1
|
Wu R, Su Y, Liao J, Shen J, Ma Y, Gao W, Dong Z, Dai Y, Yao K, Ge J. Exome Sequencing Identified Susceptible Genes for High Residual Risks in Early-Onset Coronary Atherosclerotic Disease. Clin Cardiol 2024; 47:e70066. [PMID: 39673281 PMCID: PMC11645474 DOI: 10.1002/clc.70066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/16/2024] [Accepted: 11/27/2024] [Indexed: 12/16/2024] Open
Abstract
AIMS Despite the tremendous improvement in therapeutic medication and intervention for coronary atherosclerotic disease (CAD), residual risks remain. Exome sequencing enables identification of rare variants and susceptibility genes for residual risks of early-onset coronary atherosclerotic disease (EOCAD) with well-controlled conventional risk factors. METHODS We performed whole-exome sequencing of subjects who had no conventional risk factors, defined as higher body mass index, smoking, hypertension and dyslipidemia, screened from 1950 patients with EOCAD (age ≤ 45 years, at least 50% stenosis of coronary artery by angiography), and selected control subjects from 1006 elder (age ≥ 65 years) with < 30% coronary stenosis. Gene-based association analysis and clinical phenotypic comparison were conducted. RESULTS Subjects without defined conventional risk factors accounted for 4.72% of young patients. Totally, 6 genes might be associated with residual risk of EOCAD, namely CABP1 (OR = 22.19, p = 0.02), HLA-E (OR = 22.19, p = 0.02), TOE1 (OR = 33.6, p = 0.002), HPSE2 (OR = 11.1, p = 0.04), CHST14 (OR = 22.19, p = 0.02) as well as KLHL8 (OR = 22.19, p = 0.02). Phenotypic analysis displayed the levels of low-density lipoprotein cholesterol in carriers of mutations from CABP1, HLA-E, TOE1, and HPSE2 were significantly elevated compared to noncarriers. Notably, extracellular matrix-associated CHST14 and fibrinogen-associated KLHL8 both displayed possible correlation with increased neutrophil proportion and decreased monocyte percentage (both p < 0.05), exerting potential effects on the residual inflammatory risks of EOCAD. CONCLUSION The study identified six genes related to dyslipidemia and inflammation pathways with potential association with residual risk of EOCAD, which will contribute to precision-based prevention in these patients. TRIAL REGISTRATION The GRAND study was registered at www. CLINICALTRIALS gov on July 14, 2015, and the registry number is NCT02496858.
Collapse
Affiliation(s)
- Runda Wu
- Department of Cardiology, Zhongshan HospitalFudan UniversityShanghaiP.R. China
- Shanghai Institute of Cardiovascular DiseaseShanghaiP.R. China
| | - Ya Su
- Department of CardiologyZhongshan Hospital, Qingpu BranchShanghaiP.R. China
| | - Jianquan Liao
- Department of Cardiology, Zhongshan HospitalFudan UniversityShanghaiP.R. China
- Shanghai Institute of Cardiovascular DiseaseShanghaiP.R. China
| | - Juan Shen
- Institute of Metagenomics, Qingdao‐Europe Advanced Institute for Life Sciences, BGI ResearchQingdaoP.R. China
| | - Yuanji Ma
- Department of Cardiology, Zhongshan HospitalFudan UniversityShanghaiP.R. China
- Shanghai Institute of Cardiovascular DiseaseShanghaiP.R. China
| | - Wei Gao
- Department of Cardiology, Zhongshan HospitalFudan UniversityShanghaiP.R. China
- Shanghai Institute of Cardiovascular DiseaseShanghaiP.R. China
| | - Zheng Dong
- Department of CardiologyNanjing Drum Tower HospitalNanjingP.R. China
| | - Yuxiang Dai
- Department of Cardiology, Zhongshan HospitalFudan UniversityShanghaiP.R. China
- Shanghai Institute of Cardiovascular DiseaseShanghaiP.R. China
| | - Kang Yao
- Department of Cardiology, Zhongshan HospitalFudan UniversityShanghaiP.R. China
- Shanghai Institute of Cardiovascular DiseaseShanghaiP.R. China
| | - Junbo Ge
- Department of Cardiology, Zhongshan HospitalFudan UniversityShanghaiP.R. China
- Shanghai Institute of Cardiovascular DiseaseShanghaiP.R. China
- NHC Key Laboratory of Viral Heart Diseases (Fudan University)ShanghaiP.R. China
- Key Laboratory of Viral Heart DiseasesChinese Academy of Medical SciencesBeijingP.R. China
- National Clinical Research Center for Interventional MedicineShanghaiP.R. China
| |
Collapse
|
2
|
Fry H, Mazidi M, Kartsonaki C, Clarke R, Walters RG, Chen Z, Millwood IY. The Role of Furin and Its Therapeutic Potential in Cardiovascular Disease Risk. Int J Mol Sci 2024; 25:9237. [PMID: 39273186 PMCID: PMC11394739 DOI: 10.3390/ijms25179237] [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: 07/12/2024] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 09/15/2024] Open
Abstract
Furin is an important proteolytic enzyme, converting several proteins from inactive precursors to their active forms. Recently, proteo-genomic analyses in European and East Asian populations suggested a causal association of furin with ischaemic heart disease, and there is growing interest in its role in cardiovascular disease (CVD) aetiology. In this narrative review, we present a critical appraisal of evidence from population studies to assess furin's role in CVD risk and potential as a drug target for CVD. Whilst most observational studies report positive associations between furin expression and CVD risk, some studies report opposing effects, which may reflect the complex biological roles of furin and its substrates. Genetic variation in FURIN is also associated with CVD and its risk factors. We found no evidence of current clinical development of furin as a drug target for CVD, although several phase 1 and 2 clinical trials of furin inhibitors as a type of cancer immunotherapy have been completed. The growing field of proteo-genomics in large-scale population studies may inform the future development of furin and other potential drug targets to improve the treatment and prevention of CVD.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Iona Y. Millwood
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK; (H.F.); (M.M.); (C.K.); (R.C.); (R.G.W.); (Z.C.)
| |
Collapse
|
3
|
Chua R, Wang L, Singaraja R, Ghosh S. Functional and Multi-Omics Effects of an Optimized CRISPR-Mediated FURIN Depletion in U937 Monocytes. Cells 2024; 13:588. [PMID: 38607027 PMCID: PMC11154428 DOI: 10.3390/cells13070588] [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: 12/11/2023] [Revised: 03/08/2024] [Accepted: 03/12/2024] [Indexed: 04/13/2024] Open
Abstract
The pro-protein convertase FURIN (PCSK3) is implicated in a wide range of normal and pathological biological processes such as infectious diseases, cancer and cardiovascular diseases. Previously, we performed a systemic inhibition of FURIN in a mouse model of atherosclerosis and demonstrated significant plaque reduction and alterations in macrophage function. To understand the cellular mechanisms affected by FURIN inhibition in myeloid cells, we optimized a CRISPR-mediated gene deletion protocol for successfully deriving hemizygous (HZ) and nullizygous (NZ) FURIN knockout clones in U937 monocytic cells using lipotransfection-based procedures and a dual guide RNA delivery strategy. We observed differences in monocyte and macrophage functions involving phagocytosis, lipid accumulation, cell migration, inflammatory gene expression, cytokine release patterns, secreted proteomics (cytokines) and whole-genome transcriptomics between wild-type, HZ and NZ FURIN clones. These studies provide a mechanistic basis on the possible roles of myeloid cell FURIN in cardiovascular disorders.
Collapse
Affiliation(s)
- Ruiming Chua
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore 169857, Singapore;
| | - Lijin Wang
- Centre for Computational Biology, Duke-NUS Medical School, Singapore 169857, Singapore;
| | - Roshni Singaraja
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore;
| | - Sujoy Ghosh
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore 169857, Singapore;
- Centre for Computational Biology, Duke-NUS Medical School, Singapore 169857, Singapore;
- Laboratory of Computational Biology, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| |
Collapse
|
4
|
Zeng R, Wang Y, Chen J, Liu Q. Furin knockdown inhibited EndMT and abnormal proliferation and migration of endothelial cells. Clin Hemorheol Microcirc 2024; 88:59-70. [PMID: 38820014 DOI: 10.3233/ch-242171] [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] [Indexed: 06/02/2024]
Abstract
BACKGROUND In the pathogenesis of atherosclerotic cardiovascular disorders, vascular endothelium is crucial. A critical step in the development of atherosclerosis is endothelial dysfunction. Furin may play a factor in vascular remodeling, inflammatory cell infiltration, regulation of plaque stability, and atherosclerosis by affecting the adhesion and migration of endothelial cells. It is yet unknown, though, how furin contributes to endothelial dysfunction. METHODS We stimulated endothelial cells with oxidized modified lipoprotein (ox-LDL). Endothelial-to-mesenchymal transition (EndMT) was found using immunofluorescence (IF) and western blot (WB). Furin expression level and Hippo/YAP signal activation were found using reverse transcription-quantitative PCR (RT-qPCR) and WB, respectively. To achieve the goal of furin knockdown, we transfected siRNA using the RNA transmate reagent. Following furin knockdown, cell proliferation, and migration were assessed by the CCK-8, scratch assay, and transwell gold assay, respectively. WB and IF both picked up on EndMT. WB and RT-qPCR, respectively, were used to find furin's expression level. We chose the important micrornas that can regulate furin and we then confirmed them using RT-qPCR. RESULTS EndMT was created by ox-LDL, evidenced by the up-regulation of mesenchymal cell markers and the down-regulation of endothelial cell markers. Furin expression levels in both protein and mRNA were increased, and the Hippo/YAP signaling pathway was turned on. Furin knockdown dramatically reduced the aberrant migration and proliferation of endothelial cells by ox-LDL stimulation. Furin knockdown can also suppress ox-LDL-induced EndMT, up-regulate indicators of endothelial cells, and down-regulate markers of mesenchymal cells. After ox-LDL stimulation and siRNA transfection, furin's expression level was up-regulated and down-regulated. CONCLUSION Our study demonstrated that furin knockdown could affect ox-LDL-induced abnormal endothelial cell proliferation, migration, and EndMT. This implies that furin plays an important role in endothelial dysfunction.
Collapse
Affiliation(s)
- Rui Zeng
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yimin Wang
- Rehabilitation Area of the Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jun Chen
- Rehabilitation Area of the Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Qiang Liu
- Rehabilitation Area of the Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
5
|
Kato S, Iwata O, Kato H, Fukaya S, Imai Y, Saitoh S. Furin Regulates the Alveolarization of Neonatal Lungs in a Mouse Model of Hyperoxic Lung Injury. Biomolecules 2023; 13:1656. [PMID: 38002338 PMCID: PMC10669361 DOI: 10.3390/biom13111656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Despite advances in treatment options, such as corticosteroid administration and less invasive respiratory support, bronchopulmonary dysplasia (BPD) remains an important prognostic factor in preterm infants. We previously reported that furin regulates changes in lung smooth muscle cell phenotypes, suggesting that it plays a critical role in BPD pathogenesis. Therefore, in this study, we aimed to evaluate whether it regulates the alveolarization of immature lungs through activating alveolarization-driving proteins. We first examined furin expression levels, and its functions, using an established hyperoxia-induced BPD mouse model. Thereafter, we treated mice pups, as well as primary myofibroblast cell cultures, with furin inhibitors. Finally, we administered the hyperoxia-exposed mice pups with recombinant furin. Immunofluorescence revealed the co-expression of furin with alpha-smooth muscle actin. Hyperoxia exposure for 10 d decreased alveolar formation, as well as the expression of furin and its target, IGF-1R. Hexa-D-arginine administration also significantly inhibited alveolar formation. Another furin inhibitor, decanoyl-RVKR-chloromethylketone, accumulated pro-IGF-1R, and decreased IGF-1R phosphorylation in myofibroblast primary cultures. Finally, recombinant furin treatment significantly improved alveolarization in hyperoxia-exposed mice pups. Furin regulates alveolarization in immature lungs. Therefore, this study provides novel insights regarding the involvement of furin in BPD pathogenesis, and highlights a potential treatment target for ameliorating the impact of BPD.
Collapse
Affiliation(s)
- Shin Kato
- Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya 467-8601, Japan; (O.I.); (S.F.); (S.S.)
| | - Osuke Iwata
- Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya 467-8601, Japan; (O.I.); (S.F.); (S.S.)
| | - Hiroyuki Kato
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan;
| | - Satoko Fukaya
- Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya 467-8601, Japan; (O.I.); (S.F.); (S.S.)
| | - Yukari Imai
- Department of Pharmacy, Kinjo Gakuin University, Nagoya 463-8521, Japan
| | - Shinji Saitoh
- Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya 467-8601, Japan; (O.I.); (S.F.); (S.S.)
| |
Collapse
|
6
|
Liu S, Li Y, Wei X, Adi D, Wang YT, Han M, Liu F, Chen BD, Li XM, Yang YN, Fu ZY, Ma YT. Genetic analysis of DNA methylation in dyslipidemia: a case-control study. PeerJ 2022; 10:e14590. [PMID: 36570009 PMCID: PMC9774006 DOI: 10.7717/peerj.14590] [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: 03/28/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
Background Coronary heart disease has become the leading cause of death in developed countries, and dyslipidemia is closely associated with the risk of cardiovascular disease. Dyslipidemia is caused by the abnormal regulation of several genes and signaling pathways, and dyslipidemia is influenced mainly by genetic variation. AMFR, FBXW7, INSIG1, INSIG2, and MBTPS1 genes are associated with lipid metabolism. In a recent GWAS study, the GRINA gene has been reported to be associated with dyslipidemia, but its molecular mechanism has not been thoroughly investigated. The correlation between the DNA methylation of these genes and lipid metabolism has not been studied. This study aimed to examine the relationship between the DNA methylation of these genes and the risk of dyslipidemia by comparing the methylation levels of dyslipidemia and control samples. Methods A case-control research method was used in this study. The patient's blood samples were collected at the Heart Center of the First Affiliated Hospital of Xinjiang Medical University. In the Xinjiang Han population, 100 cases of hyperlipidemia and 80 cases of the control group were selected. The two groups were age and gender-matched. Quantitative methylation analysis of CpG sites in the gene promoter regions of six genes was performed by Solexa high-throughput sequencing. Results The DNA methylation levels of 23 CpG sites in six genes were shown to be associated with hyperlipidemia, and a total of 20 DNA methylation haplotypes showed statistically significant differences between the two groups. When compared with the control group, the dyslipidemia group had significantly higher levels of methylation in the GRINA gene (2.68 vs 2.36, P = 0.04). Additionally, we also discovered a significant methylation haplotype of GRINA (P = 0.017). Conclusion The findings of this study reveal that the DNA methylation of GRINA increases the risk for dyslipidemia in humans.
Collapse
Affiliation(s)
- Shuai Liu
- First Affiliated Hospital of Xinjiang Medical University, Urumqi, China,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, China
| | - Yang Li
- First Affiliated Hospital of Xinjiang Medical University, Urumqi, China,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, China
| | - Xian Wei
- First Affiliated Hospital of Xinjiang Medical University, Urumqi, China,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, China
| | - Dilare Adi
- First Affiliated Hospital of Xinjiang Medical University, Urumqi, China,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, China
| | - Yong-Tao Wang
- First Affiliated Hospital of Xinjiang Medical University, Urumqi, China,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, China
| | - Min Han
- First Affiliated Hospital of Xinjiang Medical University, Urumqi, China,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, China
| | - Fen Liu
- First Affiliated Hospital of Xinjiang Medical University, Urumqi, China,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, China
| | - Bang-Dang Chen
- First Affiliated Hospital of Xinjiang Medical University, Urumqi, China,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, China
| | - Xiao-Mei Li
- First Affiliated Hospital of Xinjiang Medical University, Urumqi, China,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, China
| | - Yi-Ning Yang
- First Affiliated Hospital of Xinjiang Medical University, Urumqi, China,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, China
| | - Zhen-Yan Fu
- First Affiliated Hospital of Xinjiang Medical University, Urumqi, China,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, China
| | - Yi-Tong Ma
- First Affiliated Hospital of Xinjiang Medical University, Urumqi, China,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, China
| |
Collapse
|
7
|
Zhang Y, Gao X, Bai X, Yao S, Chang YZ, Gao G. The emerging role of furin in neurodegenerative and neuropsychiatric diseases. Transl Neurodegener 2022; 11:39. [PMID: 35996194 PMCID: PMC9395820 DOI: 10.1186/s40035-022-00313-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 08/10/2022] [Indexed: 12/02/2022] Open
Abstract
Furin is an important mammalian proprotein convertase that catalyzes the proteolytic maturation of a variety of prohormones and proproteins in the secretory pathway. In the brain, the substrates of furin include the proproteins of growth factors, receptors and enzymes. Emerging evidence, such as reduced FURIN mRNA expression in the brains of Alzheimer's disease patients or schizophrenia patients, has implicated a crucial role of furin in the pathophysiology of neurodegenerative and neuropsychiatric diseases. Currently, compared to cancer and infectious diseases, the aberrant expression of furin and its pharmaceutical potentials in neurological diseases remain poorly understood. In this article, we provide an overview on the physiological roles of furin and its substrates in the brain, summarize the deregulation of furin expression and its effects in neurodegenerative and neuropsychiatric disorders, and discuss the implications and current approaches that target furin for therapeutic interventions. This review may expedite future studies to clarify the molecular mechanisms of furin deregulation and involvement in the pathogenesis of neurodegenerative and neuropsychiatric diseases, and to develop new diagnosis and treatment strategies for these diseases.
Collapse
Affiliation(s)
- Yi Zhang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China
| | - Xiaoqin Gao
- Shijiazhuang People's Hospital, Hebei Medical University, Shijiazhuang, 050027, China
| | - Xue Bai
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China
| | - Shanshan Yao
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China
| | - Yan-Zhong Chang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China.
| | - Guofen Gao
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China.
| |
Collapse
|
8
|
Yamasaki G, Sakurada M, Kitagawa K, Kondo T, Takahashi M, Ueno Y. Effect of FURIN SNP rs17514846 on coronary atherosclerosis in human cardiac specimens: An autopsy study of 106 cases. Leg Med (Tokyo) 2021; 55:102006. [PMID: 35008003 DOI: 10.1016/j.legalmed.2021.102006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/08/2021] [Accepted: 12/23/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND Coronary artery disease (CAD), including coronary atherosclerosis (CAS), is one of the most common causes of death. The FURIN SNP rs17514846 is assumed to be a risk factor for CAD. We evaluated this relationship using autopsy specimens and autopsy data, such as the histopathological degree of CAS. MATERIALS AND METHODS A total of 106 samples were genotyped from obtained blood samples. Myocardial and coronary arterial FURIN levels were quantified by ELISA. The degree of CAS was classified histopathologically according to the Stary classification, and the localization of FURIN was examined by immunostaining. The obtained data were analyzed statistically. RESULTS FURIN expression was widely observed in the myocardium, vascular smooth muscle cells, endothelial cells, adipocytes, and macrophages. FURIN level in the myocardium of cases with the AA genotype at the FURIN SNP rs17514846 was higher than that in CC cases. Additionally, FURIN levels in both coronary arteries and myocardium were higher at the early stage of CAS than at the late stage microscopically. CONCLUSION Our study suggested that the A allele of rs17514846 is associated with higher FURIN level in the heart and that FURIN exhibits a higher level in the early stage of CAS. These findings deepen our understanding of the mechanism of CAS.
Collapse
Affiliation(s)
- Gentaro Yamasaki
- Division of Legal Medicine, Department of Community Medicine and Social Healthcare Science, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Makoto Sakurada
- Forensic Science Laboratory, Hyogo Prefectural Police Headquarters, Kobe, Japan
| | - Koichi Kitagawa
- Department of Advanced Medical Science, Kobe University Graduate School of Science, Technology and Innovation, Kobe, Japan
| | - Takeshi Kondo
- Division of Legal Medicine, Department of Community Medicine and Social Healthcare Science, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Motonori Takahashi
- Division of Legal Medicine, Department of Community Medicine and Social Healthcare Science, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yasuhiro Ueno
- Division of Legal Medicine, Department of Community Medicine and Social Healthcare Science, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
9
|
Abstract
The kexin-like proprotein convertases perform the initial proteolytic cleavages that ultimately generate a variety of different mature peptide and proteins, ranging from brain neuropeptides to endocrine peptide hormones, to structural proteins, among others. In this review, we present a general introduction to proprotein convertase structure and biochemistry, followed by a comprehensive discussion of each member of the kexin-like subfamily of proprotein convertases. We summarize current knowledge of human proprotein convertase insufficiency syndromes, including genome-wide analyses of convertase polymorphisms, and compare these to convertase null and mutant mouse models. These mouse models have illuminated our understanding of the roles specific convertases play in human disease and have led to the identification of convertase-specific substrates; for example, the identification of procorin as a specific PACE4 substrate in the heart. We also discuss the limitations of mouse null models in interpreting human disease, such as differential precursor cleavage due to species-specific sequence differences, and the challenges presented by functional redundancy among convertases in attempting to assign specific cleavages and/or physiological roles. However, in most cases, knockout mouse models have added substantively both to our knowledge of diseases caused by human proprotein convertase insufficiency and to our appreciation of their normal physiological roles, as clearly seen in the case of the furin, proprotein convertase 1/3, and proprotein convertase 5/6 mouse models. The creation of more sophisticated mouse models with tissue- or temporally-restricted expression of specific convertases will improve our understanding of human proprotein convertase insufficiency and potentially provide support for the emerging concept of therapeutic inhibition of convertases.
Collapse
Affiliation(s)
- Manita Shakya
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Iris Lindberg
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
10
|
Testa G, Staurenghi E, Giannelli S, Sottero B, Gargiulo S, Poli G, Gamba P, Leonarduzzi G. Up-regulation of PCSK6 by lipid oxidation products: A possible role in atherosclerosis. Biochimie 2021; 181:191-203. [PMID: 33359561 DOI: 10.1016/j.biochi.2020.12.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/11/2020] [Accepted: 12/17/2020] [Indexed: 12/26/2022]
Abstract
Atherosclerosis is a degenerative disease characterized by lesions that develop in the wall of large- and medium-sized arteries due to the accumulation of low-density lipoproteins (LDLs) in the intima. A growing bulk of evidence suggests that cholesterol oxidation products, known as oxysterols, and the aldehyde 4-hydroxy-2-nonenal (HNE), the major pro-atherogenic components of oxidized LDLs, significantly contribute to atherosclerotic plaque progression and destabilization, with eventual plaque rupture. The involvement of certain members of the protein convertase subtilisin/kexin proteases (PCSKs) in atherosclerosis has been recently hypothesized. Among them, PCSK6 has been associated with plaque instability, mainly thanks to its ability to stimulate the activity of matrix metalloproteinases (MMPs) involved in extracellular matrix remodeling and to enhance inflammation. In U937 promonocytic cells and in human umbilical vein endothelial cells, an oxysterol mixture and HNE were able to up-regulate the level and activity of PCSK6, resulting in MMP-9 activation as demonstrated by PCSK6 silencing. Inflammation, enhanced by these lipid oxidation products, plays a key role in the up-regulation of PCSK6 activity as demonstrated by cell pretreatment with NS-398, with epigallocatechin gallate or with acetylsalicylic acid, all with anti-inflammatory effects. For the first time, we demonstrated that both oxysterols and HNE, which substantially accumulate in the atherosclerotic plaque, up-regulate the activity of PCSK6. Of note, we also suggest a potential association between PCSK6 activity and MMP-9 activation, pointing out that PCSK6 could contribute to atherosclerotic plaque development.
Collapse
Affiliation(s)
- Gabriella Testa
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, 10043, Orbassano, Turin, Italy
| | - Erica Staurenghi
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, 10043, Orbassano, Turin, Italy
| | - Serena Giannelli
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, 10043, Orbassano, Turin, Italy
| | - Barbara Sottero
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, 10043, Orbassano, Turin, Italy
| | - Simona Gargiulo
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, 10043, Orbassano, Turin, Italy
| | - Giuseppe Poli
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, 10043, Orbassano, Turin, Italy
| | - Paola Gamba
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, 10043, Orbassano, Turin, Italy
| | - Gabriella Leonarduzzi
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, 10043, Orbassano, Turin, Italy.
| |
Collapse
|
11
|
Activation of proprotein convertase in the mouse habenula causes depressive-like behaviors through remodeling of extracellular matrix. Neuropsychopharmacology 2021; 46:442-454. [PMID: 32942293 PMCID: PMC7852607 DOI: 10.1038/s41386-020-00843-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 07/20/2020] [Accepted: 08/25/2020] [Indexed: 12/16/2022]
Abstract
The lateral habenula (LHb) attracts a growing interest as a regulator of monoaminergic activity which were frequently reported to be defective in depression. Here we found that chronic social defeat stress (CSDS) increased production of pro-inflammatory cytokines in LHb associated with mobilization of monocytes and remodeling of extracellular matrix by increased matrix metalloproteinase (MMP) activity. RNA-seq analysis identified proprotein convertase Pcsk5 as an upstream regulator of MMP activation, with upregulation in LHb neurons of mice with susceptibility to CSDS. PCSK5 facilitated motility of microglia in vitro by converting inactive pro-MMP14 and pro-MMP2 to their active forms, highlighting its role in mobilization of microglia and monocytes in neuroinflammation. Suppression of Pcsk5 expression via small interfering RNA (siRNA) ameliorated depressive-like behaviors and pathological mobilization of monocytes in mice with susceptibility to CSDS. PCSK5-MMPs signaling pathway could be a target for development of the antidepressants targeting the inflammatory response in specific brain regions implicated in depression.
Collapse
|
12
|
Yakala GK, Cabrera-Fuentes HA, Crespo-Avilan GE, Rattanasopa C, Burlacu A, George BL, Anand K, Mayan DC, Corlianò M, Hernández-Reséndiz S, Wu Z, Schwerk AMK, Tan ALJ, Trigueros-Motos L, Chèvre R, Chua T, Kleemann R, Liehn EA, Hausenloy DJ, Ghosh S, Singaraja RR. FURIN Inhibition Reduces Vascular Remodeling and Atherosclerotic Lesion Progression in Mice. Arterioscler Thromb Vasc Biol 2020; 39:387-401. [PMID: 30651003 PMCID: PMC6393193 DOI: 10.1161/atvbaha.118.311903] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Objective- Atherosclerotic coronary artery disease is the leading cause of death worldwide, and current treatment options are insufficient. Using systems-level network cluster analyses on a large coronary artery disease case-control cohort, we previously identified PCSK3 (proprotein convertase subtilisin/kexin family member 3; FURIN) as a member of several coronary artery disease-associated pathways. Thus, our objective is to determine the role of FURIN in atherosclerosis. Approach and Results- In vitro, FURIN inhibitor treatment resulted in reduced monocyte migration and reduced macrophage and vascular endothelial cell inflammatory and cytokine gene expression. In vivo, administration of an irreversible inhibitor of FURIN, α-1-PDX (α1-antitrypsin Portland), to hyperlipidemic Ldlr-/- mice resulted in lower atherosclerotic lesion area and a specific reduction in severe lesions. Significantly lower lesional macrophage and collagen area, as well as systemic inflammatory markers, were observed. MMP2 (matrix metallopeptidase 2), an effector of endothelial function and atherosclerotic lesion progression, and a FURIN substrate was significantly reduced in the aorta of inhibitor-treated mice. To determine FURIN's role in vascular endothelial function, we administered α-1-PDX to Apoe-/- mice harboring a wire injury in the common carotid artery. We observed significantly decreased carotid intimal thickness and lower plaque cellularity, smooth muscle cell, macrophage, and inflammatory marker content, suggesting protection against vascular remodeling. Overexpression of FURIN in this model resulted in a significant 67% increase in intimal plaque thickness, confirming that FURIN levels directly correlate with atherosclerosis. Conclusions- We show that systemic inhibition of FURIN in mice decreases vascular remodeling and atherosclerosis. FURIN-mediated modulation of MMP2 activity may contribute to the atheroprotection observed in these mice.
Collapse
Affiliation(s)
- Gopala K Yakala
- From the Translational Laboratories in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (G.K.Y., C.R., K.A., D.C.M., M.C., Z.W., A.L.J.T., L.T.-M., R.C., T.C., R.R.S.)
| | - Hector A Cabrera-Fuentes
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore (H.A.C.-F., G.E.C.-A., C.R., S.H.-R., D.J.H., S.G.).,National Heart Research Institute, National Heart Centre Singapore (H.A.C.-F., G.E.C.-A., B.L.G., S.H.-R., E.A.L., D.J.H., S.G.).,Institute of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany (H.A.C.-F.).,Department of Microbiology, Kazan Federal University, Russian Federation (H.A.C.-F.).,Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Centro de Biotecnologia-FEMSA, Nuevo Leon, México (H.A.C.-F.)
| | - Gustavo E Crespo-Avilan
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore (H.A.C.-F., G.E.C.-A., C.R., S.H.-R., D.J.H., S.G.).,National Heart Research Institute, National Heart Centre Singapore (H.A.C.-F., G.E.C.-A., B.L.G., S.H.-R., E.A.L., D.J.H., S.G.)
| | - Chutima Rattanasopa
- From the Translational Laboratories in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (G.K.Y., C.R., K.A., D.C.M., M.C., Z.W., A.L.J.T., L.T.-M., R.C., T.C., R.R.S.).,Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore (H.A.C.-F., G.E.C.-A., C.R., S.H.-R., D.J.H., S.G.)
| | - Alexandrina Burlacu
- Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania (A.B.)
| | - Benjamin L George
- National Heart Research Institute, National Heart Centre Singapore (H.A.C.-F., G.E.C.-A., B.L.G., S.H.-R., E.A.L., D.J.H., S.G.)
| | - Kaviya Anand
- From the Translational Laboratories in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (G.K.Y., C.R., K.A., D.C.M., M.C., Z.W., A.L.J.T., L.T.-M., R.C., T.C., R.R.S.)
| | - David Castaño Mayan
- From the Translational Laboratories in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (G.K.Y., C.R., K.A., D.C.M., M.C., Z.W., A.L.J.T., L.T.-M., R.C., T.C., R.R.S.)
| | - Maria Corlianò
- From the Translational Laboratories in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (G.K.Y., C.R., K.A., D.C.M., M.C., Z.W., A.L.J.T., L.T.-M., R.C., T.C., R.R.S.)
| | - Sauri Hernández-Reséndiz
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore (H.A.C.-F., G.E.C.-A., C.R., S.H.-R., D.J.H., S.G.).,National Heart Research Institute, National Heart Centre Singapore (H.A.C.-F., G.E.C.-A., B.L.G., S.H.-R., E.A.L., D.J.H., S.G.)
| | - Zihao Wu
- From the Translational Laboratories in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (G.K.Y., C.R., K.A., D.C.M., M.C., Z.W., A.L.J.T., L.T.-M., R.C., T.C., R.R.S.)
| | - Anne M K Schwerk
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden (A.M.K.S., R.K.)
| | - Amberlyn L J Tan
- From the Translational Laboratories in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (G.K.Y., C.R., K.A., D.C.M., M.C., Z.W., A.L.J.T., L.T.-M., R.C., T.C., R.R.S.)
| | - Laia Trigueros-Motos
- From the Translational Laboratories in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (G.K.Y., C.R., K.A., D.C.M., M.C., Z.W., A.L.J.T., L.T.-M., R.C., T.C., R.R.S.)
| | - Raphael Chèvre
- From the Translational Laboratories in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (G.K.Y., C.R., K.A., D.C.M., M.C., Z.W., A.L.J.T., L.T.-M., R.C., T.C., R.R.S.)
| | - Tricia Chua
- From the Translational Laboratories in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (G.K.Y., C.R., K.A., D.C.M., M.C., Z.W., A.L.J.T., L.T.-M., R.C., T.C., R.R.S.)
| | - Robert Kleemann
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden (A.M.K.S., R.K.).,Department of Vascular Surgery, Leiden University Medical Center, the Netherlands (R.K.)
| | - Elisa A Liehn
- National Heart Research Institute, National Heart Centre Singapore (H.A.C.-F., G.E.C.-A., B.L.G., S.H.-R., E.A.L., D.J.H., S.G.).,Institute of Molecular Cardiovascular Research, RWTH, Aachen, Germany (E.A.L.).,Human Genetic Laboratory, University of Medicine, Craiova, Romania (E.A.L.)
| | - Derek J Hausenloy
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore (H.A.C.-F., G.E.C.-A., C.R., S.H.-R., D.J.H., S.G.).,National Heart Research Institute, National Heart Centre Singapore (H.A.C.-F., G.E.C.-A., B.L.G., S.H.-R., E.A.L., D.J.H., S.G.).,Yong Loo Lin School of Medicine, National University Singapore (D.J.H.).,The Hatter Cardiovascular Institute, University College London, United Kingdom (D.J.H.).,The National Institute of Health Research, University College London Hospitals Biomedical Research Centre, United Kingdom (D.J.H.).,Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom (D.J.H.)
| | - Sujoy Ghosh
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore (H.A.C.-F., G.E.C.-A., C.R., S.H.-R., D.J.H., S.G.).,National Heart Research Institute, National Heart Centre Singapore (H.A.C.-F., G.E.C.-A., B.L.G., S.H.-R., E.A.L., D.J.H., S.G.)
| | - Roshni R Singaraja
- From the Translational Laboratories in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (G.K.Y., C.R., K.A., D.C.M., M.C., Z.W., A.L.J.T., L.T.-M., R.C., T.C., R.R.S.)
| |
Collapse
|
13
|
Rykaczewska U, Suur BE, Röhl S, Razuvaev A, Lengquist M, Sabater-Lleal M, van der Laan SW, Miller CL, Wirka RC, Kronqvist M, Gonzalez Diez M, Vesterlund M, Gillgren P, Odeberg J, Lindeman JH, Veglia F, Humphries SE, de Faire U, Baldassarre D, Tremoli E, Lehtiö J, Hansson GK, Paulsson-Berne G, Pasterkamp G, Quertermous T, Hamsten A, Eriksson P, Hedin U, Matic L. PCSK6 Is a Key Protease in the Control of Smooth Muscle Cell Function in Vascular Remodeling. Circ Res 2020; 126:571-585. [PMID: 31893970 DOI: 10.1161/circresaha.119.316063] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
RATIONALE PCSKs (Proprotein convertase subtilisins/kexins) are a protease family with unknown functions in vasculature. Previously, we demonstrated PCSK6 upregulation in human atherosclerotic plaques associated with smooth muscle cells (SMCs), inflammation, extracellular matrix remodeling, and mitogens. OBJECTIVE Here, we applied a systems biology approach to gain deeper insights into the PCSK6 role in normal and diseased vessel wall. METHODS AND RESULTS Genetic analyses revealed association of intronic PCSK6 variant rs1531817 with maximum internal carotid intima-media thickness progression in high-cardiovascular risk subjects. This variant was linked with PCSK6 mRNA expression in healthy aortas and plaques but also with overall plaque SMA+ cell content and pericyte fraction. Increased PCSK6 expression was found in several independent human cohorts comparing atherosclerotic lesions versus healthy arteries, using transcriptomic and proteomic datasets. By immunohistochemistry, PCSK6 was localized to fibrous cap SMA+ cells and neovessels in plaques. In human, rat, and mouse intimal hyperplasia, PCSK6 was expressed by proliferating SMA+ cells and upregulated after 5 days in rat carotid balloon injury model, with positive correlation to PDGFB (platelet-derived growth factor subunit B) and MMP (matrix metalloprotease) 2/MMP14. Here, PCSK6 was shown to colocalize and cointeract with MMP2/MMP14 by in situ proximity ligation assay. Microarrays of carotid arteries from Pcsk6-/- versus control mice revealed suppression of contractile SMC markers, extracellular matrix remodeling enzymes, and cytokines/receptors. Pcsk6-/- mice showed reduced intimal hyperplasia response upon carotid ligation in vivo, accompanied by decreased MMP14 activation and impaired SMC outgrowth from aortic rings ex vivo. PCSK6 silencing in human SMCs in vitro leads to downregulation of contractile markers and increase in MMP2 expression. Conversely, PCSK6 overexpression increased PDGFBB (platelet-derived growth factor BB)-induced cell proliferation and particularly migration. CONCLUSIONS PCSK6 is a novel protease that induces SMC migration in response to PDGFB, mechanistically via modulation of contractile markers and MMP14 activation. This study establishes PCSK6 as a key regulator of SMC function in vascular remodeling. Visual Overview: An online visual overview is available for this article.
Collapse
Affiliation(s)
- Urszula Rykaczewska
- From the Department of Molecular Medicine and Surgery, Karolinska Institute and Karolinska University Hospital Solna, Stockholm, Sweden (U.R., B.E.S., S.R., A.R., M.L., M.K., U.H., L.M.)
| | - Bianca E Suur
- From the Department of Molecular Medicine and Surgery, Karolinska Institute and Karolinska University Hospital Solna, Stockholm, Sweden (U.R., B.E.S., S.R., A.R., M.L., M.K., U.H., L.M.)
| | - Samuel Röhl
- From the Department of Molecular Medicine and Surgery, Karolinska Institute and Karolinska University Hospital Solna, Stockholm, Sweden (U.R., B.E.S., S.R., A.R., M.L., M.K., U.H., L.M.)
| | - Anton Razuvaev
- From the Department of Molecular Medicine and Surgery, Karolinska Institute and Karolinska University Hospital Solna, Stockholm, Sweden (U.R., B.E.S., S.R., A.R., M.L., M.K., U.H., L.M.)
| | - Mariette Lengquist
- From the Department of Molecular Medicine and Surgery, Karolinska Institute and Karolinska University Hospital Solna, Stockholm, Sweden (U.R., B.E.S., S.R., A.R., M.L., M.K., U.H., L.M.)
| | - Maria Sabater-Lleal
- Department of Medicine Solna, Karolinska Institute and Karolinska University Hospital Solna, Stockholm, Sweden (M.S.-L., M.G.D., G.P.-B., G.K.H., A.H., P.E., J.O.).,Unit of Genomics of Complex Diseases, Institut de Recerca Hospital de Sant Pau (IIB-Sant Pau), Barcelona, Spain (M.S.-L.)
| | - Sander W van der Laan
- Central Diagnostics Laboratory, Laboratories, Pharmacy, and Biomedical Genetics, University Medical Center Utrecht, Utrecht University, The Netherlands (S.v.d.L.)
| | - Clint L Miller
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia, Charlottesville (C.L.M.).,Division of Cardiovascular Medicine, Stanford University School of Medicine, CA (C.L.M., R.C.W., T.Q.)
| | - Robert C Wirka
- Division of Cardiovascular Medicine, Stanford University School of Medicine, CA (C.L.M., R.C.W., T.Q.)
| | - Malin Kronqvist
- From the Department of Molecular Medicine and Surgery, Karolinska Institute and Karolinska University Hospital Solna, Stockholm, Sweden (U.R., B.E.S., S.R., A.R., M.L., M.K., U.H., L.M.)
| | - Maria Gonzalez Diez
- Department of Medicine Solna, Karolinska Institute and Karolinska University Hospital Solna, Stockholm, Sweden (M.S.-L., M.G.D., G.P.-B., G.K.H., A.H., P.E., J.O.)
| | - Mattias Vesterlund
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institute, Sweden (M.V., J.L.)
| | - Peter Gillgren
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, and Department of Vascular Surgery, Södersjukhuset, Stockholm, Sweden (P.G.)
| | - Jacob Odeberg
- Department of Medicine Solna, Karolinska Institute and Karolinska University Hospital Solna, Stockholm, Sweden (M.S.-L., M.G.D., G.P.-B., G.K.H., A.H., P.E., J.O.).,Science for Life Laboratory, Department of Proteomics, School of Chemistry Biotechnology and Health (CBH), KTH, Stockholm, Sweden (J.O.)
| | - Jan H Lindeman
- Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.)
| | - Fabrizio Veglia
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (F.V., D.B., E.T.)
| | - Steve E Humphries
- Cardiovascular Genetics, Institute Cardiovascular Science, University College of London, Department of Medicine, Rayne Building, United Kingdom (S.E.H.)
| | - Ulf de Faire
- Division of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Sweden (H.d.F.)
| | - Damiano Baldassarre
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (F.V., D.B., E.T.).,Department of Medical Biotechnology and Translational Medicine, Università di Milano, Milan, Italy (D.B.)
| | - Elena Tremoli
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (F.V., D.B., E.T.)
| | | | - Janne Lehtiö
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institute, Sweden (M.V., J.L.)
| | - Göran K Hansson
- Department of Medicine Solna, Karolinska Institute and Karolinska University Hospital Solna, Stockholm, Sweden (M.S.-L., M.G.D., G.P.-B., G.K.H., A.H., P.E., J.O.)
| | - Gabrielle Paulsson-Berne
- Department of Medicine Solna, Karolinska Institute and Karolinska University Hospital Solna, Stockholm, Sweden (M.S.-L., M.G.D., G.P.-B., G.K.H., A.H., P.E., J.O.)
| | - Gerard Pasterkamp
- Laboratory of Experimental Cardiology, Division Heart & Lungs, University Medical Center Utrecht, The Netherlands (G.P.)
| | - Thomas Quertermous
- Division of Cardiovascular Medicine, Stanford University School of Medicine, CA (C.L.M., R.C.W., T.Q.)
| | - Anders Hamsten
- Department of Medicine Solna, Karolinska Institute and Karolinska University Hospital Solna, Stockholm, Sweden (M.S.-L., M.G.D., G.P.-B., G.K.H., A.H., P.E., J.O.)
| | - Per Eriksson
- Department of Medicine Solna, Karolinska Institute and Karolinska University Hospital Solna, Stockholm, Sweden (M.S.-L., M.G.D., G.P.-B., G.K.H., A.H., P.E., J.O.)
| | - Ulf Hedin
- From the Department of Molecular Medicine and Surgery, Karolinska Institute and Karolinska University Hospital Solna, Stockholm, Sweden (U.R., B.E.S., S.R., A.R., M.L., M.K., U.H., L.M.)
| | - Ljubica Matic
- From the Department of Molecular Medicine and Surgery, Karolinska Institute and Karolinska University Hospital Solna, Stockholm, Sweden (U.R., B.E.S., S.R., A.R., M.L., M.K., U.H., L.M.)
| |
Collapse
|
14
|
Löw K, Hardes K, Fedeli C, Seidah NG, Constam DB, Pasquato A, Steinmetzer T, Roulin A, Kunz S. A novel cell-based sensor detecting the activity of individual basic proprotein convertases. FEBS J 2019; 286:4597-4620. [PMID: 31276291 DOI: 10.1111/febs.14979] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 05/13/2019] [Accepted: 07/02/2019] [Indexed: 02/06/2023]
Abstract
The basic proprotein convertases (PCs) furin, PC1/3, PC2, PC5/6, PACE4, PC4, and PC7 are promising drug targets for human diseases. However, developing selective inhibitors remains challenging due to overlapping substrate recognition motifs and limited structural information. Classical drug screening approaches for basic PC inhibitors involve homogeneous biochemical assays using soluble recombinant enzymes combined with fluorogenic substrate peptides that may not accurately recapitulate the complex cellular context of the basic PC-substrate interaction. Herein we report basic PC sensor (BPCS), a novel cell-based molecular sensor that allows rapid screening of candidate inhibitors and their selectivity toward individual basic PCs within mammalian cells. BPCS consists of Gaussia luciferase linked to a sortilin-1 membrane anchor via a cleavage motif that allows efficient release of luciferase specifically if individual basic PCs are provided in the same membrane. Screening of selected candidate peptidomimetic inhibitors revealed that BPCS can readily distinguish between general and selective PC inhibitors in a high-throughput screening format. The robust and cost-effective assay format of BPCS makes it suitable to identify novel specific small-molecule inhibitors against basic PCs for therapeutic application. Its cell-based nature will allow screening for drug targets in addition to the catalytically active mature enzyme, including maturation, transport, and cellular factors that modulate the enzyme's activity. This broadened 'target range' will enhance the likelihood to identify novel small-molecule compounds that inhibit basic PCs in a direct or indirect manner and represents a conceptual advantage.
Collapse
Affiliation(s)
- Karin Löw
- Institute of Microbiology, University Hospital Center, University of Lausanne, Switzerland.,Department of Ecology and Evolution, University of Lausanne, Switzerland
| | - Kornelia Hardes
- Department of Pharmacy, Institute of Pharmaceutical Chemistry, Philipps University Marburg, Germany
| | - Chiara Fedeli
- Institute of Microbiology, University Hospital Center, University of Lausanne, Switzerland
| | - Nabil G Seidah
- Laboratory of Biochemical Neuroendocrinology, Clinical Research Institute of Montreal, (Affiliated to the University of Montreal), Canada
| | - Daniel B Constam
- Ecole Polytechnique Fédérale de Lausanne (EPFL) SV ISREC, Switzerland
| | - Antonella Pasquato
- Institute of Microbiology, University Hospital Center, University of Lausanne, Switzerland
| | - Torsten Steinmetzer
- Department of Pharmacy, Institute of Pharmaceutical Chemistry, Philipps University Marburg, Germany
| | - Alexandre Roulin
- Department of Ecology and Evolution, University of Lausanne, Switzerland
| | - Stefan Kunz
- Institute of Microbiology, University Hospital Center, University of Lausanne, Switzerland
| |
Collapse
|
15
|
Proprotein convertase furin/PCSK3 and atherosclerosis: New insights and potential therapeutic targets. Atherosclerosis 2017; 262:163-170. [DOI: 10.1016/j.atherosclerosis.2017.04.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 03/31/2017] [Accepted: 04/06/2017] [Indexed: 12/12/2022]
|
16
|
Solovyeva NI, Gureeva TA, Timoshenko OS, Moskvitina TA, Kugaevskaya EV. Furin as proprotein convertase and its role in normal and pathological biological processes. BIOCHEMISTRY (MOSCOW), SUPPLEMENT SERIES B: BIOMEDICAL CHEMISTRY 2017. [DOI: 10.1134/s1990750817020081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
17
|
Solovyeva N, Gureeva T, Timoshenko O, Moskvitina T, Kugaevskaya E. Furin as proprotein convertase and its role in normal and pathological biological processes. ACTA ACUST UNITED AC 2016; 62:609-621. [DOI: 10.18097/pbmc20166206609] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Furin belongs to serine intracellular Ca2+-dependent endopeptidases of the subtilisin family, also known as proprotein convertase (PC). Human furin is synthesized as zymogen with a molecular weight of 104 kDa, which is then activated by autocatalytic in two stages. This process can occur when zymogen migrates from the endoplasmic reticulum to the Golgi apparatus, where a large part of furin is accumulated. The molecular weigh t of the active furin is 98 kDa. Furin relates to enzymes with a narrow substrate specificity: it hydrolyzes peptide bonds at the site of paired basic amino acids and furin activity exhibits in a wide pH range 5-8. Its main biological function is activation of the functionally important protein precursors. It is accompanied by the launch of a cascade of reactions, which lead to appearance of biologically active molecules involved in realization of specific biological functions both in normal and in some patologicheskih processes. Furin substrates are biologically important proteins such as enzymes, hormones, growth factors and differentiation, receptors, adhesion proteins, proteins of blood plasma. Furin plays an important role in the development of processes such as proliferation, invasion, cell migration, survival, maintenance of homeostasis, embryogenesis, as well as the development of a number of pathologies, including cardiovascular, oncologic and neurodegenerative diseases. Furin and furin-like proprotein convertases participate as key factors in the realization of the regulatory functions of proteolytic enzymes, the value of which is currently being evaluated as most important in comparison with the degradative function of proteases.
Collapse
Affiliation(s)
| | - T.A. Gureeva
- Institute of Biomedical Chemistry, Moscow, Russia
| | | | | | | |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW The proprotein convertases subtilisin/kexin (PCSKs) are endoproteases identified as activators of precursors from hormones and peptides. On the basis of the variety of substrates and regulation in disease, they have been recognized as mediators in atherogenesis. The discovery of PCSK9, which regulates low-density lipoprotein receptor cell membrane availability, has led to a resurgence of interest in these enzymes and their function in cardiovascular diseases. RECENT FINDINGS Recent data demonstrate that PCSKs are expressed in human atheroma and are regulated in animal models of atherosclerosis. In animal models, inhibition of PCSKs, such as PCSK3, affects cell proliferation and migration as well as inflammation, reducing atherosclerosis. In addition, targeting PCSK9 lowers cholesterol levels and has now been demonstrated to lessen vascular lesion formation in mice. Experimentally investigated novel anti-PCSK9 strategies include genome editing and vaccination. Furthermore, studies show that PCSKs contribute to the initiation and progression of cardiometabolic risk factors, such as insulin resistance and obesity. SUMMARY PCSKs affect cardiovascular diseases on multiple levels, including atherosclerotic lesion formation as well as their contribution to cardiometabolic risk factors. PCSK9 is a key regulator of plasma cholesterol levels, thereby potentially affecting atherosclerosis and has rapidly emerged as a pharmacological target.
Collapse
Affiliation(s)
- Philipp Stawowy
- Deutsches Herzzentrum Berlin, Department of Medicine/Cardiology, Berlin, Germany
| |
Collapse
|
19
|
Valanti E, Tsompanidis A, Sanoudou D. Pharmacogenomics in the development and characterization of atheroprotective drugs. Methods Mol Biol 2014; 1175:259-300. [PMID: 25150873 DOI: 10.1007/978-1-4939-0956-8_11] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Atherosclerosis is the main cause of cardiovascular disease (CVD) and can lead to stroke, myocardial infarction, and death. The clinically available atheroprotective drugs aim mainly at reducing the levels of circulating low-density lipoprotein (LDL), increasing high-density lipoprotein (HDL), and attenuating inflammation. However, the cardiovascular risk remains high, along with morbidity, mortality, and incidence of adverse drug events. Pharmacogenomics is increasingly contributing towards the characterization of existing atheroprotective drugs, the evaluation of novel ones, and the identification of promising, unexplored therapeutic targets, at the global molecular pathway level. This chapter presents highlights of pharmacogenomics investigations and discoveries that have contributed towards the elucidation of pharmacological atheroprotection, while opening the way to new therapeutic approaches.
Collapse
Affiliation(s)
- Efi Valanti
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Athens, 115 27, Greece
| | | | | |
Collapse
|
20
|
Mbikay M, Mayne J, Chrétien M. Proprotein convertases subtilisin/kexin type 9, an enzyme turned escort protein: hepatic and extra hepatic functions. J Diabetes 2013; 5:391-405. [PMID: 23714205 DOI: 10.1111/1753-0407.12064] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Accepted: 05/24/2013] [Indexed: 01/07/2023] Open
Abstract
Proprotein Convertases Subtilisin/Kexin Type 9 (PCSK9) is a serine endoproteinase. Biosynthesized as a zymogen, it cleaves itself once, and then turns into an escort protein for transmembrane proteins, leading them into lysosomes for degradation. It is primarily produced and secreted by the liver. It attaches to the low-density lipoprotein receptor (LDLR) at the surface of hepatocytes and, after co-endocytosis, directs it into lysosomes where it is degraded. By downregulating LDLR, PCSK9 reduces hepatic clearance of LDL-cholesterol. Inborn or induced increase of this function causes hypercholesterolemia; its decrease causes hypocholesterolemia. This has been experimentally demonstrated ex vivo and in vivo, and corroborated by epidemiological studies associating PCSK9 genetic variations with plasma cholesterol levels. PCSK9 is now a proven target for inactivation in the treatment of hypercholesterolemia and associated atherosclerosis. However, it is still uncertain whether its severe or complete inactivation, combined with other predispositions, will be without undesirable side-effects. Some experimental data suggest that PCSK9 could contribute positively to the physiology of non-hepatic cells such as pancreatic islets β cells, adipocytes and macrophages, protecting them from excessive lipid uptake, in an endocrine, autocrine, or paracrine manner. Genetic variations that attenuate PCSK9 anti-LDLR activity are common in human populations. Their evolutionary significance still needs to be evaluated on the background of environmental pressures, such as infectious diseases, cold weather and famine, which have threatened survival and reproduction in the course of human prehistory and history.
Collapse
Affiliation(s)
- Majambu Mbikay
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario; Department of Medicine, University of Ottawa, Ottawa, Ontario; Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario; Division of Endocrinology and Metabolism, The Ottawa Hospital, Ottawa, Ontario
| | | | | |
Collapse
|
21
|
Stawowy P, Kelle S, Fleck E. PCSK9 als neues Target in der Therapie der Hypercholesterinämie. Herz 2013; 39:466-9. [DOI: 10.1007/s00059-013-3913-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 07/04/2013] [Indexed: 11/29/2022]
|
22
|
Perisic L, Hedin E, Razuvaev A, Lengquist M, Osterholm C, Folkersen L, Gillgren P, Paulsson-Berne G, Ponten F, Odeberg J, Hedin U. Profiling of atherosclerotic lesions by gene and tissue microarrays reveals PCSK6 as a novel protease in unstable carotid atherosclerosis. Arterioscler Thromb Vasc Biol 2013; 33:2432-43. [PMID: 23908247 DOI: 10.1161/atvbaha.113.301743] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Carotid plaque instability is a major cause of ischemic stroke, but detailed knowledge about underlying molecular pathways is still lacking. Here, we evaluated large-scale transcriptomic and protein expression profiling in a biobank of carotid endarterectomies followed by characterization of identified candidates, as a platform for discovery of novel proteins differentially regulated in unstable carotid lesions. APPROACH AND RESULTS Genes highly upregulated in symptomatic versus asymptomatic plaques were selected from Affymetrix microarray analyses (n=127 plaques), and tissue microarrays constructed from 34 lesions were assayed for 21 corresponding proteins by immunohistochemistry. Quantification of stainings demonstrated differential expression of CD36, CD137, and DOCK7 (P<0.05) in unstable versus stable lesions and the most significant upregulation of a proprotein convertase, PCSK6 (P<0.0001). Increased expression of PCSK6 in symptomatic lesions was verified by quantitative real-time polymerase chain reaction (n=233), and the protein was localized to smooth muscle α-actin positive cells and extracellular matrix of the fibrous cap by immunohistochemistry. PCSK6 expression positively correlated to genes associated with inflammation, matrix degradation, and mitogens in microarrays. Stimulation of human carotid smooth muscle cells in vitro with cytokines caused rapid induction of PCSK6 mRNA. CONCLUSIONS Using a combination of transcriptomic and tissue microarray profiling, we demonstrate a novel approach to identify proteins differentially expressed in unstable carotid atherosclerosis. The proprotein convertase PCSK6 was detected at increased levels in the fibrous cap of symptomatic carotid plaques, possibly associated with key processes in plaque rupture such as inflammation and extracellular matrix remodeling. Further studies are needed to clarify the role of PCSK6 in atherosclerosis.
Collapse
Affiliation(s)
- Ljubica Perisic
- From the Department of Molecular Medicine and Surgery (L.P., E.H., A.R., M.L., C.O., U.H.), and Department of Medicine (G.P.-B., J.O.), Karolinska Institute, Stockholm, Sweden; Department of Molecular Genetics, Novo Nordisk, Copenhagen, Denmark (L.F.); Department of Surgery, Södersjukhuset, Stockholm, Sweden (P.G.); Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden (F.P.); and Department of Proteomics, Royal Institute of Technology, Stockholm, Sweden (J.O.)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Time for endothelial cell proprotein convertase PC5/6 in cardiovascular medicine? J Mol Med (Berl) 2012; 89:1061-3. [PMID: 21887504 DOI: 10.1007/s00109-011-0810-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
24
|
Couture F, D'Anjou F, Day R. On the cutting edge of proprotein convertase pharmacology: from molecular concepts to clinical applications. Biomol Concepts 2011; 2:421-438. [PMID: 22308173 DOI: 10.1515/bmc.2011.034] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
There is increasing interest in the therapeutic targeting of proteases for the treatment of important diseases. Additionally new protein-based therapeutic strategies have the potential to widen the available treatments against these pathologies. In the last decade, accumulated evidence has confirmed that the family of proteases known as proprotein convertases (PCs) are potential targets for viral infections, osteoarthritis, cancer and cardiovascular disease, among others. Nevertheless, there are still many unanswered questions about the relevance of targeting PCs in a therapeutic context, especially regarding the anticipated secondary effects of treatment, considering the observed embryonic lethality of some PC knockout mice. In this review, the benefits of PCs as pharmacological targets will be discussed, with focus on concepts and strategies, as well as on the state of advancement of actual and future inhibitors.
Collapse
Affiliation(s)
- Frédéric Couture
- Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke J1H 5N4, Québec, Canada
| | | | | |
Collapse
|
25
|
Proprotein convertases in human atherosclerotic plaques: the overexpression of FURIN and its substrate cytokines BAFF and APRIL. Atherosclerosis 2011; 219:799-806. [PMID: 21889147 DOI: 10.1016/j.atherosclerosis.2011.08.011] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Revised: 08/05/2011] [Accepted: 08/10/2011] [Indexed: 12/11/2022]
Abstract
BACKGROUND Proprotein convertase subtilisin/kexin (PCSK) enzymes cleave proproteins into mature end products. Previously, MBTPS1 and PCSK9 have been shown to regulate cholesterol metabolism and LDL receptor recycling, whereas FURIN and PCSK5 have been suggested to inactivate lipases and regulate inflammation in atherosclerosis. Here, we systematically analyzed the expression of PCSKs and their targets in advanced atherosclerotic plaques. METHODS AND RESULTS Microarray and quantitative real-time PCR experiments showed that FURIN (42.86 median fold, p = 2.1e-8), but no other PCSK, is universally overexpressed in the plaques of different vascular regions. The mRNA expression screen of PCSK target proteins in plaques identified many known factors, but it also identified the significant upregulation of the previously overlooked furin-processed B cell activating cytokines APRIL (TNFSF13, 2.52 median fold, p = 3.0e-5) and BAFF (TNFSF13B, 2.97 median fold, p = 7.6e-6). The dysregulation of FURIN did not associate with its htSNPs or the previously reported regulatory SNP (-229, rs4932178) in the promoter. Immunohistochemistry experiments showed the upregulation of FURIN in the plaque lymphocytes and macrophages where it was co-expressed with BAFF/TNFSF13B and APRIL/TNFSF13. CONCLUSIONS Our data unequivocally show that FURIN is the primary PCSK that is dysregulated in the immune cells of advanced human atherosclerotic plaques, which implies a role for this enzyme in plaque pathology. Therefore, drugs that inhibit FURIN in arteries may modulate the course of this disease.
Collapse
|
26
|
Stawowy P, Kappert K. The molecular biology of furin-like proprotein convertases in vascular remodelling. Methods Mol Biol 2011; 768:191-206. [PMID: 21805243 DOI: 10.1007/978-1-61779-204-5_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Vascular smooth muscle cell (VSMC) proliferation and migration represent key features in atherosclerosis and restenosis. The proprotein convertases (PCs) furin and PC5 are highly expressed in human atheroma and are putatively involved in vascular lesion formation via the activation of precursor proteins, essential for cell proliferation and migration. In vitro assays have identified these PCs to govern cell functions via endoproteolytic cleavage of key substrates, including pro-integrins and pro-matrix metalloproteinases. In vivo gene expression studies of furin/PC5 and their substrates demonstrate their coordinated regulation in animal models of vascular remodelling and in human atherosclerotic lesions. Here we describe in vitro and in vivo models to investigate the function of furin/PC5 in VSMCs and in vascular lesion formation. In conjunction with the development of novel PC inhibitors, this should facilitate the development of new strategies targeting PCs in cardiovascular disease.
Collapse
Affiliation(s)
- Philipp Stawowy
- Department of Medicine/Cardiology, Deutsches Herzzentrum Berlin, D-13353, Berlin, Germany.
| | | |
Collapse
|
27
|
Funkelstein L, Hook V. The novel role of cathepsin L for neuropeptide production illustrated by research strategies in chemical biology with protease gene knockout and expression. Methods Mol Biol 2011; 768:107-125. [PMID: 21805239 DOI: 10.1007/978-1-61779-204-5_5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Neuropeptides are essential for cell-cell communication in the nervous and endocrine systems. Production of active neuropeptides requires proteolytic processing of proneuropeptide precursors in secretory vesicles that produce, store, and release neuropeptides that regulate physiological functions. This review describes research strategies utilizing chemical biology combined with protease gene knockout and expression to demonstrate the key role of cathepsin L for production of neuropeptides in secretory vesicles. Cathepsin L was discovered using activity-based probes and mass spectrometry to identify proenkephalin cleaving activity as cathepsin L. Significantly, in vivo protease gene knockout and expression approaches illustrate the key role of cathepsin L for neuropeptide production. Notably, cathepsin L is colocalized with neuropeptide secretory vesicles, the major site of proteolytic processing of proneuropeptides to generate active neuropeptides. Cathepsin L participates in producing opioid neuropeptides consisting of enkephalin, β-endorphin, and dynorphin, as well as in generating the POMC-derived peptide hormones ACTH and α-MSH. In addition, NPY, CCK, and catestatin neuropeptides utilize cathepsin L for their biosynthesis. The role of cathepsin L for neuropeptide production indicates its unique biological role in secretory vesicles, which contrasts with its role in lysosomes for protein degradation. Interesting evaluations of protease gene knockout studies in mice that lack cathepsin L compared to the PC1/3 and PC2 (PC, prohormone convertase) indicate the significant role of cathepsin L in neuropeptide production. Thus, dual cathepsin L and prohormone convertase protease pathways participate in neuropeptide production. These recent new findings indicate cathepsin L as a novel 'proprotein convertase' for production of neuropeptides that mediate cell-cell communication in health and disease.
Collapse
Affiliation(s)
- Lydiane Funkelstein
- Department of Neuroscience, Pharmacology, and Medicine, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, CA 93093, USA.
| | | |
Collapse
|
28
|
Funkelstein L, Beinfeld M, Minokadeh A, Zadina J, Hook V. Unique biological function of cathepsin L in secretory vesicles for biosynthesis of neuropeptides. Neuropeptides 2010; 44:457-66. [PMID: 21047684 PMCID: PMC3058267 DOI: 10.1016/j.npep.2010.08.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Revised: 08/05/2010] [Accepted: 08/30/2010] [Indexed: 11/18/2022]
Abstract
Neuropeptides are essential for cell-cell communication in the nervous and neuroendocrine systems. Production of active neuropeptides requires proteolytic processing of proneuropeptide precursors in secretory vesicles that produce, store, and release neuropeptides that regulate physiological functions. This review describes recent findings indicating the prominent role of cathepsin L in secretory vesicles for production of neuropeptides from their protein precursors. The role of cathepsin L in neuropeptide production was discovered using the strategy of activity-based probes for proenkephalin-cleaving activity for identification of the enzyme protein by mass spectrometry. The novel role of cathepsin L in secretory vesicles for neuropeptide production has been demonstrated in vivo by cathepsin L gene knockout studies, cathepsin L gene expression in neuroendocrine cells, and notably, cathepsin L localization in neuropeptide-containing secretory vesicles. Cathepsin L is involved in producing opioid neuropeptides consisting of enkephalin, β-endorphin, and dynorphin, as well as in generating the POMC-derived peptide hormones ACTH and α-MSH. In addition, NPY, CCK, and catestatin neuropeptides utilize cathepsin L for their biosynthesis. The neuropeptide-synthesizing functions of cathepsin L represent its unique activity in secretory vesicles, which contrasts with its role in lysosomes. Interesting evaluations of protease gene knockout studies in mice that lack cathepsin L compared to those lacking PC1/3 and PC2 (PC, prohormone convertase) indicate the key role of cathepsin L in neuropeptide production. Therefore, dual cathepsin L and prohormone convertase protease pathways participate in neuropeptide production. Significantly, the recent new findings indicate cathepsin L as a novel 'proprotein convertase' for production of neuropeptides that mediate cell-cell communication in health and disease.
Collapse
Affiliation(s)
- Lydiane Funkelstein
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | | | | | | | | |
Collapse
|
29
|
Rholam M, Fahy C. Processing of peptide and hormone precursors at the dibasic cleavage sites. Cell Mol Life Sci 2009; 66:2075-91. [PMID: 19300906 PMCID: PMC11115611 DOI: 10.1007/s00018-009-0007-5] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2008] [Revised: 02/11/2009] [Accepted: 02/17/2009] [Indexed: 01/31/2023]
Abstract
Many functionally important cellular peptides and proteins, including hormones, neuropeptides, and growth factors, are synthesized as inactive precursor polypeptides, which require post-translational proteolytic processing to become biologically active polypeptides. This is achieved by the action of a relatively small number of proteases that belong to a family of seven subtilisin-like proprotein convertases (PCs) including furin. In view of this, this review focuses on the importance of privileged secondary structures and of given amino acid residues around basic cleavage sites in substrate recognition by these endoproteases. In addition to their participation in normal cell functions, PCs are crucial for the initiation and progress of many important diseases. Hence, these proteases constitute potential drug targets in medicine. Accordingly, this review also discusses the approaches used to shed light on the cleavage preference and the substrate specificity of the PCs, a prerequisite to select which PCs are promising drug targets in each disease.
Collapse
Affiliation(s)
- Mohamed Rholam
- Interfaces, Traitements, Organisation et Dynamique des Systrèmes, Université Paris Diderot (Paris 7), CNRS UMR 7086, Bâtiment Lavoisier, 15 rue Jean-Antoine de Baïf, 75205, Paris Cedex 13, France.
| | | |
Collapse
|
30
|
Kappert K, Meyborg H, Baumann B, Furundzija V, Kaufmann J, Graf K, Stibenz D, Fleck E, Stawowy P. Integrin cleavage facilitates cell surface-associated proteolysis required for vascular smooth muscle cell invasion. Int J Biochem Cell Biol 2009; 41:1511-7. [DOI: 10.1016/j.biocel.2009.01.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2008] [Revised: 12/22/2008] [Accepted: 01/05/2009] [Indexed: 11/16/2022]
|
31
|
Komiyama T, Coppola JM, Larsen MJ, van Dort ME, Ross BD, Day R, Rehemtulla A, Fuller RS. Inhibition of furin/proprotein convertase-catalyzed surface and intracellular processing by small molecules. J Biol Chem 2009; 284:15729-38. [PMID: 19332539 DOI: 10.1074/jbc.m901540200] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Furin is a ubiquitously expressed proprotein convertase (PC) that plays a vital role in numerous disease processes including cancer metastasis, bacterial toxin activation (e.g. anthrax and Pseudomonas), and viral propagation (e.g. avian influenza and human immunodeficiency virus). To identify small molecule inhibitors of furin and related processing enzymes, we performed high-throughput screens of chemical diversity libraries utilizing both enzyme-based and cell-based assays. The screens identified partially overlapping sets of compounds that were further characterized for affinity, mechanism, and efficacy in additional cellular processing assays. Dicoumarols were identified as a class of compounds that inhibited furin non-competitively and reversibly with Ki values in the micromolar range. These compounds inhibited furin/furin-like activity both at the cell surface (protecting against anthrax toxin) and in the secretory pathway (blocking processing of the metastasis factor membrane-type 1 matrix metalloproteinase/MT1-MMP) at concentrations close to Ki values. Compounds tested exhibited distinct patterns of inhibition of other furin-family PCs (rat PACE4, human PC5/6 and human PC7), showing that dicoumarol derivatives might be developed as either generic or selective inhibitors of the PCs. The extensive clinical use, high bioavailability and relatively low toxicity of dicoumarols suggests that the dicoumarol structure will be a good starting point for development of drug-like inhibitors of furin and other PCs that can act both intracellularly and at the cell surface.
Collapse
Affiliation(s)
- Tomoko Komiyama
- Departments of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan 48109-0606, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Wang S, Han J, Wang Y, Lu W, Chi C. Design of peptide inhibitors for furin based on the C-terminal fragment of histone H1.2. Acta Biochim Biophys Sin (Shanghai) 2008. [PMID: 18850049 PMCID: PMC7110110 DOI: 10.1111/j.1745-7270.2008.00470.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
The mammalian proprotein convertase furin has been found to play an important role in diverse physiological and pathological events, such as the activation of viral glycoproteins and bacterial exotoxins. Small, non‐toxic and highly active, furin inhibitors are considered to be attractive drug candidates for diseases caused by virus and bacteria. In this study, a series of peptide inhibitors were designed and synthesized based on the C‐terminal fragment of histone H1.2, which has an inhibitory effect on furin. Replacing the reactive site of inhibitors with the consensus substrate recognition sequence of furin has been found to increase inhibitory activity greatly. The most potent inhibitor, I4, with 14 amino acid residues has a Ki value of 17 nM for furin. Although most of the synthesized peptides were temporary inhibitors, the inhibitor I5, with nine amino acids, retained its full potency, even after a 3 h incubation period with furin at 37 °C. These inhibitors may potentially lead to the development of anti‐viral and antibacterial drug compounds.
Collapse
Affiliation(s)
- Suming Wang
- School of Life Sciences, University of Science and Technology of China, Anhui 230027, China
| | | | | | | | | |
Collapse
|
33
|
Chrétien M, Seidah NG, Basak A, Mbikay M. Proprotein convertases as therapeutic targets. Expert Opin Ther Targets 2008; 12:1289-300. [DOI: 10.1517/14728222.12.10.1289] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
34
|
Kaludercic N, Lindsey ML, Tavazzi B, Lazzarino G, Paolocci N. Inhibiting metalloproteases with PD 166793 in heart failure: impact on cardiac remodeling and beyond. Cardiovasc Ther 2008; 26:24-37. [PMID: 18466418 DOI: 10.1111/j.1527-3466.2007.00034.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Metalloproteinases (MMPs, also called matrixins) are extracellular proteolytic enzymes involved in the degradation of both matrix and nonmatrix proteins. Currently, 25 MMPs have been identified in humans, and the overexpression of one or more MMPs has been implicated in several pathologies, spanning from cancer to rheumathoid arthritis to cardiovascular disease. While research over the past 20 years has focused on understanding MMP biology and selectively inhibiting MMP activity, key issues that remain to be addressed include MMP roles in the context of normal versus pathological conditions and whether globally inhibiting MMPs improves or deteriorates overall organ function. In terms of cardiovascular disease, increased MMP expression has been demonstrated in the setting of myocardial ischemia, reperfusion injury, and during the progression to congestive heart failure. MMPs are also major contributors to the progression of atherosclerotic lesions. In this review, we focus on cardiovascular effects produced by PD 166793, a wide-broad spectrum MMP inhibitor, originally developed by Parke-Davis (now Pfizer). We will briefly review its structure, mechanism of action, and inhibitory capacity. Finally, we will illustrate the cardiac contexts, both in vivo and in vitro, in which PD166793 administration has proven beneficial.
Collapse
Affiliation(s)
- Nina Kaludercic
- Division of Cardiology, Johns Hopkins Medical Institutions, Baltimore, MD 21205, USA
| | | | | | | | | |
Collapse
|
35
|
Remacle AG, Shiryaev SA, Oh ES, Cieplak P, Srinivasan A, Wei G, Liddington RC, Ratnikov BI, Parent A, Desjardins R, Day R, Smith JW, Lebl M, Strongin AY. Substrate cleavage analysis of furin and related proprotein convertases. A comparative study. J Biol Chem 2008; 283:20897-906. [PMID: 18505722 DOI: 10.1074/jbc.m803762200] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
We present the data and the technology, a combination of which allows us to determine the identity of proprotein convertases (PCs) related to the processing of specific protein targets including viral and bacterial pathogens. Our results, which support and extend the data of other laboratories, are required for the design of effective inhibitors of PCs because, in general, an inhibitor design starts with a specific substrate. Seven proteinases of the human PC family cleave the multibasic motifs R-X-(R/K/X)-R downward arrow and, as a result, transform proproteins, including those from pathogens, into biologically active proteins and peptides. The precise cleavage preferences of PCs have not been known in sufficient detail; hence we were unable to determine the relative importance of the individual PCs in infectious diseases, thus making the design of specific inhibitors exceedingly difficult. To determine the cleavage preferences of PCs in more detail, we evaluated the relative efficiency of furin, PC2, PC4, PC5/6, PC7, and PACE4 in cleaving over 100 decapeptide sequences representing the R-X-(R/K/X)-R downward arrow motifs of human, bacterial, and viral proteins. Our computer analysis of the data and the follow-on cleavage analysis of the selected full-length proteins corroborated our initial results thus allowing us to determine the cleavage preferences of the PCs and to suggest which PCs are promising drug targets in infectious diseases. Our results also suggest that pathogens, including anthrax PA83 and the avian influenza A H5N1 (bird flu) hemagglutinin precursor, evolved to be as sensitive to PC proteolysis as the most sensitive normal human proteins.
Collapse
Affiliation(s)
- Albert G Remacle
- Burnham Institute for Medical Research, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Baker K, Raut P, Jass JR. Colorectal cancer cells express functional cell surface-bound TGFbeta. Int J Cancer 2008; 122:1695-700. [PMID: 18076044 DOI: 10.1002/ijc.23312] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Disruptions to the TGFbeta signaling pathway have been implicated in most human adenocarcinomas. In addition to its role in cancer cell migration and metastasis, TGFbeta has been implicated in tumor-mediated immunosuppression. Membrane-bound TGFbeta has previously been reported to be expressed on a subset of regulatory T cells and was shown to be critical to their immune suppressive function. In the present study, we document expression of a signaling competent, endogenously derived form of cell surface-bound TGFbeta on colorectal cancer cells. While antibodies against only the mature form of TGFbeta failed to label cells, surface-bound TGFbeta was clearly detected by antibodies specific for both the latent and mature forms of the cytokine. Confirming the notion that the surface TGFbeta was in latent form, brief acid pulsing of the cells increased the amount of detectable membrane-associated TGFbeta. In coculture assays, this cell-bound TGFbeta could be activated and utilized in a paracrine fashion both by other cancer cells and by CD8+ intraepithelial lymphocytes. This effect was abrogated by the use of a furin inhibitor which decreased the membranous expression of TGFbeta on the tumor cells. Signaling competent membrane-bound TGFbeta on cancer cells is thus likely to be a key player in regulating tumor cell interactions with each other as well as with other cells in their microenvironment.
Collapse
Affiliation(s)
- Kristi Baker
- Department of Pathology, McGill University, Montréal, Québec, Canada.
| | | | | |
Collapse
|
37
|
Hook V, Funkelstein L, Lu D, Bark S, Wegrzyn J, Hwang SR. Proteases for processing proneuropeptides into peptide neurotransmitters and hormones. Annu Rev Pharmacol Toxicol 2008; 48:393-423. [PMID: 18184105 PMCID: PMC2731677 DOI: 10.1146/annurev.pharmtox.48.113006.094812] [Citation(s) in RCA: 195] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Peptide neurotransmitters and peptide hormones, collectively known as neuropeptides, are required for cell-cell communication in neurotransmission and for regulation of endocrine functions. Neuropeptides are synthesized from protein precursors (termed proneuropeptides or prohormones) that require proteolytic processing primarily within secretory vesicles that store and secrete the mature neuropeptides to control target cellular and organ systems. This review describes interdisciplinary strategies that have elucidated two primary protease pathways for prohormone processing consisting of the cysteine protease pathway mediated by secretory vesicle cathepsin L and the well-known subtilisin-like proprotein convertase pathway that together support neuropeptide biosynthesis. Importantly, this review discusses important areas of current and future biomedical neuropeptide research with respect to biological regulation, inhibitors, structural features of proneuropeptide and protease interactions, and peptidomics combined with proteomics for systems biological approaches. Future studies that gain in-depth understanding of protease mechanisms for generating active neuropeptides will be instrumental for translational research to develop pharmacological strategies for regulation of neuropeptide functions. Pharmacological applications for neuropeptide research may provide valuable therapeutics in health and disease.
Collapse
Affiliation(s)
- Vivian Hook
- Skaggs School of Pharmacy and Pharmaceutical Sciences, and Department of Neuroscience, Pharmacology, and Medicine, School of Medicine, University of California-San Diego, La Jolla, CA 92093-0744, USA.
| | | | | | | | | | | |
Collapse
|
38
|
Peltonen TO, Taskinen P, Soini Y, Rysä J, Ronkainen J, Ohtonen P, Satta J, Juvonen T, Ruskoaho H, Leskinen H. Distinct downregulation of C-type natriuretic peptide system in human aortic valve stenosis. Circulation 2007; 116:1283-9. [PMID: 17709640 DOI: 10.1161/circulationaha.106.685743] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Aortic valve calcification is an actively regulated process that displays hallmarks of atherosclerosis. Natriuretic peptides (A-, B-, and C-type natriuretic peptides [ANP, BNP, and CNP]) have been reported to have a role in the pathogenesis of vascular atherosclerosis, but their expression in aortic valves is not known. Here, we characterized and compared expression of natriuretic peptide system in aortic valves of patients with normal valves (n=4), aortic regurgitation (n=11), regurgitation and fibrosis (n=6), and aortic valve stenosis (n=21). METHODS AND RESULTS By reverse-transcription polymerase chain reaction, all 3 natriuretic peptides were found to be expressed in aortic valves. CNP mRNA levels were 92% lower (P<0.001) in stenotic valves, whereas no significant changes in the expression of ANP and BNP genes were found compared with valves obtained from patients with aortic regurgitation. CNP was localized by immunohistochemistry with specific CNP (32-53) antibody to valvular endothelial cells and myofibroblasts. Gene expression of furin, which proteolytically cleaves proCNP into active CNP, was 54% lower in aortic valve stenosis (P=0.04). Moreover, natriuretic peptide receptor-A and natriuretic peptide receptor-B mRNA levels were 78% and 76% lower, respectively, in stenotic valves. In contrast, gene expression of corin, a proANP- and proBNP-converting enzyme, and natriuretic peptide receptor-C did not differ between groups. CONCLUSIONS We show that natriuretic peptides, their processing enzymes, and their receptors are expressed in human aortic valves. Aortic valve stenosis is characterized by distinct downregulation of gene expression of CNP, its processing enzyme furin, and the target receptors natriuretic peptide receptor-B and natriuretic peptide receptor-A, which suggests that CNP acts as a paracrine regulator of the aortic valve calcification process.
Collapse
Affiliation(s)
- Tuomas O Peltonen
- Department of Pharmacology and Toxicology, University of Oulu, PO Box 5000, 90014 Oulu, Finland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Seidah NG, Prat A. The proprotein convertases are potential targets in the treatment of dyslipidemia. J Mol Med (Berl) 2007; 85:685-96. [PMID: 17351764 DOI: 10.1007/s00109-007-0172-7] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2006] [Revised: 01/22/2007] [Accepted: 02/09/2007] [Indexed: 10/23/2022]
Abstract
The family of the secretory proprotein convertases (PCs) comprises seven basic amino acid (aa)-specific subtilisin-like serine proteinases known as PC1/3, PC2, furin, PC4, PC5/6, PACE4 and PC7, and two other PCs, SKI-1 (subtilisin-kexin isozyme-1)/S1P (site-1 protease) and PCSK9 (proprotein convertase subtilisin kexin 9) that cleave at nonbasic residues. Except for the testicular PC4, all the other convertases are expressed in brain and peripheral organs and play a critical role in various functions including the production of diverse neuropeptides as well as growth factors and receptors, the regulation of cellular adhesion/migration, cholesterol and fatty acid homeostasis, and growth/differentiation of progenitor cells. Some of these convertases process proteins that are implicated in pathologies, including cancer malignancies, tissue regeneration, and viral infections. The implication of some of these convertases in sterol/lipid metabolism has only recently been appreciated. SKI-1/S1P activates the synthesis of cholesterol and fatty acids as well as the LDL receptor (LDLR), whereas PCSK9 inactivates the LDLR. Moreover, furin, PC5 and/or, PACE4 inactivates endothelial and lipoprotein lipases. Humans and mice exhibiting either a gain or loss of function of PCSK9 through specific point mutations or knockouts develop hypercholesterolemia and hypocholesterolemia phenotypes, respectively. A PCSK9 inhibitor in combination with statins offers a most promising therapeutic target to treat cardiovascular disorders including dyslipidemias. Specific inhibitors/modulators of the other PCs should find novel therapeutic applications in the control of PC-regulated pathologies.
Collapse
Affiliation(s)
- Nabil G Seidah
- Laboratory of Biochemical Neuroendocrinology, Clinical Research Institute of Montreal, 110 Pine Ave West, Montreal, Quebec, H2W 1R7, Canada.
| | | |
Collapse
|
40
|
Han J, Gu J, Chi C. Possible role of histone H1 in the regulation of furin-dependent proprotein processing. Acta Biochim Biophys Sin (Shanghai) 2007; 39:173-80. [PMID: 17342255 PMCID: PMC7109942 DOI: 10.1111/j.1745-7270.2007.00267.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Histone H1 and its C-terminal lysine rich fragments were recently found to be potent inhibitors of furin, a mammalian proprotein convertase. However, its role in the regulation of furin-dependent proprotein processing remains unclear. Here we report that histone H1 efficiently blocks furin-dependent pro-von Willebrand factor (pro-vWF) processing in a dose-dependent manner. Coimmunoprecipitation and immunofluorescence studies confirmed that histone H1 could interact with furin, and the interaction mainly took place on the cell surface. We noted that histone H1 was released from cells undergoing necrosis and apoptosis induced by H2O2. Our findings suggested that histone H1 might be involved in extracellular and/or intracellular furin regulation.
Collapse
Affiliation(s)
- Jinbo Han
- Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Graduate School of the Chinese Academy of SciencesShanghai 200031, China
| | - Jianxin Gu
- Gene Research Center, Medical Center of Fudan UniversityShanghai 200032, China
| | - Chengwu Chi
- Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Graduate School of the Chinese Academy of SciencesShanghai 200031, China
- Institute of Protein Research, Tongji UniversityShanghai 200092, China
- Corresponding author: Tel, 86-21-54921165; Fax, 86-21-54921011; E-mail,
| |
Collapse
|
41
|
Fischoeder A, Meyborg H, Stibenz D, Fleck E, Graf K, Stawowy P. Insulin augments matrix metalloproteinase-9 expression in monocytes. Cardiovasc Res 2006; 73:841-8. [PMID: 17234168 DOI: 10.1016/j.cardiores.2006.12.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2006] [Revised: 12/05/2006] [Accepted: 12/06/2006] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVE Insulin resistance and hyperinsulinemia are major causes of cardiovascular morbidity and mortality. Matrix metalloproteinases (MMPs), highly expressed in activated mononuclear cells in vulnerable atherosclerotic lesions, are the main proteolytic enzymes controlling plaque stability. The aim of this study was to investigate the regulation of monocyte MMP-9 by insulin. METHODS AND RESULTS Stimulation of MMP-9 expression by insulin was time- and concentration-dependent in human monocytic THP-1 cells. Inhibition of insulin receptor (IR) maturation via inhibition of its activating convertase furin with the pharmacological furin-inhibitor decanoyl-RVKR-chloromethylketone, as well as blocking of IGF-1R function with a IGF-1R blocking antibody, demonstrated that insulin mediates increases in MMP-9 via IR activation. Inhibition of insulin's "metabolic" phosphatidylinositol 3-kinase signaling with wortmannin (50 nmol/L) or LY294002 (2.5 micromol/L) did not prevent insulin-dependent MMP-9 induction. In contrast inhibition of insulin's "mitogenic" Ras-Raf-mitogen-activated protein kinase-kinase pathways with PD98059 (15 micromol/L) or U0126 (2 micromol/L) inhibited insulin-induced MMP-9 gelatinolytic activity in THP-1 cells. Likewise, PD98059 inhibited insulin augmented MMP-9 levels in primary human monocytes, whereas wortmannin had no effect. CONCLUSION This study demonstrates that insulin can induce MMP-9 via mitogenic signaling pathways in monocytes, whereas phosphatidylinositol 3-kinase-dependent signaling, typically altered in insulin resistance, is not required. Induction of MMP-9 by insulin may potentially contribute to a pro-inflammatory state and the increased cardiovascular morbidity and mortality in type 2 diabetics.
Collapse
Affiliation(s)
- Arne Fischoeder
- Department of Medicine/Cardiology, Deutsches Herzzentrum Berlin, Augustenburger Platz 1, D-13353 Berlin, Germany
| | | | | | | | | | | |
Collapse
|
42
|
Chekanov AV, Remacle AG, Golubkov VS, Akatov VS, Sikora S, Savinov AY, Fugere M, Day R, Rozanov DV, Strongin AY. Both PA63 and PA83 are endocytosed within an anthrax protective antigen mixed heptamer: A putative mechanism to overcome a furin deficiency. Arch Biochem Biophys 2006; 446:52-9. [PMID: 16384550 DOI: 10.1016/j.abb.2005.11.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2005] [Revised: 11/17/2005] [Accepted: 11/18/2005] [Indexed: 11/20/2022]
Abstract
Anthrax toxin consists of protective antigen (PA), and lethal (LF) and edema (EF) factors. A 83 kDa PA monomer (PA83) precursor binds to the cell receptor. Furin-like proprotein convertases (PCs) cleave PA83 to generate cell-bound 63 kDa protein (PA63). PA63 oligomerizes to form a ring-shaped heptamer that binds LF-EF and facilitates their entry into the cells. Several additional PCs, as opposed to furin alone, are capable of processing PA83. Following the incomplete processing of the available pool of PA83, the functional heptamer includes both PA83 and PA63. The available structures of the receptor-PA complex imply that the presence of either one or two molecules of PA83 will not impose structural limitations on the formation of the heptamer and the association of either the (PA83)(1)(PA63)(6) or (PA83)(2)(PA63)(5) heteroheptamer with LF-EF. Our data point to the intriguing mechanism of anthrax that appears to facilitate entry of the toxin into the cells which express limiting amounts of PCs and an incompletely processed PA83 pool.
Collapse
Affiliation(s)
- Alexei V Chekanov
- Infectious and Inflammatory Disease Center, The Burnham Institute for Medical Research, La Jolla, CA 92037, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|