase to NO which plays a pivotal role in host defense and vascular health, whereas the catabolism of arginine by arginase to ornithine contributes to immune suppression and vascular disease. Notably, arginase activity is upregulated in COVID-19 patients in a disease-dependent fashion, favoring the production of ornithine and its metabolites from arginine over the synthesis of NO. This rewiring of arginine metabolism in COVID-19 promotes immune and endothelial cell dysfunction, vascular smooth muscle cell proliferation and migration, inflammation, vasoconstriction, thrombosis, and arterial thickening, fibrosis, and stiffening, which can lead to vascular occlusion, muti-organ failure, and death. Strategies that restore the plasma concentration of arginine, inhibit arginase activity, and/or enhance the bioavailability and potency of NO represent promising therapeutic approaches that may preserve immune function and prevent the development of severe vascular disease in patients with COVID-19.
Collapse
|
24
|
Moughaizel M, Dagher E, Jablaoui A, Thorin C, Rhimi M, Desfontis JC, Mallem Y. Long-term high-fructose high-fat diet feeding elicits insulin resistance, exacerbates dyslipidemia and induces gut microbiota dysbiosis in WHHL rabbits. PLoS One 2022; 17:e0264215. [PMID: 35196347 PMCID: PMC8865649 DOI: 10.1371/journal.pone.0264215] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 02/04/2022] [Indexed: 01/12/2023] Open
Abstract
The metabolic syndrome (MetS) has become a global public health burden due to its link to cardiovascular disease and diabetes mellitus. The present study was designed to characterize the metabolic and cardiovascular disturbances, as well as changes in gut microbiota associated with high-fructose high-fat diet (HFFD)-induced MetS in Watanabe heritable hyperlipidemic (WHHL) rabbits. Twenty-one Watanabe rabbits were assigned to a control (n = 9) and HFFD (n = 12) groups, receiving a chow diet and a HFFD, respectively. During a 12-weeks protocol, morphological parameters were monitored; plasma fasting levels of lipids, glucose and insulin were measured and a glucose tolerance test (GTT) was performed. HOMA-IR was calculated. Cardiac function and vascular reactivity were evaluated using the Langendorff isolated heart and isolated carotid arteries methods, respectively. 16S rRNA sequencing of stool samples was used to determine gut microbial composition and abundance. HFFD-fed Watanabe rabbits exhibited increased fasting insulin (p < 0.03, 12th week vs. Baseline), HOMA-IR (p < 0.03 vs. Control), area under the curve of the GTT (p < 0.02 vs. Control), triglycerides (p < 0.05, 12th week vs. Baseline), TC (p < 0.01 vs. Control), LDL-C (p < 0.001 vs. Control). The HFFD group also displayed a significant decrease in intestinal microbial richness, evenness and diversity (FDR < 0.001, FDR < 0.0001, FDR < 0.01, respectively vs. Control group) and an increase in its Firmicutes/Bacteroidetes ratio (R = 3.39 in control vs. R = 28.24 in the HFFD group) indicating a shift in intestinal microbial composition and diversity. Our results suggest that HFFD induces insulin resistance and gut microbiota dysbiosis and accentuates dyslipidemia; and that, when subjected to HFFD, Watanabe rabbits might become a potential diet-induced MetS animal models with two main features, dyslipidemia and insulin resistance.
Collapse
Affiliation(s)
- Michelle Moughaizel
- Nutrition, PathoPhysiology and Pharmacology (NP3) Unit, Oniris, Nantes Atlantic College of Veterinary Medicine, Food Science and Engineering, Nantes, France
- * E-mail: (MM); (YM)
| | - Elie Dagher
- Laboniris, Oniris, Nantes Atlantic College of Veterinary Medicine, Food Science and Engineering, Nantes, France
| | - Amin Jablaoui
- Institut Micalis, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Chantal Thorin
- Nutrition, PathoPhysiology and Pharmacology (NP3) Unit, Oniris, Nantes Atlantic College of Veterinary Medicine, Food Science and Engineering, Nantes, France
| | - Moez Rhimi
- Institut Micalis, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Jean-Claude Desfontis
- Nutrition, PathoPhysiology and Pharmacology (NP3) Unit, Oniris, Nantes Atlantic College of Veterinary Medicine, Food Science and Engineering, Nantes, France
| | - Yassine Mallem
- Nutrition, PathoPhysiology and Pharmacology (NP3) Unit, Oniris, Nantes Atlantic College of Veterinary Medicine, Food Science and Engineering, Nantes, France
- * E-mail: (MM); (YM)
| |
Collapse
|
25
|
Trotta MC, Gesualdo C, Petrillo F, Cavasso G, Corte AD, D'Amico G, Hermenean A, Simonelli F, Rossi S. Serum Iba-1, GLUT5, and TSPO in Patients With Diabetic Retinopathy: New Biomarkers for Early Retinal Neurovascular Alterations? A Pilot Study. Transl Vis Sci Technol 2022; 11:16. [PMID: 35285861 PMCID: PMC8934554 DOI: 10.1167/tvst.11.3.16] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose This study explored the possibility of highlighting early retinal neurovascular alterations of diabetic retinopathy (DR) by monitoring in DR patients the serum levels of microglial biomarkers ionized calcium-binding adapter molecule 1 (Iba-1), glucose transporter 5 (GLUT5), and translocator protein (TSPO), along with serum changes of the endothelial dysfunction marker arginase-1. Methods Serum markers were determined by enzyme-linked immunosorbent assay in 50 patients: 12 non-diabetic subjects, 14 diabetic patients without DR, 13 patients with non-proliferative DR (NPDR), and 11 patients with proliferative DR (PDR). The results were correlated with hyperreflective retinal spots (HRS), observed with optical coherence tomography (OCT). Results Although HRS were absent in diabetic patients without DR, NPDR patients showed an average of 4 ± 1 HRS, whereas the highest presence was detected in PDR patients, with 8 ± 1 HRS (P < 0.01 vs. NPDR). HRS were positively correlated (P < 0.01) with serum levels of arginase-1 (r = 0.91), Iba-1 (r = 0.96), GLUT5 (r = 0.94), and TSPO (r = 0.88). Moreover, serum proinflammatory cytokines and chemokines showed a positive correlation (P < 0.01) with HRS number and the serum markers analyzed. Conclusions Serum markers of microglial activation positively correlate with retinal HRS in NPDR and PDR patients. Translational Relevance These data corroborate the possibility of highlighting early retinal neurovascular changes due to diabetes by monitoring circulating microglial markers.
Collapse
Affiliation(s)
- Maria Consiglia Trotta
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Carlo Gesualdo
- Eye Clinic, Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania "Luigi Vanvitelli," Naples, Italy
| | | | - Giancuomo Cavasso
- Eye Clinic, Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Alberto Della Corte
- Eye Clinic, Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Giovanbattista D'Amico
- "Aurel Ardelean" Institute of Life Sciences, Vasile Goldis Western University of Arad, Arad, Romania
| | - Anca Hermenean
- "Aurel Ardelean" Institute of Life Sciences, Vasile Goldis Western University of Arad, Arad, Romania
| | - Francesca Simonelli
- Eye Clinic, Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Settimio Rossi
- Eye Clinic, Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania "Luigi Vanvitelli," Naples, Italy
| |
Collapse
|
26
|
Effects of chronic mirabegron treatment on metabolic and cardiovascular parameters as well as on atherosclerotic lesions of WHHL rabbits with high-fructose high-fat diet-induced insulin resistance. Eur J Pharmacol 2022; 921:174870. [DOI: 10.1016/j.ejphar.2022.174870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 02/28/2022] [Accepted: 03/02/2022] [Indexed: 11/23/2022]
|
27
|
Liu Y, Li M, Lv X, Bao K, Yu Tian X, He L, Shi L, Zhu Y, Ai D. YAP Targets the TGFβ Pathway to Mediate High-Fat/High-Sucrose Diet-Induced Arterial Stiffness. Circ Res 2022; 130:851-867. [PMID: 35176871 DOI: 10.1161/circresaha.121.320464] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Metabolic syndrome is related to cardiovascular diseases, which is attributed in part, to arterial stiffness; however, the mechanisms remain unclear. The present study aimed to investigate the molecular mechanisms of metabolic syndrome-induced arterial stiffness and to identify new therapeutic targets. METHODS Arterial stiffness was induced by high-fat/high-sucrose diet in mice, which was quantified by Doppler ultrasound. Four-dimensional label-free quantitative proteomic analysis, affinity purification and mass spectrometry, and immunoprecipitation and GST pull-down experiments were performed to explore the mechanism of YAP (Yes-associated protein)-mediated TGF (transforming growth factor) β pathway activation. RESULTS YAP protein was upregulated in the aortic tunica media of mice fed a high-fat/high-sucrose diet for 2 weeks and precedes arterial stiffness. Smooth muscle cell-specific YAP knockdown attenuated high-fat/high-sucrose diet-induced arterial stiffness and activation of TGFβ-Smad2/3 signaling pathway in arteries. By contrast, Myh11CreERT2-YapTg mice exhibited exacerbated high-fat/high-sucrose diet-induced arterial stiffness and enhanced TGFβ-activated Smad2/3 phosphorylation in arteries. PPM1B (protein phosphatase, Mg2+/Mn2+-dependent 1B) was identified as a YAP-bound phosphatase that translocates into the nucleus to dephosphorylate Smads in response to TGFβ. This process was inhibited by YAP through removal of the K63-linked ubiquitin chain of PPM1B at K326. CONCLUSIONS This study provides a new mechanism by which smooth muscle cell YAP regulates the TGFβ pathway and a potential therapeutic target in metabolic syndrome-associated arterial stiffness.
Collapse
Affiliation(s)
- Yanan Liu
- Tianjin Institute of Cardiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Second Hospital of Tianjin Medical University, Tianjin Medical University, China. (Y.L., X.L., D.A.)
| | - Mengke Li
- Department of Physiology and Pathophysiology, Tianjin Medical University, China. (M.L., Y.Z., D.A.)
| | - Xue Lv
- Tianjin Institute of Cardiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Second Hospital of Tianjin Medical University, Tianjin Medical University, China. (Y.L., X.L., D.A.)
| | - Kaiwen Bao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, China. (K.B., L.S.)
| | - Xiao Yu Tian
- School of Biomedical Sciences, Chinese University of Hong Kong (X.Y.T., L.H.)
| | - Lei He
- School of Biomedical Sciences, Chinese University of Hong Kong (X.Y.T., L.H.)
| | - Lei Shi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, China. (K.B., L.S.)
| | - Yi Zhu
- Department of Physiology and Pathophysiology, Tianjin Medical University, China. (M.L., Y.Z., D.A.)
| | - Ding Ai
- Tianjin Institute of Cardiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Second Hospital of Tianjin Medical University, Tianjin Medical University, China. (Y.L., X.L., D.A.).,Department of Physiology and Pathophysiology, Tianjin Medical University, China. (M.L., Y.Z., D.A.)
| |
Collapse
|
28
|
Ugwoke CK, Cvetko E, Umek N. Skeletal Muscle Microvascular Dysfunction in Obesity-Related Insulin Resistance: Pathophysiological Mechanisms and Therapeutic Perspectives. Int J Mol Sci 2022; 23:ijms23020847. [PMID: 35055038 PMCID: PMC8778410 DOI: 10.3390/ijms23020847] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/09/2022] [Accepted: 01/11/2022] [Indexed: 02/04/2023] Open
Abstract
Obesity is a worrisomely escalating public health problem globally and one of the leading causes of morbidity and mortality from noncommunicable disease. The epidemiological link between obesity and a broad spectrum of cardiometabolic disorders has been well documented; however, the underlying pathophysiological mechanisms are only partially understood, and effective treatment options remain scarce. Given its critical role in glucose metabolism, skeletal muscle has increasingly become a focus of attention in understanding the mechanisms of impaired insulin function in obesity and the associated metabolic sequelae. We examined the current evidence on the relationship between microvascular dysfunction and insulin resistance in obesity. A growing body of evidence suggest an intimate and reciprocal relationship between skeletal muscle microvascular and glucometabolic physiology. The obesity phenotype is characterized by structural and functional changes in the skeletal muscle microcirculation which contribute to insulin dysfunction and disturbed glucose homeostasis. Several interconnected etiologic molecular mechanisms have been suggested, including endothelial dysfunction by several factors, extracellular matrix remodelling, and induction of oxidative stress and the immunoinflammatory phenotype. We further correlated currently available pharmacological agents that have deductive therapeutic relevance to the explored pathophysiological mechanisms, highlighting a potential clinical perspective in obesity treatment.
Collapse
|
29
|
Flores-Ramírez AG, Tovar-Villegas VI, Maharaj A, Garay-Sevilla ME, Figueroa A. Effects of L-Citrulline Supplementation and Aerobic Training on Vascular Function in Individuals with Obesity across the Lifespan. Nutrients 2021; 13:nu13092991. [PMID: 34578869 PMCID: PMC8466140 DOI: 10.3390/nu13092991] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/19/2021] [Accepted: 08/19/2021] [Indexed: 12/20/2022] Open
Abstract
Children with obesity are at higher risk for developing cardiometabolic diseases that once were considered health conditions of adults. Obesity is commonly associated with cardiometabolic risk factors such as dyslipidemia, hyperglycemia, hyperinsulinemia and hypertension that contribute to the development of endothelial dysfunction. Endothelial dysfunction, characterized by reduced nitric oxide (NO) production, precedes vascular abnormalities including atherosclerosis and arterial stiffness. Thus, early detection and treatment of cardiometabolic risk factors are necessary to prevent deleterious vascular consequences of obesity at an early age. Non-pharmacological interventions including L-Citrulline (L-Cit) supplementation and aerobic training stimulate endothelial NO mediated vasodilation, leading to improvements in organ perfusion, blood pressure, arterial stiffness, atherosclerosis and metabolic health (glucose control and lipid profile). Few studies suggest that the combination of L-Cit supplementation and exercise training can be an effective strategy to counteract the adverse effects of obesity on vascular function in older adults. Therefore, this review examined the efficacy of L-Cit supplementation and aerobic training interventions on vascular and metabolic parameters in obese individuals.
Collapse
Affiliation(s)
- Anaisa Genoveva Flores-Ramírez
- Department of Medical Science, Division of Health Science, University of Guanajuato, Campus León, León 37320, Mexico; (A.G.F.-R.); (V.I.T.-V.)
| | - Verónica Ivette Tovar-Villegas
- Department of Medical Science, Division of Health Science, University of Guanajuato, Campus León, León 37320, Mexico; (A.G.F.-R.); (V.I.T.-V.)
| | - Arun Maharaj
- Department of Kinesiology and Sport Management, Texas Tech University, Lubbock, TX 79409, USA;
| | - Ma Eugenia Garay-Sevilla
- Department of Medical Science, Division of Health Science, University of Guanajuato, Campus León, León 37320, Mexico; (A.G.F.-R.); (V.I.T.-V.)
- Correspondence: (M.E.G.-S.); (A.F.)
| | - Arturo Figueroa
- Department of Kinesiology and Sport Management, Texas Tech University, Lubbock, TX 79409, USA;
- Correspondence: (M.E.G.-S.); (A.F.)
| |
Collapse
|
30
|
Yanan Y, Yi J, Xiaojing L, Jing Q, Xiaohui W. Adipo-specific chemerin knockout alters the metabolomic profile of adipose tissue under normal and high-fat diet conditions: Application of an untargeted liquid chromatography-tandem mass spectrometry metabolomics method. Biomed Chromatogr 2021; 35:e5220. [PMID: 34323295 DOI: 10.1002/bmc.5220] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 06/17/2021] [Accepted: 07/25/2021] [Indexed: 12/17/2022]
Abstract
To explore the metabolic effect of chemerin, adipose-specific chemerin knockout (adipo-chemerin-/- ) male mice were established and fed with 5-week normal diet (ND) or high-fat diet (HFD), and then the glycolipid metabolism index was measured and epididymal adipose tissue metabolomics detected using untargeted LC-tandem mass spectrometry (LC-MS/MS). Under HFD, adipo-chemerin-/- mice showed improved glycolipid metabolism (decreased total cholesterol, low-density lipoprotein-cholesterol, insulin and Homeostasis Model Assessment of Insulin Resistance) compared with flox (control) mice. Furthermore, orthogonal partial least squares-discriminant analysis score plots identified separation of metabolites between adipo-chemerin-/- mice and flox mice fed ND and HFD. Under HFD, 28 metabolites were significantly enhanced in adipo-chemerin-/- mice, and pathway enrichment analysis suggested strong relationship of the differential metabolites with arginine and proline metabolism, phenylalanine metabolism, and phenylalanine, tyrosine and tryptophan biosynthesis, which were directly or indirectly related to lipid metabolism, inflammation and oxidative stress. Under ND, taurine was increased in adipo-chemerin-/- mice, resulting in taurine and hypotaurine metabolism and primary bile acid biosynthesis. In conclusion, the improved effect of chemerin knockdown on the glycolipid metabolism of HFD-feeding male mice might be associated with the increases in differential metabolites and metabolic pathways involved in lipid metabolism, inflammation and oxidative stress, which provided insights into the mechanism of chemerin from a metabolomics aspect.
Collapse
Affiliation(s)
- Yang Yanan
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Jia Yi
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Lin Xiaojing
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Qu Jing
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Wang Xiaohui
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
31
|
Ramirez-Perez FI, Woodford ML, Morales-Quinones M, Grunewald ZI, Cabral-Amador FJ, Yoshida T, Brenner DA, Manrique-Acevedo C, Martinez-Lemus LA, Chandrasekar B, Padilla J. Mutation of the 5'-untranslated region stem-loop mRNA structure reduces type I collagen deposition and arterial stiffness in male obese mice. Am J Physiol Heart Circ Physiol 2021; 321:H435-H445. [PMID: 34242094 PMCID: PMC8526337 DOI: 10.1152/ajpheart.00076.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Arterial stiffening, a characteristic feature of obesity and type 2 diabetes, contributes to the development and progression of cardiovascular diseases (CVD). Currently, no effective prophylaxis or therapeutics is available to prevent or treat arterial stiffening. A better understanding of the molecular mechanisms underlying arterial stiffening is vital to identify newer targets and strategies to reduce CVD burden. A major contributor to arterial stiffening is increased collagen deposition. In the 5'-untranslated regions of mRNAs encoding for type I collagen, an evolutionally conserved stem-loop (SL) structure plays an essential role in its stability and post-transcriptional regulation. Here, we show that feeding a high-fat/high-sucrose (HFHS) diet for 28 wk increases adiposity, insulin resistance, and blood pressure in male wild-type littermates. Moreover, arterial stiffness, assessed in vivo via aortic pulse wave velocity, and ex vivo using atomic force microscopy in aortic explants or pressure myography in isolated femoral and mesenteric arteries, was also increased in those mice. Notably, all these indices of arterial stiffness, along with collagen type I levels in the vasculature, were reduced in HFHS-fed mice harboring a mutation in the 5'SL structure, relative to wild-type littermates. This protective vascular phenotype in 5'SL-mutant mice did not associate with a reduction in insulin resistance or blood pressure. These findings implicate the 5'SL structure as a putative therapeutic target to prevent or reverse arterial stiffening and CVD associated with obesity and type 2 diabetes.NEW & NOTEWORTHY In the 5'-untranslated (UTR) regions of mRNAs encoding for type I collagen, an evolutionally conserved SL structure plays an essential role in its stability and posttranscriptional regulation. We demonstrate that a mutation of the SL mRNA structure in the 5'-UTR decreases collagen type I deposition and arterial stiffness in obese mice. Targeting this evolutionarily conserved SL structure may hold promise in the management of arterial stiffening and CVD associated with obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Francisco I Ramirez-Perez
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri.,Department of Biomedical, Biological and Chemical Engineering, University of Missouri, Columbia, Missouri
| | - Makenzie L Woodford
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri.,Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | | | - Zachary I Grunewald
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri.,Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | | | - Tadashi Yoshida
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - David A Brenner
- School of Medicine, University of California-San Diego, La Jolla, California
| | - Camila Manrique-Acevedo
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri.,Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, Missouri.,Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri
| | - Luis A Martinez-Lemus
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri.,Department of Biomedical, Biological and Chemical Engineering, University of Missouri, Columbia, Missouri.,Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Bysani Chandrasekar
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri.,Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri.,Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri.,Division of Cardiovascular Medicine, Department of Medicine, University of Missouri, Columbia, Missouri
| | - Jaume Padilla
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri.,Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| |
Collapse
|
32
|
Doghri Y, Dubreil L, Lalanne V, Hélissen O, Fleurisson R, Thorin C, Desfontis JC, Mallem MY. Soluble guanylate cyclase chronic stimulation effects on cardiovascular reactivity in cafeteria diet-induced rat model of metabolic syndrome. Eur J Pharmacol 2021; 899:173978. [PMID: 33691164 DOI: 10.1016/j.ejphar.2021.173978] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 02/05/2021] [Accepted: 02/19/2021] [Indexed: 11/19/2022]
Abstract
Metabolic syndrome is linked to an increased risk of cardiovascular complications by a mechanism involving mainly decreased nitric oxide (NO) bioavailability and impaired NO-soluble guanylate cyclase (sGC)- cyclic guanosine monophosphate (cGMP) signalling (NO-sGC-cGMP). To further develop this scientific point, this study aimed to investigate the effects of long-term treatment with BAY 41-2272 (a sGC stimulator) on cardiovascular reactivity of spontaneously hypertensive rats (SHR) as a model of metabolic syndrome. SHR were randomly divided into 3 groups: control group, cafeteria diet (CD)-fed group and CD-fed group treated daily with BAY 41-2272 (5 mg/kg) by gastric gavage for 12 weeks. In vivo measurements of body weight, abdominal circumference, blood pressure and glucose tolerance test were performed. At the end of the feeding period, ex vivo cumulative concentration-response curves were performed on isolated perfused heart (isoproterenol (0.1 nM - 1 μM)) and thoracic aorta (phenylephrine (1 nM-10 μM), acetylcholine (1 nM-10 μM), and sodium nitroprusside (SNP) (0.1 nM-0.1 μM)). We showed that chronic CD feeding induced abdominal obesity, hypertriglyceridemia, glucose intolerance and exacerbated arterial hypertension in SHR. Compared to control group, CD-fed group showed a decrease in β-adrenoceptor-induced cardiac inotropy, in coronary perfusion pressure and in aortic contraction to phenylephrine. While relaxing effects of acetylcholine and SNP were unchanged. BAY 41-2272 long-term treatment markedly prevented arterial hypertension development and glucose intolerance, enhanced the α1-adrenoceptor-induced vasoconstriction, and restored cardiac inotropy and coronary vasodilation. These findings suggest that BAY 41-2272 may be a potential novel drug for preventing metabolic and cardiovascular complications of metabolic syndrome.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/enzymology
- Aorta, Thoracic/physiopathology
- Cardiovascular Diseases/enzymology
- Cardiovascular Diseases/etiology
- Cardiovascular Diseases/physiopathology
- Cardiovascular Diseases/prevention & control
- Coronary Circulation/drug effects
- Cyclic GMP/metabolism
- Disease Models, Animal
- Enzyme Activation
- Enzyme Activators/pharmacology
- Glucose Intolerance/enzymology
- Glucose Intolerance/etiology
- Glucose Intolerance/physiopathology
- Glucose Intolerance/prevention & control
- Hypertension/enzymology
- Hypertension/etiology
- Hypertension/physiopathology
- Hypertension/prevention & control
- Hypertriglyceridemia/enzymology
- Hypertriglyceridemia/etiology
- Hypertriglyceridemia/physiopathology
- Hypertriglyceridemia/prevention & control
- Isolated Heart Preparation
- Male
- Metabolic Syndrome/enzymology
- Metabolic Syndrome/etiology
- Metabolic Syndrome/physiopathology
- Metabolic Syndrome/prevention & control
- Nitric Oxide Synthase Type II/metabolism
- Obesity, Abdominal/enzymology
- Obesity, Abdominal/etiology
- Obesity, Abdominal/physiopathology
- Obesity, Abdominal/prevention & control
- Pyrazoles/pharmacology
- Pyridines/pharmacology
- Rats, Inbred SHR
- Soluble Guanylyl Cyclase/metabolism
- Vasoconstriction/drug effects
- Vasodilation/drug effects
- Ventricular Function, Left/drug effects
- Ventricular Pressure/drug effects
- Rats
Collapse
Affiliation(s)
- Yosra Doghri
- UPSP NP3 (2017.B146), Nutrition, Pathophysiology and Pharmacology, Oniris, Nantes-Atlantic College of Veterinary Medicine Food Sciences and Engineering, 44307, Nantes Cedex 03, France
| | - Laurence Dubreil
- UMR PAnTher 703 INRA/Oniris Animal Pathophysiology and Bio Therapy for Muscle and Nervous System Diseases, Oniris, Nantes-Atlantic College of Veterinary Medicine Food Sciences and Engineering, 44307, Nantes Cedex 03, France
| | - Valérie Lalanne
- UPSP NP3 (2017.B146), Nutrition, Pathophysiology and Pharmacology, Oniris, Nantes-Atlantic College of Veterinary Medicine Food Sciences and Engineering, 44307, Nantes Cedex 03, France
| | - Ophélie Hélissen
- UPSP NP3 (2017.B146), Nutrition, Pathophysiology and Pharmacology, Oniris, Nantes-Atlantic College of Veterinary Medicine Food Sciences and Engineering, 44307, Nantes Cedex 03, France
| | - Romain Fleurisson
- UMR PAnTher 703 INRA/Oniris Animal Pathophysiology and Bio Therapy for Muscle and Nervous System Diseases, Oniris, Nantes-Atlantic College of Veterinary Medicine Food Sciences and Engineering, 44307, Nantes Cedex 03, France
| | - Chantal Thorin
- UPSP NP3 (2017.B146), Nutrition, Pathophysiology and Pharmacology, Oniris, Nantes-Atlantic College of Veterinary Medicine Food Sciences and Engineering, 44307, Nantes Cedex 03, France
| | - Jean-Claude Desfontis
- UPSP NP3 (2017.B146), Nutrition, Pathophysiology and Pharmacology, Oniris, Nantes-Atlantic College of Veterinary Medicine Food Sciences and Engineering, 44307, Nantes Cedex 03, France
| | - M Yassine Mallem
- UPSP NP3 (2017.B146), Nutrition, Pathophysiology and Pharmacology, Oniris, Nantes-Atlantic College of Veterinary Medicine Food Sciences and Engineering, 44307, Nantes Cedex 03, France.
| |
Collapse
|
33
|
Ogino N, Takahashi H, Nagaoka K, Harada Y, Kubo M, Miyagawa K, Kusanaga M, Oe S, Honma Y, Harada M, Eitoku M, Suganuma N, Ogino K. Possible contribution of hepatocyte secretion to the elevation of plasma exosomal arginase-1 in high-fat diet-fed mice. Life Sci 2021; 278:119588. [PMID: 33961860 DOI: 10.1016/j.lfs.2021.119588] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 04/16/2021] [Accepted: 04/29/2021] [Indexed: 10/21/2022]
Abstract
AIMS The elevation of arginase in vascular tissues decreases nitric oxide production, which is considered as an early step of atherosclerosis in obesity. Previously, we found that arginase-1, one of arginase isozymes, was elevated in the blood plasma of obese adults. The purpose of this study is to elucidate the mechanism by which obesity increases arginase-1 levels in the blood. MAIN METHODS C57/BL6J male mice fed a high-fat diet (HFD) for 12 weeks were analyzed for factors related to nitric oxide/arginine metabolism and plasma exosomes. To explore the arginase secretory organs, the protein expression levels were analyzed in several organs. To further investigate the relationship between exosomal arginase-1 in plasma, blood glucose levels and arginase-1 in the liver, HepG2 (the human hepatoma cell line) was analyzed after treatment with high glucose. KEY FINDINGS The increase in arginase activity in the plasma of HFD-fed mice was positively corelated with blood glucose levels and was accompanied by an increase in exosomal arginase-1 levels. Among the organs that highly express arginase, the liver of HFD-fed mice showed a significant increase in arginase-1. The expression of arginase-1 in exosomes and total lysates of HepG2 cells were increased by high glucose exposure. SIGNIFICANCE Increased exosomal arginase-1 in plasma contributes to increased plasma arginase activity in obesity. The liver is a candidate organ for the secretion of exosomal arginase-1 into plasma, and the p38 pathway induced by high glucose levels may be involved.
Collapse
Affiliation(s)
- Noriyoshi Ogino
- Department of Environmental Medicine, Kochi Medical School, Kochi University, Kohasu, Oko-cho, Nankoku, Kochi 783-8505, Japan; Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan
| | - Hidekazu Takahashi
- Division of Veterinary Medicine, Department of Public Health, Okayama University of Science, Imabari, Ehime 794-8555, Japan
| | - Kenjiro Nagaoka
- Laboratory of Hygienic Chemistry, College of Pharmaceutical Sciences, Matsuyama University, Matsuyama, Ehime 790-8578, Japan
| | - Yuki Harada
- Department of Biofunction Imaging Analysis, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama 7008530, Japan
| | - Masayuki Kubo
- Department of Environmental Medicine, Kochi Medical School, Kochi University, Kohasu, Oko-cho, Nankoku, Kochi 783-8505, Japan
| | - Koichiro Miyagawa
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan
| | - Masashi Kusanaga
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan
| | - Shinji Oe
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan
| | - Yuichi Honma
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan
| | - Masaru Harada
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan
| | - Masamitsu Eitoku
- Department of Environmental Medicine, Kochi Medical School, Kochi University, Kohasu, Oko-cho, Nankoku, Kochi 783-8505, Japan
| | - Narufumi Suganuma
- Department of Environmental Medicine, Kochi Medical School, Kochi University, Kohasu, Oko-cho, Nankoku, Kochi 783-8505, Japan
| | - Keiki Ogino
- Department of Environmental Medicine, Kochi Medical School, Kochi University, Kohasu, Oko-cho, Nankoku, Kochi 783-8505, Japan.
| |
Collapse
|
34
|
Moraes RDA, Webb RC, Silva DF. Vascular Dysfunction in Diabetes and Obesity: Focus on TRP Channels. Front Physiol 2021; 12:645109. [PMID: 33716794 PMCID: PMC7952965 DOI: 10.3389/fphys.2021.645109] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 02/09/2021] [Indexed: 01/22/2023] Open
Abstract
Transient receptor potential (TRP) superfamily consists of a diverse group of non-selective cation channels that has a wide tissue distribution and is involved in many physiological processes including sensory perception, secretion of hormones, vasoconstriction/vasorelaxation, and cell cycle modulation. In the blood vessels, TRP channels are present in endothelial cells, vascular smooth muscle cells, perivascular adipose tissue (PVAT) and perivascular sensory nerves, and these channels have been implicated in the regulation of vascular tone, vascular cell proliferation, vascular wall permeability and angiogenesis. Additionally, dysfunction of TRP channels is associated with cardiometabolic diseases, such as diabetes and obesity. Unfortunately, the prevalence of diabetes and obesity is rising worldwide, becoming an important public health problems. These conditions have been associated, highlighting that obesity is a risk factor for type 2 diabetes. As well, both cardiometabolic diseases have been linked to a common disorder, vascular dysfunction. In this review, we briefly consider general aspects of TRP channels, and we focus the attention on TRPC (canonical or classical), TRPV (vanilloid), TRPM (melastatin), and TRPML (mucolipin), which were shown to be involved in vascular alterations of diabetes and obesity or are potentially linked to vascular dysfunction. Therefore, elucidation of the functional and molecular mechanisms underlying the role of TRP channels in vascular dysfunction in diabetes and obesity is important for the prevention of vascular complications and end-organ damage, providing a further therapeutic target in the treatment of these metabolic diseases.
Collapse
Affiliation(s)
- Raiana Dos Anjos Moraes
- Laboratory of Cardiovascular Physiology and Pharmacology, Institute of Health Sciences, Federal University of Bahia, Salvador, Brazil.,Postgraduate Course in Biotechnology in Health and Investigative Medicine, Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
| | - R Clinton Webb
- Department of Cell Biology and Anatomy and Cardiovascular Translational Research Center, University of South Carolina, Columbia, SC, United States
| | - Darízy Flávia Silva
- Laboratory of Cardiovascular Physiology and Pharmacology, Institute of Health Sciences, Federal University of Bahia, Salvador, Brazil.,Postgraduate Course in Biotechnology in Health and Investigative Medicine, Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
| |
Collapse
|
35
|
Oxidative Stress and Vascular Damage in the Context of Obesity: The Hidden Guest. Antioxidants (Basel) 2021; 10:antiox10030406. [PMID: 33800427 PMCID: PMC7999611 DOI: 10.3390/antiox10030406] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 02/07/2023] Open
Abstract
The vascular system plays a central role in the transport of cells, oxygen and nutrients between different regions of the body, depending on the needs, as well as of metabolic waste products for their elimination. While the structure of different components of the vascular system varies, these structures, especially those of main arteries and arterioles, can be affected by the presence of different cardiovascular risk factors, including obesity. This vascular remodeling is mainly characterized by a thickening of the media layer as a consequence of changes in smooth muscle cells or excessive fibrosis accumulation. These vascular changes associated with obesity can trigger functional alterations, with endothelial dysfunction and vascular stiffness being especially common features of obese vessels. These changes can also lead to impaired tissue perfusion that may affect multiple tissues and organs. In this review, we focus on the role played by perivascular adipose tissue, the activation of the renin-angiotensin-aldosterone system and endoplasmic reticulum stress in the vascular dysfunction associated with obesity. In addition, the participation of oxidative stress in this vascular damage, which can be produced in the perivascular adipose tissue as well as in other components of the vascular wall, is updated.
Collapse
|
36
|
Sohouli MH, Fatahi S, Lari A, Lotfi M, Seifishahpar M, Găman MA, Rahideh ST, AlBatati SK, AlHossan AM, Alkhalifa SA, Alomar SA, Abu-Zaid A. The effect of paleolithic diet on glucose metabolism and lipid profile among patients with metabolic disorders: a systematic review and meta-analysis of randomized controlled trials. Crit Rev Food Sci Nutr 2021; 62:4551-4562. [PMID: 33492173 DOI: 10.1080/10408398.2021.1876625] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Several randomized clinical trials (RCTs) have investigated the effects of the Paleolithic diet (PD) in adult patients suffering from metabolic disorders. However, the results of these RCTs are conflicting. Therefore, we conducted a systematic review and meta-analysis to assess the effects of the PD in patients with metabolic disorders. METHODS We searched the PubMed/Medline, Scopus, Cochrane Databases, Google Scholar, Web of Science, and Embase databases up to June, 2020. The data were pooled using a random-effects model. From the eligible publications, 10 articles were selected for inclusion in this systematic review and meta-analysis. The meta-analysis was performed using a random-effects model. The heterogeneity was determined using the I2 statistics and the Cochrane Q test. RESULTS The pooled results from the random-effects model showed a significant reduction of the homeostatic model assessment of insulin resistance (HOMA-IR) (weighted mean difference, WMD: -0.39, 95% CI: -0.70, -0.08), fasting insulin (WMD: -12.17 μU/mL, 95% CI: -24.26, -0.08), total cholesterol (WMD: -0.32 mmol/l, 95% CI: -0.49, -0.15), triglycerides (WMD: -0.29 mmol/L, 95% CI: -0.42, -0.16), low-density lipoprotein cholesterol (WMD: -0.35 mmol/L, 95% CI: -0.67, -0.03), blood pressure (BP)(WMD - 5.89 mmHg; 95% CI - 9.973 to - 1.86 for the systolic BP and WMD - 4.01 mmHg; 95% CI - 6.21 to - 1.80 for the diastolic BP values) and C-reactive protein (CRP) levels (WMD: -0.84, mg/L, 95% CI: -1.62, -0.06) in the PD group versus control group. CONCLUSIONS Our findings provide better insights into the effect of the PD on the modulation of the glucose and lipid metabolism factors in patients with metabolic disorders, providing comprehensive information for the development of future RCTs with a high quality design.
Collapse
Affiliation(s)
- Mohammad Hassan Sohouli
- Faculty of Public Health Branch, Student Research Committee, Iran University of Medical Sciences, Tehran, Iran.,Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Somaye Fatahi
- Faculty of Public Health Branch, Student Research Committee, Iran University of Medical Sciences, Tehran, Iran.,Pediatric Gastroenterology, Hepatology and Nutrition Research Center, Research Institute for Children's Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abolfazl Lari
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Mojtaba Lotfi
- Department of Pediatric Endocrinology and Metabolism, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Seifishahpar
- Faculty of Nutrition and Food Technology, Department of Clinical Nutrition and Dietetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mihnea-Alexandru Găman
- "Carol Davila" University of Medicine and Pharmacy, 8 Eroii Sanitari Boulevard, Bucharest, Romania & Center of Hematology and Bone Marrow Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| | - Seyedeh Tayebeh Rahideh
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Saud K AlBatati
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | | | | | - Sara A Alomar
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Ahmed Abu-Zaid
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia.,College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
37
|
Wang Z, Liao W, Liu F, Yang T, Xie W, Liao M, Gu D, Zhang Y. Downregulation of lncRNA EPB41L4A-AS1 Mediates Activation of MYD88-Dependent NF-κB Pathway in Diabetes-Related Inflammation. Diabetes Metab Syndr Obes 2021; 14:265-277. [PMID: 33505165 PMCID: PMC7829128 DOI: 10.2147/dmso.s280765] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 12/30/2020] [Indexed: 12/13/2022] Open
Abstract
PURPOSE Long non-coding RNAs (lncRNAs) have been shown to be involved in many human diseases. In this study, we aimed to reveal the role and molecular mechanism of lncRNA EPB41L4A-AS1 in type 2 diabetic mellitus (T2DM)-related inflammation. METHODS To explore the relationships between the expression of EPB41L4A-AS1 and inflammatory factors in the blood of T2DM patients, we analyzed peripheral blood mononuclear cell (PBMC) expression microarrays of T2DM patients and expression microarrays of PBMC treated with lipopolysaccharide (LPS) from the GEO database. The relationship between EPB41L4A-AS1 and phospho-p65 was explored by Western blotting (WB) and immunofluorescence. The interactions between EPB41L4A-AS1 and myeloid differentiation factor 88 (MYD88) were also verified through quantitative real-time PCR, WB, and chromatin immunoprecipitation. Glycolysis and mitochondrial stress were detected by Seahorse. RESULTS EPB41L4A-AS1 showed very low expression, which was significantly negatively correlated with levels of inflammatory factors in PBMCs of T2DM patients and PBMCs treated with LPS. These results were verified by cell experiments on PBMC and THP-1 cells. Knockdown of EPB41L4A-AS1 led to the phosphorylation and nuclear translocation of p65 and thus activated the NF-κB signaling pathway; it also reduced the enrichment of H3K9me3 in the MYD88 promoter and increased expression of MYD88. Overall, EPB41L4A-AS1 knockdown promoted the level of glycolysis and ultimately enhanced the inflammatory response. CONCLUSION EPB41L4A-AS1 knockdown activated the NF-κB signaling pathway through a MYD88-dependent regulatory mechanism, promoted glycolysis, and ultimately enhanced the inflammatory response. These results demonstrate that EPB41L4A-AS1 is closely associated with inflammation in T2DM, and that low expression of EPB41L4A-AS1 may be used as an indicator of chronic inflammation and possible diabetic vascular complications in T2DM patients.
Collapse
Affiliation(s)
- Ziqing Wang
- School of Chemistry, Tsinghua University, Beijing100084, People’s Republic of China
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen518055, People’s Republic of China
| | - Weijie Liao
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen518055, People’s Republic of China
- Key Laboratory in Healthy Science and Technology, Division of Life Science, Tsinghua Shenzhen International Graduate School, Shenzhen518055, People’s Republic of China
| | - Fuhai Liu
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen518055, People’s Republic of China
- Department of Pathology, Xuzhou Medical University, Xuzhou221104, People’s Republic of China
| | - Tingpeng Yang
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen518055, People’s Republic of China
| | - Weidong Xie
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen518055, People’s Republic of China
- Key Laboratory in Healthy Science and Technology, Division of Life Science, Tsinghua Shenzhen International Graduate School, Shenzhen518055, People’s Republic of China
| | - Meijian Liao
- Key Laboratory in Healthy Science and Technology, Division of Life Science, Tsinghua Shenzhen International Graduate School, Shenzhen518055, People’s Republic of China
- Department of Pathology, Xuzhou Medical University, Xuzhou221104, People’s Republic of China
| | - Dayong Gu
- Department of Laboratory Medicine, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen518035, People’s Republic of China
- Dayong Gu Department of Laboratory Medicine, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen518035, People’s Republic of ChinaTel +86-13602601597 Email
| | - Yaou Zhang
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen518055, People’s Republic of China
- Key Laboratory in Healthy Science and Technology, Division of Life Science, Tsinghua Shenzhen International Graduate School, Shenzhen518055, People’s Republic of China
- Correspondence: Yaou Zhang State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen518055, People’s Republic of ChinaTel +86-755-2603-6884 Email
| |
Collapse
|
38
|
Gunawardena T, Merinopoulos I, Wickramarachchi U, Vassiliou V, Eccleshall S. Endothelial Dysfunction and Coronary Vasoreactivity - A Review of the History, Physiology, Diagnostic Techniques, and Clinical Relevance. Curr Cardiol Rev 2021; 17:85-100. [PMID: 32552654 PMCID: PMC8142375 DOI: 10.2174/1573403x16666200618161942] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 03/23/2020] [Accepted: 04/09/2020] [Indexed: 01/08/2023] Open
Abstract
The fervency for advancement and evolution in percutaneous coronary intervention has revolutionised the treatment of coronary artery disease. Historically, the focus of the interventional cardiologist was directed at the restoration of luminal patency of the major epicardial coronary arteries, yet whilst this approach is evolving with much greater utilisation of physiological assessment, it often neglects consideration of the role of the coronary microcirculation, which has been shown to clearly influence prognosis. In this review, we explore the narrative of the coronary circulation as more than just a simple conduit for blood but an organ with functional significance. We review organisation and physiology of the coronary circulation, as well as the current methods and techniques used to examine it. We discuss the studies exploring coronary artery endothelial function, appreciating that coronary artery disease occurs on a spectrum of disorder and that percutaneous coronary intervention has a latent effect on the coronary circulation with long-term consequences. It is concluded that greater recognition of the coronary artery endothelium and mechanisms of the coronary circulation should further guide revascularisation strategies.
Collapse
Affiliation(s)
- Tharusha Gunawardena
- Address correspondence to this author at the Department of Cardiology, Norfolk and Norwich University Hospital, Colney Lane NR4 7UY, Norwich, England; E-mail:
| | | | | | | | | |
Collapse
|
39
|
Obesity-Related Endothelial Dysfunction: moving from classical to emerging mechanisms. ENDOCRINE AND METABOLIC SCIENCE 2020. [DOI: 10.1016/j.endmts.2020.100063] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
40
|
Stein RA, Young LM. From ACE2 to COVID-19: A multiorgan endothelial disease. Int J Infect Dis 2020; 100:425-430. [PMID: 32896660 PMCID: PMC7832810 DOI: 10.1016/j.ijid.2020.08.083] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 08/31/2020] [Indexed: 02/06/2023] Open
Affiliation(s)
- Richard A Stein
- NYU Tandon School of Engineering, Department of Chemical and Biomolecular Engineering, 6 MetroTech Center, Brooklyn, NY 11201, USA; LaGuardia Community College, Department of Natural Sciences, City University of New York, New York, NY 11101, USA.
| | - Lauren M Young
- University of Chicago, Department of Internal Medicine, 5841 S Maryland Ave, Chicago, IL 60637, USA.
| |
Collapse
|
41
|
Mietus-Snyder M, Narayanan N, Krauss RM, Laine-Graves K, McCann JC, Shigenaga MK, McHugh TH, Ames BN, Suh JH. Randomized nutrient bar supplementation improves exercise-associated changes in plasma metabolome in adolescents and adult family members at cardiometabolic risk. PLoS One 2020; 15:e0240437. [PMID: 33079935 PMCID: PMC7575082 DOI: 10.1371/journal.pone.0240437] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 09/26/2020] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Poor diets contribute to metabolic complications of obesity, insulin resistance and dyslipidemia. Metabolomic biomarkers may serve as early nutrition-sensitive health indicators. This family-based lifestyle change program compared metabolic outcomes in an intervention group (INT) that consumed 2 nutrient bars daily for 2-months and a control group (CONT). METHODS Overweight, predominantly minority and female adolescent (Teen)/parent adult caretaker (PAC) family units were recruited from a pediatric obesity clinic. CONT (8 Teen, 8 PAC) and INT (10 Teen, 10 PAC) groups randomized to nutrient bar supplementation attended weekly classes that included group nutrition counseling and supervised exercise. Pre-post physical and behavioral parameters, fasting traditional biomarkers, plasma sphingolipids and amino acid metabolites were measured. RESULTS In the full cohort, a baseline sphingolipid ceramide principal component composite score correlated with adiponectin, triglycerides, triglyceride-rich very low density lipoproteins, and atherogenic small low density lipoprotein (LDL) sublasses. Inverse associations were seen between a sphingomyelin composite score and C-reactive protein, a dihydroceramide composite score and diastolic blood pressure, and the final principal component that included glutathionone with fasting insulin and the homeostatic model of insulin resistance. In CONT, plasma ceramides, sphinganine, sphingosine and amino acid metabolites increased, presumably due to increased physical activity. Nutrient bar supplementation (INT) blunted this rise and significantly decreased ureagenic, aromatic and gluconeogenic amino acid metabolites. Metabolomic changes were positively correlated with improvements in clinical biomarkers of dyslipidemia. CONCLUSION Nutrient bar supplementation with increased physical activity in obese Teens and PAC elicits favorable metabolomic changes that correlate with improved dyslipidemia. The trial from which the analyses reported upon herein was part of a series of nutrient bar clinical trials registered at clinicaltrials.gov as NCT02239198.
Collapse
Affiliation(s)
- Michele Mietus-Snyder
- Division of Cardiology, Department of Pediatrics, Children’s National Hospital, George Washington University School of Medicine and Health Sciences, Washington DC, United States of America
| | - Nisha Narayanan
- Weill Cornell Medical College, Cornell University, New York, New York, United States of America
| | - Ronald M. Krauss
- University of California Benioff Children’s Hospital San Francisco, San Francisco, California, United States of America
- Children’s Hospital Oakland Research Institute, Oakland, California, United States of America
| | - Kirsten Laine-Graves
- University of California Benioff Children’s Hospital Oakland Research Institute, Oakland, California, United States of America
| | - Joyce C. McCann
- University of California Benioff Children’s Hospital Oakland Research Institute, Oakland, California, United States of America
| | - Mark K. Shigenaga
- University of California Benioff Children’s Hospital Oakland Research Institute, Oakland, California, United States of America
| | - Tara H. McHugh
- United States Department of Agriculture, Western Regional Research Center, Albany, California, United States of America
| | - Bruce N. Ames
- University of California Benioff Children’s Hospital Oakland Research Institute, Oakland, California, United States of America
| | - Jung H. Suh
- University of California Benioff Children’s Hospital Oakland Research Institute, Oakland, California, United States of America
| |
Collapse
|
42
|
Xia Y, Zhang Z, Lin W, Yan J, Zhu C, Yin D, He S, Su Y, Xu N, Caldwell RW, Yao L, Chen Y. Modulating microglia activation prevents maternal immune activation induced schizophrenia-relevant behavior phenotypes via arginase 1 in the dentate gyrus. Neuropsychopharmacology 2020; 45:1896-1908. [PMID: 32599605 PMCID: PMC7608378 DOI: 10.1038/s41386-020-0743-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 06/10/2020] [Accepted: 06/16/2020] [Indexed: 12/31/2022]
Abstract
Prenatal infection during pregnancy increases the risk for developing neuropsychiatric disorders such as schizophrenia. This is linked to an inflammatory microglial phenotype in the offspring induced by maternal immune activation (MIA). Microglia are crucial for brain development and maintenance of neuronal niches, however, whether and how their activation is involved in the regulation of neurodevelopment remains unclear. Here, we used a MIA rodent model in which polyinosinic: polycytidylic acid (poly (I:C)) was injected into pregnant mice. We found fewer parvalbumin positive (PV+) cells and impaired GABAergic transmission in the dentate gyrus (DG), accompanied by schizophrenia-like behavior in the adult offspring. Minocycline, a potent inhibitor of microglia activation, successfully prevented the above-mentioned deficits in the offspring. Furthermore, by using microglia-specific arginase 1 (Arg1) ablation as well as overexpression in DG, we identified a critical role of Arg1 in microglia activation to protect against poly (I:C) imparted neuropathology and altered behavior in offspring. Taken together, our results highlight that Arg1-mediated alternative activation of microglia are potential therapeutic targets for psychiatric disorders induced by MIA.
Collapse
Affiliation(s)
- Yucen Xia
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006 China
| | - Zhiqing Zhang
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006 China
| | - Weipeng Lin
- grid.22069.3f0000 0004 0369 6365Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, Shanghai, 200062 China
| | - Jinglan Yan
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006 China
| | - Chuan’an Zhu
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006 China
| | - Dongmin Yin
- grid.22069.3f0000 0004 0369 6365Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, Shanghai, 200062 China
| | - Su He
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006 China
| | - Yang Su
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006 China
| | - Nenggui Xu
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006 China
| | - Robert William Caldwell
- grid.410427.40000 0001 2284 9329Department of Pharmacology and Toxicology, Augusta University, Augusta, GA 30912 USA
| | - Lin Yao
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China. .,School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Yongjun Chen
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China. .,Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao Greater Bay Area, Guangzhou, 510515, China. .,Guangdong Province Key Laboratory of Psychiatric Disorders, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
43
|
Acacetin improves endothelial dysfunction and aortic fibrosis in insulin-resistant SHR rats by estrogen receptors. Mol Biol Rep 2020; 47:6899-6918. [PMID: 32892299 PMCID: PMC7561596 DOI: 10.1007/s11033-020-05746-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 08/28/2020] [Indexed: 12/20/2022]
Abstract
The aim of the work was to investigate the effects of acacetin on endothelial dysfunction and aortic fibrosis in insulin-resistant SHR rats and explore its mechanism. Seven-week-old male spontaneously hypertensive rats (SHR) were selected to establish a rat model of hypertension with insulin resistance induced by 10% fructose. The nuclear factor kappa B p65 (NF-κB p65) and Collagen I were observed by Immunohistochemistry. Immunofluorescence was used to observe estrogen receptor-alpha (ERα), estrogen receptor-beta (ERβ), and G protein-coupled receptor 30 (GPR30). Western blotting was used to detect interleukin (IL-1β), Arginase 2 (ARG2), Nostrin, endothelial nitric oxide synthase (eNOS), TGF-β, Smad3, ERK pathway proteins such as p-c-Raf, p-MEK1/2, p-ERK, ERK, p-P90RSK and p-MSK1. We found that acacetin did have an improvement on endothelial dysfunction and fibrosis. Meanwhile, it was also found to have a significant effect on the level of estrogen in this model by accident. Then, the experiment of uterine weight gain in mice confirmed that acacetin had a certain estrogen-like effect in vivo and played its role through the estrogen receptors pathway. In vitro experience HUVEC cells were stimulated with 30 mM/L glucose and 100 mM/L NaCl for 24 h to establish the endothelial cell injury model. HUVEC cells were treated with 1 μM/L estrogen receptors antagonist (ICI 182780) for 30 min before administration. Cell experiments showed that acacetin could reduce the apoptosis of HUVEC cells, the levels of inflammatory cytokines and the expression of TGF-β, Collagen I and Smad3 in endothelial cell injury model. After treatment with ICI 182780, the improvement of acacetin was significantly reversed. The results showed that acacetin relieved endothelial dysfunction and reduced the aortic fibrosis in insulin-resistant SHR rats by reducing the release of inflammatory factors and improving vasodilatory function through estrogen signaling pathway.
Collapse
|
44
|
Grunewald ZI, Ramirez-Perez FI, Woodford ML, Morales-Quinones M, Mejia S, Manrique-Acevedo C, Siebenlist U, Martinez-Lemus LA, Chandrasekar B, Padilla J. TRAF3IP2 (TRAF3 Interacting Protein 2) Mediates Obesity-Associated Vascular Insulin Resistance and Dysfunction in Male Mice. Hypertension 2020; 76:1319-1329. [PMID: 32829657 DOI: 10.1161/hypertensionaha.120.15262] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Insulin resistance in the vasculature is a characteristic feature of obesity and contributes to the pathogenesis of vascular dysfunction and disease. However, the molecular mechanisms underlying obesity-associated vascular insulin resistance and dysfunction remain poorly understood. We hypothesized that TRAF3IP2 (TRAF3 interacting protein 2), a proinflammatory adaptor molecule known to activate pathological stress pathways and implicated in cardiovascular diseases, plays a causal role in obesity-associated vascular insulin resistance and dysfunction. We tested this hypothesis by employing genetic-manipulation in endothelial cells in vitro, in isolated arteries ex vivo, and diet-induced obesity in a mouse model of TRAF3IP2 ablation in vivo. We show that ectopic expression of TRAF3IP2 blunts insulin signaling in endothelial cells and diminishes endothelium-dependent vasorelaxation in isolated aortic rings. Further, 16 weeks of high fat/high sucrose feeding impaired glucose tolerance, aortic insulin-induced vasorelaxation, and hindlimb postocclusive reactive hyperemia, while increasing blood pressure and arterial stiffness in wild-type male mice. Notably, TRAF3IP2 ablation protected mice from such high fat/high sucrose feeding-induced metabolic and vascular defects. Interestingly, wild-type female mice expressed markedly reduced levels of TRAF3IP2 mRNA independent of diet and were protected against high fat/high sucrose diet-induced vascular dysfunction. These data indicate that TRAF3IP2 plays a causal role in vascular insulin resistance and dysfunction. Specifically, the present findings highlight a sexual dimorphic role of TRAF3IP2 in vascular control and identify it as a promising therapeutic target in vasculometabolic derangements associated with obesity, particularly in males.
Collapse
Affiliation(s)
- Zachary I Grunewald
- From the Department of Nutrition and Exercise Physiology (Z.I.G., M.L.W., J.P.), University of Missouri, Columbia.,Dalton Cardiovascular Research Center (Z.I.G., F.I.R.-P., M.L.W., M.M.-Q., S.M., C.M.-A., L.A.M.-L., B.C., J.P.), University of Missouri, Columbia
| | - Francisco I Ramirez-Perez
- Dalton Cardiovascular Research Center (Z.I.G., F.I.R.-P., M.L.W., M.M.-Q., S.M., C.M.-A., L.A.M.-L., B.C., J.P.), University of Missouri, Columbia.,Department of Biological Engineering (F.I.R.-P., L.A.M.-L.), University of Missouri, Columbia
| | - Makenzie L Woodford
- From the Department of Nutrition and Exercise Physiology (Z.I.G., M.L.W., J.P.), University of Missouri, Columbia.,Dalton Cardiovascular Research Center (Z.I.G., F.I.R.-P., M.L.W., M.M.-Q., S.M., C.M.-A., L.A.M.-L., B.C., J.P.), University of Missouri, Columbia
| | - Mariana Morales-Quinones
- Dalton Cardiovascular Research Center (Z.I.G., F.I.R.-P., M.L.W., M.M.-Q., S.M., C.M.-A., L.A.M.-L., B.C., J.P.), University of Missouri, Columbia
| | - Salvador Mejia
- Dalton Cardiovascular Research Center (Z.I.G., F.I.R.-P., M.L.W., M.M.-Q., S.M., C.M.-A., L.A.M.-L., B.C., J.P.), University of Missouri, Columbia
| | - Camila Manrique-Acevedo
- Dalton Cardiovascular Research Center (Z.I.G., F.I.R.-P., M.L.W., M.M.-Q., S.M., C.M.-A., L.A.M.-L., B.C., J.P.), University of Missouri, Columbia.,Division of Endocrinology and Metabolism, Department of Medicine (C.M.-A.), University of Missouri, Columbia.,Harry S. Truman Memorial Veterans' Hospital, Columbia, MO (C.M.-A., B.C.)
| | | | - Luis A Martinez-Lemus
- Dalton Cardiovascular Research Center (Z.I.G., F.I.R.-P., M.L.W., M.M.-Q., S.M., C.M.-A., L.A.M.-L., B.C., J.P.), University of Missouri, Columbia.,Department of Biological Engineering (F.I.R.-P., L.A.M.-L.), University of Missouri, Columbia.,Department of Medical Pharmacology and Physiology (L.A.M.-L., B.C.), University of Missouri, Columbia
| | - Bysani Chandrasekar
- Dalton Cardiovascular Research Center (Z.I.G., F.I.R.-P., M.L.W., M.M.-Q., S.M., C.M.-A., L.A.M.-L., B.C., J.P.), University of Missouri, Columbia.,Division of Cardiovascular Medicine, Department of Medicine (B.C.), University of Missouri, Columbia.,Department of Medical Pharmacology and Physiology (L.A.M.-L., B.C.), University of Missouri, Columbia.,Harry S. Truman Memorial Veterans' Hospital, Columbia, MO (C.M.-A., B.C.)
| | - Jaume Padilla
- From the Department of Nutrition and Exercise Physiology (Z.I.G., M.L.W., J.P.), University of Missouri, Columbia.,Dalton Cardiovascular Research Center (Z.I.G., F.I.R.-P., M.L.W., M.M.-Q., S.M., C.M.-A., L.A.M.-L., B.C., J.P.), University of Missouri, Columbia
| |
Collapse
|
45
|
S. Clemente G, van Waarde A, F. Antunes I, Dömling A, H. Elsinga P. Arginase as a Potential Biomarker of Disease Progression: A Molecular Imaging Perspective. Int J Mol Sci 2020; 21:E5291. [PMID: 32722521 PMCID: PMC7432485 DOI: 10.3390/ijms21155291] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 12/11/2022] Open
Abstract
Arginase is a widely known enzyme of the urea cycle that catalyzes the hydrolysis of L-arginine to L-ornithine and urea. The action of arginase goes beyond the boundaries of hepatic ureogenic function, being widespread through most tissues. Two arginase isoforms coexist, the type I (Arg1) predominantly expressed in the liver and the type II (Arg2) expressed throughout extrahepatic tissues. By producing L-ornithine while competing with nitric oxide synthase (NOS) for the same substrate (L-arginine), arginase can influence the endogenous levels of polyamines, proline, and NO•. Several pathophysiological processes may deregulate arginase/NOS balance, disturbing the homeostasis and functionality of the organism. Upregulated arginase expression is associated with several pathological processes that can range from cardiovascular, immune-mediated, and tumorigenic conditions to neurodegenerative disorders. Thus, arginase is a potential biomarker of disease progression and severity and has recently been the subject of research studies regarding the therapeutic efficacy of arginase inhibitors. This review gives a comprehensive overview of the pathophysiological role of arginase and the current state of development of arginase inhibitors, discussing the potential of arginase as a molecular imaging biomarker and stimulating the development of novel specific and high-affinity arginase imaging probes.
Collapse
Affiliation(s)
- Gonçalo S. Clemente
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (G.S.C.); (A.v.W.); (I.F.A.)
| | - Aren van Waarde
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (G.S.C.); (A.v.W.); (I.F.A.)
| | - Inês F. Antunes
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (G.S.C.); (A.v.W.); (I.F.A.)
| | - Alexander Dömling
- Department of Drug Design, Groningen Research Institute of Pharmacy, University of Groningen, 9713 AV Groningen, The Netherlands;
| | - Philip H. Elsinga
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (G.S.C.); (A.v.W.); (I.F.A.)
| |
Collapse
|
46
|
Almeida JFQ, Shults N, de Souza AMA, Ji H, Wu X, Woods J, Sandberg K. Short-term very low caloric intake causes endothelial dysfunction and increased susceptibility to cardiac arrhythmias and pathology in male rats. Exp Physiol 2020; 105:1172-1184. [PMID: 32410300 PMCID: PMC7496402 DOI: 10.1113/ep088434] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 05/05/2020] [Indexed: 01/03/2023]
Abstract
New Findings What is the central question of this study? What are the effects of a 2 week period of severe food restriction on vascular reactivity of resistance arteries and on cardiac structure and function? What is the main finding and its importance? This study showed, for the first time, that a 2 week period of severe food restriction in adult male Fischer rats caused endothelial dysfunction in mesenteric arteries and increased the susceptibility to ischaemia–reperfusion‐induced arrhythmias and cardiac pathology. Our findings might have ramifications for cardiovascular risk in people who experience periods of inadequate caloric intake.
Abstract Severe food restriction (sFR) is a common dieting strategy for rapid weight loss. Male Fischer rats were maintained on a control (CT) or sFR (40% of CT food intake) diet for 14 days to mimic low‐calorie crash diets. The sFR diet reduced body weight by 16%. Haematocrits were elevated by 10% in the sFR rats, which was consistent with the reduced plasma volume. Mesenteric arteries from sFR rats had increased sensitivity to vasoconstrictors, including angiotensin II [maximum (%): CT, 1.30 ± 0.46 versus sFR, 11.5 ± 1.6; P < 0.0001; n = 7] and phenylephrine [maximum (%): CT, 78.5 ± 2.8 versus sFR, 94.5 ± 1.7; P < 0.001; n = 7] and reduced sensitivity to the vasodilator acetylcholine [EC50 (nm): CT, 49.2 ± 5.2 versus sFR, 71.6 ± 6.8; P < 0.05; n = 7]. Isolated hearts from sFR rats had a 1.7‐fold increase in the rate of cardiac arrhythmias in response to ischaemia–reperfusion and more cardiac pathology, including myofibrillar disarray with contractions and cardiomyocyte lysis, than hearts from CT rats. The sFR dietary regimen is similar to very low‐calorie commercial and self‐help weight‐loss programmes, which provide ∼800–1000 kcal day−1. Therefore, these findings in rats warrant the study of cardiovascular function in individuals who engage in extreme dieting or are subjected to bouts of very low caloric intake for other reasons, such as socioeconomic factors and natural disasters.
Collapse
Affiliation(s)
| | - Nataliia Shults
- Department of Pharmacology & Physiology, Georgetown University, Washington, DC, USA
| | | | - Hong Ji
- Department of Medicine, Georgetown University, Washington, DC, USA
| | - Xie Wu
- Department of Medicine, Georgetown University, Washington, DC, USA
| | - James Woods
- Department of Biostatistics, Bioinformatics and Biomathematics, Georgetown University, Washington, DC, USA
| | - Kathryn Sandberg
- Department of Medicine, Georgetown University, Washington, DC, USA
| |
Collapse
|
47
|
Su E, Zhao L, Yang X, Zhu B, Liu Y, Zhao W, Wang X, Qi D, Zhu L, Gao C. Aggravated endothelial endocrine dysfunction and intimal thickening of renal artery in high-fat diet-induced obese pigs following renal denervation. BMC Cardiovasc Disord 2020; 20:176. [PMID: 32295540 PMCID: PMC7161153 DOI: 10.1186/s12872-020-01472-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 04/07/2020] [Indexed: 12/11/2022] Open
Abstract
Background Renal denervation (RDN) targeting the sympathetic nerves in the renal arterial adventitia as a treatment of resistant hypertension can cause endothelial injury and vascular wall injury. This study aims to evaluate the risk of atherosclerosis induced by RDN in renal arteries. Methods A total of 15 minipigs were randomly assigned to 3 groups: (1) control group, (2) sham group, and (3) RDN group (n = 5 per group). All pigs were fed a high-fat diet (HFD) for 6 months after appropriate treatment. The degree of intimal thickening of renal artery and the conversion of endothelin 1 (ET-1) receptors were evaluated by histological staining. Western blot was used to assess the expression of nitric oxide (NO) synthesis signaling pathway, ET-1 and its receptors, NADPH oxidase 2 (NOX2) and 4-hydroxynonenal (4-HNE) proteins, and the activation of NF-kappa B (NF-κB). Results The histological staining results suggested that compared to the sham treatment, RDN led to significant intimal thickening and significantly promoted the production of endothelin B receptor (ETBR) in vascular smooth muscle cells (VSMCs). Western blotting analysis indicated that RDN significantly suppressed the expression of AMPK/Akt/eNOS signaling pathway proteins, and decreased the production of NO, and increased the expression of endothelin system proteins including endothelin-1 (ET-1), endothelin converting enzyme 1 (ECE1), endothelin A receptor (ETAR) and ETBR; and upregulated the expression of NOX2 and 4-HNE proteins and enhanced the activation of NF-kappa B (NF-κB) when compared with the sham treatment (all p < 0.05). There were no significant differences between the control and sham groups (all p > 0.05). Conclusions RDN aggravated endothelial endocrine dysfunction and intimal thickening, and increased the risk of atherosclerosis in renal arteries of HFD-fed pigs.
Collapse
Affiliation(s)
- Enyong Su
- Department of Cardiology, Zhengzhou University People's Hospital, No.7 Weiwu road, Jinshui District, Zhengzhou, 450003, Henan, China.,Department of Cardiology, Huazhong Fuwai Hospital, Zhengzhou, 451464, Henan, China
| | - Linwei Zhao
- Department of Cardiology, Zhengzhou University People's Hospital, No.7 Weiwu road, Jinshui District, Zhengzhou, 450003, Henan, China.,Department of Cardiology, Huazhong Fuwai Hospital, Zhengzhou, 451464, Henan, China
| | - Xiaohang Yang
- Department of Cardiology, Zhengzhou University People's Hospital, No.7 Weiwu road, Jinshui District, Zhengzhou, 450003, Henan, China
| | - Binbin Zhu
- Department of Cardiology, Zhengzhou University People's Hospital, No.7 Weiwu road, Jinshui District, Zhengzhou, 450003, Henan, China.,Department of Cardiology, Huazhong Fuwai Hospital, Zhengzhou, 451464, Henan, China
| | - Yahui Liu
- Department of Cardiology, Huazhong Fuwai Hospital, Zhengzhou, 451464, Henan, China
| | - Wen Zhao
- Zhengzhou University School of Pharmaceutical Sciences, Zhengzhou, 450001, Henan, China
| | - Xianpei Wang
- Department of Cardiology, Zhengzhou University People's Hospital, No.7 Weiwu road, Jinshui District, Zhengzhou, 450003, Henan, China.,Department of Cardiology, Huazhong Fuwai Hospital, Zhengzhou, 451464, Henan, China
| | - Datun Qi
- Department of Cardiology, Zhengzhou University People's Hospital, No.7 Weiwu road, Jinshui District, Zhengzhou, 450003, Henan, China.,Department of Cardiology, Huazhong Fuwai Hospital, Zhengzhou, 451464, Henan, China
| | - Lijie Zhu
- Department of Cardiology, Zhengzhou University People's Hospital, No.7 Weiwu road, Jinshui District, Zhengzhou, 450003, Henan, China.,Department of Cardiology, Huazhong Fuwai Hospital, Zhengzhou, 451464, Henan, China
| | - Chuanyu Gao
- Department of Cardiology, Zhengzhou University People's Hospital, No.7 Weiwu road, Jinshui District, Zhengzhou, 450003, Henan, China. .,Department of Cardiology, Huazhong Fuwai Hospital, Zhengzhou, 451464, Henan, China.
| |
Collapse
|
48
|
Ye G, Gao H, Huang Q, Lin Y, Liao X, Zhang H, Yang BC. Metabolomic Characterization of Metabolic Disturbances in the Extracellular Microenvironment of Oleate-Treated Macrophages Using Gas Chromatography–Mass Spectrometry. ANAL LETT 2020. [DOI: 10.1080/00032719.2020.1750623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Guozhu Ye
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, China
| | - Han Gao
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qiansheng Huang
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, China
| | - Yi Lin
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, China
| | - Xu Liao
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, China
| | - Han Zhang
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, China
| | - Bi-cheng Yang
- Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| |
Collapse
|
49
|
Sun Y, Venugopal J, Guo C, Fan Y, Li J, Gong Y, Chen YE, Zhang H, Eitzman DT. Clopidogrel Resistance in a Murine Model of Diet-Induced Obesity Is Mediated by the Interleukin-1 Receptor and Overcome With DT-678. Arterioscler Thromb Vasc Biol 2020; 40:1533-1542. [PMID: 32268786 DOI: 10.1161/atvbaha.120.314146] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Clopidogrel is a commonly used P2Y12 inhibitor to treat and prevent arterial thrombotic events. Clopidogrel is a prodrug that requires bioactivation by CYP (cytochrome P450) enzymes to exert antiplatelet activity. Diabetes mellitus is associated with an increased risk of ischemic events, and impaired ability to generate the active metabolite (AM) from clopidogrel. The objective of this study is to identify the mechanism of clopidogrel resistance in a murine model of diet-induced obesity (DIO). Approach and Results: C57BL/6J mice and IL-1R-/- mice were given high-fat diet for 10 weeks to generate a murine model of diet-induced obesity. Platelet aggregation and carotid arterial thrombosis were assessed in response to clopidogrel treatment. Wild-type DIO mice exhibited resistance to antiplatelet and antithrombotic effects of clopidogrel that was associated with reduced hepatic expression of CYP genes and reduced generation of the AM. IL (Interleukin)-1 receptor-deficient DIO (IL1R-/- DIO) mice showed no resistance to clopidogrel. Lack of resistance was accompanied by increased exposure of the clopidogrel AM. This resistance was also absent when wild-type DIO mice were treated with the conjugate of the clopidogrel AM, DT-678. CONCLUSIONS These findings indicate that antiplatelet effects of clopidogrel may be impaired in the setting of diabetes mellitus due to reduced prodrug bioactivation related to IL-1 receptor signaling. Therapeutic targeting of P2Y12 in patients with diabetes mellitus using the conjugate of clopidogrel AM may lead to improved outcomes.
Collapse
Affiliation(s)
- Yifang Sun
- From the Department of Internal Medicine, Cardiovascular Research Center, University of Michigan Medical Center, Ann Arbor (Y.S., J.V., C.G., Y.F., Y.E.C., D.T.E.).,Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China (Y.S.)
| | - Jessica Venugopal
- From the Department of Internal Medicine, Cardiovascular Research Center, University of Michigan Medical Center, Ann Arbor (Y.S., J.V., C.G., Y.F., Y.E.C., D.T.E.)
| | - Chiao Guo
- From the Department of Internal Medicine, Cardiovascular Research Center, University of Michigan Medical Center, Ann Arbor (Y.S., J.V., C.G., Y.F., Y.E.C., D.T.E.)
| | - Yanbo Fan
- From the Department of Internal Medicine, Cardiovascular Research Center, University of Michigan Medical Center, Ann Arbor (Y.S., J.V., C.G., Y.F., Y.E.C., D.T.E.)
| | - Jianping Li
- Department of Cardiology, Peking University First Hospital, Beijing, China (J.L., Y.G.)
| | - Yanjun Gong
- Department of Cardiology, Peking University First Hospital, Beijing, China (J.L., Y.G.)
| | - Y Eugene Chen
- From the Department of Internal Medicine, Cardiovascular Research Center, University of Michigan Medical Center, Ann Arbor (Y.S., J.V., C.G., Y.F., Y.E.C., D.T.E.)
| | - Haoming Zhang
- Department of Pharmacology, University of Michigan, Ann Arbor (H.Z.)
| | - Daniel T Eitzman
- From the Department of Internal Medicine, Cardiovascular Research Center, University of Michigan Medical Center, Ann Arbor (Y.S., J.V., C.G., Y.F., Y.E.C., D.T.E.)
| |
Collapse
|
50
|
Cheng H, Lu T, Wang J, Xia Y, Chai X, Zhang M, Yao Y, Zhou N, Zhou S, Chen X, Su W, Liu C, Yi W, Chen Y, Yao L. HuangqiGuizhiWuwu Decoction Prevents Vascular Dysfunction in Diabetes via Inhibition of Endothelial Arginase 1. Front Physiol 2020; 11:201. [PMID: 32269530 PMCID: PMC7109290 DOI: 10.3389/fphys.2020.00201] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 02/21/2020] [Indexed: 12/12/2022] Open
Abstract
Hyperglycemia induces vascular endothelial dysfunction, which contributes to the development of vascular complication of diabetes. A classic prescription of traditional medicine, HuangqiGuizhiWuwu Decoction (HGWWD) has been used for the treatment of various cardiovascular and cerebrovascular diseases, which all are related with vascular pathology. The present study investigated the effect of HGWWD treatment in streptozocin (STZ)-induced vascular dysfunction in mouse models. In vivo studies were performed using wild type mice as well as arginase 1 knockout specific in endothelial cells (EC-A1-/-) of control mice, diabetes mice and diabetes mice treated with HGWWD (60 g crude drugs/kg/d) for 2 weeks. For in vitro studies, aortic tissues were treated with mice serum containing HGWWD with or without adenoviral arginase 1 (Ad-A1) transduction in high glucose (HG) medium. We found that HGWWD treatment restored STZ-induced impaired mean velocity and pulsatility index of mouse left femoral arteries, aortic pulse wave velocity and vascular endothelial relaxation accompanied by elevated NO production in the aorta and plasma, as well as reduced endothelial arginase activity and aortic arginase 1 expression. The protective effect of HGWWD is reversed by an inhibitor of nitric oxide synthesis. Meanwhile, the preventive effect of serum containing HGWWD in endothelial vascular dysfunction is completely blocked by Ad-A1 transduction in HG incubated aortas. HGWWD treatment further improved endothelial vascular dysfunction in STZ induced EC-A1-/- mice. This study demonstrates that HGWWD improved STZ-induced vascular dysfunction through arginase 1 - NO signaling, specifically targeting endothelial arginase 1.
Collapse
Affiliation(s)
- Hong Cheng
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tian Lu
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jingya Wang
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yucen Xia
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoshu Chai
- Department of Oncology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Minyi Zhang
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yutong Yao
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Na Zhou
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Sisi Zhou
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinyi Chen
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Weiwei Su
- Guangdong Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Cunzhi Liu
- Acupuncture Research Center, Beijing University of Chinese Medicine, Beijing, China
| | - Wei Yi
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yongjun Chen
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lin Yao
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|