1
|
Narayanan S, Vuckovic S, Bergman O, Wirka R, Verdezoto Mosquera J, Chen QS, Baldassarre D, Tremoli E, Veglia F, Lengquist M, Aherahrrou R, Razuvaev A, Gigante B, Björck HM, Miller CL, Quertermous T, Hedin U, Matic L. Atheroma transcriptomics identifies ARNTL as a smooth muscle cell regulator and with clinical and genetic data improves risk stratification. Eur Heart J 2024:ehae768. [PMID: 39552248 DOI: 10.1093/eurheartj/ehae768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 02/10/2024] [Accepted: 10/23/2024] [Indexed: 11/19/2024] Open
Abstract
BACKGROUND AND AIMS The role of vascular smooth muscle cells (SMCs) in atherosclerosis has evolved to indicate causal genetic links with the disease. Single cell RNA sequencing (scRNAseq) studies have identified multiple cell populations of mesenchymal origin within atherosclerotic lesions, including various SMC sub-phenotypes, but it is unknown how they relate to patient clinical parameters and genetics. Here, mesenchymal cell populations in atherosclerotic plaques were correlated with major coronary artery disease (CAD) genetic variants and functional analyses performed to identify SMC markers involved in the disease. METHODS Bioinformatic deconvolution was done on bulk microarrays from carotid plaques in the Biobank of Karolinska Endarterectomies (BiKE, n = 125) using public plaque scRNAseq data and associated with patient clinical data and follow-up information. BiKE patients were clustered based on the deconvoluted cell fractions. Quantitative trait loci (QTLs) analyses were performed to predict the effect of CAD associated genetic variants on mesenchymal cell fractions (cfQTLs) and gene expression (eQTLs) in plaques. RESULTS Lesions from symptomatic patients had higher fractions of Type 1 macrophages and pericytes, but lower fractions of classical and modulated SMCs compared with asymptomatic ones, particularly females. Presence of diabetes or statin treatment did not affect the cell fraction distribution. Clustering based on plaque cell fractions, revealed three patient groups, with relative differences in their stability profiles and associations to stroke, even during long-term follow-up. Several single nucleotide polymorphisms associated with plaque mesenchymal cell fractions, upstream of the circadian rhythm gene ARNTL were identified. In vitro silencing of ARNTL in human carotid SMCs increased the expression of contractile markers and attenuated cell proliferation. CONCLUSIONS This study shows the potential of combining scRNAseq data with vertically integrated clinical, genetic, and transcriptomic data from a large biobank of human plaques, for refinement of patient vulnerability and risk prediction stratification. The study revealed novel CAD-associated variants that may be functionally linked to SMCs in atherosclerotic plaques. Specifically, variants in the ARNTL gene may influence SMC ratios and function, and its role as a regulator of SMC proliferation should be further investigated.
Collapse
Affiliation(s)
- Sampath Narayanan
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska University Hospital and Karolinska Institutet, BioClinicum J8:20, Visionsgatan 4, SE-171 76 Stockholm, Sweden
| | - Sofija Vuckovic
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska University Hospital and Karolinska Institutet, BioClinicum J8:20, Visionsgatan 4, SE-171 76 Stockholm, Sweden
| | - Otto Bergman
- Division of Cardiovascular Medicine, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Karolinska University Hospital, Solna, Sweden
| | - Robert Wirka
- Department of Medicine and Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | - Qiao Sen Chen
- Division of Cardiovascular Medicine, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Karolinska University Hospital, Solna, Sweden
| | - Damiano Baldassarre
- Centro Cardiologico Monzino, IRCCS, Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, Università di Milano, Milan, Italy
| | | | | | - Mariette Lengquist
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska University Hospital and Karolinska Institutet, BioClinicum J8:20, Visionsgatan 4, SE-171 76 Stockholm, Sweden
| | - Redouane Aherahrrou
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Institute for Cardiogenetics, Universität zu Lübeck; DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany
- University Heart Centre Lübeck, Lübeck, Germany
| | - Anton Razuvaev
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska University Hospital and Karolinska Institutet, BioClinicum J8:20, Visionsgatan 4, SE-171 76 Stockholm, Sweden
| | - Bruna Gigante
- Division of Cardiovascular Medicine, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Karolinska University Hospital, Solna, Sweden
| | - Hanna M Björck
- Division of Cardiovascular Medicine, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Karolinska University Hospital, Solna, Sweden
| | - Clint L Miller
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Thomas Quertermous
- Division of Cardiovascular Medicine, Stanford University, Stanford, CA, USA
| | - Ulf Hedin
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska University Hospital and Karolinska Institutet, BioClinicum J8:20, Visionsgatan 4, SE-171 76 Stockholm, Sweden
| | - Ljubica Matic
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska University Hospital and Karolinska Institutet, BioClinicum J8:20, Visionsgatan 4, SE-171 76 Stockholm, Sweden
| |
Collapse
|
2
|
Han X, Xu S, Hu K, Yu Y, Wang X, Qu C, Yang B, Liu X. Early growth response 1 exacerbates thoracic aortic aneurysm and dissection of mice by inducing the phenotypic switching of vascular smooth muscle cell through the activation of Krüppel-like factor 5. Acta Physiol (Oxf) 2024; 240:e14237. [PMID: 39345002 DOI: 10.1111/apha.14237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 08/15/2024] [Accepted: 09/14/2024] [Indexed: 10/01/2024]
Abstract
AIM Vascular smooth muscle cell (VSMC) phenotypic switching has been reported to regulate vascular function and thoracic aortic aneurysm and dissection (TAAD) progression. Early growth response 1 (Egr1) is associated with the differentiation of VSMCs. However, the mechanisms through which Egr1 participates in the regulation of VSMCs and progression of TAAD remain unknown. This study aimed to investigate the role of Egr1 in the phenotypic switching of VSMCs and the development of TAAD. METHODS Wild-type C57BL/6 and SMC-specific Egr1-knockout mice were used as experimental subjects and fed β-aminopropionitrile for 4 weeks to construct the TAAD model. Ultrasound and aortic staining were performed to examine the pathological features in thoracic aortic tissues. Transwell, wound healing, CCK8, and immunofluorescence assays detected the migration and proliferation of synthetic VSMCs. Egr1 was directly bound to the promoter of Krüppel-like factor 5 (KLF5) and promoted the expression of KLF5, which was validated by JASPAR database and dual-luciferase reporter assay. RESULTS Egr1 expression increased and was partially co-located with VSMCs in aortic tissues of mice with TAAD. SMC-specific Egr1 deficiency alleviated TAAD and inhibited the phenotypic switching of VSMC. Egr1 knockdown prevented the phenotypic switching of VSMCs and subsequently suppressed the migration and proliferation of synthetic VSMCs. The inhibitory effects of Egr1 deficiency on VSMCs were blunted once KLF5 was overexpressed. CONCLUSION Egr1 aggravated the development of TAAD by promoting the phenotypic switching of VSMCs via enhancing the transcriptional activation of KLF5. These results suggest that inhibition of SMC-specific Egr1 expression is a promising therapy for TAAD.
Collapse
MESH Headings
- Animals
- Early Growth Response Protein 1/metabolism
- Early Growth Response Protein 1/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Kruppel-Like Transcription Factors/metabolism
- Kruppel-Like Transcription Factors/genetics
- Mice
- Aortic Aneurysm, Thoracic/metabolism
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/pathology
- Aortic Dissection/metabolism
- Aortic Dissection/pathology
- Aortic Dissection/genetics
- Mice, Inbred C57BL
- Mice, Knockout
- Phenotype
- Male
- Myocytes, Smooth Muscle/metabolism
- Disease Models, Animal
- Cell Proliferation
Collapse
Affiliation(s)
- Xueyu Han
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Shengnan Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Ke Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Xiukun Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Chuan Qu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Bo Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Xin Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| |
Collapse
|
3
|
da Silveira EJD, Barros CCDS, Bottino MC, Castilho RM, Squarize C. The rhythms of histones in regeneration: The epigenetic modifications determined by clock genes. Exp Dermatol 2024; 33:e15005. [PMID: 38284199 PMCID: PMC10865818 DOI: 10.1111/exd.15005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 10/30/2023] [Accepted: 12/18/2023] [Indexed: 01/30/2024]
Abstract
The evolutionary establishment of an internal biological clock is a primordial event tightly associated with a 24-h period. Changes in the circadian rhythm can affect cellular functions, including proliferation, DNA repair and redox state. Even isolated organs, tissues and cells can maintain an autonomous circadian rhythm. These cell-autonomous molecular mechanisms are driven by intracellular clock genes, such as BMAL1. Little is known about the role of core clock genes and epigenetic modifications in the skin. Our focus was to identify BMAL1-driven epigenetic modifications associated with gene transcription by mapping the acetylation landscape of histones in epithelial cells responding to injury. We explored the role of BMAL1 in epidermal wound and tissue regeneration using a loss-of-function approach in vivo. We worked with BMAL1 knockout mice and a contraction-resistance wound healing protocol, determining the histone modifications using specific antibodies to detect the acetylation levels of histones H3 and H4. We found significant differences in the acetylation levels of histones in both homeostatic and injured skin with deregulated BMAL1. The intact skin displayed varied acetylation levels of histones H3 and H4, including hyperacetylation of H3 Lys 9 (H3K9). The most pronounced changes were observed at the repair site, with notable alterations in the acetylation pattern of histone H4. These findings reveal the importance of histone modifications in response to injury and indicate that modulation of BMAL1 and its associated epigenetic events could be therapeutically harnessed to improve skin regeneration.
Collapse
Affiliation(s)
- Ericka J. D. da Silveira
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
- Department of Dentistry, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Caio C. D. S. Barros
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Marco C. Bottino
- Department of Cariology, Restorative Sciences, and Endodontics, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Rogerio M. Castilho
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
- Michigan Medicine, University of Michigan Rogel Cancer Center, Ann Arbor, Michigan, USA
| | - Cristiane Squarize
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
- Michigan Medicine, University of Michigan Rogel Cancer Center, Ann Arbor, Michigan, USA
| |
Collapse
|
4
|
Khachigian LM. The MEK-ERK-Egr-1 axis and its regulation in cardiovascular disease. Vascul Pharmacol 2023; 153:107232. [PMID: 37734428 DOI: 10.1016/j.vph.2023.107232] [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: 08/10/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 09/23/2023]
Abstract
Cardiovascular disease (CVD) is the primary cause of morbidity and mortality in the Western world. Multiple molecular and cellular processes underpinning the pathogenesis of CVD are regulated by the zinc finger transcription factor and product of an immediate-early gene, early growth response-1 (Egr-1). Egr-1 regulates multiple pro-inflammatory processes that underpin the manifestation of CVD. The activity of Egr-1 itself is influenced by a range of post-translational modifications including sumoylation, ubiquitination and acetylation. Egr-1 also undergoes phosphorylation by protein kinases, such as extracellular-signal regulated kinase (ERK) which is itself phosphorylated by MEK. This article reviews recent progress on the MEK-ERK-Egr-1 cascade, notably regulation in conjunction with factors and agents such as TET2, TRIB2, MIAT, SphK1, cAMP, teneligliptin, cholinergic drugs, red wine and flavonoids, wogonin, febuxostat, docosahexaenoic acid and AT1R blockade. Such insights should provide new opportunity for therapeutic intervention in CVD.
Collapse
Affiliation(s)
- Levon M Khachigian
- Vascular Biology and Translational Research, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
5
|
Rios FJ, Montezano AC, Camargo LL, Touyz RM. Impact of Environmental Factors on Hypertension and Associated Cardiovascular Disease. Can J Cardiol 2023; 39:1229-1243. [PMID: 37422258 DOI: 10.1016/j.cjca.2023.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/24/2023] [Accepted: 07/02/2023] [Indexed: 07/10/2023] Open
Abstract
Hypertension is the primary cause of cardiovascular diseases and is responsible for nearly 9 million deaths worldwide annually. Increasing evidence indicates that in addition to pathophysiologic processes, numerous environmental factors, such as geographic location, lifestyle choices, socioeconomic status, and cultural practices, influence the risk, progression, and severity of hypertension, even in the absence of genetic risk factors. In this review, we discuss the impact of some environmental determinants on hypertension. We focus on clinical data from large population studies and discuss some potential molecular and cellular mechanisms. We highlight how these environmental determinants are interconnected, as small changes in one factor might affect others, and further affect cardiovascular health. In addition, we discuss the crucial impact of socioeconomic factors and how these determinants influence diverse communities with economic disparities. Finally, we address opportunities and challenges for new research to address gaps in knowledge on understanding molecular mechanisms whereby environmental factors influence development of hypertension and associated cardiovascular disease.
Collapse
Affiliation(s)
- Francisco J Rios
- Research Institute of the McGill University Health Centre, Montréal, Québec, Canada.
| | - Augusto C Montezano
- Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Livia L Camargo
- Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Rhian M Touyz
- Research Institute of the McGill University Health Centre, Montréal, Québec, Canada.
| |
Collapse
|
6
|
Jin W, Tian Y, Ding Y, Zhou D, Li L, Yuan M, Wu Y, Ye M, Luan J, Yang K. Pers reverse angiotensin II -induced vascular smooth muscle cell proliferation by targeting cyclin E expression via inhibition of the MAPK signaling pathway. Chronobiol Int 2023; 40:903-917. [PMID: 37338051 DOI: 10.1080/07420528.2023.2224904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 06/01/2023] [Accepted: 06/08/2023] [Indexed: 06/21/2023]
Abstract
The circadian rhythm of blood pressure (BP) is believed to be regulated by the clock system, which is closely linked to levels of angiotensin II (Ang II). This study aimed to investigate whether Ang II mediates the proliferation of vascular smooth muscle cells (VSMCs) through the interaction between the clock system and the mitogen-activated protein kinase (MAPK) signaling pathway. Primary rat aortic VSMCs were treated with Ang II, with or without MAPK inhibitors. VSMC proliferation, expression of clock genes, CYCLIN E, and MAPK pathways were assessed. Ang II treatment resulted in increased VSMC proliferation and rapid upregulation of clock gene Periods (Pers) expression. Compared to the non-diseased control (NC) group, VSMCs incubated with Ang II displayed a noticeable delay in the G1/S phase transition and downregulation of CYCLIN E upon silencing of Per1 and Per2 genes. Importantly, silencing Per1 or Per2 in VSMCs led to decreased expression of key MAPK pathway proteins, including RAS, phosphorylated mitogen-activated protein kinase (P-MEK), and phosphorylated extracellular signal-regulated protein kinase (P-ERK). Moreover, the MEK and ERK inhibitors, U0126 and SCH772986, significantly attenuated the Ang II-induced proliferation of VSMCs, as evidenced by an increased G1/S phase transition and decreased CYCLIN E expression. The MAPK pathway plays a critical role in regulating VSMC proliferation in response to Ang II stimulation. This regulation is controlled by the expression of circadian clock genes involved in the cell cycle. These findings provide novel insights for further research on diseases associated with abnormal VSMC proliferation.
Collapse
Affiliation(s)
- Wan Jin
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui, P.R. China
- Department of Pharmacy, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, P.R. China
| | - Yu Tian
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui, P.R. China
- Department of Pharmacy, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, P.R. China
| | - Yanyun Ding
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui, P.R. China
- Department of Pharmacy, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, P.R. China
| | - Deixi Zhou
- Department of Pharmacy, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, P.R. China
| | - Long Li
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui, P.R. China
- Department of Pharmacy, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, P.R. China
| | - Meng Yuan
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui, P.R. China
- Department of Pharmacy, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, P.R. China
| | - Yuanzhu Wu
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui, P.R. China
- Department of Pharmacy, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, P.R. China
| | - Mingqi Ye
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui, P.R. China
- Department of Pharmacy, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, P.R. China
| | - Jiajie Luan
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui, P.R. China
- Department of Pharmacy, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, P.R. China
| | - Kui Yang
- Department of Pharmacy, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, P.R. China
| |
Collapse
|
7
|
Uruski P, Matuszewska J, Leśniewska A, Rychlewski D, Niklas A, Mikuła-Pietrasik J, Tykarski A, Książek K. An integrative review of nonobvious puzzles of cellular and molecular cardiooncology. Cell Mol Biol Lett 2023; 28:44. [PMID: 37221467 DOI: 10.1186/s11658-023-00451-y] [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: 02/22/2023] [Accepted: 04/17/2023] [Indexed: 05/25/2023] Open
Abstract
Oncologic patients are subjected to four major treatment types: surgery, radiotherapy, chemotherapy, and immunotherapy. All nonsurgical forms of cancer management are known to potentially violate the structural and functional integrity of the cardiovascular system. The prevalence and severity of cardiotoxicity and vascular abnormalities led to the emergence of a clinical subdiscipline, called cardiooncology. This relatively new, but rapidly expanding area of knowledge, primarily focuses on clinical observations linking the adverse effects of cancer therapy with deteriorated quality of life of cancer survivors and their increased morbidity and mortality. Cellular and molecular determinants of these relations are far less understood, mainly because of several unsolved paths and contradicting findings in the literature. In this article, we provide a comprehensive view of the cellular and molecular etiology of cardiooncology. We pay particular attention to various intracellular processes that arise in cardiomyocytes, vascular endothelial cells, and smooth muscle cells treated in experimentally-controlled conditions in vitro and in vivo with ionizing radiation and drugs representing diverse modes of anti-cancer activity.
Collapse
Affiliation(s)
- Paweł Uruski
- Department of Hypertensiology, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Julia Matuszewska
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Aleksandra Leśniewska
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Daniel Rychlewski
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Arkadiusz Niklas
- Department of Hypertensiology, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Justyna Mikuła-Pietrasik
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Andrzej Tykarski
- Department of Hypertensiology, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Krzysztof Książek
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland.
| |
Collapse
|
8
|
Yang C, Deng Z, Zeng Q, Chang X, Wu X, Li G. BMAL1 involved in autophagy and injury of thoracic aortic endothelial cells of rats induced by intermittent heat stress through the AMPK/mTOR/ULK1 pathway. Biochem Biophys Res Commun 2023; 661:34-41. [PMID: 37086572 DOI: 10.1016/j.bbrc.2023.04.035] [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: 02/21/2023] [Revised: 03/30/2023] [Accepted: 04/14/2023] [Indexed: 04/24/2023]
Abstract
Physiological activities of the body exhibit an obvious biological rhythm. At the core of the circadian rhythm, BMAL1 is the only clock gene whose deletion leads to abnormal physiological functions. However, whether intermittent heat stress influences cardiovascular function by altering the circadian rhythm of clock genes has not been reported. This study aimed to investigate whether intermittent heat stress induces autophagy and apoptosis, and the effects of BMAL1 on thoracic aortic autophagy and apoptosis. An intermittent heat stress model was established in vitro, and western blotting and immunofluorescence were used to detect the expression of autophagy, apoptosis, the AMPK/mTOR/ULK1 pathway, and BMAL1. After BMAL1 silencing, RT-qPCR was performed to detect the expression levels of autophagy and apoptosis-related genes. Our results suggest that heat stress induces autophagy and apoptosis in RTAECs. In addition, intermittent heat stress increased the phosphorylation of AMPK and ULK1, but reduced the phosphorylation of mTOR, AMPK inhibitor Compound C reversed the phosphorylation of AMPK, mTOR, and ULK1, and Beclin1 and LC3-II/LC3-I were downregulated. Furthermore, BMAL1 expression was elevated in vitro and shBMAL1 decreased autophagy and apoptosis. We revealed that intermittent heat stress induces autophagy and apoptosis, and that BMAL1 may be involved in the occurrence of autophagy and apoptosis.
Collapse
Affiliation(s)
- Chunli Yang
- School of Public Health, Ningxia Medical University, Yinchuan, Ningxia, 750004, China; Nursing Department, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, 750004, China
| | - Ziwei Deng
- School of Public Health, Ningxia Medical University, Yinchuan, Ningxia, 750004, China
| | - Qihang Zeng
- Institute of Basic Medical Science, Ningxia Medical University, Yinchuan, Ningxia, 750004, China
| | - Xiaoyu Chang
- School of Public Health, Ningxia Medical University, Yinchuan, Ningxia, 750004, China
| | - Xiaomin Wu
- Institute of Basic Medical Science, Ningxia Medical University, Yinchuan, Ningxia, 750004, China
| | - Guanghua Li
- School of Public Health, Ningxia Medical University, Yinchuan, Ningxia, 750004, China; Institute of Basic Medical Science, Ningxia Medical University, Yinchuan, Ningxia, 750004, China.
| |
Collapse
|
9
|
Takaguri A, Ishizaka R, Maki S, Satoh K. The role of tribbles homolog 2 in vascular smooth muscle cell proliferation. Cell Biol Int 2023; 47:787-795. [PMID: 36626273 DOI: 10.1002/cbin.11982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 12/09/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023]
Abstract
Tribbles homolog 2 (TRIB2) functions as an adapter protein that regulates signal transductions involved in a variety of cellular functions, including tumorigenesis. However, the role of TRIB2 in the proliferation of vascular smooth muscle cells (VSMCs) and the underlying expression mechanisms remain unclear. The present study investigated the role of TRIB2 in VSMC proliferation and revealed that TRIB2 expression increases following vascular injury and platelet-derived growth factor (PDGF)-BB-stimulated VSMCs. We found that pretreatment with diphenyleneiodonium (a nicotinamide adenine dinucleotide phosphate oxidase inhibitor), U0126 (an inhibitor of mitogen-activated protein kinase kinase 1 [MEK1]), or siRNA targeting the gene encoding early growth response 1 (EGR-1) significantly inhibits PDGF-BB-induced TRIB2 expression in VSMCs. Furthermore, TRIB2 knockdown significantly inhibits PDGF-BB-induced proliferation of VSMCs but does not affect the phosphorylation of AKT. However, phosphorylation of ERK1 and expression of proliferating cell nuclear antibody are significantly suppressed in VSMCs by PDGF-BB stimulation. Thus, PDGF-BB-induced TRIB2 expression is mediated by ROS/ERK/EGR-1 pathways and plays a critical role in VSMC proliferation via modulation of ERK activity. We propose TRIB2 as a promising therapeutic target for the prevention of neointima formation and vascular disease.
Collapse
Affiliation(s)
- Akira Takaguri
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, Sapporo, Teine-ku, Japan
| | - Rena Ishizaka
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, Sapporo, Teine-ku, Japan
| | - Shota Maki
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, Sapporo, Teine-ku, Japan
| | - Kumi Satoh
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, Sapporo, Teine-ku, Japan
| |
Collapse
|
10
|
Yao J, Liang J, Li H. Screening for key genes in circadian regulation in advanced atherosclerosis: A bioinformatic analysis. Front Cardiovasc Med 2023; 9:990757. [PMID: 36712250 PMCID: PMC9878187 DOI: 10.3389/fcvm.2022.990757] [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: 07/10/2022] [Accepted: 12/28/2022] [Indexed: 01/15/2023] Open
Abstract
Background Atherosclerosis (AS) is the most important cardiovascular disease threatening human health, leading to adverse events such as myocardial infarction and stroke. The research on the pathogenesis and causes of AS is being improved step by step, and many factors are associated with AS. However, the relationship between circadian regulation and the pathogenesis of AS is still unclear. Our study identified 2 key genes of circadian regulation in AS by bioinformatics analysis, which provides new perspectives to understand the relationship between circadian rhythm and AS. Methods We downloaded samples of early and advanced AS from public databases, screened key genes by weighted gene co-expression network analysis (WGCNA) and Lasso, calculated the immune cell content of the samples using "CIBERSORT," and analyzed the relationship between key genes and immune cells. Results We obtained the most relevant core modules for advanced AS and analyzed the functions of these modules. Two circadian rhythm-related genes were obtained, which influence the immune infiltration of this late AS. ROC curves demonstrated the efficacy of key genes to differentiate between early and advanced AS. Conclusion We identified 2 genes most associated with circadian rhythms in advanced AS, whose association with AS has not been elucidated and may become the next therapeutic target.
Collapse
Affiliation(s)
- Jiali Yao
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Jingyan Liang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China,Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou, Jiangsu, China
| | - Hongliang Li
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China,Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou, Jiangsu, China,*Correspondence: Hongliang Li,
| |
Collapse
|
11
|
Bax M, Romanov V, Junday K, Giannoulatou E, Martinac B, Kovacic JC, Liu R, Iismaa SE, Graham RM. Arterial dissections: Common features and new perspectives. Front Cardiovasc Med 2022; 9:1055862. [PMID: 36561772 PMCID: PMC9763901 DOI: 10.3389/fcvm.2022.1055862] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/16/2022] [Indexed: 12/12/2022] Open
Abstract
Arterial dissections, which involve an abrupt tear in the wall of a major artery resulting in the intramural accumulation of blood, are a family of catastrophic disorders causing major, potentially fatal sequelae. Involving diverse vascular beds, including the aorta or coronary, cervical, pulmonary, and visceral arteries, each type of dissection is devastating in its own way. Traditionally they have been studied in isolation, rather than collectively, owing largely to the distinct clinical consequences of dissections in different anatomical locations - such as stroke, myocardial infarction, and renal failure. Here, we review the shared and unique features of these arteriopathies to provide a better understanding of this family of disorders. Arterial dissections occur commonly in the young to middle-aged, and often in conjunction with hypertension and/or migraine; the latter suggesting they are part of a generalized vasculopathy. Genetic studies as well as cellular and molecular investigations of arterial dissections reveal striking similarities between dissection types, particularly their pathophysiology, which includes the presence or absence of an intimal tear and vasa vasorum dysfunction as a cause of intramural hemorrhage. Pathway perturbations common to all types of dissections include disruption of TGF-β signaling, the extracellular matrix, the cytoskeleton or metabolism, as evidenced by the finding of mutations in critical genes regulating these processes, including LRP1, collagen genes, fibrillin and TGF-β receptors, or their coupled pathways. Perturbances in these connected signaling pathways contribute to phenotype switching in endothelial and vascular smooth muscle cells of the affected artery, in which their physiological quiescent state is lost and replaced by a proliferative activated phenotype. Of interest, dissections in various anatomical locations are associated with distinct sex and age predilections, suggesting involvement of gene and environment interactions in disease pathogenesis. Importantly, these cellular mechanisms are potentially therapeutically targetable. Consideration of arterial dissections as a collective pathology allows insight from the better characterized dissection types, such as that involving the thoracic aorta, to be leveraged to inform the less common forms of dissections, including the potential to apply known therapeutic interventions already clinically available for the former.
Collapse
Affiliation(s)
- Monique Bax
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Valentin Romanov
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Keerat Junday
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Eleni Giannoulatou
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Boris Martinac
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Jason C. Kovacic
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
- St. Vincent’s Hospital, Darlinghurst, NSW, Australia
- Icahn School of Medicine at Mount Sinai, Cardiovascular Research Institute, New York, NY, United States
| | - Renjing Liu
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Siiri E. Iismaa
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Robert M. Graham
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
- St. Vincent’s Hospital, Darlinghurst, NSW, Australia
| |
Collapse
|
12
|
Shen Y, Xu LR, Yan D, Zhou M, Han TL, Lu C, Tang X, Lin CP, Qian RZ, Guo DQ. BMAL1 modulates smooth muscle cells phenotypic switch towards fibroblast-like cells and stabilizes atherosclerotic plaques by upregulating YAP1. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166450. [PMID: 35598770 DOI: 10.1016/j.bbadis.2022.166450] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 05/03/2022] [Accepted: 05/13/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Ischemic heart diseases and ischemic stroke are closely related to circadian clock and unstable atherosclerotic plaques. Vascular smooth muscle cells (VSMCs) can stabilize or destabilize an atherosclerotic lesion through phenotypic switch. BMAL1 is not only an indispensable core component in circadian clock but also an important regulator in atherosclerosis and VSMCs proliferation. However, little is known about the modulation mechanisms of BMAL1 in VSMCs phenotypic switch and atherosclerotic plaque stability. METHODS We integrated histological analysis of human plaques, in vivo experiments of VSMC-specific Bmal1-/- mice, in vitro experiments, and gene set enrichment analysis (GSEA) of public datasets of human plaques to explore the function of BMAL1 in VSMCs phonotypic switch and plaque stability. FINDINGS Comparing to human unstable plaques, BMAL1 was higher in stable plaques, accompanied by elevated YAP1 and fibroblast maker FSP1 which were positively correlated with BMAL1. In response to Methyl-β-cyclodextrin-cholesterol, oxidized-low-density-lipoprotein and platelet-derived-growth-factor-BB, VSMCs embarked on phenotypic switch and upregulated BMAL, YAP1 and FSP1. Besides, BMAL1 overexpression promoted VSMCs phonotypic switch towards fibroblast-like cells by transcriptionally upregulating the expression of YAP1. BMAL1 or YAP1 knock-down inhibited VSMCs phonotypic switch and downregulated FSP1. Furthermore, VSMC-specific Bmal1-/- mice exhibited VSMCs with lower YAP1 and FSP1 levels, and more vulnerable plaques with less collagen content. In addition, BMAL1 suppressed the migration of VSMCs. The GSEA results of public datasets were consistent with our laboratory findings. INTERPRETATION Our results highlight the importance of BMAL1 as a major regulator in VSMCs phenotypic switch towards fibroblast-like cells which stabilize an atherosclerotic plaque.
Collapse
Affiliation(s)
- Yang Shen
- Department of Vascular Surgery, Institute of Vascular Surgery, National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China
| | - Li-Rong Xu
- Department of Pathology, School of Basic Medical Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Dong Yan
- Department of Vascular Surgery, Institute of Vascular Surgery, National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China
| | - Min Zhou
- Department of Vascular Surgery, Institute of Vascular Surgery, National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China
| | - Tong-Lei Han
- Department of Vascular Surgery, Institute of Vascular Surgery, National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China
| | - Chao Lu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, 138 Yixueyuan Rd., Shanghai 200032, China
| | - Xiao Tang
- Department of Vascular Surgery, Institute of Vascular Surgery, National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China
| | - Chang-Po Lin
- Department of Vascular Surgery, Institute of Vascular Surgery, National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China.
| | - Rui-Zhe Qian
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, 138 Yixueyuan Rd., Shanghai 200032, China.
| | - Da-Qiao Guo
- Department of Vascular Surgery, Institute of Vascular Surgery, National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China.
| |
Collapse
|
13
|
Zhang Y, Liu L, Zhao X, Yan S, Zeng F, Zhou D. New insight into ischemic stroke: Circadian rhythm in post-stroke angiogenesis. Front Pharmacol 2022; 13:927506. [PMID: 36016550 PMCID: PMC9395980 DOI: 10.3389/fphar.2022.927506] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/07/2022] [Indexed: 11/29/2022] Open
Abstract
The circadian rhythm is an endogenous clock system that coordinates and optimizes various physiological and pathophysiological processes, which accord with the master and the peripheral clock. Increasing evidence indicates that endogenous circadian rhythm disruption is involved in the lesion volume and recovery of ischemic stroke. As a critical recovery mechanism in post-stroke, angiogenesis reestablishes the regional blood supply and enhances cognitive and behavioral abilities, which is mainly composed of the following processes: endothelial cell proliferation, migration, and pericyte recruitment. The available evidence revealed that the circadian governs many aspects of angiogenesis. This study reviews the mechanism by which circadian rhythms regulate the process of angiogenesis and its contribution to functional recovery in post-stroke at the aspects of the molecular level. A comprehensive understanding of the circadian clock regulating angiogenesis in post-stroke is expected to develop new strategies for the treatment of cerebral infarction.
Collapse
Affiliation(s)
- Yuxing Zhang
- The Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, China
| | - Lijuan Liu
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xin Zhao
- The Medical School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Siyang Yan
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Fukang Zeng
- The Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, China
| | - Desheng Zhou
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- *Correspondence: Desheng Zhou,
| |
Collapse
|
14
|
Sun Z, Li L, Yan Z, Zhang L, Zang G, Qian Y, Wang Z. Circadian rhythm disorders elevate macrophages cytokines release and promote multiple tissues/organs dysfunction in mice. Physiol Behav 2022; 249:113772. [PMID: 35247442 DOI: 10.1016/j.physbeh.2022.113772] [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: 09/20/2021] [Revised: 02/02/2022] [Accepted: 03/01/2022] [Indexed: 10/19/2022]
Abstract
BACKGROUND Circadian rhythm disorders are severe threats to human health. The negative impact of circadian rhythm disorders on tissues/organs has not been systematically analyzed. Therefore, there is an urgent need to evaluate the damage caused by circadian rhythm disorders and explore the possible mechanisms. METHODS Six-week-old male mice were divided into the control (Con) group (normal circadian rhythm), L24 group (constant light), D12L12 group (weekly shift light/dark cycle), and D24 group (constant dark). Body weight was recorded every 10 days. Ninety days after model construction, the serum lipid and cytokine level, liver function, fat accumulation, carotid artery stenosis, and cardiomyopathological changes were detected in mice. Macrophages in the liver, subscapular fat, and heart tissues were labeled with immunofluorescence staining. Mouse peritoneal macrophages were then isolated. Inflammatory cytokine levels were measured in the macrophage supernatant. The ability of macrophages to form foam cells was also tested. The supernatant from macrophages in different groups was added to AML12 (hepatocytes), 3T3-L1 (preadipocytes), or HL-1 (cardiomyocytes). Effects of conditioned media on recipient cells were determined. RESULTS Body weight, serum lipids and cytokines, subscapular fat accumulation, liver enzymes, carotid artery stenosis, and myocardial fibrosis levels of the L24, D12L12, and D24 groups mice were significantly higher than those in the Con group. Macrophages were significantly increased in the liver, heart, and subscapular fat of mice with circadian rhythmdisorders. Cytokine secretion by peritoneal macrophages was enhanced in the L24, D12L12, and D24 groups. Under oxidized low density lipoprotein (oxLDL) stimulation, macrophages with circadian rhythm disorders are more likely to form foam cells. Conditioned media from the L24, D12L12, and D24 groups significantly promoted AML12 apoptosis and lipid intake, accelerated the adipogenic differentiation of 3T3-L1, and up-regulated collagen I in HL-1. CONCLUSION These findings reveal that macrophages are increased in the tissues/organs under circadian rhythm disorders, and these macrophages could aggravate obesity, promote liver disease, accelerate atherosclerosis, and increase myocardial fibrosis through the paracrine effect.
Collapse
Affiliation(s)
- Zhen Sun
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Lihua Li
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhixin Yan
- Department of Burn Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Lili Zhang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Guangyao Zang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yongjiang Qian
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| |
Collapse
|
15
|
Bryant AJ, Ebrahimi E, Nguyen A, Wolff CA, Gumz ML, Liu AC, Esser KA. A wrinkle in time: circadian biology in pulmonary vascular health and disease. Am J Physiol Lung Cell Mol Physiol 2022; 322:L84-L101. [PMID: 34850650 PMCID: PMC8759967 DOI: 10.1152/ajplung.00037.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
An often overlooked element of pulmonary vascular disease is time. Cellular responses to time, which are regulated directly by the core circadian clock, have only recently been elucidated. Despite an extensive collection of data regarding the role of rhythmic contribution to disease pathogenesis (such as systemic hypertension, coronary artery, and renal disease), the roles of key circadian transcription factors in pulmonary hypertension remain understudied. This is despite a large degree of overlap in the pulmonary hypertension and circadian rhythm fields, not only including shared signaling pathways, but also cell-specific effects of the core clock that are known to result in both protective and adverse lung vessel changes. Therefore, the goal of this review is to summarize the current dialogue regarding common pathways in circadian biology, with a specific emphasis on its implications in the progression of pulmonary hypertension. In this work, we emphasize specific proteins involved in the regulation of the core molecular clock while noting the circadian cell-specific changes relevant to vascular remodeling. Finally, we apply this knowledge to the optimization of medical therapy, with a focus on sleep hygiene and the role of chronopharmacology in patients with this disease. In dissecting the unique relationship between time and cellular biology, we aim to provide valuable insight into the practical implications of considering time as a therapeutic variable. Armed with this information, physicians will be positioned to more efficiently use the full four dimensions of patient care, resulting in improved morbidity and mortality of pulmonary hypertension patients.
Collapse
Affiliation(s)
- Andrew J. Bryant
- 1Department of Medicine, University of Florida College of Medicine, Gainesville, Florida
| | - Elnaz Ebrahimi
- 1Department of Medicine, University of Florida College of Medicine, Gainesville, Florida
| | - Amy Nguyen
- 1Department of Medicine, University of Florida College of Medicine, Gainesville, Florida
| | - Christopher A. Wolff
- 2Department of Physiology, University of Florida College of Medicine, Gainesville, Florida
| | - Michelle L. Gumz
- 2Department of Physiology, University of Florida College of Medicine, Gainesville, Florida
| | - Andrew C. Liu
- 2Department of Physiology, University of Florida College of Medicine, Gainesville, Florida
| | - Karyn A. Esser
- 2Department of Physiology, University of Florida College of Medicine, Gainesville, Florida
| |
Collapse
|
16
|
Potential Role of Melatonin as an Adjuvant for Atherosclerotic Carotid Arterial Stenosis. Molecules 2021; 26:molecules26040811. [PMID: 33557283 PMCID: PMC7914857 DOI: 10.3390/molecules26040811] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/27/2021] [Accepted: 02/02/2021] [Indexed: 12/23/2022] Open
Abstract
Carotid artery stenosis (CAS) is an atherosclerotic disease characterized by a narrowing of the artery lumen and a high risk of ischemic stroke. Risk factors of atherosclerosis, including smoking, hypertension, hyperglycemia, hyperlipidemia, aging, and disrupted circadian rhythm, may potentiate atherosclerosis in the carotid artery and further reduce the arterial lumen. Ischemic stroke due to severe CAS and cerebral ischemic/reperfusion (I/R) injury after the revascularization of CAS also adversely affect clinical outcomes. Melatonin is a pluripotent agent with potent anti-inflammatory, anti-oxidative, and neuroprotective properties. Although there is a shortage of direct clinical evidence demonstrating the benefits of melatonin in CAS patients, previous studies have shown that melatonin may be beneficial for patients with CAS in terms of reducing endothelial damage, stabilizing arterial plaque, mitigating the harm from CAS-related ischemic stroke and cerebral I/R injury, and alleviating the adverse effects of the related risk factors. Additional pre-clinical and clinical are required to confirm this speculation.
Collapse
|
17
|
Chen Z, Xiong ZF, Liu X. Research progress on the interaction between circadian clock and early vascular aging. Exp Gerontol 2021; 146:111241. [PMID: 33453324 DOI: 10.1016/j.exger.2021.111241] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 12/15/2022]
Abstract
Considerable researches implicate that the circadian clock regulates the responsive rhythms of organs and sets the orderly aging process of cells indirectly. It influences an array of diverse biological process including intestinal flora, peripheral inflammatory responses, and redox homeostasis. People with sleep disoders and other kinds of circadian disruptions are prone to have vascular aging earlier. Meanwhile, those people are always faced with chronic vascular inflammation. It has not been elucidated that the specific mechanism of the interaction between the circadian system and early vascular aging. To explore the biphasic relationship between vascular aging and the circadian system, we summarize what is linking circadian clock with early vascular aging through four major prospect: inflammatory process, oxidative stress response, intestinal flora, and cellular senescence. Meanwhile, we discuss the hypothesis that the deterioration of circadian rhythms may exacerbate the process of early vascular aging, leading to the cardiovascular diseases. It will help us to provide new ideas for understanding the process of vascular aging and exploring the possible ways to design personalized chronotherapies.
Collapse
Affiliation(s)
- Zhuoying Chen
- Department of Geriatrics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, China
| | - Zhi-Fan Xiong
- Department of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, China
| | - Xiangjie Liu
- Department of Geriatrics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, China.
| |
Collapse
|
18
|
Zhang Z, Yu B, Wang X, Luo C, Zhou T, Zheng X, Ding J. Circadian rhythm and atherosclerosis (Review). Exp Ther Med 2020; 20:96. [PMID: 32973945 PMCID: PMC7506962 DOI: 10.3892/etm.2020.9224] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 08/11/2020] [Indexed: 12/28/2022] Open
Abstract
Atherosclerosis is the leading cause of morbidity and mortality worldwide. The underlying pathogenesis involves multiple metabolic disorders, endothelial dysfunction and a maladaptive immune response, and leads to chronic arterial wall inflammation. Numerous normal physiological activities exhibit daily rhythmicity, including energy metabolism, vascular function and inflammatory immunoreactions, and disrupted or misaligned circadian rhythms may promote the progression of atherosclerosis. However, the association between the circadian rhythm and atherosclerosis remains to be fully elucidated. In the present review, the effects of the circadian rhythm on atherosclerosis progression are discussed.
Collapse
Affiliation(s)
- Zaiqiang Zhang
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, Hubei 443000, P.R. China.,Institute of Cardiovascular Diseases, China Three Gorges University, Yichang, Hubei 443000, P.R. China
| | - Bin Yu
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, Hubei 443000, P.R. China.,Institute of Cardiovascular Diseases, China Three Gorges University, Yichang, Hubei 443000, P.R. China
| | - Xinan Wang
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, Hubei 443000, P.R. China.,Institute of Cardiovascular Diseases, China Three Gorges University, Yichang, Hubei 443000, P.R. China
| | - Caiyun Luo
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, Hubei 443000, P.R. China.,Institute of Cardiovascular Diseases, China Three Gorges University, Yichang, Hubei 443000, P.R. China
| | - Tian Zhou
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, Hubei 443000, P.R. China.,Institute of Cardiovascular Diseases, China Three Gorges University, Yichang, Hubei 443000, P.R. China
| | - Xiaxia Zheng
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, Hubei 443000, P.R. China.,Institute of Cardiovascular Diseases, China Three Gorges University, Yichang, Hubei 443000, P.R. China
| | - Jiawang Ding
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, Hubei 443000, P.R. China.,Institute of Cardiovascular Diseases, China Three Gorges University, Yichang, Hubei 443000, P.R. China
| |
Collapse
|