1
|
Wang J, Zhao R, Xu S, Zhou XY, Cai K, Chen YL, Zhou ZY, Sun X, Shi Y, Wang F, Gui YH, Tao H, Zhao JY. NOTCH1 mitochondria localization during heart development promotes mitochondrial metabolism and the endothelial-to-mesenchymal transition in mice. Nat Commun 2024; 15:9945. [PMID: 39550366 PMCID: PMC11569218 DOI: 10.1038/s41467-024-54407-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 11/07/2024] [Indexed: 11/18/2024] Open
Abstract
Notch signaling activation drives an endothelial-to-mesenchymal transition (EndMT) critical for heart development, although evidence suggests that the reprogramming of endothelial cell metabolism can regulate endothelial function independent of canonical cell signaling. Herein, we investigated the crosstalk between Notch signaling and metabolic reprogramming in the EndMT process. Biochemically, we find that the NOTCH1 intracellular domain (NICD1) localizes to endothelial cell mitochondria, where it interacts with and activates the complex to enhance mitochondrial metabolism. Targeting NICD1 to mitochondria induces more EndMT compared with wild-type NICD1, and small molecule activation of PDH during pregnancy improves the phenotype in a mouse model of congenital heart defect. A NOTCH1 mutation observed in non-syndromic tetralogy of Fallot patients decreases NICD1 mitochondrial localization and subsequent PDH activity in heart tissues. Altogether, our findings demonstrate NICD1 enrichment in mitochondria of the developing mouse heart, which induces EndMT by activating PDH and subsequently improving mitochondrial metabolism.
Collapse
Affiliation(s)
- Jie Wang
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Cardiovascular Center, Children's Hospital of Fudan University, Shanghai, China
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases (Fudan University), Children's Hospital of Fudan University, Shanghai, China
| | - Rui Zhao
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sha Xu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Xiang-Yu Zhou
- Obstetrics & Gynecology Hospital of Fudan University, Fudan University, Shanghai, China
| | - Ke Cai
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu-Ling Chen
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ze-Yu Zhou
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Sun
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Shi
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Wang
- Cardiovascular Center, Children's Hospital of Fudan University, Shanghai, China.
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases (Fudan University), Children's Hospital of Fudan University, Shanghai, China.
| | - Yong-Hao Gui
- Cardiovascular Center, Children's Hospital of Fudan University, Shanghai, China.
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases (Fudan University), Children's Hospital of Fudan University, Shanghai, China.
| | - Hui Tao
- Department of Cardiothoracic Surgery, Second Hospital of Anhui Medical University, Anhui Medical University, Hefei, China.
| | - Jian-Yuan Zhao
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- International Human Phenome Institutes (Shanghai), Shanghai, China.
| |
Collapse
|
2
|
Liu F, Schrack JA, Walston J, Mathias RA, Windham BG, Grams ME, Coresh J, Walker KA. Mid-life plasma proteins associated with late-life prefrailty and frailty: a proteomic analysis. GeroScience 2024; 46:5247-5265. [PMID: 38856871 PMCID: PMC11336072 DOI: 10.1007/s11357-024-01219-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 05/21/2024] [Indexed: 06/11/2024] Open
Abstract
Physical frailty is a syndrome that typically manifests in later life, although the pathogenic process causing physical frailty likely begins decades earlier. To date, few studies have examined the biological signatures in mid-life associated with physical frailty later in life. Among 4,189 middle-aged participants (57.8 ± 5.0 years, 55.8% women) from the Atherosclerosis Risk in Community (ARIC) study, we evaluated the associations of 4,955 plasma proteins (log 2-transformed and standardized) measured using the SomaScan platform with their frailty status approximately 20 years later. Using multinomial logistic regression models adjusting for demographics, health behaviors, kidney function, total cholesterol, and comorbidities, 12 and 221 proteins were associated with prefrailty and frailty in later life, respectively (FDR p < 0.05). Top frailty-associated proteins included neurocan core protein (NCAN, OR = 0.66), fatty acid-binding protein heart (FABP3, OR = 1.62) and adipocyte (FABP4, OR = 1.65), as well proteins involved in the contactin-1 (CNTN1), toll-like receptor 5 (TLR5), and neurogenic locus notch homolog protein 1 (NOTCH1) signaling pathway relevant to skeletal muscle regeneration, myelination, and inflammation. Pathway analyses suggest midlife dysregulation of inflammation, metabolism, extracellular matrix, angiogenesis, and lysosomal autophagy among those at risk for late-life frailty. After further adjusting for midlife body mass index (BMI) - an established frailty risk factor - only CNTN1 (OR = 0.75) remained significantly associated with frailty. Post-hoc analyses demonstrated that the top 41 midlife frailty-associated proteins mediate 32% of the association between mid-life BMI and late-life frailty. Our findings provide new insights into frailty etiology earlier in the life course, enhancing the potential for prevention.
Collapse
Affiliation(s)
- Fangyu Liu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| | - Jennifer A Schrack
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Center On Aging and Health, Johns Hopkins University, Baltimore, MD, USA
| | - Jeremy Walston
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Rasika A Mathias
- Genomics and Precision Health Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infection Disease, Bethesda, MD, USA
| | - B Gwen Windham
- Department of Medicine, MIND Center, University of Mississippi Medical Center, Jackson, MS, USA
| | - Morgan E Grams
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Division of Precision Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Population Health and Medicine, Optimal Aging Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Keenan A Walker
- Laboratory of Behavioral Neuroscience, National Institute On Aging, Baltimore, MD, USA
| |
Collapse
|
3
|
Petersen TB, Suthahar N, Asselbergs FW, de Bakker M, Akkerhuis KM, Constantinescu AA, van Ramshorst J, Katsikis PD, van der Spek PJ, Umans VA, de Boer RA, Boersma E, Rizopoulos D, Kardys I. Proteomic biomarkers related to obesity in heart failure with reduced ejection fraction and their associations with outcome. Obesity (Silver Spring) 2024; 32:1658-1669. [PMID: 39039788 DOI: 10.1002/oby.24094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 07/24/2024]
Abstract
OBJECTIVE Heart failure (HF) pathophysiology in patients with obesity may be distinct. To study these features, we identified obesity-related biomarkers from 4210 circulating proteins in patients with HF with reduced ejection fraction (HFrEF) and examined associations of these proteins with HF prognosis and biological mechanisms. METHODS In 373 patients with trimonthly blood sampling during a median follow-up of 2.1 (25th-75th percentile: 1.1-2.6) years, we applied an aptamer-based multiplex approach measuring 4210 proteins in baseline samples and the last two samples before study end. Associations between obesity (BMI > 30 kg/m2) and baseline protein levels were analyzed. Subsequently, associations of serially measured obesity-related proteins with biological mechanisms and the primary endpoint (PEP; composite of cardiovascular mortality, HF hospitalization, left ventricular assist device implantation, and heart transplantation) were examined. RESULTS Obesity was identified in 26% (96/373) of patients. A total of 30% (112/373) experienced a PEP (with obesity: 26% [25/96] vs. without obesity: 31% [87/277]). A total of 141/4210 proteins were linked to obesity, reflecting mechanisms of neuron projection development, cell adhesion, and muscle cell migration. A total of 50/141 proteins were associated with the PEP, of which 12 proteins related to atherosclerosis or hypertrophy provided prognostic information beyond clinical characteristics, N-terminal pro-B-type natriuretic peptide, and high-sensitivity troponin T. CONCLUSIONS Patients with HFrEF and obesity show distinct proteomic profiles compared to patients with HFrEF without obesity. Obesity-related proteins are independently associated with HF outcome. These proteins carry potential to improve management of obesity-related HF and could be leads for future research.
Collapse
Affiliation(s)
- Teun B Petersen
- Department of Cardiology, Thorax Center Cardiovascular Institute, Erasmus MC, Rotterdam, the Netherlands
- Department of Biostatistics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Navin Suthahar
- Department of Cardiology, Thorax Center Cardiovascular Institute, Erasmus MC, Rotterdam, the Netherlands
| | - Folkert W Asselbergs
- Department of Cardiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Marie de Bakker
- Department of Cardiology, Thorax Center Cardiovascular Institute, Erasmus MC, Rotterdam, the Netherlands
| | - K Martijn Akkerhuis
- Department of Cardiology, Thorax Center Cardiovascular Institute, Erasmus MC, Rotterdam, the Netherlands
| | - Alina A Constantinescu
- Department of Cardiology, Thorax Center Cardiovascular Institute, Erasmus MC, Rotterdam, the Netherlands
| | - Jan van Ramshorst
- Department of Cardiology, Northwest Clinics, Alkmaar, the Netherlands
| | - Peter D Katsikis
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Peter J van der Spek
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Victor A Umans
- Department of Cardiology, Northwest Clinics, Alkmaar, the Netherlands
| | - Rudolf A de Boer
- Department of Cardiology, Thorax Center Cardiovascular Institute, Erasmus MC, Rotterdam, the Netherlands
| | - Eric Boersma
- Department of Cardiology, Thorax Center Cardiovascular Institute, Erasmus MC, Rotterdam, the Netherlands
| | - Dimitris Rizopoulos
- Department of Biostatistics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Isabella Kardys
- Department of Cardiology, Thorax Center Cardiovascular Institute, Erasmus MC, Rotterdam, the Netherlands
| |
Collapse
|
4
|
Qin T, Ma TY, Huang K, Lu SJ, Zhong JH, Li JJ. Lipoprotein (a)-Related Inflammatory Imbalance: A Novel Horizon for the Development of Atherosclerosis. Curr Atheroscler Rep 2024; 26:383-394. [PMID: 38878139 PMCID: PMC11236888 DOI: 10.1007/s11883-024-01215-5] [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] [Accepted: 05/15/2024] [Indexed: 07/11/2024]
Abstract
PURPOSE OF REVIEW The primary objective of this review is to explore the pathophysiological roles and clinical implications of lipoprotein(a) [Lp(a)] in the context of atherosclerotic cardiovascular disease (ASCVD). We seek to understand how Lp(a) contributes to inflammation and arteriosclerosis, aiming to provide new insights into the mechanisms of ASCVD progression. RECENT FINDINGS Recent research highlights Lp(a) as an independent risk factor for ASCVD. Studies show that Lp(a) not only promotes the inflammatory processes but also interacts with various cellular components, leading to endothelial dysfunction and smooth muscle cell proliferation. The dual role of Lp(a) in both instigating and, under certain conditions, mitigating inflammation is particularly noteworthy. This review finds that Lp(a) plays a complex role in the development of ASCVD through its involvement in inflammatory pathways. The interplay between Lp(a) levels and inflammatory responses highlights its potential as a target for therapeutic intervention. These insights could pave the way for novel approaches in managing and preventing ASCVD, urging further investigation into Lp(a) as a therapeutic target.
Collapse
Affiliation(s)
- Ting Qin
- Department of Cardiology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Hainan, 570208, China
| | - Tian-Yi Ma
- Department of Cardiology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Hainan, 570208, China
| | - Kang Huang
- Department of Cardiology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Hainan, 570208, China
| | - Shi-Juan Lu
- Department of Cardiology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Hainan, 570208, China.
| | - Jiang-Hua Zhong
- Department of Cardiology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Hainan, 570208, China.
| | - Jian-Jun Li
- Cadiometabolic Center, State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| |
Collapse
|
5
|
Zhao J, Sormani L, Jacquelin S, Li H, Styke C, Zhou C, Beesley J, Oon L, Kaur S, Sim SL, Wong HY, Dight J, Hashemi G, Shafiee A, Roy E, Patel J, Khosrotehrani K. Distinct roles of SOX9 in self-renewal of progenitors and mesenchymal transition of the endothelium. Angiogenesis 2024; 27:545-560. [PMID: 38733496 PMCID: PMC11303482 DOI: 10.1007/s10456-024-09927-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 04/24/2024] [Indexed: 05/13/2024]
Abstract
Regenerative capabilities of the endothelium rely on vessel-resident progenitors termed endothelial colony forming cells (ECFCs). This study aimed to investigate if these progenitors are impacted by conditions (i.e., obesity or atherosclerosis) characterized by increased serum levels of oxidized low-density lipoprotein (oxLDL), a known inducer of Endothelial-to-Mesenchymal Transition (EndMT). Our investigation focused on understanding the effects of EndMT on the self-renewal capabilities of progenitors and the associated molecular alterations. In the presence of oxLDL, ECFCs displayed classical features of EndMT, through reduced endothelial gene and protein expression, function as well as increased mesenchymal genes, contractility, and motility. Additionally, ECFCs displayed a dramatic loss in self-renewal capacity in the presence of oxLDL. RNA-sequencing analysis of ECFCs exposed to oxLDL validated gene expression changes suggesting EndMT and identified SOX9 as one of the highly differentially expressed genes. ATAC sequencing analysis identified SOX9 binding sites associated with regions of dynamic chromosome accessibility resulting from oxLDL exposure, further pointing to its importance. EndMT phenotype and gene expression changes induced by oxLDL in vitro or high fat diet (HFD) in vivo were reversed by the silencing of SOX9 in ECFCs or the endothelial-specific conditional knockout of Sox9 in murine models. Overall, our findings support that EndMT affects vessel-resident endothelial progenitor's self-renewal. SOX9 activation is an early transcriptional event that drives the mesenchymal transition of endothelial progenitor cells. The identification of the molecular network driving EndMT in vessel-resident endothelial progenitors presents a new avenue in understanding and preventing a range of condition where this process is involved.
Collapse
Affiliation(s)
- Jilai Zhao
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - Laura Sormani
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - Sebastien Jacquelin
- Mater Research, Translational Research Institute, Macrophage Biology Laboratory, Brisbane, QLD, 4102, Australia
| | - Haiming Li
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - Cassandra Styke
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - Chenhao Zhou
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - Jonathan Beesley
- Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Linus Oon
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - Simranpreet Kaur
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
- Mater Research, Translational Research Institute, Macrophage Biology Laboratory, Brisbane, QLD, 4102, Australia
| | - Seen-Ling Sim
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - Ho Yi Wong
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - James Dight
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - Ghazaleh Hashemi
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - Abbas Shafiee
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - Edwige Roy
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - Jatin Patel
- Centre for Ageing Research Program, Queensland University of Technology, Brisbane, QLD, 4102, Australia
| | - Kiarash Khosrotehrani
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia.
| |
Collapse
|
6
|
Zhang Q, Meng H, Wang X, Chen Y, Yan Z, Ruan J, Meng F. Low expression of Notch1 may be associated with acute myocardial infarction. Front Cardiovasc Med 2024; 11:1367675. [PMID: 38841263 PMCID: PMC11150703 DOI: 10.3389/fcvm.2024.1367675] [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: 02/11/2024] [Accepted: 05/03/2024] [Indexed: 06/07/2024] Open
Abstract
Background The transmembrane protein Notch1 is associated with cell growth, development, differentiation, proliferation, apoptosis, adhesion, and the epithelial mesenchymal transition. Proteomics, as a research method, uses a series of sequencing techniques to study the composition, expression levels, and modifications of proteins. Here, the association between Notch1 and acute myocardial infarction (AMI) was investigated using proteomics, to assess the possibility of using Notch1 as a biomarker for the disease. Methods Fifty-five eligible patients with AMI and 74 with chronic coronary syndrome (CCS) were enrolled, representing the experimental and control groups, respectively. The mRNA levels were assessed using RT-qPCR and proteins were measured using ELISA, and the results were compared and analyzed. Results Notch1 mRNA levels were 0.52 times higher in the peripheral blood mononuclear cells of the AMI group relative to the CCS group (p < 0.05) while Notch1 protein levels were 0.63 times higher in peripheral blood plasma in AMI patients (p < 0.05). Notch1 levels were not associated with older age, hypertension, smoking, high abdominal-blood glucose, high total cholesterol, and high LDL in AMI. Logistic regression indicated associations between AMI and reduced Notch1 expression, hypertension, smoking, and high fasting glucose. Conclusions Notch1 expression was reduced in the peripheral blood of patients with AMI relative to those with CCS. The low expression of Notch1 was found to be an independent risk factor for AMI and may thus be an indicator of the disease.
Collapse
Affiliation(s)
- Qing Zhang
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
- Jilin Provincial Engineering Laboratory for Endothelial Function and Genetic Diagnosis of Cardiovascular Disease, Changchun, Jilin, China
- Jilin Provincial Molecular Biology Research Center for Precision Medicine of Major Cardiovascular Disease, Jilin Provincial Cardiovascular Research Institute, Changchun, Jilin, China
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Changchun, Jilin, China
| | - Heyu Meng
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
- Jilin Provincial Engineering Laboratory for Endothelial Function and Genetic Diagnosis of Cardiovascular Disease, Changchun, Jilin, China
- Jilin Provincial Molecular Biology Research Center for Precision Medicine of Major Cardiovascular Disease, Jilin Provincial Cardiovascular Research Institute, Changchun, Jilin, China
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Changchun, Jilin, China
| | - Xue Wang
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
- Jilin Provincial Engineering Laboratory for Endothelial Function and Genetic Diagnosis of Cardiovascular Disease, Changchun, Jilin, China
- Jilin Provincial Molecular Biology Research Center for Precision Medicine of Major Cardiovascular Disease, Jilin Provincial Cardiovascular Research Institute, Changchun, Jilin, China
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Changchun, Jilin, China
| | - Yanqiu Chen
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
- Jilin Provincial Engineering Laboratory for Endothelial Function and Genetic Diagnosis of Cardiovascular Disease, Changchun, Jilin, China
- Jilin Provincial Molecular Biology Research Center for Precision Medicine of Major Cardiovascular Disease, Jilin Provincial Cardiovascular Research Institute, Changchun, Jilin, China
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Changchun, Jilin, China
| | - Zhaohan Yan
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
- Jilin Provincial Engineering Laboratory for Endothelial Function and Genetic Diagnosis of Cardiovascular Disease, Changchun, Jilin, China
- Jilin Provincial Molecular Biology Research Center for Precision Medicine of Major Cardiovascular Disease, Jilin Provincial Cardiovascular Research Institute, Changchun, Jilin, China
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Changchun, Jilin, China
| | - Jianjun Ruan
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
- Jilin Provincial Engineering Laboratory for Endothelial Function and Genetic Diagnosis of Cardiovascular Disease, Changchun, Jilin, China
- Jilin Provincial Molecular Biology Research Center for Precision Medicine of Major Cardiovascular Disease, Jilin Provincial Cardiovascular Research Institute, Changchun, Jilin, China
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Changchun, Jilin, China
| | - Fanbo Meng
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
- Jilin Provincial Engineering Laboratory for Endothelial Function and Genetic Diagnosis of Cardiovascular Disease, Changchun, Jilin, China
- Jilin Provincial Molecular Biology Research Center for Precision Medicine of Major Cardiovascular Disease, Jilin Provincial Cardiovascular Research Institute, Changchun, Jilin, China
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Changchun, Jilin, China
| |
Collapse
|
7
|
Awad KS, Wang S, Dougherty EJ, Keshavarz A, Demirkale CY, Yu ZX, Miller L, Elinoff JM, Danner RL. BMPR2 Loss Activates AKT by Disrupting DLL4/NOTCH1 and PPARγ Signaling in Pulmonary Arterial Hypertension. Int J Mol Sci 2024; 25:5403. [PMID: 38791441 PMCID: PMC11121464 DOI: 10.3390/ijms25105403] [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: 04/16/2024] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 05/26/2024] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive cardiopulmonary disease characterized by pathologic vascular remodeling of small pulmonary arteries. Endothelial dysfunction in advanced PAH is associated with proliferation, apoptosis resistance, and endothelial to mesenchymal transition (EndoMT) due to aberrant signaling. DLL4, a cell membrane associated NOTCH ligand, plays a pivotal role maintaining vascular integrity. Inhibition of DLL4 has been associated with the development of pulmonary hypertension, but the mechanism is incompletely understood. Here we report that BMPR2 silencing in pulmonary artery endothelial cells (PAECs) activated AKT and suppressed the expression of DLL4. Consistent with these in vitro findings, increased AKT activation and reduced DLL4 expression was found in the small pulmonary arteries of patients with PAH. Increased NOTCH1 activation through exogenous DLL4 blocked AKT activation, decreased proliferation and reversed EndoMT. Exogenous and overexpression of DLL4 induced BMPR2 and PPRE promoter activity, and BMPR2 and PPARG mRNA in idiopathic PAH (IPAH) ECs. PPARγ, a nuclear receptor associated with EC homeostasis, suppressed by BMPR2 loss was induced and activated by DLL4/NOTCH1 signaling in both BMPR2-silenced and IPAH ECs, reversing aberrant phenotypic changes, in part through AKT inhibition. Directly blocking AKT or restoring DLL4/NOTCH1/PPARγ signaling may be beneficial in preventing or reversing the pathologic vascular remodeling of PAH.
Collapse
MESH Headings
- Humans
- Proto-Oncogene Proteins c-akt/metabolism
- Signal Transduction
- Bone Morphogenetic Protein Receptors, Type II/metabolism
- Bone Morphogenetic Protein Receptors, Type II/genetics
- PPAR gamma/metabolism
- PPAR gamma/genetics
- Receptor, Notch1/metabolism
- Receptor, Notch1/genetics
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Endothelial Cells/metabolism
- Adaptor Proteins, Signal Transducing/metabolism
- Adaptor Proteins, Signal Transducing/genetics
- Calcium-Binding Proteins/metabolism
- Calcium-Binding Proteins/genetics
- Pulmonary Arterial Hypertension/metabolism
- Pulmonary Arterial Hypertension/genetics
- Pulmonary Arterial Hypertension/pathology
- Male
- Cell Proliferation
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/pathology
- Female
- Cells, Cultured
Collapse
Affiliation(s)
- Keytam S. Awad
- Critical Care Medicine Department, Clinical Center, NIH, Bethesda, MD 20892, USA; (S.W.); (E.J.D.); (A.K.); (C.Y.D.); (L.M.); (R.L.D.)
| | - Shuibang Wang
- Critical Care Medicine Department, Clinical Center, NIH, Bethesda, MD 20892, USA; (S.W.); (E.J.D.); (A.K.); (C.Y.D.); (L.M.); (R.L.D.)
| | - Edward J. Dougherty
- Critical Care Medicine Department, Clinical Center, NIH, Bethesda, MD 20892, USA; (S.W.); (E.J.D.); (A.K.); (C.Y.D.); (L.M.); (R.L.D.)
| | - Ali Keshavarz
- Critical Care Medicine Department, Clinical Center, NIH, Bethesda, MD 20892, USA; (S.W.); (E.J.D.); (A.K.); (C.Y.D.); (L.M.); (R.L.D.)
| | - Cumhur Y. Demirkale
- Critical Care Medicine Department, Clinical Center, NIH, Bethesda, MD 20892, USA; (S.W.); (E.J.D.); (A.K.); (C.Y.D.); (L.M.); (R.L.D.)
| | - Zu Xi Yu
- Critical Care Medicine and Pulmonary Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892, USA; (Z.X.Y.); (J.M.E.)
| | - Latonia Miller
- Critical Care Medicine Department, Clinical Center, NIH, Bethesda, MD 20892, USA; (S.W.); (E.J.D.); (A.K.); (C.Y.D.); (L.M.); (R.L.D.)
| | - Jason M. Elinoff
- Critical Care Medicine and Pulmonary Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892, USA; (Z.X.Y.); (J.M.E.)
| | - Robert L. Danner
- Critical Care Medicine Department, Clinical Center, NIH, Bethesda, MD 20892, USA; (S.W.); (E.J.D.); (A.K.); (C.Y.D.); (L.M.); (R.L.D.)
- Critical Care Medicine and Pulmonary Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892, USA; (Z.X.Y.); (J.M.E.)
| |
Collapse
|
8
|
Awad KS, Wang S, Dougherty EJ, Keshavarz A, Demirkale CY, Yu ZX, Miller L, Elinoff JM, Danner RL. Disruption of DLL4/NOTCH1 Causes Dysregulated PPARγ/AKT Signaling in Pulmonary Arterial Hypertension. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.31.578230. [PMID: 38903104 PMCID: PMC11188078 DOI: 10.1101/2024.01.31.578230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive cardiopulmonary disease characterized by vascular remodeling of small pulmonary arteries. Endothelial dysfunction in advanced PAH is associated with proliferation, apoptosis resistance, and endothelial to mesenchymal transition (EndoMT) due to aberrant signaling. DLL4, a cell membrane associated NOTCH ligand, activates NOTCH1 signaling and plays a pivotal role maintaining vascular integrity. Inhibition of DLL4 has been associated with the development of pulmonary hypertension, but the mechanism is incompletely understood. Here we report that BMPR2 silencing in PAECs activated AKT and decreased DLL4 expression. DLL4 loss was also seen in lungs of patients with IPAH and HPAH. Over-expression of DLL4 in PAECs induced BMPR2 promoter activity and exogenous DLL4 increased BMPR2 mRNA through NOTCH1 activation. Furthermore, DLL4/NOTCH1 signaling blocked AKT activation, decreased proliferation and reversed EndoMT in BMPR2-silenced PAECs and ECs from IPAH patients. PPARγ, suppressed by BMPR2 loss, was induced and activated by DLL4/NOTCH1 signaling in both BMPR2-silenced and IPAH PAECs, reversing aberrant phenotypic changes, in part through AKT inhibition. Finally, leniolisib, a well-tolerated oral PI3Kδ/AKT inhibitor, decreased cell proliferation, induced apoptosis and reversed markers of EndoMT in BMPR2-silenced PAECs. Restoring DLL4/NOTCH1/PPARγ signaling and/or suppressing AKT activation may be beneficial in preventing or reversing the pathologic vascular remodeling of PAH.
Collapse
Affiliation(s)
- Keytam S Awad
- Critical Care Medicine Department, NIH Clinical Center, Bethesda, MD, US, 20892
| | - Shuibang Wang
- Critical Care Medicine Department, NIH Clinical Center, Bethesda, MD, US, 20892
| | - Edward J Dougherty
- Critical Care Medicine Department, NIH Clinical Center, Bethesda, MD, US, 20892
| | - Ali Keshavarz
- Critical Care Medicine Department, NIH Clinical Center, Bethesda, MD, US, 20892
| | - Cumhur Y Demirkale
- Critical Care Medicine Department, NIH Clinical Center, Bethesda, MD, US, 20892
| | - Zu Xi Yu
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, US, 20892
| | - Latonia Miller
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, US, 20892
| | - Jason M Elinoff
- Critical Care Medicine Department, NIH Clinical Center, Bethesda, MD, US, 20892
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, US, 20892
| | - Robert L Danner
- Critical Care Medicine Department, NIH Clinical Center, Bethesda, MD, US, 20892
| |
Collapse
|
9
|
Sunshine HL, Cicchetto AC, Kaczor-Urbanowicz KE, Ma F, Pi D, Symons C, Turner M, Shukla V, Christofk HR, Vallim TA, Iruela-Arispe ML. Endothelial Jagged1 levels and distribution are post-transcriptionally controlled by ZFP36 decay proteins. Cell Rep 2024; 43:113627. [PMID: 38157296 PMCID: PMC10884959 DOI: 10.1016/j.celrep.2023.113627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 11/02/2023] [Accepted: 12/12/2023] [Indexed: 01/03/2024] Open
Abstract
Vascular morphogenesis requires a delicate gradient of Notch signaling controlled, in part, by the distribution of ligands (Dll4 and Jagged1). How Jagged1 (JAG1) expression is compartmentalized in the vascular plexus remains unclear. Here, we show that Jag1 mRNA is a direct target of zinc-finger protein 36 (ZFP36), an RNA-binding protein involved in mRNA decay that we find robustly induced by vascular endothelial growth factor (VEGF). Endothelial cells lacking ZFP36 display high levels of JAG1 and increase angiogenic sprouting in vitro. Furthermore, mice lacking Zfp36 in endothelial cells display mispatterned and increased levels of JAG1 in the developing retinal vascular plexus. Abnormal levels of JAG1 at the sprouting front alters NOTCH1 signaling, increasing the number of tip cells, a phenotype that is rescued by imposing haploinsufficiency of Jag1. Our findings reveal an important feedforward loop whereby VEGF stimulates ZFP36, consequently suppressing Jag1 to enable adequate levels of Notch signaling during sprouting angiogenesis.
Collapse
Affiliation(s)
- Hannah L Sunshine
- Molecular, Cellular, and Integrative Physiology Graduate Program, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Andrew C Cicchetto
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Karolina Elżbieta Kaczor-Urbanowicz
- Center for Oral and Head/Neck Oncology Research, UCLA Biosystems & Function, UCLA School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095-1668, USA; UCLA Section of Orthodontics, UCLA School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; UCLA Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Feiyang Ma
- Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Danielle Pi
- Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Chloe Symons
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Martin Turner
- Immunology Programme, The Babraham Institute, CB22 3AT Cambridge, UK
| | - Vipul Shukla
- Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Center for Human Immunobiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Heather R Christofk
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095-1606, USA
| | - Thomas A Vallim
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095-1606, USA
| | - M Luisa Iruela-Arispe
- Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
10
|
Jiang Q, Wang L, Si X, Bian Y, Zhang W, Cui H, Gui H, Zhang Y, Li B, Tan D. Pterostilbene antagonizes homocysteine-induced oxidative stress, apoptosis and lipid deposition in vascular endothelial cells. FOOD SCIENCE AND HUMAN WELLNESS 2023. [DOI: 10.1016/j.fshw.2023.02.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
|
11
|
Katsi V, Papakonstantinou I, Tsioufis K. Atherosclerosis, Diabetes Mellitus, and Cancer: Common Epidemiology, Shared Mechanisms, and Future Management. Int J Mol Sci 2023; 24:11786. [PMID: 37511551 PMCID: PMC10381022 DOI: 10.3390/ijms241411786] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/03/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
The involvement of cardiovascular disease in cancer onset and development represents a contemporary interest in basic science. It has been recognized, from the most recent research, that metabolic syndrome-related conditions, ranging from atherosclerosis to diabetes, elicit many pathways regulating lipid metabolism and lipid signaling that are also linked to the same framework of multiple potential mechanisms for inducing cancer. Otherwise, dyslipidemia and endothelial cell dysfunction in atherosclerosis may present common or even interdependent changes, similar to oncogenic molecules elevated in many forms of cancer. However, whether endothelial cell dysfunction in atherosclerotic disease provides signals that promote the pre-clinical onset and proliferation of malignant cells is an issue that requires further understanding, even though more questions are presented with every answer. Here, we highlight the molecular mechanisms that point to a causal link between lipid metabolism and glucose homeostasis in metabolic syndrome-related atherosclerotic disease with the development of cancer. The knowledge of these breakthrough mechanisms may pave the way for the application of new therapeutic targets and for implementing interventions in clinical practice.
Collapse
Affiliation(s)
- Vasiliki Katsi
- Department of Cardiology, Hippokration Hospital, 11527 Athens, Greece
| | | | - Konstantinos Tsioufis
- Department of Cardiology, Hippokration Hospital, 11527 Athens, Greece
- School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
12
|
Franco-Acevedo A, Comes J, Mack JJ, Valenzuela NM. New insights into maladaptive vascular responses to donor specific HLA antibodies in organ transplantation. FRONTIERS IN TRANSPLANTATION 2023; 2:1146040. [PMID: 38993843 PMCID: PMC11235244 DOI: 10.3389/frtra.2023.1146040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 04/03/2023] [Indexed: 07/13/2024]
Abstract
Transplant vasculopathy (TV) causes thickening of donor blood vessels in transplanted organs, and is a significant cause of graft loss and mortality in allograft recipients. It is known that patients with repeated acute rejection and/or donor specific antibodies are predisposed to TV. Nevertheless, the exact molecular mechanisms by which alloimmune injury culminates in this disease have not been fully delineated. As a result of this incomplete knowledge, there is currently a lack of effective therapies for this disease. The immediate intracellular signaling and the acute effects elicited by anti-donor HLA antibodies are well-described and continuing to be revealed in deeper detail. Further, advances in rejection diagnostics, including intragraft gene expression, provide clues to the inflammatory changes within allografts. However, mechanisms linking these events with long-term outcomes, particularly the maladaptive vascular remodeling seen in transplant vasculopathy, are still being delineated. New evidence demonstrates alterations in non-coding RNA profiles and the occurrence of endothelial to mesenchymal transition (EndMT) during acute antibody-mediated graft injury. EndMT is also readily apparent in numerous settings of non-transplant intimal hyperplasia, and lessons can be learned from advances in those fields. This review will provide an update on these recent developments and remaining questions in our understanding of HLA antibody-induced vascular damage, framed within a broader consideration of manifestations and implications across transplanted organ types.
Collapse
Affiliation(s)
- Adriana Franco-Acevedo
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA, United States
| | - Johanna Comes
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Julia J Mack
- Department of Medicine, Division of Cardiology, University of California, Los Angeles, CA, United States
| | - Nicole M Valenzuela
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA, United States
| |
Collapse
|
13
|
Bruserud Ø, Mosevoll KA, Bruserud Ø, Reikvam H, Wendelbo Ø. The Regulation of Neutrophil Migration in Patients with Sepsis: The Complexity of the Molecular Mechanisms and Their Modulation in Sepsis and the Heterogeneity of Sepsis Patients. Cells 2023; 12:cells12071003. [PMID: 37048076 PMCID: PMC10093057 DOI: 10.3390/cells12071003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Sepsis is defined as life-threatening organ dysfunction caused by a dysregulated host response to infection. Common causes include gram-negative and gram-positive bacteria as well as fungi. Neutrophils are among the first cells to arrive at an infection site where they function as important effector cells of the innate immune system and as regulators of the host immune response. The regulation of neutrophil migration is therefore important both for the infection-directed host response and for the development of organ dysfunctions in sepsis. Downregulation of CXCR4/CXCL12 stimulates neutrophil migration from the bone marrow. This is followed by transmigration/extravasation across the endothelial cell barrier at the infection site; this process is directed by adhesion molecules and various chemotactic gradients created by chemotactic cytokines, lipid mediators, bacterial peptides, and peptides from damaged cells. These mechanisms of neutrophil migration are modulated by sepsis, leading to reduced neutrophil migration and even reversed migration that contributes to distant organ failure. The sepsis-induced modulation seems to differ between neutrophil subsets. Furthermore, sepsis patients should be regarded as heterogeneous because neutrophil migration will possibly be further modulated by the infecting microorganisms, antimicrobial treatment, patient age/frailty/sex, other diseases (e.g., hematological malignancies and stem cell transplantation), and the metabolic status. The present review describes molecular mechanisms involved in the regulation of neutrophil migration; how these mechanisms are altered during sepsis; and how bacteria/fungi, antimicrobial treatment, and aging/frailty/comorbidity influence the regulation of neutrophil migration.
Collapse
Affiliation(s)
- Øystein Bruserud
- Leukemia Research Group, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
- Section for Hematology, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
- Correspondence:
| | - Knut Anders Mosevoll
- Section for Infectious Diseases, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
- Section for Infectious Diseases, Department of Clinical Research, University of Bergen, 5021 Bergen, Norway
| | - Øyvind Bruserud
- Department for Anesthesiology and Intensive Care, Haukeland University Hospital, 5021 Bergen, Norway
| | - Håkon Reikvam
- Leukemia Research Group, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
- Section for Hematology, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
| | - Øystein Wendelbo
- Section for Infectious Diseases, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
- Faculty of Health, VID Specialized University, Ulriksdal 10, 5009 Bergen, Norway
| |
Collapse
|
14
|
Hasan SS, Fischer A. Notch Signaling in the Vasculature: Angiogenesis and Angiocrine Functions. Cold Spring Harb Perspect Med 2023; 13:a041166. [PMID: 35667708 PMCID: PMC9899647 DOI: 10.1101/cshperspect.a041166] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Formation of a functional blood vessel network is a complex process tightly controlled by pro- and antiangiogenic signals released within the local microenvironment or delivered through the bloodstream. Endothelial cells precisely integrate such temporal and spatial changes in extracellular signals and generate an orchestrated response by modulating signaling transduction, gene expression, and metabolism. A key regulator in vessel formation is Notch signaling, which controls endothelial cell specification, proliferation, migration, adhesion, and arteriovenous differentiation. This review summarizes the molecular biology of endothelial Notch signaling and how it controls angiogenesis and maintenance of the established, quiescent vasculature. In addition, recent progress in the understanding of Notch signaling in endothelial cells for controlling organ homeostasis by transcriptional regulation of angiocrine factors and its relevance to disease will be discussed.
Collapse
Affiliation(s)
- Sana S Hasan
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Andreas Fischer
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Institute for Clinical Chemistry, University Medical Center Göttingen, 37075 Göttingen, Germany
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| |
Collapse
|
15
|
Liu T, Zhang C, Ying J, Wang Y, Yan G, Zhou Y, Lu G. Inhibition of the intracellular domain of Notch1 results in vascular endothelial cell dysfunction in sepsis. Front Immunol 2023; 14:1134556. [PMID: 37205094 PMCID: PMC10185824 DOI: 10.3389/fimmu.2023.1134556] [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: 12/30/2022] [Accepted: 04/21/2023] [Indexed: 05/21/2023] Open
Abstract
Background Notch signaling is critical for regulating the function of vascular endothelial cells (ECs). However, the effect of the intracellular domain of Notch1 (NICD) on EC injury in sepsis remains unclear. Methods We established a cell model of vascular endothelial dysfunction and induced sepsis in a mouse model via lipopolysaccharide (LPS) injection and cecal ligation and puncture (CLP). Endothelial barrier function and expression of endothelial-related proteins were determined using CCK-8, permeability, flow cytometry, immunoblot, and immunoprecipitation assays. The effect of NICD inhibition or activation on endothelial barrier function was evaluated in vitro. Melatonin was used for NICD activation in sepsis mice. The survival rate, Evans blue dye of organs, vessel relaxation assay, immunohistochemistry, ELISA, immunoblot were used to explore the specific role of melatonin for sepsis induced vascular dysfunction in vivo. Results We found that LPS, interleukin 6, and serum collected from septic children could inhibit the expression of NICD and its downstream regulator Hes1, which impaired endothelial barrier function and led to EC apoptosis through the AKT pathway. Mechanistically, LPS decreased the stability of NICD by inhibiting the expression of a deubiquitylating enzyme, ubiquitin-specific proteases 8 (USP8). Melatonin, however, upregulated USP8 expression, thus maintaining the stability of NICD and Notch signaling, which ultimately reduced EC injury in our sepsis model and elevated the survival rate of septic mice. Conclusions We found a previously uncharacterized role of Notch1 in mediating vascular permeability during sepsis, and we showed that inhibition of NICD resulted in vascular EC dysfunction in sepsis, which was reversed by melatonin. Thus, the Notch1 signaling pathway is a potential target for the treatment of sepsis.
Collapse
Affiliation(s)
- Tingyan Liu
- Department of Critical Care Medicine, Children’s Hospital of Fudan University, Shanghai, China
| | - Caiyan Zhang
- Department of Critical Care Medicine, Children’s Hospital of Fudan University, Shanghai, China
| | - Jiayun Ying
- Department of Critical Care Medicine, Children’s Hospital of Fudan University, Shanghai, China
| | - Yaodong Wang
- Department of Critical Care Medicine, Children’s Hospital of Fudan University, Shanghai, China
| | - Gangfeng Yan
- Department of Critical Care Medicine, Children’s Hospital of Fudan University, Shanghai, China
| | - Yufeng Zhou
- Institute of Pediatrics, Children’s Hospital of Fudan University, National Children’s Medical Center, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, Shanghai, China
- *Correspondence: Yufeng Zhou, ; Guoping Lu,
| | - Guoping Lu
- Department of Critical Care Medicine, Children’s Hospital of Fudan University, Shanghai, China
- *Correspondence: Yufeng Zhou, ; Guoping Lu,
| |
Collapse
|
16
|
Li S, Han Y, Zhang Q, Tang D, Li J, Weng L. Comprehensive molecular analyses of an autoimmune-related gene predictive model and immune infiltrations using machine learning methods in moyamoya disease. Front Mol Biosci 2022; 9:991425. [PMID: 36605987 PMCID: PMC9808060 DOI: 10.3389/fmolb.2022.991425] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
Background: Growing evidence suggests the links between moyamoya disease (MMD) and autoimmune diseases. However, the molecular mechanism from genetic perspective remains unclear. This study aims to clarify the potential roles of autoimmune-related genes (ARGs) in the pathogenesis of MMD. Methods: Two transcription profiles (GSE157628 and GSE141025) of MMD were downloaded from GEO databases. ARGs were obtained from the Gene and Autoimmune Disease Association Database (GAAD) and DisGeNET databases. Differentially expressed ARGs (DEARGs) were identified using "limma" R packages. GO, KEGG, GSVA, and GSEA analyses were conducted to elucidate the underlying molecular function. There machine learning methods (LASSO logistic regression, random forest (RF), support vector machine-recursive feature elimination (SVM-RFE)) were used to screen out important genes. An artificial neural network was applied to construct an autoimmune-related signature predictive model of MMD. The immune characteristics, including immune cell infiltration, immune responses, and HLA gene expression in MMD, were explored using ssGSEA. The miRNA-gene regulatory network and the potential therapeutic drugs for hub genes were predicted. Results: A total of 260 DEARGs were identified in GSE157628 dataset. These genes were involved in immune-related pathways, infectious diseases, and autoimmune diseases. We identified six diagnostic genes by overlapping the three machine learning algorithms: CD38, PTPN11, NOTCH1, TLR7, KAT2B, and ISG15. A predictive neural network model was constructed based on the six genes and presented with great diagnostic ability with area under the curve (AUC) = 1 in the GSE157628 dataset and further validated by GSE141025 dataset. Immune infiltration analysis showed that the abundance of eosinophils, natural killer T (NKT) cells, Th2 cells were significant different between MMD and controls. The expression levels of HLA-A, HLA-B, HLA-C, HLA-DMA, HLA-DRB6, HLA-F, and HLA-G were significantly upregulated in MMD. Four miRNAs (mir-26a-5p, mir-1343-3p, mir-129-2-3p, and mir-124-3p) were identified because of their interaction at least with four hub DEARGs. Conclusion: Machine learning was used to develop a reliable predictive model for the diagnosis of MMD based on ARGs. The uncovered immune infiltration and gene-miRNA and gene-drugs regulatory network may provide new insight into the pathogenesis and treatment of MMD.
Collapse
Affiliation(s)
- Shifu Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, Hunan, China
| | - Ying Han
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital of Central South University, Changsha, Hunan, China,Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Qian Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, Hunan, China
| | - Dong Tang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, Hunan, China
| | - Jian Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, Hunan, China,Hydrocephalus Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ling Weng
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, Hunan, China,Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China,Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China,Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China,*Correspondence: Ling Weng,
| |
Collapse
|
17
|
Konduracka E, Krawczyk K, Surmiak M, Pudełek M, Malinowski KP, Mastalerz L, Zimnoch M, Samek L, Styszko K, Furman L, Gałkowski M, Nessler J, Różański K, Sanak M. Monocyte exposure to fine particulate matter results in miRNA release: A link between air pollution and potential clinical complication. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2022; 96:103996. [PMID: 36228992 DOI: 10.1016/j.etap.2022.103996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 09/26/2022] [Accepted: 10/03/2022] [Indexed: 06/16/2023]
Abstract
Chronic exposure to PM2.5 contributes to the pathogenesis of numerous disorders, although the underlying mechanisms remain unknown. The study investigated whether exposure of human monocytes to PM2.5 is associated with alterations in miRNAs. Monocytes were exposed in vitro to PM2.5 collected during winter and summer, followed by miRNA isolation from monocytes. Additionally, in 140 persons chronically exposed to air pollution, some miRNA patterns were isolated from serum seasonally. Between-season differences in chemical PM2.5 composition were observed. Some miRNAs were expressed both in monocytes and in human serum. MiR-34c-5p and miR-223-5p expression was more pronounced in winter. Bioinformatics analyses showed that selected miRNAs were involved in the regulation of several pathways. The expression of the same miRNA species in monocytes and serum suggests that these cells are involved in the production of miRNAs implicated in the development of disorders mediated by inflammation, oxidative stress, proliferation, and apoptosis after exposure to PM2.5.
Collapse
Affiliation(s)
- Ewa Konduracka
- Jagiellonian University Medical College, Coronary Disease Department and Heart Failure, John Paul II Hospital, Kraków, Poland.
| | - Krzysztof Krawczyk
- Jagiellonian University Medical College, Faculty of Health Sciences, Department of Emergency Medicine, Kraków, Poland
| | - Marcin Surmiak
- Jagiellonian University Medical College, 2nd Department of Internal Medicine, Kraków, Poland
| | - Maciej Pudełek
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Krzysztof Piotr Malinowski
- Jagiellonian University Medical College, Faculty of Medicine, Department of Bioinformatics and Telemedicine, Kraków, Poland
| | - Lucyna Mastalerz
- Jagiellonian University Medical College, 2nd Department of Internal Medicine, Kraków, Poland
| | - Mirosław Zimnoch
- AGH University of Science and Technology, Faculty of Physics and Applied Computer Science, Kraków, Poland; Max Planck Institute for Biogeochemistry in Jena, Hans-Knöll Jena, Germany
| | - Lucyna Samek
- AGH University of Science and Technology, Faculty of Physics and Applied Computer Science, Kraków, Poland; Max Planck Institute for Biogeochemistry in Jena, Hans-Knöll Jena, Germany
| | - Katarzyna Styszko
- AGH University of Science and Technology, Faculty of Energy and Fuels, Kraków, Poland
| | - Leszek Furman
- AGH University of Science and Technology, Faculty of Physics and Applied Computer Science, Kraków, Poland; Max Planck Institute for Biogeochemistry in Jena, Hans-Knöll Jena, Germany
| | - Michał Gałkowski
- AGH University of Science and Technology, Faculty of Physics and Applied Computer Science, Kraków, Poland; Max Planck Institute for Biogeochemistry in Jena, Hans-Knöll Jena, Germany; Department of Biogeochemical Signals, Max Planck Institute for Biogeochemistry, Hans-Knöll Str. 10, 07745 Jena, Germany
| | - Jadwiga Nessler
- Jagiellonian University Medical College, Coronary Disease Department and Heart Failure, John Paul II Hospital, Kraków, Poland
| | - Kazimierz Różański
- AGH University of Science and Technology, Faculty of Physics and Applied Computer Science, Kraków, Poland; Max Planck Institute for Biogeochemistry in Jena, Hans-Knöll Jena, Germany
| | - Marek Sanak
- Jagiellonian University Medical College, 2nd Department of Internal Medicine, Kraków, Poland
| |
Collapse
|
18
|
Fleig S, Kapanadze T, Bernier-Latmani J, Lill JK, Wyss T, Gamrekelashvili J, Kijas D, Liu B, Hüsing AM, Bovay E, Jirmo AC, Halle S, Ricke-Hoch M, Adams RH, Engel DR, von Vietinghoff S, Förster R, Hilfiker-Kleiner D, Haller H, Petrova TV, Limbourg FP. Loss of vascular endothelial notch signaling promotes spontaneous formation of tertiary lymphoid structures. Nat Commun 2022; 13:2022. [PMID: 35440634 PMCID: PMC9018798 DOI: 10.1038/s41467-022-29701-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 03/21/2022] [Indexed: 12/20/2022] Open
Abstract
Tertiary lymphoid structures (TLS) are lymph node-like immune cell clusters that emerge during chronic inflammation in non-lymphoid organs like the kidney, but their origin remains not well understood. Here we show, using conditional deletion strategies of the canonical Notch signaling mediator Rbpj, that loss of endothelial Notch signaling in adult mice induces the spontaneous formation of bona fide TLS in the kidney, liver and lung, based on molecular, cellular and structural criteria. These TLS form in a stereotypical manner around parenchymal arteries, while secondary lymphoid structures remained largely unchanged. This effect is mediated by endothelium of blood vessels, but not lymphatics, since a lymphatic endothelial-specific targeting strategy did not result in TLS formation, and involves loss of arterial specification and concomitant acquisition of a high endothelial cell phenotype, as shown by transcriptional analysis of kidney endothelial cells. This indicates a so far unrecognized role for vascular endothelial cells and Notch signaling in TLS initiation. Loss of canonical Notch signaling in vascular endothelial cells induces spontaneous formation of proto-typical tertiary lymphoid structures in mouse kidney, liver and lungs, which form around central arteries that acquire a high endothelial cell signature
Collapse
Affiliation(s)
- Susanne Fleig
- Vascular Medicine Research, Hannover Medical School, 30625, Hannover, Germany.,Department of Nephrology and Hypertension, Hannover Medical School, 30625, Hannover, Germany.,Department of Geriatric Medicine (Medical Clinic VI), RWTH Aachen University Hospital, 52074, Aachen, Germany
| | - Tamar Kapanadze
- Vascular Medicine Research, Hannover Medical School, 30625, Hannover, Germany.,Department of Nephrology and Hypertension, Hannover Medical School, 30625, Hannover, Germany
| | - Jeremiah Bernier-Latmani
- Vascular and Tumor Biology Laboratory, Department of Oncology UNIL CHUV and Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | - Julia K Lill
- Department of Immunodynamics, Institute for Experimental Immunology and Imaging, Medical Research Centre, University Hospital Essen, 45147, Essen, Germany
| | - Tania Wyss
- Vascular and Tumor Biology Laboratory, Department of Oncology UNIL CHUV and Ludwig Institute for Cancer Research, Lausanne, Switzerland.,SIB Swiss Institute of Bioinformatics, Lausanne, 1015, Switzerland
| | - Jaba Gamrekelashvili
- Vascular Medicine Research, Hannover Medical School, 30625, Hannover, Germany.,Department of Nephrology and Hypertension, Hannover Medical School, 30625, Hannover, Germany
| | - Dustin Kijas
- Vascular Medicine Research, Hannover Medical School, 30625, Hannover, Germany.,Department of Nephrology and Hypertension, Hannover Medical School, 30625, Hannover, Germany
| | - Bin Liu
- Hannover Medical School, Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Anne M Hüsing
- Department of Nephrology and Hypertension, Hannover Medical School, 30625, Hannover, Germany
| | - Esther Bovay
- Max-Planck-Institute for Molecular Biomedicine, 48149, Muenster, Germany
| | - Adan Chari Jirmo
- Hannover Medical School, Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany.,Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Stephan Halle
- Institute of Immunology, Hannover Medical School, 30625, Hannover, Germany
| | - Melanie Ricke-Hoch
- Department of Cardiology and Angiology, Hannover Medical School, 30625, Hannover, Germany
| | - Ralf H Adams
- Max-Planck-Institute for Molecular Biomedicine, 48149, Muenster, Germany
| | - Daniel R Engel
- Department of Immunodynamics, Institute for Experimental Immunology and Imaging, Medical Research Centre, University Hospital Essen, 45147, Essen, Germany
| | - Sibylle von Vietinghoff
- Department of Nephrology and Hypertension, Hannover Medical School, 30625, Hannover, Germany.,Division of Medicine I, Nephrology section, UKB Bonn University Hospital, Bonn, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, 30625, Hannover, Germany
| | - Denise Hilfiker-Kleiner
- Department of Cardiology and Angiology, Hannover Medical School, 30625, Hannover, Germany.,Department of Cardiovascular Complications of Oncologic Therapies, Medical Faculty of the Philipps University Marburg, 35037, Marburg, Germany
| | - Hermann Haller
- Department of Nephrology and Hypertension, Hannover Medical School, 30625, Hannover, Germany
| | - Tatiana V Petrova
- Vascular and Tumor Biology Laboratory, Department of Oncology UNIL CHUV and Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | - Florian P Limbourg
- Vascular Medicine Research, Hannover Medical School, 30625, Hannover, Germany. .,Department of Nephrology and Hypertension, Hannover Medical School, 30625, Hannover, Germany.
| |
Collapse
|
19
|
Chen M, Li F, Jiang Q, Zhang W, Li Z, Tang W. Role of miR-181b/Notch1 Axis in circ_TNPO1 Promotion of Proliferation and Migration of Atherosclerotic Vascular Smooth Muscle Cells. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:4086935. [PMID: 35388333 PMCID: PMC8977319 DOI: 10.1155/2022/4086935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 02/19/2022] [Accepted: 03/04/2022] [Indexed: 11/17/2022]
Abstract
Background The role and expression level change in circ_TNPO1 (hsa_circ_0072951) in atherosclerosis (AS) and VSMC dysfunction remain unknown. In this study, we try to explore the effects of circ_TNPO1 on oxidized low-density lipoprotein (ox-LDL)-induced human vascular smooth muscle cell (VSMC) excessive proliferation and migration, and the potential molecular mechanism. Methods Quantitative real-time polymerase chain reaction (RT-qPCR) and western blot experiment were used to detect the serum samples from AS patients and healthy controls. CCK-8, Transwell, and the dual-luciferase reporter gene assay were used to detect the cell biology. Results In human AS serum and ox-LDL-induced VSMCs, circ_TNPO1 was increased, whereas miR-181b was decreased. Silencing circ_TNPO1 inhibited proliferation and migration activity and reduced protein expression of PCNA, Ki-67, MMP2, and E-cadherin and promoted N-cadherin protein expression in ox-LDL induced VSMCs. Remarkably, miR-181b knockdown or Notch1 overexpression could efficiently offset the proliferation and migration inhibiting effect of circ_TNPO1 knockdown in ox-LDL-induced VSMCs. Furthermore, a molecular mechanism study pointed out that circ_TNPO1 and Notch1 are direct-acting targets of miR-181b. Conclusions In conclusion, our study indicated that circ_TNPO1 promotes the proliferation and migration progression of VSMCs in atherosclerosis through the miR-181b/Notch1 axis.
Collapse
Affiliation(s)
- Mingxiang Chen
- Department of Cardiovascular Surgery, The Third Affiliated Hospital Of Chongqing Medical University, Chongqing, China
| | - Fuping Li
- Department of Cardiovascular Surgery, The Third Affiliated Hospital Of Chongqing Medical University, Chongqing, China
| | - Qilong Jiang
- Department of Cardiovascular Surgery, The Third Affiliated Hospital Of Chongqing Medical University, Chongqing, China
| | - WeiMin Zhang
- Department of Cardiovascular Surgery, The Third Affiliated Hospital Of Chongqing Medical University, Chongqing, China
| | - Zhiping Li
- Department of Cardiovascular Surgery, The Third Affiliated Hospital Of Chongqing Medical University, Chongqing, China
| | - Wenshuai Tang
- Department of Cardiovascular Surgery, The Third Affiliated Hospital Of Chongqing Medical University, Chongqing, China
| |
Collapse
|
20
|
Singh V, Akash R, Chaudhary G, Singh R, Choudhury S, Shukla A, Prabhu SN, Gangwar N, Garg SK. Sepsis downregulates aortic Notch signaling to produce vascular hyporeactivity in mice. Sci Rep 2022; 12:2941. [PMID: 35190630 PMCID: PMC8861011 DOI: 10.1038/s41598-022-06949-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 02/02/2022] [Indexed: 11/25/2022] Open
Abstract
Inhibition of Notch signaling in macrophages is known to reduce inflammation, however, its role in regulating vascular hyporeactivity in sepsis is unknown. Thus we aimed to evaluate the effect of sepsis on vascular Notch signaling. Polymicrobial sepsis was induced by caecal ligation and puncture (CLP) in mice. mRNA expressions of Notch receptors (Notch1,3) and ligands (Jag1, Dll4), and downstream effector genes (Hey1, MLCK, MYPT1) were assessed by RT-qPCR. Protein level of activated Notch (NICD) was assessed by Western blot and immuno-histochemistry. Isometric tension in isolated aortic rings was measured by wire myography.CLP down-regulated aortic expression of Notch3, Jag1 and Dll4 as compared to control mice. Additionally, the protein level of NICD was found to be lesser in aortic tissue sections from CLP mice. Expression of Hey1 and MLCK were attenuated whereas MYPT1 expression was increased in septic mouse aorta. DAPT pretreatment did not improve CLP-induced vascular hyporeactivity to NA, CaCl2 and high K+ (80 mM), rather significantly attenuated the aortic response to these vasoconstrictors in control mice. Treatment with 1400 W reversed attenuated Notch3 (but not Jag1 and MLCK) expression in septic mouse aorta. In conclusion, sepsis significantly attenuated the Notch (especially Notch3) signaling in mouse aorta along with reduction in contractile gene expression and vasoconstriction response. Further, iNOS/NO pathway was involved in sepsis-induced down-regulation of Notch3 receptor. Thus systemic inhibition of Notch signaling during sepsis may have serious impact on sepsis-induced vascular hyporeactivity.
Collapse
Affiliation(s)
- Vandana Singh
- Smooth Muscle Pharmacology and Molecular Pharmacology Laboratory, Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science and Animal Husbandry, U.P. Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go-Anusandhan Sansthan (DUVASU), Mathura, 281001, India
| | - Raut Akash
- Smooth Muscle Pharmacology and Molecular Pharmacology Laboratory, Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science and Animal Husbandry, U.P. Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go-Anusandhan Sansthan (DUVASU), Mathura, 281001, India
| | - Gaurav Chaudhary
- Smooth Muscle Pharmacology and Molecular Pharmacology Laboratory, Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science and Animal Husbandry, U.P. Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go-Anusandhan Sansthan (DUVASU), Mathura, 281001, India
| | - Rajneesh Singh
- Smooth Muscle Pharmacology and Molecular Pharmacology Laboratory, Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science and Animal Husbandry, U.P. Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go-Anusandhan Sansthan (DUVASU), Mathura, 281001, India
| | - Soumen Choudhury
- Smooth Muscle Pharmacology and Molecular Pharmacology Laboratory, Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science and Animal Husbandry, U.P. Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go-Anusandhan Sansthan (DUVASU), Mathura, 281001, India.
| | - Amit Shukla
- Smooth Muscle Pharmacology and Molecular Pharmacology Laboratory, Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science and Animal Husbandry, U.P. Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go-Anusandhan Sansthan (DUVASU), Mathura, 281001, India
| | - Shyama N Prabhu
- Department of Veterinary Pathology, College of Veterinary Science and Animal Husbandry, U.P. Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go-Anusandhan Sansthan, Mathura, 281001, India
| | - Neeraj Gangwar
- Department of Veterinary Pathology, College of Veterinary Science and Animal Husbandry, U.P. Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go-Anusandhan Sansthan, Mathura, 281001, India
| | - Satish K Garg
- Smooth Muscle Pharmacology and Molecular Pharmacology Laboratory, Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science and Animal Husbandry, U.P. Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go-Anusandhan Sansthan (DUVASU), Mathura, 281001, India
| |
Collapse
|
21
|
Liu M, Xu K, Saaoud F, Shao Y, Zhang R, Lu Y, Sun Y, Drummer C, Li L, Wu S, Kunapuli SP, Criner GJ, Sun J, Shan H, Jiang X, Wang H, Yang X. 29 m 6A-RNA Methylation (Epitranscriptomic) Regulators Are Regulated in 41 Diseases including Atherosclerosis and Tumors Potentially via ROS Regulation - 102 Transcriptomic Dataset Analyses. J Immunol Res 2022; 2022:1433323. [PMID: 35211628 PMCID: PMC8863469 DOI: 10.1155/2022/1433323] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/31/2021] [Indexed: 12/12/2022] Open
Abstract
We performed a database mining on 102 transcriptomic datasets for the expressions of 29 m6A-RNA methylation (epitranscriptomic) regulators (m6A-RMRs) in 41 diseases and cancers and made significant findings: (1) a few m6A-RMRs were upregulated; and most m6A-RMRs were downregulated in sepsis, acute respiratory distress syndrome, shock, and trauma; (2) half of 29 m6A-RMRs were downregulated in atherosclerosis; (3) inflammatory bowel disease and rheumatoid arthritis modulated m6A-RMRs more than lupus and psoriasis; (4) some organ failures shared eight upregulated m6A-RMRs; end-stage renal failure (ESRF) downregulated 85% of m6A-RMRs; (5) Middle-East respiratory syndrome coronavirus infections modulated m6A-RMRs the most among viral infections; (6) proinflammatory oxPAPC modulated m6A-RMRs more than DAMP stimulation including LPS and oxLDL; (7) upregulated m6A-RMRs were more than downregulated m6A-RMRs in cancer types; five types of cancers upregulated ≥10 m6A-RMRs; (8) proinflammatory M1 macrophages upregulated seven m6A-RMRs; (9) 86% of m6A-RMRs were differentially expressed in the six clusters of CD4+Foxp3+ immunosuppressive Treg, and 8 out of 12 Treg signatures regulated m6A-RMRs; (10) immune checkpoint receptors TIM3, TIGIT, PD-L2, and CTLA4 modulated m6A-RMRs, and inhibition of CD40 upregulated m6A-RMRs; (11) cytokines and interferons modulated m6A-RMRs; (12) NF-κB and JAK/STAT pathways upregulated more than downregulated m6A-RMRs whereas TP53, PTEN, and APC did the opposite; (13) methionine-homocysteine-methyl cycle enzyme Mthfd1 downregulated more than upregulated m6A-RMRs; (14) m6A writer RBM15 and one m6A eraser FTO, H3K4 methyltransferase MLL1, and DNA methyltransferase, DNMT1, regulated m6A-RMRs; and (15) 40 out of 165 ROS regulators were modulated by m6A eraser FTO and two m6A writers METTL3 and WTAP. Our findings shed new light on the functions of upregulated m6A-RMRs in 41 diseases and cancers, nine cellular and molecular mechanisms, novel therapeutic targets for inflammatory disorders, metabolic cardiovascular diseases, autoimmune diseases, organ failures, and cancers.
Collapse
Affiliation(s)
- Ming Liu
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
- Department of Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Keman Xu
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Fatma Saaoud
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Ying Shao
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Ruijing Zhang
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
- Department of Nephrology, The Affiliated People's Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Yifan Lu
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Yu Sun
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Charles Drummer
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Li Li
- Department of Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Sheng Wu
- Metabolic Disease Research; Inflammation, Translational & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Satya P. Kunapuli
- Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Gerard J. Criner
- Thoracic Medicine and Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Jianxin Sun
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Huimin Shan
- Metabolic Disease Research; Inflammation, Translational & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Xiaohua Jiang
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
- Metabolic Disease Research; Inflammation, Translational & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Hong Wang
- Metabolic Disease Research; Inflammation, Translational & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Xiaofeng Yang
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
- Metabolic Disease Research; Inflammation, Translational & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
- Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
22
|
Lu L, Wang Z, Zhang H, Liu T, Fang H. Drynaria fortunei improves lipid profiles of elderly patients with postmenopausal osteoporosis via regulation of Notch1-NLRP3 inflammasome-mediated inflammation. Gynecol Endocrinol 2022; 38:176-180. [PMID: 34907823 DOI: 10.1080/09513590.2021.2015760] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
BACKGROUND Dyslipidemia is a common comorbidity in elderly patients with postmenopausal osteoporosis (PMOP). Drynaria fortunei (Rhizoma drynariae) is well-known in traditional Chinese medicine for its ability to improve bone mineral density (BMD). However, whether and how Drynaria fortunei improves plasma lipid profiles in elderly PMOP patients remains unclear. METHODS Eighty elderly female patients with concurrent PMOP and hyperlipemia were randomly assigned to Drynaria fortunei 2(n = 40) or control (n = 40) groups. The clinical efficacies of Drynaria fortunei were evaluated. At 0, 3-, 6-, 9-, and 12-month of follow-up, plasma levels of IL-1β, IL-18, TNF-α, IL-6, IL-8, and IL-10 were measured using ELISA, whereas PBMC levels of NLRP3, ASC, caspase-1, NF-κB, SIRT1, and Notch1 were measured using RT-qPCR. PBMC isolated from PMOP patients were cultured and treated with Drynaria fortunei to determine its influence on NLRP3 inflammasome and associated cytokines. RESULTS Drynaria fortunei effectively improved patients' BMD and lipid profiles. IL-1β, IL-18, TNF-α, IL-6, IL-8 levels, as well as inflammasome-molecules of NLRP3, ASC, caspase-1, and NF-κB increased over time in the control group, but were significantly attenuated with Drynaria fortunei administration. In vitro, Drynaria fortunei suppressed NLRP3 inflammasome and associated cytokines by increasing SIRT1 or decreasing Notch1. Drynaria fortunei had inhibitory effects on NLRP3 inflammasome and Notch1 even when SIRT1 expression was suppressed. CONCLUSIONS Drynaria fortunei has been demonstrated to significantly improve lipid profiles for elderly PMOP patients. Drynaria fortunei may down-regulate Notch1 independently of SIRT1 to suppress NLRP3 inflammasome-mediated inflammation, thus improving plasma lipid profile.
Collapse
Affiliation(s)
- Lin Lu
- Department of Orthopedics, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, PR China
| | - Zhi Wang
- Department of Orthopedics, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, PR China
| | - Hanqing Zhang
- Department of Orthopedics, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, PR China
| | - Tongou Liu
- Department of Gynecology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, PR China
- First Clinical Medical College, Hubei University of Traditional Chinese Medicine, Wuhan, PR China
- Hubei Province Academy of Traditional Chinese Medicine, Wuhan, PR China
| | - Hong Fang
- Department of Gynecology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, PR China
- First Clinical Medical College, Hubei University of Traditional Chinese Medicine, Wuhan, PR China
- Hubei Province Academy of Traditional Chinese Medicine, Wuhan, PR China
| |
Collapse
|
23
|
De Rosa S, Iaconetti C, Eyileten C, Yasuda M, Albanese M, Polimeni A, Sabatino J, Sorrentino S, Postula M, Indolfi C. Flow-Responsive Noncoding RNAs in the Vascular System: Basic Mechanisms for the Clinician. J Clin Med 2022; 11:jcm11020459. [PMID: 35054151 PMCID: PMC8777617 DOI: 10.3390/jcm11020459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/11/2022] [Accepted: 01/14/2022] [Indexed: 12/10/2022] Open
Abstract
The vascular system is largely exposed to the effect of changing flow conditions. Vascular cells can sense flow and its changes. Flow sensing is of pivotal importance for vascular remodeling. In fact, it influences the development and progression of atherosclerosis, controls its location and has a major influx on the development of local complications. Despite its importance, the research community has traditionally paid scarce attention to studying the association between different flow conditions and vascular biology. More recently, a growing body of evidence has been accumulating, revealing that ncRNAs play a key role in the modulation of several biological processes linking flow-sensing to vascular pathophysiology. This review summarizes the most relevant evidence on ncRNAs that are directly or indirectly responsive to flow conditions to the benefit of the clinician, with a focus on the underpinning mechanisms and their potential application as disease biomarkers.
Collapse
Affiliation(s)
- Salvatore De Rosa
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (C.I.); (M.Y.); (M.A.); (A.P.); (J.S.); (S.S.)
- Correspondence: (S.D.R.); (C.I.)
| | - Claudio Iaconetti
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (C.I.); (M.Y.); (M.A.); (A.P.); (J.S.); (S.S.)
| | - Ceren Eyileten
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology CePT, Medical University of Warsaw, 02-097 Warsaw, Poland; (C.E.); (M.P.)
| | - Masakazu Yasuda
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (C.I.); (M.Y.); (M.A.); (A.P.); (J.S.); (S.S.)
| | - Michele Albanese
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (C.I.); (M.Y.); (M.A.); (A.P.); (J.S.); (S.S.)
| | - Alberto Polimeni
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (C.I.); (M.Y.); (M.A.); (A.P.); (J.S.); (S.S.)
| | - Jolanda Sabatino
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (C.I.); (M.Y.); (M.A.); (A.P.); (J.S.); (S.S.)
| | - Sabato Sorrentino
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (C.I.); (M.Y.); (M.A.); (A.P.); (J.S.); (S.S.)
| | - Marek Postula
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology CePT, Medical University of Warsaw, 02-097 Warsaw, Poland; (C.E.); (M.P.)
| | - Ciro Indolfi
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (C.I.); (M.Y.); (M.A.); (A.P.); (J.S.); (S.S.)
- Mediterranea Cardiocentro, 80122 Naples, Italy
- Correspondence: (S.D.R.); (C.I.)
| |
Collapse
|
24
|
Mukherjee P, Chattopadhyay A, Grijalva V, Dorreh N, Lagishetty V, Jacobs JP, Clifford BL, Vallim T, Mack JJ, Navab M, Reddy ST, Fogelman AM. Oxidized phospholipids cause changes in jejunum mucus that induce dysbiosis and systemic inflammation. J Lipid Res 2022; 63:100153. [PMID: 34808192 PMCID: PMC8953663 DOI: 10.1016/j.jlr.2021.100153] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 10/26/2021] [Accepted: 11/16/2021] [Indexed: 12/18/2022] Open
Abstract
We previously reported that adding a concentrate of transgenic tomatoes expressing the apoA-I mimetic peptide 6F (Tg6F) to a Western diet (WD) ameliorated systemic inflammation. To determine the mechanism(s) responsible for these observations, Ldlr-/- mice were fed chow, a WD, or WD plus Tg6F. We found that a WD altered the taxonomic composition of bacteria in jejunum mucus. For example, Akkermansia muciniphila virtually disappeared, while overall bacteria numbers and lipopolysaccharide (LPS) levels increased. In addition, gut permeability increased, as did the content of reactive oxygen species and oxidized phospholipids in jejunum mucus in WD-fed mice. Moreover, gene expression in the jejunum decreased for multiple peptides and proteins that are secreted into the mucous layer of the jejunum that act to limit bacteria numbers and their interaction with enterocytes including regenerating islet-derived proteins, defensins, mucin 2, surfactant A, and apoA-I. Following WD, gene expression also decreased for Il36γ, Il23, and Il22, cytokines critical for antimicrobial activity. WD decreased expression of both Atoh1 and Gfi1, genes required for the formation of goblet and Paneth cells, and immunohistochemistry revealed decreased numbers of goblet and Paneth cells. Adding Tg6F ameliorated these WD-mediated changes. Adding oxidized phospholipids ex vivo to the jejunum from mice fed a chow diet reproduced the changes in gene expression in vivo that occurred when the mice were fed WD and were prevented with addition of 6F peptide. We conclude that Tg6F ameliorates the WD-mediated increase in oxidized phospholipids that cause changes in jejunum mucus, which induce dysbiosis and systemic inflammation.
Collapse
Affiliation(s)
- Pallavi Mukherjee
- Division of Cardiology, Department of Medicine, Los Angeles, CA, USA
| | | | - Victor Grijalva
- Division of Cardiology, Department of Medicine, Los Angeles, CA, USA
| | - Nasrin Dorreh
- Division of Cardiology, Department of Medicine, Los Angeles, CA, USA
| | - Venu Lagishetty
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Los Angeles, CA, USA; UCLA Microbiome Center, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jonathan P Jacobs
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Los Angeles, CA, USA; UCLA Microbiome Center, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; The Division of Gastroenterology, Hepatology and Parenteral Nutrition, Veterans Administration Greater Los Angeles Healthcare System Los Angeles, Los Angeles, CA, USA
| | | | - Thomas Vallim
- Division of Cardiology, Department of Medicine, Los Angeles, CA, USA; Department of Biological Chemistry, Los Angeles, CA, USA
| | - Julia J Mack
- Division of Cardiology, Department of Medicine, Los Angeles, CA, USA
| | - Mohamad Navab
- Division of Cardiology, Department of Medicine, Los Angeles, CA, USA
| | - Srinivasa T Reddy
- Division of Cardiology, Department of Medicine, Los Angeles, CA, USA; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
| | - Alan M Fogelman
- Division of Cardiology, Department of Medicine, Los Angeles, CA, USA
| |
Collapse
|
25
|
Katakia YT, Thakkar NP, Thakar S, Sakhuja A, Goyal R, Sharma H, Dave R, Mandloi A, Basu S, Nigam I, Kuncharam BVR, Chowdhury S, Majumder S. Dynamic alterations of H3K4me3 and H3K27me3 at ADAM17 and Jagged-1 gene promoters cause an inflammatory switch of endothelial cells. J Cell Physiol 2021; 237:992-1012. [PMID: 34520565 DOI: 10.1002/jcp.30579] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/25/2021] [Accepted: 08/28/2021] [Indexed: 01/01/2023]
Abstract
Histone protein modifications control the inflammatory state of many immune cells. However, how dynamic alteration in histone methylation causes endothelial inflammation and apoptosis is not clearly understood. To examine this, we explored two contrasting histone methylations; an activating histone H3 lysine 4 trimethylation (H3K4me3) and a repressive histone H3 lysine 27 trimethylation (H3K27me3) in endothelial cells (EC) undergoing inflammation. Through computer-aided reconstruction and 3D printing of the human coronary artery, we developed a unique model where EC were exposed to a pattern of oscillatory/disturbed flow as similar to in vivo conditions. Upon induction of endothelial inflammation, we detected a significant rise in H3K4me3 caused by an increase in the expression of SET1/COMPASS family of H3K4 methyltransferases, including MLL1, MLL2, and SET1B. In contrast, EC undergoing inflammation exhibited truncated H3K27me3 level engendered by EZH2 cytosolic translocation through threonine 367 phosphorylation and an increase in the expression of histone demethylating enzyme JMJD3 and UTX. Additionally, many SET1/COMPASS family of proteins, including MLL1 (C), MLL2, and WDR5, were associated with either UTX or JMJD3 or both and such association was elevated in EC upon exposure to inflammatory stimuli. Dynamic enrichment of H3K4me3 and loss of H3K27me3 at Notch-associated gene promoters caused ADAM17 and Jagged-1 derepression and abrupt Notch activation. Conversely, either reducing H3K4me3 or increasing H3K27me3 in EC undergoing inflammation attenuated Notch activation, endothelial inflammation, and apoptosis. Together, these findings indicate that dynamic chromatin modifications may cause an inflammatory and apoptotic switch of EC and that epigenetic reprogramming can potentially improve outcomes in endothelial inflammation-associated cardiovascular diseases.
Collapse
Affiliation(s)
- Yash T Katakia
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, India
| | - Niyati P Thakkar
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, India
| | - Sumukh Thakar
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, India
| | - Ashima Sakhuja
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, India
| | - Raghav Goyal
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, India
| | - Harshita Sharma
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, India
| | - Rakshita Dave
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, India
| | - Ayushi Mandloi
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, India
| | - Sayan Basu
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, India
| | - Ishan Nigam
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, India
| | - Bhanu V R Kuncharam
- Department of Chemical Engineering, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, India
| | - Shibasish Chowdhury
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, India
| | - Syamantak Majumder
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, India
| |
Collapse
|
26
|
Martos-Rodríguez CJ, Albarrán-Juárez J, Morales-Cano D, Caballero A, MacGrogan D, de la Pompa JL, Carramolino L, Bentzon JF. Fibrous Caps in Atherosclerosis Form by Notch-Dependent Mechanisms Common to Arterial Media Development. Arterioscler Thromb Vasc Biol 2021; 41:e427-e439. [PMID: 34261328 DOI: 10.1161/atvbaha.120.315627] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Objective Atheromatous fibrous caps are produced by smooth muscle cells (SMCs) that are recruited to the subendothelial space. We tested whether the recruitment mechanisms are the same as in embryonic artery development, which relies prominently on Notch signaling to form the subendothelial medial SMC layers. Approach and Results Notch elements were expressed in regions of fibrous cap in human and mouse plaques. To assess the causal role of Notch signaling in cap formation, we studied atherosclerosis in mice where the Notch pathway was inactivated in SMCs by conditional knockout of the essential effector transcription factor RBPJ (recombination signal-binding protein for immunoglobulin kappa J region). The recruitment of cap SMCs was significantly reduced without major effects on plaque size. Lineage tracing revealed the accumulation of SMC-derived plaque cells in the cap region was unaltered but that Notch-defective cells failed to re-acquire the SMC phenotype in the cap. Conversely, to analyze whether the loss of Notch signaling is required for SMC-derived cells to accumulate in atherogenesis, we studied atherosclerosis in mice with constitutive activation of Notch signaling in SMCs achieved by conditional expression of the Notch intracellular domain. Forced Notch signaling inhibited the ability of medial SMCs to contribute to plaque cells, including both cap SMCs and osteochondrogenic cells, and significantly reduced atherosclerosis development. Conclusions Sequential loss and gain of Notch signaling is needed to build the cap SMC population. The shared mechanisms with embryonic arterial media assembly suggest that the cap forms as a neo-media that restores the connection between endothelium and subendothelial SMCs, transiently disrupted in early atherogenesis.
Collapse
MESH Headings
- Actins/genetics
- Actins/metabolism
- Animals
- Arteries/metabolism
- Arteries/pathology
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Cell Lineage
- Cells, Cultured
- Disease Progression
- Fibrosis
- Humans
- Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics
- Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism
- Jagged-1 Protein/genetics
- Jagged-1 Protein/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phenotype
- Plaque, Atherosclerotic
- Rats
- Receptors, Notch/genetics
- Receptors, Notch/metabolism
- Signal Transduction
- Tunica Media/metabolism
- Tunica Media/pathology
- Mice
Collapse
Affiliation(s)
- Carlos J Martos-Rodríguez
- Experimental Pathology of Atherosclerosis Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (C.J.M.-R., D.M.-C., A.C., L.C., J.F.B.)
| | - Julián Albarrán-Juárez
- Heart Diseases, Department of Clinical Medicine (J.A.-J., A.C., J.F.B.), Aarhus University, Denmark
| | - Daniel Morales-Cano
- Experimental Pathology of Atherosclerosis Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (C.J.M.-R., D.M.-C., A.C., L.C., J.F.B.)
| | - Ainoa Caballero
- Experimental Pathology of Atherosclerosis Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (C.J.M.-R., D.M.-C., A.C., L.C., J.F.B.)
- Heart Diseases, Department of Clinical Medicine (J.A.-J., A.C., J.F.B.), Aarhus University, Denmark
| | - Donal MacGrogan
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (D.M., J.L.d.l.P.)
- Ciber de Enfermedades Cardiovasculares, Madrid, Spain (D.M., J.L.d.l.P.)
| | - José Luis de la Pompa
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (D.M., J.L.d.l.P.)
- Ciber de Enfermedades Cardiovasculares, Madrid, Spain (D.M., J.L.d.l.P.)
| | - Laura Carramolino
- Experimental Pathology of Atherosclerosis Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (C.J.M.-R., D.M.-C., A.C., L.C., J.F.B.)
| | - Jacob F Bentzon
- Experimental Pathology of Atherosclerosis Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (C.J.M.-R., D.M.-C., A.C., L.C., J.F.B.)
- Heart Diseases, Department of Clinical Medicine (J.A.-J., A.C., J.F.B.), Aarhus University, Denmark
- Steno Diabetes Center Aarhus, Department of Clinical Medicine (J.F.B.), Aarhus University, Denmark
| |
Collapse
|
27
|
Marracino L, Fortini F, Bouhamida E, Camponogara F, Severi P, Mazzoni E, Patergnani S, D’Aniello E, Campana R, Pinton P, Martini F, Tognon M, Campo G, Ferrari R, Vieceli Dalla Sega F, Rizzo P. Adding a "Notch" to Cardiovascular Disease Therapeutics: A MicroRNA-Based Approach. Front Cell Dev Biol 2021; 9:695114. [PMID: 34527667 PMCID: PMC8435685 DOI: 10.3389/fcell.2021.695114] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 08/09/2021] [Indexed: 12/18/2022] Open
Abstract
Dysregulation of the Notch pathway is implicated in the pathophysiology of cardiovascular diseases (CVDs), but, as of today, therapies based on the re-establishing the physiological levels of Notch in the heart and vessels are not available. A possible reason is the context-dependent role of Notch in the cardiovascular system, which would require a finely tuned, cell-specific approach. MicroRNAs (miRNAs) are short functional endogenous, non-coding RNA sequences able to regulate gene expression at post-transcriptional levels influencing most, if not all, biological processes. Dysregulation of miRNAs expression is implicated in the molecular mechanisms underlying many CVDs. Notch is regulated and regulates a large number of miRNAs expressed in the cardiovascular system and, thus, targeting these miRNAs could represent an avenue to be explored to target Notch for CVDs. In this Review, we provide an overview of both established and potential, based on evidence in other pathologies, crosstalks between miRNAs and Notch in cellular processes underlying atherosclerosis, myocardial ischemia, heart failure, calcification of aortic valve, and arrhythmias. We also discuss the potential advantages, as well as the challenges, of using miRNAs for a Notch-based approach for the diagnosis and treatment of the most common CVDs.
Collapse
Affiliation(s)
- Luisa Marracino
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | | | - Esmaa Bouhamida
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Francesca Camponogara
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Paolo Severi
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Elisa Mazzoni
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Simone Patergnani
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Emanuele D’Aniello
- Cardiovascular Institute, Azienda Ospedaliero-Universitaria di Ferrara, Ferrara, Italy
| | - Roberta Campana
- Cardiovascular Institute, Azienda Ospedaliero-Universitaria di Ferrara, Ferrara, Italy
| | - Paolo Pinton
- Maria Cecilia Hospital, GVM Care & Research, Ravenna, Italy
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Fernanda Martini
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Mauro Tognon
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Gianluca Campo
- Maria Cecilia Hospital, GVM Care & Research, Ravenna, Italy
- Cardiovascular Institute, Azienda Ospedaliero-Universitaria di Ferrara, Ferrara, Italy
| | - Roberto Ferrari
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, Ferrara, Italy
- Maria Cecilia Hospital, GVM Care & Research, Ravenna, Italy
| | | | - Paola Rizzo
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, Ferrara, Italy
- Maria Cecilia Hospital, GVM Care & Research, Ravenna, Italy
| |
Collapse
|
28
|
Luo C, Zhou X, Wang L, Zeng Q, Fan J, He S, Zhang H, Wei A. Screening and identification of NOTCH1, CDKN2A, and NOS3 as differentially expressed autophagy-related genes in erectile dysfunction. PeerJ 2021; 9:e11986. [PMID: 34447638 PMCID: PMC8366525 DOI: 10.7717/peerj.11986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 07/26/2021] [Indexed: 12/19/2022] Open
Abstract
Background Loss of function of key autophagy genes are associated with a variety of diseases. However specific role of autophagy-related genes in erectile dysfunction ED remains unclear. This study explores the autophagy-related differentially expressed genes (ARGs) profiles and related molecular mechanisms in Corpus Cavernosum endothelial dysfunction, which is a leading cause of ED. Methods The Gene Expression Omnibus (GEO) database was used to identify the key genes and pathways. Differentially expressed genes (DEGs) were mined using the limma package in R language. Next, ARGs were obtained by matching DEGs and autophagy-related genes from GeneCard using Venn diagrams. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses of ARGs were described using clusterProfiler and org.Hs.eg.db in R. Moreover, hub ARGs were screened out through protein-protein interaction (PPI), gene-microRNAs, and gene-transcription factors (TFs) networks then visualized using Cytoscape. Of note, the rat model of diabetic ED was established to validate some hub ARGs with qRT-PCR and Western blots. Results Twenty ARGs were identified from four ED samples and eight non-ED samples. GO analysis revealed that molecular functions (MF) of upregulated ARGs were mainly enriched in nuclear receptor activity. Also, MF of downregulated ARGs were mainly enriched in oxidoreductase activity, acting on NAD(P)H and heme proteins as acceptors. Moreover, six hub ARGs were identified by setting high degrees in the network. Additionally, hsa-mir-24-3p and hsa-mir-335-5p might play a central role in several ARGs regulation, and the transcription factors-hub genes network was centered with 13 ARGs. The experimental results further showed that the expression of Notch1, NOS3, and CDKN2A in the diabetic ED group was downregulated compared to the control. Conclusions Our study deepens the autophagy-related mechanistic understanding of endothelial dysfunction of ED. NOTCH1, CDKN2A, and NOS3 are involved in the regulation of endothelial dysfunction and may be potential therapeutic targets for ED by modulating autophagy.
Collapse
Affiliation(s)
- Chao Luo
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiongcai Zhou
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Department of Urology, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Li Wang
- School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong, China
| | - Qinyu Zeng
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Junhong Fan
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Shuhua He
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Haibo Zhang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Anyang Wei
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
29
|
Fortini F, Vieceli Dalla Sega F, Marracino L, Severi P, Rapezzi C, Rizzo P, Ferrari R. Well-Known and Novel Players in Endothelial Dysfunction: Updates on a Notch(ed) Landscape. Biomedicines 2021; 9:biomedicines9080997. [PMID: 34440201 PMCID: PMC8393382 DOI: 10.3390/biomedicines9080997] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/03/2021] [Accepted: 08/06/2021] [Indexed: 12/27/2022] Open
Abstract
Endothelial dysfunction characterizes every aspect of the so-called cardiovascular continuum, a series of events ranging from hypertension to the development of atherosclerosis and, finally, to coronary heart disease, thrombus formation, myocardial infarction, and heart failure. Endothelial dysfunction is the main prognostic factor for the progression of vascular disorders, which responds to drug intervention and lifestyle changes. Virtually all of the drugs used to prevent cardiovascular disorders, such as long-used and new antilipidemic agents and inhibitors of angiotensin enzyme (ACEi), exert an important effect on the endothelium. Endothelial dysfunction is a central feature of coronavirus disease -19 (COVID-19), and it is now clear that life-risk complications of the disease are prompted by alterations of the endothelium induced by viral infection. As a consequence, the progression of COVID-19 is worse in the subjects in whom endothelial dysfunction is already present, such as elderly, diabetic, obese, and hypertensive patients. Importantly, circulating biomarkers of endothelial activation and injury predict the severity and mortality of the disease and can be used to evaluate the efficacy of treatments. The purpose of this review is to provide updates on endothelial function by discussing its clinical relevance in the cardiovascular continuum, the latest insights from molecular and cellular biology, and their implications for clinical practice, with a focus on new actors, such as the Notch signaling and emerging therapies for cardiovascular disease.
Collapse
Affiliation(s)
- Francesca Fortini
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (F.F.); (F.V.D.S.); (C.R.); (P.R.)
| | | | - Luisa Marracino
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (P.S.)
| | - Paolo Severi
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (P.S.)
| | - Claudio Rapezzi
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (F.F.); (F.V.D.S.); (C.R.); (P.R.)
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (P.S.)
| | - Paola Rizzo
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (F.F.); (F.V.D.S.); (C.R.); (P.R.)
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (P.S.)
| | - Roberto Ferrari
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (F.F.); (F.V.D.S.); (C.R.); (P.R.)
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (P.S.)
- Correspondence: ; Tel.: +39-053-229-3707
| |
Collapse
|
30
|
Kapoor A, Nation DA. Role of Notch signaling in neurovascular aging and Alzheimer's disease. Semin Cell Dev Biol 2021; 116:90-97. [PMID: 33384205 PMCID: PMC8236496 DOI: 10.1016/j.semcdb.2020.12.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/03/2020] [Accepted: 12/21/2020] [Indexed: 12/13/2022]
Abstract
The Notch signaling pathway is an evolutionarily conserved cell signaling system known to be involved in vascular development and function. Recent evidence suggests that dysfunctional Notch signaling could play a critical role in the pathophysiology of neurodegenerative diseases. We reviewed current literature on the role of Notch signaling pathway, and specifically Notch receptor genes and proteins, in aging, cerebrovascular disease and Alzheimer's disease. We hypothesize that Notch signaling may represent a key point of overlap between age-related vascular and Alzheimer's pathophysiology contributing to their comorbidity and combined influence on cognitive decline and dementia. Numerous findings from studies of genetics, neuropathology and cell culture models all suggest a link between altered Notch signaling and Alzheimer's pathophysiology. Age-related changes in Notch signaling may also trigger neurovascular dysfunction, contributing to the development of neurodegenerative diseases; however, additional studies are warranted. Future research directly exploring the influence of aberrant Notch signaling in the development of Alzheimer's disease is needed to better understand this mechanism.
Collapse
Affiliation(s)
- Arunima Kapoor
- Department of Psychological Science, University of California, Irvine, Irvine, CA, USA
| | - Daniel A Nation
- Department of Psychological Science, University of California, Irvine, Irvine, CA, USA; Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA.
| |
Collapse
|
31
|
Liu M, Wu N, Xu K, Saaoud F, Vasilopoulos E, Shao Y, Zhang R, Wang J, Shen H, Yang WY, Lu Y, Sun Y, Drummer C, Liu L, Li L, Hu W, Yu J, Praticò D, Sun J, Jiang X, Wang H, Yang X. Organelle Crosstalk Regulators Are Regulated in Diseases, Tumors, and Regulatory T Cells: Novel Classification of Organelle Crosstalk Regulators. Front Cardiovasc Med 2021; 8:713170. [PMID: 34368262 PMCID: PMC8339352 DOI: 10.3389/fcvm.2021.713170] [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: 05/21/2021] [Accepted: 06/14/2021] [Indexed: 12/15/2022] Open
Abstract
To examine whether the expressions of 260 organelle crosstalk regulators (OCRGs) in 16 functional groups are modulated in 23 diseases and 28 tumors, we performed extensive -omics data mining analyses and made a set of significant findings: (1) the ratios of upregulated vs. downregulated OCRGs are 1:2.8 in acute inflammations, 1:1 in metabolic diseases, 1:1.2 in autoimmune diseases, and 1:3.8 in organ failures; (2) sepsis and trauma-upregulated OCRG groups such as vesicle, mitochondrial (MT) fission, and mitophagy but not others, are termed as the cell crisis-handling OCRGs. Similarly, sepsis and trauma plus organ failures upregulated seven OCRG groups including vesicle, MT fission, mitophagy, sarcoplasmic reticulum–MT, MT fusion, autophagosome–lysosome fusion, and autophagosome/endosome–lysosome fusion, classified as the cell failure-handling OCRGs; (3) suppression of autophagosome–lysosome fusion in endothelial and epithelial cells is required for viral replications, which classify this decreased group as the viral replication-suppressed OCRGs; (4) pro-atherogenic damage-associated molecular patterns (DAMPs) such as oxidized low-density lipoprotein (oxLDL), lipopolysaccharide (LPS), oxidized-1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine (oxPAPC), and interferons (IFNs) totally upregulated 33 OCRGs in endothelial cells (ECs) including vesicle, MT fission, mitophagy, MT fusion, endoplasmic reticulum (ER)–MT contact, ER– plasma membrane (PM) junction, autophagosome/endosome–lysosome fusion, sarcoplasmic reticulum–MT, autophagosome–endosome/lysosome fusion, and ER–Golgi complex (GC) interaction as the 10 EC-activation/inflammation-promoting OCRG groups; (5) the expression of OCRGs is upregulated more than downregulated in regulatory T cells (Tregs) from the lymph nodes, spleen, peripheral blood, intestine, and brown adipose tissue in comparison with that of CD4+CD25− T effector controls; (6) toll-like receptors (TLRs), reactive oxygen species (ROS) regulator nuclear factor erythroid 2-related factor 2 (Nrf2), and inflammasome-activated regulator caspase-1 regulated the expressions of OCRGs in diseases, virus-infected cells, and pro-atherogenic DAMP-treated ECs; (7) OCRG expressions are significantly modulated in all the 28 cancer datasets, and the upregulated OCRGs are correlated with tumor immune infiltrates in some tumors; (8) tumor promoter factor IKK2 and tumor suppressor Tp53 significantly modulate the expressions of OCRGs. Our findings provide novel insights on the roles of upregulated OCRGs in the pathogenesis of inflammatory diseases and cancers, and novel pathways for the future therapeutic interventions for inflammations, sepsis, trauma, organ failures, autoimmune diseases, metabolic cardiovascular diseases (CVDs), and cancers.
Collapse
Affiliation(s)
- Ming Liu
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Department of Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, China
| | - Na Wu
- Departments of Endocrinology and Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Keman Xu
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Fatma Saaoud
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Eleni Vasilopoulos
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Ying Shao
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Ruijing Zhang
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Department of Nephrology, The Affiliated People's Hospital of Shanxi Medical University, Taiyuan, China
| | - Jirong Wang
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Department of Cardiology, The Affiliated People's Hospital of Shanxi Medical University, Taiyuan, China
| | - Haitao Shen
- Departments of Endocrinology and Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | | | - Yifan Lu
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Yu Sun
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Charles Drummer
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Lu Liu
- Metabolic Disease Research, Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Li Li
- Department of Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, China
| | - Wenhui Hu
- Metabolic Disease Research, Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Jun Yu
- Metabolic Disease Research, Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Domenico Praticò
- Alzheimer's Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Jianxin Sun
- Department of Medicine, Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| | - Xiaohua Jiang
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Metabolic Disease Research, Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Hong Wang
- Metabolic Disease Research, Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Xiaofeng Yang
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Metabolic Disease Research, Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| |
Collapse
|
32
|
Wang Y, Fang Y, Lu P, Wu B, Zhou B. NOTCH Signaling in Aortic Valve Development and Calcific Aortic Valve Disease. Front Cardiovasc Med 2021; 8:682298. [PMID: 34239905 PMCID: PMC8259786 DOI: 10.3389/fcvm.2021.682298] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/14/2021] [Indexed: 01/05/2023] Open
Abstract
NOTCH intercellular signaling mediates the communications between adjacent cells involved in multiple biological processes essential for tissue morphogenesis and homeostasis. The NOTCH1 mutations are the first identified human genetic variants that cause congenital bicuspid aortic valve (BAV) and calcific aortic valve disease (CAVD). Genetic variants affecting other genes in the NOTCH signaling pathway may also contribute to the development of BAV and the pathogenesis of CAVD. While CAVD occurs commonly in the elderly population with tri-leaflet aortic valve, patients with BAV have a high risk of developing CAVD at a young age. This observation indicates an important role of NOTCH signaling in the postnatal homeostasis of the aortic valve, in addition to its prenatal functions during aortic valve development. Over the last decade, animal studies, especially with the mouse models, have revealed detailed information in the developmental etiology of congenital aortic valve defects. In this review, we will discuss the molecular and cellular aspects of aortic valve development and examine the embryonic pathogenesis of BAV. We will focus our discussions on the NOTCH signaling during the endocardial-to-mesenchymal transformation (EMT) and the post-EMT remodeling of the aortic valve. We will further examine the involvement of the NOTCH mutations in the postnatal development of CAVD. We will emphasize the deleterious impact of the embryonic valve defects on the homeostatic mechanisms of the adult aortic valve for the purpose of identifying the potential therapeutic targets for disease intervention.
Collapse
Affiliation(s)
- Yidong Wang
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Yuan Fang
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Pengfei Lu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Bingruo Wu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Bin Zhou
- Departments of Genetics, Pediatrics (Pediatric Genetic Medicine), and Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, United States
- The Einstein Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
33
|
Allen F, Maillard I. Therapeutic Targeting of Notch Signaling: From Cancer to Inflammatory Disorders. Front Cell Dev Biol 2021; 9:649205. [PMID: 34124039 PMCID: PMC8194077 DOI: 10.3389/fcell.2021.649205] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 04/26/2021] [Indexed: 12/20/2022] Open
Abstract
Over the past two decades, the Notch signaling pathway has been investigated as a therapeutic target for the treatment of cancers, and more recently in the context of immune and inflammatory disorders. Notch is an evolutionary conserved pathway found in all metazoans that is critical for proper embryonic development and for the postnatal maintenance of selected tissues. Through cell-to-cell contacts, Notch orchestrates cell fate decisions and differentiation in non-hematopoietic and hematopoietic cell types, regulates immune cell development, and is integral to shaping the amplitude as well as the quality of different types of immune responses. Depriving some cancer types of Notch signals has been shown in preclinical studies to stunt tumor growth, consistent with an oncogenic function of Notch signaling. In addition, therapeutically antagonizing Notch signals showed preclinical potential to prevent or reverse inflammatory disorders, including autoimmune diseases, allergic inflammation and immune complications of life-saving procedures such allogeneic bone marrow and solid organ transplantation (graft-versus-host disease and graft rejection). In this review, we discuss some of these unique approaches, along with the successes and challenges encountered so far to target Notch signaling in preclinical and early clinical studies. Our goal is to emphasize lessons learned to provide guidance about emerging strategies of Notch-based therapeutics that could be deployed safely and efficiently in patients with immune and inflammatory disorders.
Collapse
Affiliation(s)
- Frederick Allen
- Division of Hematology and Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Ivan Maillard
- Division of Hematology and Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
34
|
Additive contribution of microRNA-34a/b/c to human arterial ageing and atherosclerosis. Atherosclerosis 2021; 327:49-58. [PMID: 34038763 DOI: 10.1016/j.atherosclerosis.2021.05.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 05/05/2021] [Accepted: 05/12/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS Preclinical data suggest that the ageing-induced miR-34a regulates vascular senescence. Herein we sought to assess whether the miR-34 family members miR-34a, miR-34b and miR-34c are involved in human arterial disease. METHODS Expression levels of miR-34a/b/c were quantified by TaqMan assay in peripheral blood mononuclear cells (PBMCs) derived from a consecutive cohort of 221 subjects who underwent cardiovascular risk assessment and thorough vascular examination for aortic stiffness and extent of arterial atherosclerosis. RESULTS High miR-34a was independently associated with the presence of CAD [OR (95%C.I.): 3.87 (1.56-9.56); p = 0.003] and high miR-34c with the number of diseased arterial beds [OR (95%C.I.): 1.88 (1.034-3.41); p = 0.038], while concurrent high expression of miR-34-a/c or all three miR-34a/b/c was associated with aortic stiffening (miR-34a/c: p = 0.022; miR-34a/b/c: p = 0.041) and with the extent of atherosclerosis [OR (95%C.I.) for number of coronary arteries [miR-34a/c: 3.29 (1.085-9.95); miR-34a/b/c: 6.06 (1.74-21.2)] and number of diseased arterial beds [miR-34a/c: 3.51 (1.45-8.52); miR-34a/b/c: 2.89 (1.05-7.92)] after controlling for possible confounders (p < 0.05 for all). Mechanistically, the increased levels of miR-34a or miR-34c were inversely associated with expression of SIRT1 or JAG1, NOTCH2, CTNNB1 and ATF1, respectively. The association of miR-34a/c or miR-34a/b/c with CAD was mainly mediated through SIRT1 and to a lesser extent through JAG1 as revealed by generalized structural equation modeling. Leukocyte-specific ablation of miR-34a/b/c ameliorates atherosclerotic plaque development and increases Sirt1 and Jag1 expression in an atherosclerosis mouse model confirming the human findings. CONCLUSIONS The present study reveals the clinical significance of the additive role of miR-34a/b/c in vascular ageing and atherosclerotic vascular disease.
Collapse
|
35
|
Novel Therapeutical Approaches to Managing Atherosclerotic Risk. Int J Mol Sci 2021; 22:ijms22094633. [PMID: 33924893 PMCID: PMC8125277 DOI: 10.3390/ijms22094633] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/23/2021] [Accepted: 04/26/2021] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis is a multifactorial vascular disease that leads to inflammation and stiffening of the arteries and decreases their elasticity due to the accumulation of calcium, small dense Low Density Lipoproteins (sdLDL), inflammatory cells, and fibrotic material. A review of studies pertaining to cardiometabolic risk factors, lipids alterations, hypolipidemic agents, nutraceuticals, hypoglycaemic drugs, atherosclerosis, endothelial dysfunction, and inflammation was performed. There are several therapeutic strategies including Proprotein Convertase Subtilisin/Kexin 9 (PCSK9) inhibitors, inclisiran, bempedoic acid, Glucagon-Like Peptide-1 Receptor agonists (GLP-1 RAs), and nutraceuticals that promise improvement in the atheromatous plaque from a molecular point of view, because have actions on the exposure of the LDL-Receptor (LDL-R), on endothelial dysfunction, activation of macrophages, on lipid oxidation, formations on foam cells, and deposition extracellular lipids. Atheroma plaque reduction both as a result of LDL-Cholesterol (LDL-C) intensive lowering and reducing inflammation and other residual risk factors is an integral part of the management of atherosclerotic disease, and the use of valid therapeutic alternatives appear to be appealing avenues to solving the problem.
Collapse
|
36
|
The molecular mechanism of mechanotransduction in vascular homeostasis and disease. Clin Sci (Lond) 2021; 134:2399-2418. [PMID: 32936305 DOI: 10.1042/cs20190488] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/14/2020] [Accepted: 09/02/2020] [Indexed: 12/12/2022]
Abstract
Blood vessels are constantly exposed to mechanical stimuli such as shear stress due to flow and pulsatile stretch. The extracellular matrix maintains the structural integrity of the vessel wall and coordinates with a dynamic mechanical environment to provide cues to initiate intracellular signaling pathway(s), thereby changing cellular behaviors and functions. However, the precise role of matrix-cell interactions involved in mechanotransduction during vascular homeostasis and disease development remains to be fully determined. In this review, we introduce hemodynamics forces in blood vessels and the initial sensors of mechanical stimuli, including cell-cell junctional molecules, G-protein-coupled receptors (GPCRs), multiple ion channels, and a variety of small GTPases. We then highlight the molecular mechanotransduction events in the vessel wall triggered by laminar shear stress (LSS) and disturbed shear stress (DSS) on vascular endothelial cells (ECs), and cyclic stretch in ECs and vascular smooth muscle cells (SMCs)-both of which activate several key transcription factors. Finally, we provide a recent overview of matrix-cell interactions and mechanotransduction centered on fibronectin in ECs and thrombospondin-1 in SMCs. The results of this review suggest that abnormal mechanical cues or altered responses to mechanical stimuli in EC and SMCs serve as the molecular basis of vascular diseases such as atherosclerosis, hypertension and aortic aneurysms. Collecting evidence and advancing knowledge on the mechanotransduction in the vessel wall can lead to a new direction of therapeutic interventions for vascular diseases.
Collapse
|
37
|
Lin A, Peiris NJ, Dhaliwal H, Hakim M, Li W, Ganesh S, Ramaswamy Y, Patel S, Misra A. Mural Cells: Potential Therapeutic Targets to Bridge Cardiovascular Disease and Neurodegeneration. Cells 2021; 10:cells10030593. [PMID: 33800271 PMCID: PMC7999039 DOI: 10.3390/cells10030593] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/01/2021] [Accepted: 03/04/2021] [Indexed: 02/06/2023] Open
Abstract
Mural cells collectively refer to the smooth muscle cells and pericytes of the vasculature. This heterogenous population of cells play a crucial role in the regulation of blood pressure, distribution, and the structural integrity of the vascular wall. As such, dysfunction of mural cells can lead to the pathogenesis and progression of a number of diseases pertaining to the vascular system. Cardiovascular diseases, particularly atherosclerosis, are perhaps the most well-described mural cell-centric case. For instance, atherosclerotic plaques are most often described as being composed of a proliferative smooth muscle cap accompanied by a necrotic core. More recently, the role of dysfunctional mural cells in neurodegenerative diseases, such as Alzheimer’s and Parkinson’s disease, is being recognized. In this review, we begin with an exploration of the mechanisms underlying atherosclerosis and neurodegenerative diseases, such as mural cell plasticity. Next, we highlight a selection of signaling pathways (PDGF, Notch and inflammatory signaling) that are conserved across both diseases. We propose that conserved mural cell signaling mechanisms can be exploited for the identification or development of dual-pronged therapeutics that impart both cardio- and neuroprotective qualities.
Collapse
MESH Headings
- Alzheimer Disease/drug therapy
- Alzheimer Disease/genetics
- Alzheimer Disease/metabolism
- Alzheimer Disease/pathology
- Animals
- Atherosclerosis/drug therapy
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Cardiotonic Agents/pharmacology
- Disease Models, Animal
- Gene Expression Regulation
- Humans
- Mice
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Neuroprotective Agents/pharmacology
- Parkinson Disease/drug therapy
- Parkinson Disease/genetics
- Parkinson Disease/metabolism
- Parkinson Disease/pathology
- Pericytes/drug effects
- Pericytes/metabolism
- Pericytes/pathology
- Plaque, Atherosclerotic/drug therapy
- Plaque, Atherosclerotic/genetics
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/pathology
- Platelet-Derived Growth Factor/genetics
- Platelet-Derived Growth Factor/metabolism
- Receptors, Notch/genetics
- Receptors, Notch/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Alexander Lin
- Heart Research Institute, Sydney, NSW 2042, Australia; (A.L.); (N.J.P.); (H.D.); (M.H.); (W.L.); (S.P.)
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, NSW 2006, Australia;
| | - Niridu Jude Peiris
- Heart Research Institute, Sydney, NSW 2042, Australia; (A.L.); (N.J.P.); (H.D.); (M.H.); (W.L.); (S.P.)
- Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia
| | - Harkirat Dhaliwal
- Heart Research Institute, Sydney, NSW 2042, Australia; (A.L.); (N.J.P.); (H.D.); (M.H.); (W.L.); (S.P.)
- Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia
| | - Maria Hakim
- Heart Research Institute, Sydney, NSW 2042, Australia; (A.L.); (N.J.P.); (H.D.); (M.H.); (W.L.); (S.P.)
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Weizhen Li
- Heart Research Institute, Sydney, NSW 2042, Australia; (A.L.); (N.J.P.); (H.D.); (M.H.); (W.L.); (S.P.)
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, NSW 2006, Australia;
| | - Subramaniam Ganesh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India;
- The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
| | - Yogambha Ramaswamy
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, NSW 2006, Australia;
| | - Sanjay Patel
- Heart Research Institute, Sydney, NSW 2042, Australia; (A.L.); (N.J.P.); (H.D.); (M.H.); (W.L.); (S.P.)
- Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia
- Cardiac Catheterization Laboratory, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia
| | - Ashish Misra
- Heart Research Institute, Sydney, NSW 2042, Australia; (A.L.); (N.J.P.); (H.D.); (M.H.); (W.L.); (S.P.)
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
- Correspondence: ; Tel.: +61-18-0065-1373
| |
Collapse
|
38
|
Huang C, Yang D, Ye GW, Powell CA, Guo P. Vascular Notch Signaling in Stress Hematopoiesis. Front Cell Dev Biol 2021; 8:606448. [PMID: 33585446 PMCID: PMC7873850 DOI: 10.3389/fcell.2020.606448] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 12/07/2020] [Indexed: 12/22/2022] Open
Abstract
Canonical Notch signaling is one of the most conserved signaling cascades. It regulates cell proliferation, cell differentiation, and cell fate maintenance in a variety of biological systems during development and cancer (Fortini, 2009; Kopan and Ilagan, 2009; Andersson et al., 2011; Ntziachristos et al., 2014). For the hematopoietic system, during embryonic development, Notch1 is essential for the emergence of hematopoietic stem cells (HSCs) at the aorta-gornado-mesonephro regions of the dorsal aorta. At adult stage, Notch receptors and Notch targets are expressed at different levels in diverse hematopoietic cell types and influence lineage choices. For example, Notch specifies T cell lineage over B cells. However, there has been a long-lasting debate on whether Notch signaling is required for the maintenance of adult HSCs, utilizing transgenic animals inactivating different components of the Notch signaling pathway in HSCs or niche cells. The aims of the current mini-review are to summarize the evidence that disapproves or supports such hypothesis and point at imperative questions waiting to be addressed; hence, some of the seemingly contradictory findings could be reconciled. We need to better delineate the Notch signaling events using biochemical assays to identify direct Notch targets within HSCs or niche cells in specific biological context. More importantly, we call for more elaborate studies that pertain to whether niche cell type (vascular endothelial cells or other stromal cell)-specific Notch ligands regulate the differentiation of T cells in solid tumors during the progression of T-lymphoblastic lymphoma (T-ALL) or chronic myelomonocytic leukemia (CMML). We believe that the investigation of vascular endothelial cells' or other stromal cell types' interaction with hematopoietic cells during homeostasis and stress can offer insights toward specific and effective Notch-related therapeutics.
Collapse
Affiliation(s)
- Can Huang
- McCann Health Medical Communications, New York, NY, United States
| | - Dawei Yang
- Zhongshan Hospital Fudan University, Zhongshan Hospital Institute for Clinical Science, Shanghai Medical College, Fudan University; Shanghai Engineering Research Center of AI Technology for Cardiopulmonary Disease, Shanghai, China.,Division of Pulmonary, Critical Care, and Sleep Medicine, Fibrosis Research Center, Icahn School of Medicine at Mount Sinai, Mount Sinai-National Jewish Respiratory Institute, New York, NY, United States
| | - George W Ye
- Division of Pulmonary, Critical Care, and Sleep Medicine, Fibrosis Research Center, Icahn School of Medicine at Mount Sinai, Mount Sinai-National Jewish Respiratory Institute, New York, NY, United States
| | - Charles A Powell
- Division of Pulmonary, Critical Care, and Sleep Medicine, Fibrosis Research Center, Icahn School of Medicine at Mount Sinai, Mount Sinai-National Jewish Respiratory Institute, New York, NY, United States
| | - Peipei Guo
- Division of Pulmonary, Critical Care, and Sleep Medicine, Fibrosis Research Center, Icahn School of Medicine at Mount Sinai, Mount Sinai-National Jewish Respiratory Institute, New York, NY, United States
| |
Collapse
|
39
|
MicroRNA-34a: the bad guy in age-related vascular diseases. Cell Mol Life Sci 2021; 78:7355-7378. [PMID: 34698884 PMCID: PMC8629897 DOI: 10.1007/s00018-021-03979-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/08/2021] [Accepted: 10/12/2021] [Indexed: 12/12/2022]
Abstract
The age-related vasculature alteration is the prominent risk factor for vascular diseases (VD), namely, atherosclerosis, abdominal aortic aneurysm, vascular calcification (VC) and pulmonary arterial hypertension (PAH). The chronic sterile low-grade inflammation state, alias inflammaging, characterizes elderly people and participates in VD development. MicroRNA34-a (miR-34a) is emerging as an important mediator of inflammaging and VD. miR-34a increases with aging in vessels and induces senescence and the acquisition of the senescence-associated secretory phenotype (SASP) in vascular smooth muscle (VSMCs) and endothelial (ECs) cells. Similarly, other VD risk factors, including dyslipidemia, hyperglycemia and hypertension, modify miR-34a expression to promote vascular senescence and inflammation. miR-34a upregulation causes endothelial dysfunction by affecting ECs nitric oxide bioavailability, adhesion molecules expression and inflammatory cells recruitment. miR-34a-induced senescence facilitates VSMCs osteoblastic switch and VC development in hyperphosphatemia conditions. Conversely, atherogenic and hypoxic stimuli downregulate miR-34a levels and promote VSMCs proliferation and migration during atherosclerosis and PAH. MiR34a genetic ablation or miR-34a inhibition by anti-miR-34a molecules in different experimental models of VD reduce vascular inflammation, senescence and apoptosis through sirtuin 1 Notch1, and B-cell lymphoma 2 modulation. Notably, pleiotropic drugs, like statins, liraglutide and metformin, affect miR-34a expression. Finally, human studies report that miR-34a levels associate to atherosclerosis and diabetes and correlate with inflammatory factors during aging. Herein, we comprehensively review the current knowledge about miR-34a-dependent molecular and cellular mechanisms activated by VD risk factors and highlight the diagnostic and therapeutic potential of modulating its expression in order to reduce inflammaging and VD burn and extend healthy lifespan.
Collapse
|
40
|
Zheng PF, Yin RX, Guan YZ, Wei BL, Liu CX, Deng GX. Association between SLC44A4-NOTCH4 SNPs and serum lipid levels in the Chinese Han and Maonan ethnic groups. Nutr Metab (Lond) 2020; 17:105. [PMID: 33317561 PMCID: PMC7737288 DOI: 10.1186/s12986-020-00533-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 12/03/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The current research was to assess the relationship of the solute carrier family 44 member 4 (SLC44A4) rs577272, notch receptor 4 (NOTCH4) rs3134931 SNPs and serum lipid levels in the Han and Maonan ethnic groups. METHODS The genetic makeup of the SLC44A4 rs577272 and NOTCH4 rs3134931 SNPs in 2467 unrelated subjects (Han, 1254; Maonan,1213) was obtained by using polymerase chain reaction and restriction fragment length polymorphism technique, combined with gel electrophoresis, and confirmed by direct sequencing. RESULTS The genotype frequencies of SLC44A4 rs577272 and NOTCH4 rs3134931 SNPs were different between Han and Maonan populations (P < 0.05); respectively. The SLC44A4 rs577272 SNP was associated with total cholesterol (TC) and high-density lipoprotein cholesterol (HDL-C) levels in Maonan group. The NOTCH4 rs3134931 SNP was associated with triglyceride (TG) in Han; and TG and low-density lipoprotein cholesterol (LDL-C) levels in Maonan groups (P < 0.025-0.001). Stratified analysis according to gender showed that the SLC44A4 rs577272 SNP was associated with TC and HDL-C in Han and Maonan females; TC in Maonan males, meanwhile, the NOTCH4 rs3134931 SNP was associated with TG and HDL-C in Han males; TG in Han females; TG and LDL-C in Maonan males; and TG, HDL-C and LDL-C in Maonan females. Linkage disequilibrium analysis showed that the most common haplotype was rs577272G-rs3134931A (> 50%) in both Han and Maonan groups. The haplotype of rs577272G-rs3134931A was associated with TG and HDL-C in Han; and TC, TG and HDL-C in Maonan ethnic groups. CONCLUSIONS These results suggest that the relationship among SLC44A4 rs577272, NOTCH4 rs3134931 SNPs and serum lipid parameters may vary depending on the gender and/or ethnicity/race in some populations. Haplotypes could explain more changes in serum lipid parameters than any single SNP alone particularly for TC, TG and HDL-C.
Collapse
Affiliation(s)
- Peng-Fei Zheng
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, Guangxi, People's Republic of China
| | - Rui-Xing Yin
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, Guangxi, People's Republic of China. .,Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Disease Control and Prevention, Nanning, 530021, Guangxi, People's Republic of China. .,Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning, 530021, Guangxi, People's Republic of China.
| | - Yao-Zong Guan
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, Guangxi, People's Republic of China
| | - Bi-Liu Wei
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, Guangxi, People's Republic of China
| | - Chun-Xiao Liu
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, Guangxi, People's Republic of China
| | - Guo-Xiong Deng
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, Guangxi, People's Republic of China
| |
Collapse
|
41
|
Guo Q, Guo Q, Xiao Y, Li C, Huang Y, Luo X. Regulation of bone marrow mesenchymal stem cell fate by long non-coding RNA. Bone 2020; 141:115617. [PMID: 32853852 DOI: 10.1016/j.bone.2020.115617] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 08/19/2020] [Accepted: 08/21/2020] [Indexed: 12/20/2022]
Abstract
Bone mesenchymal stem cells (BMSCs) are progenitor cells isolated from bone marrow, which keep potential to differentiate into several kinds of cells including osteoblasts and adipocytes. A dynamic mutual regulation exists between osteogenesis and adipogenesis processes. Long non-coding RNA (lncRNA) performs diverse functions in biological activities including regulation of BMSCs commitment. Evidence has shown that lncRNA regulates key signaling pathways including TGFβ/BMP, Wnt and Notch pathways, and several transcription factors in BMSCs differention. Dysregulation of lncRNA in BMSCs leads to disruption of osteo-adipogenesis difffrentiation and results in impairment of bone homeostasis. In this review, we focus on the role of lncRNA in several critical signaling pathways that involved in regulation of osteo-adipogenesis of BMSC and prospects the potential clinical application of lncRNA.
Collapse
Affiliation(s)
- Qiaoyue Guo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 87# Xiangya Road, Changsha, Hunan, PR China
| | - Qi Guo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 87# Xiangya Road, Changsha, Hunan, PR China
| | - Ye Xiao
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 87# Xiangya Road, Changsha, Hunan, PR China
| | - Changjun Li
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 87# Xiangya Road, Changsha, Hunan, PR China
| | - Yan Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 87# Xiangya Road, Changsha, Hunan, PR China
| | - Xianghang Luo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 87# Xiangya Road, Changsha, Hunan, PR China.
| |
Collapse
|
42
|
Zohorsky K, Mequanint K. Designing Biomaterials to Modulate Notch Signaling in Tissue Engineering and Regenerative Medicine. TISSUE ENGINEERING PART B-REVIEWS 2020; 27:383-410. [PMID: 33040694 DOI: 10.1089/ten.teb.2020.0182] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The design of cell-instructive biomaterials for tissue engineering and regenerative medicine is at a crossroads. Although the conventional tissue engineering approach is top-down (cells seeded to macroporous scaffolds and mature to form tissues), bottom-up tissue engineering strategies are becoming appealing. With such developments, we can study cell signaling events, thus enabling functional tissue assembly in physiologic and diseased models. Among many important signaling pathways, the Notch signaling pathway is the most diverse in its influence during tissue morphogenesis and repair following injury. Although Notch signaling is extensively studied in developmental biology and cancer biology, our knowledge of designing biomaterial-based Notch signaling platforms and incorporating Notch signaling components into engineered tissue systems is limited. By incorporating Notch signaling to tissue engineering scaffolds, we can direct cell-specific responses and improve engineered tissue maturation. This review will discuss recent progress in the development of Notch signaling biomaterials as a promising target to control cellular fate decisions, including the influences of ligand identity, biophysical material cues, ligand presentation strategies, and mechanotransduction. Notch signaling is consequently of interest to direct, control, and reprogram cellular behavior on a biomaterial surface. We anticipate that discussions in this article will allow for enhanced knowledge and insight into designing Notch targeted biomaterials for various tissue engineering and cell fate determinations. Impact statement Notch signaling is recognized as an important pathway in tissue engineering and regenerative medicine; however, there is no systematic review on this topic. The comprehensive review and perspectives presented here provide an in-depth discussion on ligand presentation strategies both in 2D and in 3D cell culture environments involving biomaterials/scaffolds. In addition, this review article provides insight into the challenges in designing cell surrogate biomaterials capable of providing Notch signals. To the best of the authors' knowledge, this is the first review relevant to the fields of tissue engineering.
Collapse
Affiliation(s)
- Kathleen Zohorsky
- School of Biomedical Engineering and The University of Western Ontario, London, Canada
| | - Kibret Mequanint
- School of Biomedical Engineering and The University of Western Ontario, London, Canada.,Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, Canada
| |
Collapse
|
43
|
Synergy between 15-lipoxygenase and secreted PLA 2 promotes inflammation by formation of TLR4 agonists from extracellular vesicles. Proc Natl Acad Sci U S A 2020; 117:25679-25689. [PMID: 32973091 DOI: 10.1073/pnas.2005111117] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Damage-associated endogenous molecules induce innate immune response, thus making sterile inflammation medically relevant. Stress-derived extracellular vesicles (stressEVs) released during oxidative stress conditions were previously found to activate Toll-like receptor 4 (TLR4), resulting in expression of a different pattern of immune response proteins in comparison to lipopolysaccharide (LPS), underlying the differences between pathogen-induced and sterile inflammation. Here we report that synergistic activities of 15-lipoxygenase (15-LO) and secreted phospholipase A2 (sPLA2) are needed for the formation of TLR4 agonists, which were identified as lysophospholipids (lysoPLs) with oxidized unsaturated acyl chain. Hydroxy, hydroperoxy, and keto products of 2-arachidonoyl-lysoPI oxidation by 15-LO were identified by mass spectrometry (MS), and they activated the same gene pattern as stressEVs. Extracellular PLA2 activity was detected in the synovial fluid from rheumatoid arthritis and gout patients. Furthermore, injection of sPLA2 promoted K/BxN serum-induced arthritis in mice, whereby ankle swelling was partially TLR4 dependent. Results confirm the role of oxidized lysoPL of stressEVs in sterile inflammation that promotes chronic diseases. Both 15-LO and sPLA2 enzymes are induced during inflammation, which opens the opportunity for therapy without compromising innate immunity against pathogens.
Collapse
|
44
|
Hasan SS, Jabs M, Taylor J, Wiedmann L, Leibing T, Nordström V, Federico G, Roma LP, Carlein C, Wolff G, Ekim-Üstünel B, Brune M, Moll I, Tetzlaff F, Gröne HJ, Fleming T, Géraud C, Herzig S, Nawroth PP, Fischer A. Endothelial Notch signaling controls insulin transport in muscle. EMBO Mol Med 2020; 12:e09271. [PMID: 32187826 PMCID: PMC7136962 DOI: 10.15252/emmm.201809271] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 02/21/2020] [Accepted: 02/25/2020] [Indexed: 12/26/2022] Open
Abstract
The role of the endothelium is not just limited to acting as an inert barrier for facilitating blood transport. Endothelial cells (ECs), through expression of a repertoire of angiocrine molecules, regulate metabolic demands in an organ‐specific manner. Insulin flux across the endothelium to muscle cells is a rate‐limiting process influencing insulin‐mediated lowering of blood glucose. Here, we demonstrate that Notch signaling in ECs regulates insulin transport to muscle. Notch signaling activity was higher in ECs isolated from obese mice compared to non‐obese. Sustained Notch signaling in ECs lowered insulin sensitivity and increased blood glucose levels. On the contrary, EC‐specific inhibition of Notch signaling increased insulin sensitivity and improved glucose tolerance and glucose uptake in muscle in a high‐fat diet‐induced insulin resistance model. This was associated with increased transcription of Cav1, Cav2, and Cavin1, higher number of caveolae in ECs, and insulin uptake rates, as well as increased microvessel density. These data imply that Notch signaling in the endothelium actively controls insulin sensitivity and glucose homeostasis and may therefore represent a therapeutic target for diabetes.
Collapse
Affiliation(s)
- Sana S Hasan
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Markus Jabs
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jacqueline Taylor
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Lena Wiedmann
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Thomas Leibing
- Department of Dermatology, Venereology, and Allergology, University Medical Center and Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Section of Clinical and Molecular Dermatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Viola Nordström
- Division of Cellular and Molecular Pathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Giuseppina Federico
- Division of Cellular and Molecular Pathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Leticia P Roma
- Biophysics Department, Center for Human and Molecular Biology (ZHMB), Saarland University, Homburg, Germany
| | - Christopher Carlein
- Biophysics Department, Center for Human and Molecular Biology (ZHMB), Saarland University, Homburg, Germany
| | - Gretchen Wolff
- Institute for Diabetes and Cancer (IDC) and Joint Heidelberg-IDC Translational Diabetes Program, Helmholtz Center Munich, Neuherberg, Germany
| | - Bilgen Ekim-Üstünel
- Institute for Diabetes and Cancer (IDC) and Joint Heidelberg-IDC Translational Diabetes Program, Helmholtz Center Munich, Neuherberg, Germany
| | - Maik Brune
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, Heidelberg, Germany
| | - Iris Moll
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Fabian Tetzlaff
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hermann-Josef Gröne
- Division of Cellular and Molecular Pathology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Institute of Pharmacology, Philipps University of Marburg, Marburg, Germany
| | - Thomas Fleming
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, Heidelberg, Germany
| | - Cyrill Géraud
- Department of Dermatology, Venereology, and Allergology, University Medical Center and Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Section of Clinical and Molecular Dermatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stephan Herzig
- Institute for Diabetes and Cancer (IDC) and Joint Heidelberg-IDC Translational Diabetes Program, Helmholtz Center Munich, Neuherberg, Germany.,Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, Heidelberg, Germany
| | - Peter P Nawroth
- Institute for Diabetes and Cancer (IDC) and Joint Heidelberg-IDC Translational Diabetes Program, Helmholtz Center Munich, Neuherberg, Germany.,Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, Heidelberg, Germany
| | - Andreas Fischer
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, Heidelberg, Germany.,European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
45
|
Atherosclerosis: Insights into Vascular Pathobiology and Outlook to Novel Treatments. J Cardiovasc Transl Res 2020; 13:744-757. [PMID: 32072564 DOI: 10.1007/s12265-020-09961-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 01/22/2020] [Indexed: 12/14/2022]
Abstract
The pathobiology of atherosclerosis and its current and potential future treatments are summarized, with a spotlight on three central cell types involved: (i) endothelial cells (ECs), (ii) macrophages, and (iii) vascular smooth muscle cells (VSMCs). (i) EC behaviour is regulated by the central transcription factors YAP/TAZ in reaction to biomechanical forces, such as hemodynamic shear stress. (ii) VSMC transdifferentiation (phenotype switching) to a macrophage-like phenotype contributes to the majority of cells positive for common cell surface macrophage markers in atherosclerotic plaques. (iii) Intra-plaque macrophages originate in a significant number from vascular resident macrophages. They can be activated via pattern recognition receptors on cell membrane (e.g. toll-like receptors) and inside cells (e.g. inflammasomes), requiring priming by neutrophil extracellular traps (NETs). ECs and macrophages can also be characterized by single-cell RNA sequencing. Adaptive immunity plays an important role in the inflammatory process. Future therapeutic options include vaccination, TRAF-STOPs, senolysis, or CD47 blockade. Graphical Abstract.
Collapse
|
46
|
Malashicheva A, Kostina A, Kostareva A, Irtyuga O, Gordeev M, Uspensky V. Notch signaling in the pathogenesis of thoracic aortic aneurysms: A bridge between embryonic and adult states. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165631. [PMID: 31816439 DOI: 10.1016/j.bbadis.2019.165631] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 11/23/2019] [Accepted: 12/02/2019] [Indexed: 12/17/2022]
Abstract
Aneurysms of the thoracic aorta are a "silent killer" with no evident clinical signs until the fatal outcome. Molecular and genetic bases of thoracic aortic aneurysms mainly include transforming growth factor beta signaling, smooth muscle contractile units and metabolism genes, and extracellular matrix genes. In recent studies, a role of Notch signaling, among other pathways, has emerged in disease pathogenesis. Notch is a highly conserved signaling pathway that regulates the development and differentiation of many types of tissues and influences major cellular processes such as cell proliferation, differentiation and apoptosis. Mutations in several Notch signaling components have been associated with a number of heart defects, demonstrating an essential role of Notch signaling both in cardiovascular system development and its maintenance during postnatal life. This review discusses the role of Notch signaling in the pathogenesis of thoracic aortic aneurysms considering development and maintenance of the aortic root and how developmental regulations by Notch signaling may influence thoracic aortic aneurysms.
Collapse
Affiliation(s)
- Anna Malashicheva
- Almazov National Medical Research Centre, Akkuratova, 2, 197341 Saint Petersburg, Russia; Institute of Cytology, Russian Academy of Sciences, Tikhoretskiy, 4, 194064 Saint Petersburg, Russia; Saint Petersburg State University, Department of Embryology, Universitetskaya nab., 7/9, 199034, Saint Petersburg, Russia.
| | - Aleksandra Kostina
- Almazov National Medical Research Centre, Akkuratova, 2, 197341 Saint Petersburg, Russia; Institute of Cytology, Russian Academy of Sciences, Tikhoretskiy, 4, 194064 Saint Petersburg, Russia
| | - Anna Kostareva
- Almazov National Medical Research Centre, Akkuratova, 2, 197341 Saint Petersburg, Russia
| | - Olga Irtyuga
- Almazov National Medical Research Centre, Akkuratova, 2, 197341 Saint Petersburg, Russia
| | - Mikhail Gordeev
- Almazov National Medical Research Centre, Akkuratova, 2, 197341 Saint Petersburg, Russia
| | - Vladimir Uspensky
- Almazov National Medical Research Centre, Akkuratova, 2, 197341 Saint Petersburg, Russia
| |
Collapse
|
47
|
KRIT1 Deficiency Promotes Aortic Endothelial Dysfunction. Int J Mol Sci 2019; 20:ijms20194930. [PMID: 31590384 PMCID: PMC6801783 DOI: 10.3390/ijms20194930] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/20/2019] [Accepted: 09/30/2019] [Indexed: 01/07/2023] Open
Abstract
Loss-of-function mutations of the gene encoding Krev interaction trapped protein 1 (KRIT1) are associated with the pathogenesis of Cerebral Cavernous Malformation (CCM), a major cerebrovascular disease characterized by abnormally enlarged and leaky capillaries and affecting 0.5% of the human population. However, growing evidence demonstrates that KRIT1 is implicated in the modulation of major redox-sensitive signaling pathways and mechanisms involved in adaptive responses to oxidative stress and inflammation, suggesting that its loss-of-function mutations may have pathological effects not limited to CCM disease. The aim of this study was to address whether KRIT1 loss-of-function predisposes to the development of pathological conditions associated with enhanced endothelial cell susceptibility to oxidative stress and inflammation, such as arterial endothelial dysfunction (ED) and atherosclerosis. Silencing of KRIT1 in human aortic endothelial cells (HAECs), coronary artery endothelial cells (HCAECs), and umbilical vein endothelial cells (HUVECs) resulted in increased expression of endothelial proinflammatory adhesion molecules vascular cell adhesion molecule 1 (VCAM-1) and intercellular adhesion molecule 1 (ICAM-1) and in enhanced susceptibility to tumor necrosis factor alpha (TNF-α)-induced apoptosis. These effects were associated with a downregulation of Notch1 activation that could be rescued by antioxidant treatment, suggesting that they are consequent to altered intracellular redox homeostasis induced by KRIT1 loss-of-function. Furthermore, analysis of the aorta of heterozygous KRIT1+/- mice fed a high-fructose diet to induce systemic oxidative stress and inflammation demonstrated a 1.6-fold increased expression of VCAM-1 and an approximately 2-fold enhanced fat accumulation (7.5% vs 3.6%) in atherosclerosis-prone regions, including the aortic arch and aortic root, as compared to corresponding wild-type littermates. In conclusion, we found that KRIT1 deficiency promotes ED, suggesting that, besides CCM, KRIT1 may be implicated in genetic susceptibility to the development of atherosclerotic lesions.
Collapse
|
48
|
Aquila G, Kostina A, Vieceli Dalla Sega F, Shlyakhto E, Kostareva A, Marracino L, Ferrari R, Rizzo P, Malaschicheva A. The Notch pathway: a novel therapeutic target for cardiovascular diseases? Expert Opin Ther Targets 2019; 23:695-710. [PMID: 31304807 DOI: 10.1080/14728222.2019.1641198] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: The Notch pathway is involved in determining cell fate during development and postnatally in continuously renewing tissues, such as the endothelium, the epithelium, and in the stem cells pool. The dysregulation of the Notch pathway is one of the causes of limited response, or resistance, to available cancer treatments and novel therapeutic strategies based on Notch inhibition are being investigated in preclinical and clinical studies in oncology. A large body of evidence now shows that the dysregulation of the Notch pathway is also involved in the pathophysiology of cardiovascular diseases (CVDs). Areas covered: This review discusses the molecular mechanisms involving Notch which underlie heart failure, aortic valve calcification, and aortic aneurysm. Expert opinion: Despite the existence of preventive, pharmacological and surgical interventions approaches, CVDs are the first causes of mortality worldwide. The Notch pathway is becoming increasingly recognized as being involved in heart failure, aortic aneurysm and aortic valve calcification, which are among the most common global causes of mortality due to CVDs. As already shown in cancer, the dissection of the biological processes and molecular mechanisms involving Notch should pave the way for new strategies to prevent and cure these diseases.
Collapse
Affiliation(s)
- Giorgio Aquila
- Department of Medical Sciences, University of Ferrara , Ferrara , Italy
| | - Aleksandra Kostina
- Laboratory of Molecular Cardiology, Almazov National Medical Research Centre , St-Petersburg , Russia.,Laboratory of Regenerative Biomedicine, Institute of Cytology, Russian Academy of Sciences , St-Petersburg , Russia
| | | | - Eugeniy Shlyakhto
- Laboratory of Molecular Cardiology, Almazov National Medical Research Centre , St-Petersburg , Russia
| | - Anna Kostareva
- Laboratory of Molecular Cardiology, Almazov National Medical Research Centre , St-Petersburg , Russia
| | - Luisa Marracino
- Department of Morphology, Surgery and Experimental Medicine and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara , Ferrara , Italy
| | - Roberto Ferrari
- Department of Medical Sciences, University of Ferrara , Ferrara , Italy.,Maria Cecilia Hospital, GVM Care & Research , Cotignola , Italy
| | - Paola Rizzo
- Maria Cecilia Hospital, GVM Care & Research , Cotignola , Italy.,Department of Morphology, Surgery and Experimental Medicine and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara , Ferrara , Italy
| | - Anna Malaschicheva
- Laboratory of Molecular Cardiology, Almazov National Medical Research Centre , St-Petersburg , Russia.,Laboratory of Regenerative Biomedicine, Institute of Cytology, Russian Academy of Sciences , St-Petersburg , Russia.,Department of Embryology, Faculty of Biology, Saint-Petersburg State University , St. Petersburg , Russia
| |
Collapse
|
49
|
Vieceli Dalla Sega F, Fortini F, Aquila G, Campo G, Vaccarezza M, Rizzo P. Notch Signaling Regulates Immune Responses in Atherosclerosis. Front Immunol 2019; 10:1130. [PMID: 31191522 PMCID: PMC6540611 DOI: 10.3389/fimmu.2019.01130] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 05/03/2019] [Indexed: 01/05/2023] Open
Abstract
Atherosclerosis is a chronic autoimmune inflammatory disease that can cause coronary artery disease, stroke, peripheral artery disease, depending on which arteries are affected. At the beginning of atherosclerosis plasma lipoproteins accumulate in the sub-endothelial space. In response, monocytes migrate from the circulation through the endothelium into the intima where they differentiate into macrophages. These early events trigger a complex immune response that eventually involves many cellular subtypes of both innate and adaptive immunity. The Notch signaling pathway is an evolutionary conserved cell signaling system that mediates cell-to-cell communication. Recent studies have revealed that Notch modulate atherosclerosis by controlling macrophages polarization into M1 or M2 subtypes. Furthermore, it is known that Notch signaling controls differentiation and activity of T-helper and cytotoxic T-cells in inflammatory diseases. In this review, we will discuss the role of Notch in modulating immunity in the context of atherosclerosis and whether targeting Notch may represent a therapeutic strategy.
Collapse
Affiliation(s)
| | - Francesca Fortini
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, Cotignola, Italy
| | - Giorgio Aquila
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Gianluca Campo
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, Cotignola, Italy.,Cardiovascular Center, Azienda Ospedaliero-Universitaria di Ferrara, Cona, Italy
| | - Mauro Vaccarezza
- Faculty of Health Sciences, School of Pharmacy and Biomedical Sciences, Curtin University, Perth, WA, Australia
| | - Paola Rizzo
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, Cotignola, Italy.,Department of Morphology, Surgery, and Experimental Medicine, University of Ferrara, Ferrara, Italy.,Laboratory for Technologies of Advanced Therapies, University of Ferrara, Ferrara, Italy
| |
Collapse
|
50
|
The Use of Nutraceuticals to Counteract Atherosclerosis: The Role of the Notch Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:5470470. [PMID: 31915510 PMCID: PMC6935452 DOI: 10.1155/2019/5470470] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 03/13/2019] [Indexed: 12/13/2022]
Abstract
Despite the currently available pharmacotherapies, today, thirty percent of worldwide deaths are due to cardiovascular diseases (CVDs), whose primary cause is atherosclerosis, an inflammatory disorder characterized by the buildup of lipid deposits on the inside of arteries. Multiple cellular signaling pathways have been shown to be involved in the processes underlying atherosclerosis, and evidence has been accumulating for the crucial role of Notch receptors in regulating the functions of the diverse cell types involved in atherosclerosis onset and progression. Several classes of nutraceuticals have potential benefits for the prevention and treatment of atherosclerosis and CVDs, some of which could in part be due to their ability to modulate the Notch pathway. In this review, we summarize the current state of knowledge on the role of Notch in vascular health and its modulation by nutraceuticals for the prevention of atherosclerosis and/or treatment of related CVDs.
Collapse
|