1
|
Shi W, Tang H, Tan S, Wang L, Yu Z, Gao S, Zhou J. MMA-induced LOXL2 + PSCs promote linear ECM alignment in the aging pancreas leading to pancreatic cancer progression. Cell Death Dis 2025; 16:419. [PMID: 40425551 PMCID: PMC12116754 DOI: 10.1038/s41419-025-07751-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 05/02/2025] [Accepted: 05/21/2025] [Indexed: 05/29/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an age-associated malignancy closely linked to the extracellular matrix (ECM). However, the impact of age-related ECM changes in the normal pancreas on PDAC progression remains unclear. Here, we find that increased linear ECM alignment in normal pancreatic tissues from aged PDAC patients is associated with PDAC progression and worse outcomes. Furthermore, serum methylmalonic acid (MMA) levels are elevated in aged PDAC patients and associated with increased linear ECM alignment in normal pancreatic tissues of PDAC patients. Functionally, MMA promotes LOXL2 expression in pancreatic stellate cells (PSCs), increases linear ECM alignment in normal pancreatic tissues, and facilitates tumor progression. Mechanistically, MMA upregulates KLF10, which forms a transcriptional complex with SP1 to enhance LOXL2 expression in PSCs. Our study demonstrates the role of MMA-induced LOXL2+PSCs in ECM remodeling, thus serving as a potential therapeutic target to mitigate PDAC progression in aged patients. Schematic diagram showing the molecular mechanism by which MMA-induced LOXL+PSCs promote PDAC progression in the aging pancreas. In aged individuals, elevated levels of MMA in the blood induce the activation of the KLF10/SP1‒LOXL2 axis in PSCs to increase linear ECM alignment. Following the initiation of pancreatic cancer, this increased linear ECM alignment leads to increased tumor invasion into surrounding tissues, resulting in a greater proportion of stage T3/T4 tumors and a greater incidence of LVI and PNI in aged patients, ultimately leading to poorer outcomes (This schematic was created with www.figdraw.com ,export id: PRPRS4e268).
Collapse
Affiliation(s)
- Wenyuan Shi
- Department of Hepato-Pancreatico-Biliary Surgery, Zhongda Hospital Southeast University, Nanjing, China
- Department of Surgery, School of Medicine, Southeast University, Nanjing, China
| | - Haodong Tang
- Department of Hepato-Pancreatico-Biliary Surgery, Zhongda Hospital Southeast University, Nanjing, China
- Department of Surgery, School of Medicine, Southeast University, Nanjing, China
| | - Siyuan Tan
- Department of Hepato-Pancreatico-Biliary Surgery, Zhongda Hospital Southeast University, Nanjing, China
- Department of Surgery, School of Medicine, Southeast University, Nanjing, China
| | - Lishan Wang
- Department of Hepato-Pancreatico-Biliary Surgery, Zhongda Hospital Southeast University, Nanjing, China
- Department of Surgery, School of Medicine, Southeast University, Nanjing, China
| | - Zeqian Yu
- Department of Hepato-Pancreatico-Biliary Surgery, Zhongda Hospital Southeast University, Nanjing, China
- Department of Surgery, School of Medicine, Southeast University, Nanjing, China
| | - Shan Gao
- Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing, China
| | - Jiahua Zhou
- Department of Hepato-Pancreatico-Biliary Surgery, Zhongda Hospital Southeast University, Nanjing, China.
- Department of Surgery, School of Medicine, Southeast University, Nanjing, China.
| |
Collapse
|
2
|
Passang T, Wang S, Zhang H, Zeng F, Hsu PC, Wang W, Li JM, Liu Y, Ravindranathan S, Lesinski GB, Waller EK. VPAC2 Receptor Signaling Promotes Growth and Immunosuppression in Pancreatic Cancer. Cancer Res 2024; 84:2954-2967. [PMID: 38809694 PMCID: PMC11458156 DOI: 10.1158/0008-5472.can-23-3628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 03/29/2024] [Accepted: 05/20/2024] [Indexed: 05/31/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) harbors a complex tumor microenvironment, and cross-talk among cells in the tumor microenvironment can contribute to drug resistance and relapse. Vasoactive intestinal peptide (VIP) is overexpressed in PDAC, and VIP receptors expressed on T cells are a targetable pathway that sensitizes PDAC to immunotherapy. In this study, we showed that pancreatic cancer cells engage in autocrine VIP signaling through VIP receptor 2 (VPAC2). High coexpression of VIP with VPAC2 correlated with reduced relapse-free survival in patients with PDAC. VPAC2 activation in PDAC cells upregulated Piwi-like RNA-mediated gene silencing 2, which stimulated cancer cell clonogenic growth. In addition, VPAC2 signaling increased expression of TGFβ1 to inhibit T-cell function. Loss of VPAC2 on PDAC cells led to reduced tumor growth and increased sensitivity to anti-PD-1 immunotherapy in mouse models of PDAC. Overall, these findings expand our understanding of the role of VIP/VPAC2 signaling in PDAC and provide the rationale for developing potent VPAC2-specific antagonists for treating patients with PDAC. Significance: Autocrine VIP signaling via VPAC2 promotes cancer cell growth and inhibits T-cell function in pancreatic ductal adenocarcinoma, highlighting its potential as a therapeutic target to improve pancreatic cancer treatment.
Collapse
Affiliation(s)
- Tenzin Passang
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Shuhua Wang
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Hanwen Zhang
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Fanyuan Zeng
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Po-Chih Hsu
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Wenxi Wang
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Jian Ming Li
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Yuan Liu
- Winship Cancer Institute Emory University, Atlanta, GA, USA
| | - Sruthi Ravindranathan
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Gregory B. Lesinski
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Winship Cancer Institute Emory University, Atlanta, GA, USA
| | - Edmund K. Waller
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Winship Cancer Institute Emory University, Atlanta, GA, USA
| |
Collapse
|
3
|
Li N, Xue Y, Zhu C, Chen N, Qi M, Fang M, Huang S. The zinc-finger transcription factor KLF6 regulates cardiac fibrosis. Life Sci 2024; 351:122805. [PMID: 38851422 DOI: 10.1016/j.lfs.2024.122805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 05/09/2024] [Accepted: 06/04/2024] [Indexed: 06/10/2024]
Abstract
AIMS Heart failure (HF) is one of the most devastating consequences of cardiovascular diseases. Regardless of etiology, cardiac fibrosis is present and promotes the loss of heart function in HF patients. Cardiac resident fibroblasts, in response to a host of pro-fibrogenic stimuli, trans-differentiate into myofibroblasts to mediate cardiac fibrosis, the underlying mechanism of which remains incompletely understood. METHODS Fibroblast-myofibroblast transition was induced in vitro by exposure to transforming growth factor (TGF-β). Cardiac fibrosis was induced in mice by either transverse aortic constriction (TAC) or by chronic infusion with angiotensin II (Ang II). RESULTS Through bioinformatic screening, we identified Kruppel-like factor 6 (KLF6) as a transcription factor preferentially up-regulated in cardiac fibroblasts from individuals with non-ischemic cardiomyopathy (NICM) compared to the healthy donors. Further analysis showed that nuclear factor kappa B (NF-κB) bound to the KLF6 promoter and mediated KLF6 trans-activation by pro-fibrogenic stimuli. KLF6 knockdown attenuated whereas KLF6 over-expression enhanced TGF-β induced fibroblast-myofibroblast transition in vitro. More importantly, myofibroblast-specific KLF6 depletion ameliorated cardiac fibrosis and rescued heart function in mice subjected to the TAC procedure or chronic Ang II infusion. SIGNIFICANCE In conclusion, our data support a role for KLF6 in cardiac fibrosis.
Collapse
Affiliation(s)
- Nan Li
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Human Anatomy, Nanjing Medical University, Nanjing, China
| | - Yujia Xue
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Human Anatomy, Nanjing Medical University, Nanjing, China
| | - Chenghao Zhu
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Naxia Chen
- Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research and Key Laboratory of Emergency and Trauma of Ministry of Education, Institute of Cardiovascular Research, Department of Cardiology, the First Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Mengwen Qi
- Center for Experimental Medicine, Jiangsu Health Vocational College, Nanjing, China
| | - Mingming Fang
- Center for Experimental Medicine, Jiangsu Health Vocational College, Nanjing, China.
| | - Shan Huang
- Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research and Key Laboratory of Emergency and Trauma of Ministry of Education, Institute of Cardiovascular Research, Department of Cardiology, the First Affiliated Hospital, Hainan Medical University, Haikou, China.
| |
Collapse
|
4
|
Eni-Aganga I, Lanaghan ZM, Ismail F, Korolkova O, Goodwin JS, Balasubramaniam M, Dash C, Pandhare J. KLF6 activates Sp1-mediated prolidase transcription during TGF-β 1 signaling. J Biol Chem 2024; 300:105605. [PMID: 38159857 PMCID: PMC10847167 DOI: 10.1016/j.jbc.2023.105605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 12/09/2023] [Accepted: 12/18/2023] [Indexed: 01/03/2024] Open
Abstract
Prolidase (PEPD) is the only hydrolase that cleaves the dipeptides containing C-terminal proline or hydroxyproline-the rate-limiting step in collagen biosynthesis. However, the molecular regulation of prolidase expression remains largely unknown. In this study, we have identified overlapping binding sites for the transcription factors Krüppel-like factor 6 (KLF6) and Specificity protein 1 (Sp1) in the PEPD promoter and demonstrate that KLF6/Sp1 transcriptionally regulate prolidase expression. By cloning the PEPD promoter into a luciferase reporter and through site-directed deletion, we pinpointed the minimal sequences required for KLF6 and Sp1-mediated PEPD promoter-driven transcription. Interestingly, Sp1 inhibition abrogated KLF6-mediated PEPD promoter activity, suggesting that Sp1 is required for the basal expression of prolidase. We further studied the regulation of PEPD by KLF6 and Sp1 during transforming growth factor β1 (TGF-β1) signaling, since both KLF6 and Sp1 are key players in TGF-β1 mediated collagen biosynthesis. Mouse and human fibroblasts exposed to TGF-β1 resulted in the induction of PEPD transcription and prolidase expression. Inhibition of TGF-β1 signaling abrogated PEPD promoter-driven transcriptional activity of KLF6 and Sp1. Knock-down of KLF6 as well as Sp1 inhibition also reduced prolidase expression. Chromatin immunoprecipitation assay supported direct binding of KLF6 and Sp1 to the PEPD promoter and this binding was enriched by TGF-β1 treatment. Finally, immunofluorescence studies showed that KLF6 co-operates with Sp1 in the nucleus to activate prolidase expression and enhance collagen biosynthesis. Collectively, our results identify functional elements of the PEPD promoter for KLF6 and Sp1-mediated transcriptional activation and describe the molecular mechanism of prolidase expression.
Collapse
Affiliation(s)
- Ireti Eni-Aganga
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, Tennessee, USA; School of Graduate Studies, Meharry Medical College, Nashville, Tennessee, USA; Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, Tennessee, USA
| | - Zeljka Miletic Lanaghan
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, Tennessee, USA; Neuroscience Graduate Program, Vanderbilt University, Nashville, Tennessee, USA
| | - Farah Ismail
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, Tennessee, USA
| | - Olga Korolkova
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, Tennessee, USA; Department of Biochemistry, Cancer Biology, Pharmacology and Neuroscience, Meharry Medical College, Nashville, Tennessee, USA
| | - Jeffery Shawn Goodwin
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, Tennessee, USA; Department of Biochemistry, Cancer Biology, Pharmacology and Neuroscience, Meharry Medical College, Nashville, Tennessee, USA
| | - Muthukumar Balasubramaniam
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, Tennessee, USA; Department of Biochemistry, Cancer Biology, Pharmacology and Neuroscience, Meharry Medical College, Nashville, Tennessee, USA
| | - Chandravanu Dash
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, Tennessee, USA; Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, Tennessee, USA; Department of Biochemistry, Cancer Biology, Pharmacology and Neuroscience, Meharry Medical College, Nashville, Tennessee, USA.
| | - Jui Pandhare
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, Tennessee, USA; School of Graduate Studies, Meharry Medical College, Nashville, Tennessee, USA; Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, Tennessee, USA.
| |
Collapse
|
5
|
Alve S, Gramolelli S, Jukonen J, Juteau S, Pink A, Manninen AA, Hänninen S, Monto E, Lackman MH, Carpén O, Saharinen P, Karaman S, Vaahtomeri K, Ojala PM. DLL4/Notch3/WNT5B axis mediates bidirectional prometastatic crosstalk between melanoma and lymphatic endothelial cells. JCI Insight 2024; 9:e171821. [PMID: 37971882 PMCID: PMC10906450 DOI: 10.1172/jci.insight.171821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 11/15/2023] [Indexed: 11/19/2023] Open
Abstract
Despite strong indications that interactions between melanoma and lymphatic vessels actively promote melanoma progression, the molecular mechanisms are not yet completely understood. To characterize molecular factors of this crosstalk, we established human primary lymphatic endothelial cell (LEC) cocultures with human melanoma cell lines. Here, we show that coculture with melanoma cells induced transcriptomic changes in LECs and led to multiple changes in their function. WNT5B, a paracrine signaling molecule upregulated in melanoma cells upon LEC interaction, was found to contribute to the functional changes in LECs. Moreover, WNT5B transcription was regulated by Notch3 in melanoma cells following the coculture with LECs, and Notch3 and WNT5B were coexpressed in melanoma patient primary tumor and metastasis samples. Moreover, melanoma cells derived from LEC coculture escaped efficiently from the primary site to the proximal tumor-draining lymph nodes, which was impaired upon WNT5B depletion. This supported the role of WNT5B in promoting the metastatic potential of melanoma cells through its effects on LECs. Finally, DLL4, a Notch ligand expressed in LECs, was identified as an upstream inducer of the Notch3/WNT5B axis in melanoma. This study elucidated WNT5B as a key molecular factor mediating bidirectional crosstalk between melanoma cells and lymphatic endothelium and promoting melanoma metastasis.
Collapse
Affiliation(s)
- Sanni Alve
- Translational Cancer Medicine Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Silvia Gramolelli
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Joonas Jukonen
- Translational Cancer Medicine Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Susanna Juteau
- Department of Pathology, Helsinki University Hospital (HUS), University of Helsinki, Helsinki, Finland
| | - Anne Pink
- Translational Cancer Medicine Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Atte A. Manninen
- Department of Plastic Surgery, Park Hospital, Helsinki University Hospital (HUS), and
| | - Satu Hänninen
- Department of Pathology, Helsinki University Hospital (HUS), University of Helsinki, Helsinki, Finland
| | - Elisa Monto
- Translational Cancer Medicine Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Madeleine H. Lackman
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Olli Carpén
- Helsinki Biobank, and
- Department of Pathology and Research Program in Systems Oncology, University of Helsinki, HUS Diagnostic Center, Helsinki University Hospital, Finland
| | - Pipsa Saharinen
- Translational Cancer Medicine Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Wihuri Research Institute, Biomedicum, Helsinki, Finland
- Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Sinem Karaman
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Wihuri Research Institute, Biomedicum, Helsinki, Finland
| | - Kari Vaahtomeri
- Translational Cancer Medicine Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Wihuri Research Institute, Biomedicum, Helsinki, Finland
| | - Päivi M. Ojala
- Translational Cancer Medicine Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Pathology, Helsinki University Hospital (HUS), University of Helsinki, Helsinki, Finland
| |
Collapse
|
6
|
Liu Y, Han B, Zheng W, Peng P, Yang C, Jiang G, Ma Y, Li J, Ni J, Sun D. Identification of genetic associations and functional SNPs of bovine KLF6 gene on milk production traits in Chinese holstein. BMC Genom Data 2023; 24:72. [PMID: 38017423 PMCID: PMC10685595 DOI: 10.1186/s12863-023-01175-w] [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: 05/23/2023] [Accepted: 11/13/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Our previous research identified the Kruppel like factor 6 (KLF6) gene as a prospective candidate for milk production traits in dairy cattle. The expression of KLF6 in the livers of Holstein cows during the peak of lactation was significantly higher than that during the dry and early lactation periods. Notably, it plays an essential role in activating peroxisome proliferator-activated receptor α (PPARα) signaling pathways. The primary aim of this study was to further substantiate whether the KLF6 gene has significant genetic effects on milk traits in dairy cattle. RESULTS Through direct sequencing of PCR products with pooled DNA, we totally identified 12 single nucleotide polymorphisms (SNPs) within the KLF6 gene. The set of SNPs encompasses 7 located in 5' flanking region, 2 located in exon 2 and 3 located in 3' untranslated region (UTR). Of these, the g.44601035G > A is a missense mutation that resulting in the replacement of arginine (CGG) with glutamine (CAG), consequently leading to alterations in the secondary structure of the KLF6 protein, as predicted by SOPMA. The remaining 7 regulatory SNPs significantly impacted the transcriptional activity of KLF6 following mutation (P < 0.005), manifesting as changes in transcription factor binding sites. Additionally, 4 SNPs located in both the UTR and exons were predicted to influence the secondary structure of KLF6 mRNA using the RNAfold web server. Furthermore, we performed the genotype-phenotype association analysis using SAS 9.2 which found all the 12 SNPs were significantly correlated to milk yield, fat yield, fat percentage, protein yield and protein percentage within both the first and second lactations (P < 0.0001 ~ 0.0441). Also, with Haploview 4.2 software, we found the 12 SNPs linked closely and formed a haplotype block, which was strongly associated with five milk traits (P < 0.0001 ~ 0.0203). CONCLUSIONS In summary, our study represented the KLF6 gene has significant impacts on milk yield and composition traits in dairy cattle. Among the identified SNPs, 7 were implicated in modulating milk traits by impacting transcriptional activity, 4 by altering mRNA secondary structure, and 1 by affecting the protein secondary structure of KLF6. These findings provided valuable molecular insights for genomic selection program of dairy cattle.
Collapse
Affiliation(s)
- Yanan Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, National Engineering Laboratory for Animal Breeding, Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, China Agricultural University, No. 2 Yuanmingyuan West Road, Haidian District, Beijing, 100193, China
| | - Bo Han
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, National Engineering Laboratory for Animal Breeding, Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, China Agricultural University, No. 2 Yuanmingyuan West Road, Haidian District, Beijing, 100193, China
| | - Weijie Zheng
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, National Engineering Laboratory for Animal Breeding, Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, China Agricultural University, No. 2 Yuanmingyuan West Road, Haidian District, Beijing, 100193, China
| | - Peng Peng
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, National Engineering Laboratory for Animal Breeding, Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, China Agricultural University, No. 2 Yuanmingyuan West Road, Haidian District, Beijing, 100193, China
| | - Chendong Yang
- Hebei Province Animal Husbandry and Fine Breeds Work Station, No. 7 Xuefu Road, Changan District, Shijiazhuang, 050000, China
| | - Guie Jiang
- Hebei Province Animal Husbandry and Fine Breeds Work Station, No. 7 Xuefu Road, Changan District, Shijiazhuang, 050000, China
| | - Yabin Ma
- Hebei Province Animal Husbandry and Fine Breeds Work Station, No. 7 Xuefu Road, Changan District, Shijiazhuang, 050000, China
| | - Jianming Li
- Hebei Province Animal Husbandry and Fine Breeds Work Station, No. 7 Xuefu Road, Changan District, Shijiazhuang, 050000, China
| | - Junqing Ni
- Hebei Province Animal Husbandry and Fine Breeds Work Station, No. 7 Xuefu Road, Changan District, Shijiazhuang, 050000, China.
| | - Dongxiao Sun
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, National Engineering Laboratory for Animal Breeding, Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, China Agricultural University, No. 2 Yuanmingyuan West Road, Haidian District, Beijing, 100193, China.
| |
Collapse
|
7
|
Wang Q, Chang Y, Yang X, Han Z. Deep sequencing of circulating miRNAs and target mRNAs level in deep venous thrombosis patients. IET Syst Biol 2023; 17:212-227. [PMID: 37466160 PMCID: PMC10439493 DOI: 10.1049/syb2.12071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 06/15/2023] [Accepted: 06/26/2023] [Indexed: 07/20/2023] Open
Abstract
Deep venous thrombosis is one of the most common peripheral vascular diseases that lead to major morbidity and mortality. The authors aimed to identify potential differentially expressed miRNAs and target mRNAs, which were helpful in understanding the potential molecule mechanism of deep venous thrombosis. The plasma samples of patients with deep venous thrombosis were obtained for the RNA sequencing. Differentially expressed miRNAs were identified, followed by miRNA-mRNA target analysis. Enrichment analysis was used to analyze the potential biological function of target mRNAs. GSE19151 and GSE173461 datasets were used for expression validation of mRNAs and miRNAs. 131 target mRNAs of 21 differentially expressed miRNAs were identified. Among which, 8 differentially expressed miRNAs including hsa-miR-150-5p, hsa-miR-326, hsa-miR-144-3p, hsa-miR-199a-5p, hsa-miR-199b-5p, hsa-miR-125a-5p, hsa-let-7e-5p and hsa-miR-381-3p and their target mRNAs (PRKCA, SP1, TP53, SLC27A4, PDE1B, EPHB3, IRS1, HIF1A, MTUS1 and ZNF652) were found associated with deep venous thrombosis for the first time. Interestingly, PDE1B and IRS1 had a potential diagnostic value for patients. Additionally, 3 important signaling pathways including p53, PI3K-Akt and MAPK were identified in the enrichment analysis of target mRNAs (TP53, PRKCA and IRS1). Identified circulating miRNAs and target mRNAs and related signaling pathways may be involved in the process of deep venous thrombosis.
Collapse
Affiliation(s)
- Qingxian Wang
- Department of Orthopedic Trauma, Orthopedic Research Institution of Hebei ProvinceKey Labratory of Biomechanics of Hebei ProvinceThe Third Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Yunhe Chang
- Department of Orthopedic Trauma, Orthopedic Research Institution of Hebei ProvinceKey Labratory of Biomechanics of Hebei ProvinceThe Third Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Xuqing Yang
- Department of Orthopedic Trauma, Orthopedic Research Institution of Hebei ProvinceKey Labratory of Biomechanics of Hebei ProvinceThe Third Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Ziwang Han
- Department of Orthopedic Trauma, Orthopedic Research Institution of Hebei ProvinceKey Labratory of Biomechanics of Hebei ProvinceThe Third Hospital of Hebei Medical UniversityShijiazhuangChina
| |
Collapse
|
8
|
Parab S, Setten E, Astanina E, Bussolino F, Doronzo G. The tissue-specific transcriptional landscape underlines the involvement of endothelial cells in health and disease. Pharmacol Ther 2023; 246:108418. [PMID: 37088448 DOI: 10.1016/j.pharmthera.2023.108418] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 03/23/2023] [Accepted: 04/17/2023] [Indexed: 04/25/2023]
Abstract
Endothelial cells (ECs) that line vascular and lymphatic vessels are being increasingly recognized as important to organ function in health and disease. ECs participate not only in the trafficking of gases, metabolites, and cells between the bloodstream and tissues but also in the angiocrine-based induction of heterogeneous parenchymal cells, which are unique to their specific tissue functions. The molecular mechanisms regulating EC heterogeneity between and within different tissues are modeled during embryogenesis and become fully established in adults. Any changes in adult tissue homeostasis induced by aging, stress conditions, and various noxae may reshape EC heterogeneity and induce specific transcriptional features that condition a functional phenotype. Heterogeneity is sustained via specific genetic programs organized through the combinatory effects of a discrete number of transcription factors (TFs) that, at the single tissue-level, constitute dynamic networks that are post-transcriptionally and epigenetically regulated. This review is focused on outlining the TF-based networks involved in EC specialization and physiological and pathological stressors thought to modify their architecture.
Collapse
Affiliation(s)
- Sushant Parab
- Department of Oncology, University of Torino, IT, Italy; Candiolo Cancer Institute-IRCCS-FPO, Candiolo, Torino, IT, Italy
| | - Elisa Setten
- Department of Oncology, University of Torino, IT, Italy; Candiolo Cancer Institute-IRCCS-FPO, Candiolo, Torino, IT, Italy
| | - Elena Astanina
- Candiolo Cancer Institute-IRCCS-FPO, Candiolo, Torino, IT, Italy
| | - Federico Bussolino
- Department of Oncology, University of Torino, IT, Italy; Candiolo Cancer Institute-IRCCS-FPO, Candiolo, Torino, IT, Italy.
| | - Gabriella Doronzo
- Department of Oncology, University of Torino, IT, Italy; Candiolo Cancer Institute-IRCCS-FPO, Candiolo, Torino, IT, Italy
| |
Collapse
|
9
|
Thapa P, Jiang H, Ding N, Hao Y, Alshahrani A, Lee EY, Fujii J, Wei Q. Loss of Peroxiredoxin IV Protects Mice from Azoxymethane/Dextran Sulfate Sodium-Induced Colorectal Cancer Development. Antioxidants (Basel) 2023; 12:677. [PMID: 36978925 PMCID: PMC10045277 DOI: 10.3390/antiox12030677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/20/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Peroxiredoxin IV (Prx4), a typical two-cysteine-containing member of the peroxidase family, functions as an antioxidant to maintain cellular redox homeostasis through the reduction of reactive oxygen species (ROS) via cycles of oxidation-reduction reactions. Under oxidative stress, all Prxs including Prx4 are inactivated as their catalytic cysteines undergo hyperoxidation, and hyperoxidized two-cysteine Prxs can be exclusively repaired and revitalized through the reduction cycle catalyzed by sulfiredoxin (Srx). Previously, we showed that Prx4 is a preferred substrate of Srx, and knockout of Srx in mice leads to resistance to azoxymethane/dextran sulfate sodium (AOM/DSS)-induced colon carcinogenesis. To further understand the significance of the Srx/Prx4 axis in colorectal cancer development, Prx4-/- mice were established and subjected to standard AOM/DSS protocol. Compared with wildtype littermates, mice with Prx4-/- genotype had significantly fewer and smaller tumors. Histopathological analysis revealed that loss of Prx4 leads to increased cell death through lipid peroxidation and lower infiltration of inflammatory cells in the knockout tumors compared to wildtype. Treatment with DSS alone also showed decreased infiltration of macrophages and lymphocytes in the colon of knockout mice, suggesting a role for Prx4 in inflammatory response. In addition, loss of Prx4 caused alterations in plasma cytokines and chemokines after DSS and AOM/DSS treatments. These findings suggest that loss of Prx4 protects mice from AOM/DSS-induced colon tumorigenesis. Thus, targeting Prx4 may provide novel strategies for colon cancer prevention and treatment.
Collapse
Affiliation(s)
- Pratik Thapa
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, KY 40506, USA
| | - Hong Jiang
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, KY 40506, USA
| | - Na Ding
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, KY 40506, USA
| | - Yanning Hao
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, KY 40506, USA
| | - Aziza Alshahrani
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, KY 40506, USA
| | - Eun Y. Lee
- Department of Pathology and Laboratory Medicine, University of Kentucky College of Medicine, Lexington, KY 40506, USA
| | - Junichi Fujii
- Department of Biomolecular Function, Yamagata University, Yamagata 990-9585, Japan
| | - Qiou Wei
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, KY 40506, USA
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| |
Collapse
|
10
|
Ximerakis M, Holton KM, Giadone RM, Ozek C, Saxena M, Santiago S, Adiconis X, Dionne D, Nguyen L, Shah KM, Goldstein JM, Gasperini C, Gampierakis IA, Lipnick SL, Simmons SK, Buchanan SM, Wagers AJ, Regev A, Levin JZ, Rubin LL. Heterochronic parabiosis reprograms the mouse brain transcriptome by shifting aging signatures in multiple cell types. NATURE AGING 2023; 3:327-345. [PMID: 37118429 PMCID: PMC10154248 DOI: 10.1038/s43587-023-00373-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 01/30/2023] [Indexed: 04/30/2023]
Abstract
Aging is a complex process involving transcriptomic changes associated with deterioration across multiple tissues and organs, including the brain. Recent studies using heterochronic parabiosis have shown that various aspects of aging-associated decline are modifiable or even reversible. To better understand how this occurs, we performed single-cell transcriptomic profiling of young and old mouse brains after parabiosis. For each cell type, we cataloged alterations in gene expression, molecular pathways, transcriptional networks, ligand-receptor interactions and senescence status. Our analyses identified gene signatures, demonstrating that heterochronic parabiosis regulates several hallmarks of aging in a cell-type-specific manner. Brain endothelial cells were found to be especially malleable to this intervention, exhibiting dynamic transcriptional changes that affect vascular structure and function. These findings suggest new strategies for slowing deterioration and driving regeneration in the aging brain through approaches that do not rely on disease-specific mechanisms or actions of individual circulating factors.
Collapse
Affiliation(s)
- Methodios Ximerakis
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Kristina M Holton
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Richard M Giadone
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Ceren Ozek
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Monika Saxena
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Samara Santiago
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Xian Adiconis
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Danielle Dionne
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Lan Nguyen
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kavya M Shah
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Jill M Goldstein
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Caterina Gasperini
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Ioannis A Gampierakis
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Scott L Lipnick
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sean K Simmons
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sean M Buchanan
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Amy J Wagers
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Joslin Diabetes Center, Boston, MA, USA
- Paul F. Glenn Center for the Biology of Aging, Harvard Medical School, Boston, MA, USA
| | - Aviv Regev
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Howard Hughes Medical Institute, Koch Institute of Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Joshua Z Levin
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Lee L Rubin
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
11
|
Samak M, Kues A, Kaltenborn D, Klösener L, Mietsch M, Germena G, Hinkel R. Dysregulation of Krüppel-like Factor 2 and Myocyte Enhancer Factor 2D Drive Cardiac Microvascular Inflammation and Dysfunction in Diabetes. Int J Mol Sci 2023; 24:2482. [PMID: 36768805 PMCID: PMC9916909 DOI: 10.3390/ijms24032482] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 02/03/2023] Open
Abstract
Cardiovascular complications are the main cause of morbidity and mortality from diabetes. Herein, vascular inflammation is a major pathological manifestation. We previously characterized the cardiac microvascular inflammatory phenotype in diabetic patients and highlighted micro-RNA 92a (miR-92a) as a driver of endothelial dysfunction. In this article, we further dissect the molecular underlying of these findings by addressing anti-inflammatory Krüppel-like factors 2 and 4 (KLF2 and KLF4). We show that KLF2 dysregulation in diabetes correlates with greater monocyte adhesion as well as migratory defects in cardiac microvascular endothelial cells. We also describe, for the first time, a role for myocyte enhancer factor 2D (MEF2D) in cardiac microvascular dysfunction in diabetes. We show that both KLFs 2 and 4, as well as MEF2D, are dysregulated in human and porcine models of diabetes. Furthermore, we prove a direct interaction between miR-92a and all three targets. Altogether, our data strongly qualify miR-92a as a potential therapeutic target for diabetes-associated cardiovascular disease.
Collapse
Affiliation(s)
- Mostafa Samak
- Laboratory Animal Science Unit, Leibniz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Kellnerweg 4, 37077 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Andreas Kues
- Laboratory Animal Science Unit, Leibniz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Kellnerweg 4, 37077 Göttingen, Germany
| | - Diana Kaltenborn
- Laboratory Animal Science Unit, Leibniz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Kellnerweg 4, 37077 Göttingen, Germany
| | - Lina Klösener
- Laboratory Animal Science Unit, Leibniz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Kellnerweg 4, 37077 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
- Institute for Animal Hygiene, Animal Welfare and Farm Animal Behaviour, University of Veterinary Medicine, 30173 Hannover, Germany
| | - Matthias Mietsch
- Laboratory Animal Science Unit, Leibniz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Kellnerweg 4, 37077 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Giulia Germena
- Laboratory Animal Science Unit, Leibniz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Kellnerweg 4, 37077 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Rabea Hinkel
- Laboratory Animal Science Unit, Leibniz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Kellnerweg 4, 37077 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
- Institute for Animal Hygiene, Animal Welfare and Farm Animal Behaviour, University of Veterinary Medicine, 30173 Hannover, Germany
| |
Collapse
|
12
|
Tripska K, Igreja Sá IC, Vasinova M, Vicen M, Havelek R, Eissazadeh S, Svobodova Z, Vitverova B, Theuer C, Bernabeu C, Nachtigal P. Monoclonal anti-endoglin antibody TRC105 (carotuximab) prevents hypercholesterolemia and hyperglycemia-induced endothelial dysfunction in human aortic endothelial cells. Front Med (Lausanne) 2022; 9:845918. [PMID: 36160139 PMCID: PMC9490272 DOI: 10.3389/fmed.2022.845918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
Endoglin (Eng) is a co-receptor of the transforming growth factor β superfamily playing an important role in endothelial dysfunction. TRC105 (carotuximab) is a monoclonal antibody that blocks Eng and its downstream Smad signaling pathway. Here we have investigated for the first time the effects of TRC105 treatment on the development of endothelial dysfunction induced by 7-ketocholesterol (7K) or high glucose (HG), focusing on Eng expression, signaling, and function. In the hypercholesterolemia study, human aortic endothelial cells (HAoECs) were treated with TRC105 (300 μg/ml) for 1 h, followed by the addition of 7K (10 μg/ml) for another 12 h. In the hyperglycemia study, HAoECs were exposed to HG (45 mM) for 60 h, followed by the addition of TRC105 for another 12 h, and cells treated with 5mM glucose and 40 mM mannitol served as control. Protein levels, adhesion, and transmigration of monocytes were assessed by flow cytometry, mRNA expression was measured by qRT-PCR. 7K and HG treatment increased protein levels of NF-κB and Eng and adhesion and transmigration of monocytes through HAoECs monolayer. TRC105 pretreatment reduced the 7K- or HG-induced Eng protein levels and pSmad1/5 and pSmad2/3 signaling. Despite increased protein levels of P-selectin and VCAM-1, TRC105 mediated blockage of Eng prevented 7K- and HG-induced adhesion and transmigration of monocytes through endothelial monolayers. These results suggest that TRC105-mediated Eng blockage can counteract the hypercholesterolemia- and hyperglycemia-induced endothelial dysfunction in HAoECs, suggesting that Eng might be a potential therapeutic target in disorders associated with elevated cholesterol and glucose levels.
Collapse
Affiliation(s)
- Katarina Tripska
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Ivone Cristina Igreja Sá
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Martina Vasinova
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Matej Vicen
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Radim Havelek
- Department of Medical Biochemistry, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Samira Eissazadeh
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Zuzana Svobodova
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Barbora Vitverova
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Charles Theuer
- Tracon Pharmaceuticals, Inc., San Diego, CA, United States
| | - Carmelo Bernabeu
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Petr Nachtigal
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czechia
- *Correspondence: Petr Nachtigal,
| |
Collapse
|
13
|
Bioinformatics and Experimental Analyses Reveal NFIC as an Upstream Transcriptional Regulator for Ischemic Cardiomyopathy. Genes (Basel) 2022; 13:genes13061051. [PMID: 35741813 PMCID: PMC9222441 DOI: 10.3390/genes13061051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/29/2022] [Accepted: 05/30/2022] [Indexed: 02/06/2023] Open
Abstract
Ischemic cardiomyopathy (ICM) caused by coronary artery disease always leads to myocardial infarction and heart failure. Identification of novel transcriptional regulators in ICM is an effective method to establish new diagnostic and therapeutic strategies. In this study, we used two RNA-seq datasets and one microarray dataset from different studies, including 25 ICM and 21 non-failing control (NF) samples of human left ventricle tissues for further analysis. In total, 208 differentially expressed genes (DEGs) were found by combining two RNA-seq datasets with batch effects removed. GO and KEGG analyses of DEGs indicated that the response to wounding, positive regulation of smooth muscle contraction, chromatin, PI3K-Akt signaling pathway, and transporters pathways are involved in ICM. Simple Enrichment Analysis found that NFIC-binding motifs are enriched in promoter regions of downregulated genes. The Gene Importance Calculator further proved that NFIC is vital. NFIC and its downstream genes were verified in the validating microarray dataset. Meanwhile, in rat cardiomyocyte cell line H9C2 cells, two genes (Tspan1 and Hopx) were confirmed, which decreased significantly along with knocking down Nfic expression. In conclusion, NFIC participates in the ICM process by regulating TSPAN1 and HOPX. NFIC and its downstream genes may be marker genes and potential diagnostic and therapeutic targets for ICM.
Collapse
|
14
|
Karaman K, Celik A, Aytac S, Bakar-Ates F. Increased endoglin levels correlated with angiogenesis-associated angiopoietin-2 in haemophilia patients. Haemophilia 2021; 27:e747-e753. [PMID: 34614537 DOI: 10.1111/hae.14430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/21/2021] [Accepted: 09/23/2021] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Haemophilia is a bleeding disorder that occurs due to the deficiency of coagulation factors, and the angiogenesis process is an important process underlying the pathophysiology of haemophilic arthropathy. The role of the new adipocytokine endoglin (ENG) in patients with haemophilia is not yet known. AIM The aim of this study is to evaluate the association between ENG protein and angiogenesis-related cytokines in patients with haemophilia for the first time. METHODS Plasma protein levels and mRNA expressions of ENG and various angiogenesis-associated cytokines were compared in blood samples collected from 28 patients with haemophilia A or B and 29 healthy volunteers. The relationship between the cytokines and ENG were determined by correlation analysis. RESULTS Plasma ENG levels and angiogenic markers were found to be significantly higher in patients with haemophilia compared to controls. Real-time PCR studies showed that mRNA expressions of ENG, vascular endothelial growth factor A, hypoxia-inducible factor A, and prostaglandin E2 increased in patients with haemophilia. Correlation analysis showed a significant positive correlation between ENG and angiopoietin-2 levels in the haemophilia group. Besides, a significant decrease in annexin-V binding to platelets in haemophilia patients compared to control was found to be related to the bleeding profiles in the patients. CONCLUSIONS This study determined that ENG protein may be involved in the formation of angiogenesis in haemophilia patients and its effects may be related to angiogenetic marker angiopoietin-2 in this process. Our findings contribute to the literature during the determination of target proteins in haemophilia treatment.
Collapse
Affiliation(s)
- Kubra Karaman
- Ankara University, Faculty of Pharmacy, Department of Biochemistry, Ankara, Turkey
| | - Aybuke Celik
- Ankara University, Faculty of Pharmacy, Department of Biochemistry, Ankara, Turkey
| | - Selin Aytac
- Faculty of Medicine, Department of Paediatrics, Division of Paediatric Haematology, Hacettepe University, Ankara, Turkey
| | - Filiz Bakar-Ates
- Ankara University, Faculty of Pharmacy, Department of Biochemistry, Ankara, Turkey
| |
Collapse
|
15
|
Wei G, Zhu D, Sun Y, Zhang L, Liu X, Li M, Gu J. The protective effects of azilsartan against oscillatory shear stress-induced endothelial dysfunction and inflammation are mediated by KLF6. J Biochem Mol Toxicol 2021; 35:1-8. [PMID: 33793019 DOI: 10.1002/jbt.22766] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 12/04/2020] [Accepted: 03/02/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND AND PURPOSE Atherosclerosis is a common cardiovascular disease with high morbidity and mortality. It is reported to be related to oscillatory shear stress (OSS)-induced endothelial dysfunction and excessive production of inflammatory factors. Azilsartan, a specific antagonist of the angiotensin II receptor, has been approved for the management of hypertensive subjects with diabetes mellitus type II (DMII). The present study will investigate the effects of azilsartan against OSS-induced endothelial dysfunction and inflammation, as well as the underlying mechanism. MATERIALS AND METHODS Cell viability was detected using an MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide) assay. Quantitative reverse-transcription polymerase chain reaction (qRT-PCR) and enzyme-linked immunosorbent assay were used to determine the expression levels of IL-6, TNF-α, IL-1β, VCAM-1, and ICAM-1 in human aortic endothelial cells (HAECs). Generation of reactive oxygen species (ROS) was measured using 2'-7'dichlorofluorescin diacetate (DCFH-DA) staining, and the level of reduced glutathione (GSH) was evaluated using a commercial kit. The adhesion of THP-1 monocytes to HAECs was evaluated using calcein-AM staining. The expression level of KLF6 was determined using qRT-PCR and Western blot analysis. RESULTS According to the result of the MTT assay, 5 and 10 μM azilsartan were considered as the optimized concentrations applied in the present study. The elevated production of IL-6, TNF-α, and IL-1β, increased levels of ROS, decreased levels of reduced GSH, upregulated VCAM-1, ICAM-1, and E-selectin, and the aggravated adhesion of THP-1 cells to HAECs induced by OSS were all reversed by the introduction of azilsartan. The downregulation of KLF6 induced by OSS was significantly reversed by azilsartan. By knocking down the expression of KLF6, the suppressed adhesion of THP-1 cells to the HAECs, and the downregulation of VCAM-1 and ICAM-1 induced by azilsartan in OSS-stimulated HAECs were greatly reversed. CONCLUSION The protective effects of azilsartan against OSS-induced endothelial dysfunction and inflammation might be mediated by KLF6.
Collapse
Affiliation(s)
- Guoqian Wei
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Dayong Zhu
- Department of General Surgery, Heilongjiang Provincial Hospital, Harbin, Heilongjiang Province, China
| | - Yongtao Sun
- Department of Imaging, Heilongjiang Provincial Hospital, Harbin, Heilongjiang Province, China
| | - Lan Zhang
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xian Liu
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Ming Li
- Department of Endocrinology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Jinxia Gu
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| |
Collapse
|
16
|
Hu K, Zheng QK, Ma RJ, Ma C, Sun ZG, Zhang N. Krüppel-Like Factor 6 Splice Variant 1: An Oncogenic Transcription Factor Involved in the Progression of Multiple Malignant Tumors. Front Cell Dev Biol 2021; 9:661731. [PMID: 33816511 PMCID: PMC8017371 DOI: 10.3389/fcell.2021.661731] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 02/23/2021] [Indexed: 01/03/2023] Open
Abstract
Krüppel-like factor 6 (KLF6) is one of the most studied members of the specificity protein/Krüppel-like factor (SP/KLF) transcription factor family. It has a typical zinc finger structure and plays a pivotal role in regulating the biological processes of cells. Recently, it has been considered to play a role in combatting cancer. Krüppel-like factor 6 splice variant 1 (KLF6-SV1), being one of the alternative KLF6 splicing isoforms, participates in tumor occurrence and development and has the potential to become a new target for molecular targeted therapy, although its action mechanism remains to be determined. The purpose of this article is to provide a comprehensive and systematic review of the important role of KLF6-SV1 in human malignant tumors to provide novel insights for oncotherapy.
Collapse
Affiliation(s)
- Kang Hu
- School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Qing-Kang Zheng
- School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Rui-Jie Ma
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chao Ma
- School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Zhi-Gang Sun
- Department of Thoracic Surgery, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Nan Zhang
- Department of Oncology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
17
|
Vicen M, Igreja Sá IC, Tripská K, Vitverová B, Najmanová I, Eissazadeh S, Micuda S, Nachtigal P. Membrane and soluble endoglin role in cardiovascular and metabolic disorders related to metabolic syndrome. Cell Mol Life Sci 2021; 78:2405-2418. [PMID: 33185696 PMCID: PMC11072708 DOI: 10.1007/s00018-020-03701-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 10/05/2020] [Accepted: 10/31/2020] [Indexed: 02/07/2023]
Abstract
Membrane endoglin (Eng, CD105) is a transmembrane glycoprotein essential for the proper function of vascular endothelium. It might be cleaved by matrix metalloproteinases to form soluble endoglin (sEng), which is released into the circulation. Metabolic syndrome comprises conditions/symptoms that usually coincide (endothelial dysfunction, arterial hypertension, hyperglycemia, obesity-related insulin resistance, and hypercholesterolemia), and are considered risk factors for cardiometabolic disorders such as atherosclerosis, type II diabetes mellitus, and liver disorders. The purpose of this review is to highlight current knowledge about the role of Eng and sEng in the disorders mentioned above, in vivo and in vitro extent, where we can find a wide range of contradictory results. We propose that reduced Eng expression is a hallmark of endothelial dysfunction development in chronic pathologies related to metabolic syndrome. Eng expression is also essential for leukocyte transmigration and acute inflammation, suggesting that Eng is crucial for the regulation of endothelial function during the acute phase of vascular defense reaction to harmful conditions. sEng was shown to be a circulating biomarker of preeclampsia, and we propose that it might be a biomarker of metabolic syndrome-related symptoms and pathologies, including hypercholesterolemia, hyperglycemia, arterial hypertension, and diabetes mellitus as well, despite the fact that some contradictory findings have been reported. Besides, sEng can participate in the development of endothelial dysfunction and promote the development of arterial hypertension, suggesting that high levels of sEng promote metabolic syndrome symptoms and complications. Therefore, we suggest that the treatment of metabolic syndrome should take into account the importance of Eng in the endothelial function and levels of sEng as a biomarker and risk factor of related pathologies.
Collapse
Affiliation(s)
- Matej Vicen
- Faculty of Pharmacy in Hradec Kralove, Department of Biological and Medical Sciences, Charles University, Heyrovskeho 1203, Hradec Kralove, 500 03, Czech Republic
| | - Ivone Cristina Igreja Sá
- Faculty of Pharmacy in Hradec Kralove, Department of Biological and Medical Sciences, Charles University, Heyrovskeho 1203, Hradec Kralove, 500 03, Czech Republic
| | - Katarína Tripská
- Faculty of Pharmacy in Hradec Kralove, Department of Biological and Medical Sciences, Charles University, Heyrovskeho 1203, Hradec Kralove, 500 03, Czech Republic
| | - Barbora Vitverová
- Faculty of Pharmacy in Hradec Kralove, Department of Biological and Medical Sciences, Charles University, Heyrovskeho 1203, Hradec Kralove, 500 03, Czech Republic
| | - Iveta Najmanová
- Faculty of Pharmacy in Hradec Kralove, Department of Biological and Medical Sciences, Charles University, Heyrovskeho 1203, Hradec Kralove, 500 03, Czech Republic
| | - Samira Eissazadeh
- Faculty of Pharmacy in Hradec Kralove, Department of Biological and Medical Sciences, Charles University, Heyrovskeho 1203, Hradec Kralove, 500 03, Czech Republic
| | - Stanislav Micuda
- Faculty of Medicine in Hradec Kralove, Department of Pharmacology, Charles University, Simkova 870, Hradec Kralove, 500 03, Czech Republic
| | - Petr Nachtigal
- Faculty of Pharmacy in Hradec Kralove, Department of Biological and Medical Sciences, Charles University, Heyrovskeho 1203, Hradec Kralove, 500 03, Czech Republic.
| |
Collapse
|
18
|
Potential Second-Hits in Hereditary Hemorrhagic Telangiectasia. J Clin Med 2020; 9:jcm9113571. [PMID: 33167572 PMCID: PMC7694477 DOI: 10.3390/jcm9113571] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 10/29/2020] [Accepted: 11/02/2020] [Indexed: 12/13/2022] Open
Abstract
Hereditary hemorrhagic telangiectasia (HHT) is an autosomal dominant genetic disorder that presents with telangiectases in skin and mucosae, and arteriovenous malformations (AVMs) in internal organs such as lungs, liver, and brain. Mutations in ENG (endoglin), ACVRL1 (ALK1), and MADH4 (Smad4) genes account for over 95% of HHT. Localized telangiectases and AVMs are present in different organs, with frequencies which differ among affected individuals. By itself, HHT gene heterozygosity does not account for the focal nature and varying presentation of the vascular lesions leading to the hypothesis of a “second-hit” that triggers the lesions. Accumulating research has identified a variety of triggers that may synergize with HHT gene heterozygosity to generate the vascular lesions. Among the postulated second-hits are: mechanical trauma, light, inflammation, vascular injury, angiogenic stimuli, shear stress, modifier genes, and somatic mutations in the wildtype HHT gene allele. The aim of this review is to summarize these triggers, as well as the functional mechanisms involved.
Collapse
|
19
|
Syafruddin SE, Mohtar MA, Wan Mohamad Nazarie WF, Low TY. Two Sides of the Same Coin: The Roles of KLF6 in Physiology and Pathophysiology. Biomolecules 2020; 10:biom10101378. [PMID: 32998281 PMCID: PMC7601070 DOI: 10.3390/biom10101378] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/26/2020] [Accepted: 09/26/2020] [Indexed: 12/12/2022] Open
Abstract
The Krüppel-like factors (KLFs) family of proteins control several key biological processes that include proliferation, differentiation, metabolism, apoptosis and inflammation. Dysregulation of KLF functions have been shown to disrupt cellular homeostasis and contribute to disease development. KLF6 is a relevant example; a range of functional and expression assays suggested that the dysregulation of KLF6 contributes to the onset of cancer, inflammation-associated diseases as well as cardiovascular diseases. KLF6 expression is either suppressed or elevated depending on the disease, and this is largely due to alternative splicing events producing KLF6 isoforms with specialised functions. Hence, the aim of this review is to discuss the known aspects of KLF6 biology that covers the gene and protein architecture, gene regulation, post-translational modifications and functions of KLF6 in health and diseases. We put special emphasis on the equivocal roles of its full-length and spliced variants. We also deliberate on the therapeutic strategies of KLF6 and its associated signalling pathways. Finally, we provide compelling basic and clinical questions to enhance the knowledge and research on elucidating the roles of KLF6 in physiological and pathophysiological processes.
Collapse
Affiliation(s)
- Saiful E. Syafruddin
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (M.A.M.); (T.Y.L.)
- Correspondence: ; Tel.: +60-3-9145-9040
| | - M. Aiman Mohtar
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (M.A.M.); (T.Y.L.)
| | - Wan Fahmi Wan Mohamad Nazarie
- Biotechnology Programme, Faculty of Science and Natural Resources, Universiti Malaysia Sabah, Kota Kinabalu 88400, Malaysia;
| | - Teck Yew Low
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (M.A.M.); (T.Y.L.)
| |
Collapse
|
20
|
Cetın N, Sav NM, Kıraz ZK, Gencler A. Transforming Growth Factor-Beta 1 and Endoglin Levels in Congenital Solitary Functioning Kidney. Indian J Nephrol 2020; 30:270-276. [PMID: 33273793 PMCID: PMC7699669 DOI: 10.4103/ijn.ijn_111_19] [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: 04/01/2019] [Revised: 05/28/2019] [Accepted: 11/17/2019] [Indexed: 11/09/2022] Open
Abstract
Introduction: Glomerular hyperfiltration leads to hypertension, microalbuminuria, and impaired renal function in children with congenital solitary functioning kidney (cSFK). The purpose of this study was to investigate the associations between serum transforming growth factor β-1 (TGF) and endoglin levels and hypertension, renal function or microalbuminuria in children with cSFK. Materials and Methods: 63 patients and 36 controls were included in the study. Serum endoglin and TGF-β1 level was measured using ELISA commercial kits. Results: Serum TGF-β1 and endoglin levels were higher in patients than those of controls (P = 0.04 and P < 0.001, respectively). The prevalence of hypertension was found to be 45.6%. There was a positive association between endoglin levels and the presence of masked hypertension (odds ratio: 1.121, P = 0.04). TGF-β1 and endoglin levels were positively associated with microalbuminuria (OR: 1.17, P = 0.04; OR: 1.836, P = 0.01). ROC curve analysis showed that serum endoglin and TGF-β1 levels had predictive value for microalbuminuria (cut-off value: 4.86 ng/mL, sensitivity: 94.7%, specificity: 54.5%, area under the curve ± standard error [AUC ± SE]: 0.888 ± 0.025, P = 0.01 for endoglin; cut-off value 561.24 pg/mL, sensitivity: 89.5%, specificity: 73%, AUC ± SE: 0.995 ± 0.334, P = 0.02 for TGF-β1). There were no significant relationships between glomerular filtration rate and serum TGF-β1 or endoglin levels. Conclusions: Endoglin and TGF-β1 may play an important role in the pathophysiology of microalbuminuria in cSFK. Endoglin may have a role in the development of hypertension in children with cSFK.
Collapse
Affiliation(s)
- Nuran Cetın
- Department of Pediatric Nephrology, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Nadide Melike Sav
- Department of Pediatric Nephrology, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Zeynep Kusku Kıraz
- Department of Biochemistry, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Aylin Gencler
- Department of Pediatric Nephrology, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| |
Collapse
|
21
|
Calvo-Sánchez MI, Fernández-Martos S, Carrasco E, Moreno-Bueno G, Bernabéu C, Quintanilla M, Espada J. A role for the Tgf-β/Bmp co-receptor Endoglin in the molecular oscillator that regulates the hair follicle cycle. J Mol Cell Biol 2020; 11:39-52. [PMID: 30239775 PMCID: PMC6359924 DOI: 10.1093/jmcb/mjy051] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 09/18/2018] [Indexed: 12/18/2022] Open
Abstract
The hair follicle is a biological oscillator that alternates growth, regression, and rest phases driven by the sequential activation of the proliferation/differentiation programs of resident stem cell populations. The activation of hair follicle stem cell niches and subsequent entry into the growing phase is mainly regulated by Wnt/β-catenin signalling, while regression and resting phases are mainly regulated by Tgf-β/Bmp/Smad activity. A major question still unresolved is the nature of the molecular switch that dictates the coordinated transition between both signalling pathways. Here we have focused on the role of Endoglin (Eng), a key co-receptor for members of the Tgf-β/Bmp family of growth factors. Using an Eng haploinsufficient mouse model, we report that Eng is required to maintain a correct follicle cycling pattern and for an adequate stimulation of hair follicle stem cell niches. We further report that β-catenin binds to the Eng promoter depending on Bmp signalling. Moreover, we show that β-catenin interacts with Smad4 in a Bmp/Eng-dependent context and both proteins act synergistically to activate Eng promoter transcription. These observations point to the existence of a growth/rest switching mechanism in the hair follicle that is based on an Eng-dependent feedback cross-talk between Wnt/β-catenin and Bmp/Smad signals.
Collapse
Affiliation(s)
- María I Calvo-Sánchez
- Instituto de Investigaciones Biomédicas 'Alberto Sols', Consejo Superior de Investigaciones Científicas (CSIC)-Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Madrid, Spain.,Instituto de Investigaciones Biosanitarias, Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria (UFV), Pozuelo de Alarcon, Spain
| | | | - Elisa Carrasco
- Instituto de Investigaciones Biomédicas 'Alberto Sols', Consejo Superior de Investigaciones Científicas (CSIC)-Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Gema Moreno-Bueno
- Instituto de Investigaciones Biomédicas 'Alberto Sols', Consejo Superior de Investigaciones Científicas (CSIC)-Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Spain
| | - Carmelo Bernabéu
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Miguel Quintanilla
- Instituto de Investigaciones Biomédicas 'Alberto Sols', Consejo Superior de Investigaciones Científicas (CSIC)-Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Jesús Espada
- Instituto de Investigaciones Biomédicas 'Alberto Sols', Consejo Superior de Investigaciones Científicas (CSIC)-Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Madrid, Spain.,Centro Integrativo de Biología y Química Aplicada, Universidad Bernardo O´Higgins, Santiago, Chile
| |
Collapse
|
22
|
MiR-429 regulates blood−spinal cord barrier permeability by targeting Krüppel-like factor 6. Biochem Biophys Res Commun 2020; 525:740-746. [DOI: 10.1016/j.bbrc.2020.02.138] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 02/22/2020] [Indexed: 11/17/2022]
|
23
|
Zhang X, Tang X, Ma F, Fan Y, Sun P, Zhu T, Zhang J, Hamblin MH, Chen YE, Yin KJ. Endothelium-targeted overexpression of Krüppel-like factor 11 protects the blood-brain barrier function after ischemic brain injury. Brain Pathol 2020; 30:746-765. [PMID: 32196819 DOI: 10.1111/bpa.12831] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 02/07/2020] [Accepted: 03/15/2020] [Indexed: 12/22/2022] Open
Abstract
Microvascular endothelial cell (EC) injury and the subsequent blood-brain barrier (BBB) breakdown are frequently seen in many neurological disorders, including stroke. We have previously documented that peroxisome proliferator-activated receptor gamma (PPARγ)-mediated cerebral protection during ischemic insults needs Krüppel-like factor 11 (KLF11) as a critical coactivator. However, the role of endothelial KLF11 in cerebrovascular function and stroke outcome is unclear. This study is aimed at investigating the regulatory role of endothelial KLF11 in BBB preservation and neurovascular protection after ischemic stroke. EC-targeted overexpression of KLF11 significantly mitigated BBB leakage in ischemic brains, evidenced by significantly reduced extravasation of BBB tracers and infiltration of peripheral immune cells, and less brain water content. Endothelial cell-selective KLF11 transgenic (EC-KLF11 Tg) mice also exhibited smaller brain infarct and improved neurological function in response to ischemic insults. Furthermore, EC-targeted transgenic overexpression of KLF11 preserved cerebral tight junction (TJ) levels and attenuated the expression of pro-inflammatory factors in mice after ischemic stroke. Mechanistically, we demonstrated that KLF11 directly binds to the promoter of major endothelial TJ proteins including occludin and ZO-1 to promote their activities. Our data indicate that KLF11 functions at the EC level to preserve BBB structural and functional integrity, and therefore, confers brain protection in ischemic stroke. KLF11 may be a novel therapeutic target for the treatment of ischemic stroke and other neurological conditions involving BBB breakdown and neuroinflammation.
Collapse
Affiliation(s)
- Xuejing Zhang
- Pittsburgh Institute of Brain Disorders & Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213
| | - Xuelian Tang
- Pittsburgh Institute of Brain Disorders & Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213
| | - Feifei Ma
- Pittsburgh Institute of Brain Disorders & Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213
| | - Yanbo Fan
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109
| | - Ping Sun
- Pittsburgh Institute of Brain Disorders & Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213
| | - Tianqing Zhu
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109
| | - Jifeng Zhang
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109
| | - Milton H Hamblin
- Department of Pharmacology, Tulane University School of Medicine, 1430 Tulane Avenue SL83, New Orleans, LA, 70112
| | - Y Eugene Chen
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109
| | - Ke-Jie Yin
- Pittsburgh Institute of Brain Disorders & Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213.,Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, 15261
| |
Collapse
|
24
|
Muenzner P, Hauck CR. Neisseria gonorrhoeae Blocks Epithelial Exfoliation by Nitric-Oxide-Mediated Metabolic Cross Talk to Promote Colonization in Mice. Cell Host Microbe 2020; 27:793-808.e5. [PMID: 32289262 DOI: 10.1016/j.chom.2020.03.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 12/19/2019] [Accepted: 03/17/2020] [Indexed: 12/18/2022]
Abstract
Several pathogens suppress exfoliation, a key defense of epithelia against microbial colonization. Common among these pathogens, exemplified by Neisseria gonorrhoeae, is their ability to bind carcinoembryonic antigen-related cell adhesion molecules (CEACAMs). Gonococcal CEACAM engagement triggers the expression of CD105, which is necessary to block epithelial exfoliation, whereas homotypic CEACAM-CEACAM interactions or antibody-mediated CEACAM clustering does not lead to CD105 expression. Here, we show that CEACAM-associated bacteria release nitric oxide (NO) during anaerobic respiration, and membrane-permeable NO initiates a eukaryotic signaling pathway involving soluble guanylate cyclase (sGC), protein kinase G, and the transcription factor CREB to upregulate CD105 expression. A murine vaginal infection model with N. gonorrhoeae reveals this metabolic cross communication allows bacterial suppression of epithelial exfoliation to facilitate mucosal colonization. Disrupting NO-initiated responses in host cells re-establishes epithelial exfoliation and inhibits mouse genital tract colonization by N. gonorrhoeae, suggesting a host-directed approach to prevent bacterial infections.
Collapse
Affiliation(s)
- Petra Muenzner
- Lehrstuhl Für Zellbiologie, Fachbereich Biologie, Universität Konstanz, 78457 Konstanz, Germany
| | - Christof R Hauck
- Lehrstuhl Für Zellbiologie, Fachbereich Biologie, Universität Konstanz, 78457 Konstanz, Germany; Konstanz Research School Chemical Biology, Universität Konstanz, 78457 Konstanz, Germany.
| |
Collapse
|
25
|
Sabatino ME, Castellaro A, Racca AC, Carbajosa González S, Pansa MF, Soria G, Bocco JL. Krüppel-Like Factor 6 Is Required for Oxidative and Oncogene-Induced Cellular Senescence. Front Cell Dev Biol 2019; 7:297. [PMID: 31824948 PMCID: PMC6882731 DOI: 10.3389/fcell.2019.00297] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 11/07/2019] [Indexed: 12/24/2022] Open
Abstract
Krüppel-like factor 6 (KLF6) is a transcription factor involved in the regulation of several cellular processes. Regarding its role in tumorigenesis, KLF6 is considered a tumor suppressor. Numerous reports demonstrate its frequent genomic loss or down-regulation, implying a functional inactivation in a broad range of human cancers. Previous work from our laboratory showed that the down-regulation of KLF6 expression in normal fibroblasts leads to cellular transformation, while its ectopic expression interferes with the oncogenic transformation triggered by activated Ras through a cell cycle arrest. We hypothesize that the growth suppressor activity of KLF6 may involve the induction of cellular senescence thereby helping to prevent the proliferation of cells at risk of neoplastic transformation. Here, we explored the association of KLF6 up-regulation in two different cellular senescence scenarios. We found that KLF6 silencing bypasses both oxidative and oncogene-induced senescence. In this context, KLF6 expression per se was capable to trigger cellular senescence in both normal and tumoral contexts. As such, the findings presented in this report provide insights into a potential mechanism by which KLF6 may play a suppressing role of uncontrolled or damaged cell proliferation.
Collapse
Affiliation(s)
- Maria Eugenia Sabatino
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Andrés Castellaro
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Ana C Racca
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Sofía Carbajosa González
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Maria Florencia Pansa
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Gastón Soria
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Jose Luis Bocco
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
26
|
Yang Y, Yu H, Yang C, Zhang Y, Ai X, Wang X, Lu K, Yi B. Krüppel-like factor 6 mediates pulmonary angiogenesis in rat experimental hepatopulmonary syndrome and is aggravated by bone morphogenetic protein 9. Biol Open 2019; 8:bio.040121. [PMID: 31189661 PMCID: PMC6602319 DOI: 10.1242/bio.040121] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Hepatopulmonary syndrome (HPS) is a serious pulmonary vascular disease derived from chronic liver disease, and its key pathogenesis is angiogenesis. Krüppel-like factor 6 (KLF6) mediates physiological repair and remodeling during vascular injury. However, the role of KLF6 in pulmonary microvascular endothelial cells (PMVECs) during angiogenesis of HPS and its underlying mechanism in HPS have not been investigated. Common bile duct ligation (CBDL) in rats can replicate pulmonary vascular abnormalities of human HPS. Here, we found that advanced pulmonary angiogenesis and pulmonary injury score coincided with the increase of KLF6 level in PMVECs of CBDL rat; KLF6 in PMVECs was also induced while cultured with CBDL rat serum in vitro. Inhibition of KLF6 dramatically suppressed PMVEC-mediated proliferation, migration and tube formation in vivo; this may be related to the downregulation of activin receptor-like kinase-1 (ALK1) and endoglin (ENG), which are transacted by KLF6. Bone morphogenetic protein 9 (BMP9) enhanced the expression of KLF6 in PMVECs and was involved in the angiogenesis of HPS. These results suggest that KLF6 triggers PMVEC-mediated angiogenesis of HPS and is aggravated by BMP9, and the inhibition of the BMP9/KLF6 axis may be an effective strategy for HPS treatment. Summary: Krüppel-like factor 6, which is triggered by pulmonary injury and promoted by bone morphogenetic protein 9, mediates pulmonary angiogenesis in rat experimental hepatopulmonary syndrome and then aggravates lung dysfunction.
Collapse
Affiliation(s)
- Yihui Yang
- Department of Anaesthesia, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038 China.,Department of Anesthesia, The Third Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563000 China
| | - Hongfu Yu
- Department of Anaesthesia, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038 China
| | - Congwen Yang
- Department of Anaesthesia, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038 China
| | - Yunfei Zhang
- Department of Anaesthesia, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038 China.,Department of Anesthesia, The Third Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563000 China
| | - Xiangfa Ai
- Department of Anaesthesia, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038 China
| | - Xiaobo Wang
- Department of LBCMCP, Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 31062 Toulouse, France
| | - Kaizhi Lu
- Department of Anaesthesia, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038 China
| | - Bin Yi
- Department of Anaesthesia, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038 China
| |
Collapse
|
27
|
Abstract
Osler's disease is an autosomal dominant hereditary syndrome which belongs to the group of orphan diseases. Affected patients suffer primarily from severe epistaxis. Diagnosis is based on the Curaçao criteria and molecular genetic tests. Organ manifestations can be found in the form of arteriovenous shunts in the lung, liver, and gastrointestinal tract; more rarely also in the central nervous system (CNS) and other parts of the body. Many patients with gastrointestinal and other organ manifestations are frequently clinically asymptomatic; therefore, organ screening is essential to avoid later complications and should be performed in centers with particular expertise. No curative therapy currently exists. From the otolaryngologist's perspective, nasal mucosa treatments and endonasal laser applications are important and effective therapeutic approaches to epistaxis. Pharmacological interventions are focused on compensation of haploinsufficiency as well as antiangiogenetic approaches. Severe side effects have to be considered.
Collapse
|
28
|
Buda V, Andor M, Baibata DE, Cozlac R, Radu G, Coricovac D, Danciu C, Ledeti I, Cheveresan A, Nica C, Tuduce P, Tomescu MC. Decreased sEng plasma levels in hypertensive patients with endothelial dysfunction under chronic treatment with Perindopril. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:1915-1925. [PMID: 31239642 PMCID: PMC6556213 DOI: 10.2147/dddt.s186378] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 03/15/2019] [Indexed: 12/25/2022]
Abstract
Purpose: Endoglin is a transmembrane glycoprotein which plays an important role in maintaining cardiovascular homeostasis. One of its forms, soluble endoglin (sEng), a molecule with antiangiogenic properties, has been found overexpressed in patients suffering from hypercholesterolemia, diabetes mellitus and hypertension, and is proposed as a marker of endothelial damage. Accordingly, we aimed to quantify the efficacy of various antihypertensive regimens on sEng levels, in hypertensive patients with endothelial dysfunction. Patients and methods: 323 patients were enrolled, and there were 99 patients with normal blood pressure values, 106 hypertensive patients under chronic treatment with different types of antihypertensive molecules (beta blockers, calcium channel blockers, and diuretics) in monotherapy, and 118 hypertensive patients under chronic treatment with perindopril. sEng plasma levels were quantified and were correlated with classical methods of assessing the endothelial damage. Results: Patients under chronic treatment with perindopril had lower sEng plasma levels compared with the other group of hypertensive patients under different regimens of antihypertensive treatment (sEng: 4.73±1.39 versus 5.63±2.33, p<0.01). Conclusion: Decreased sEng plasma levels were found in patients under chronic treatment with perindopril, when compared with other antihypertensive regimens of treatment (beta blockers, calcium channel blockers, and/or diuretics).
Collapse
Affiliation(s)
- Valentina Buda
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, "Victor Babeş" University of Medicine and Pharmacy, Timisoara, Romania
| | - Minodora Andor
- Department of Medical Semiotics, Faculty of Medicine, "Victor Babeş" University of Medicine and Pharmacy, Timisoara, Romania
| | - Dana Emilia Baibata
- Department of Cardiology VI, Faculty of Medicine, "Victor Babeş" University of Medicine and Pharmacy, Timisoara, Romania.,Cardiovascular Diseases Institute, Timisoara, Romania
| | - Ramona Cozlac
- Department of Cardiology VI, Faculty of Medicine, "Victor Babeş" University of Medicine and Pharmacy, Timisoara, Romania.,Cardiovascular Diseases Institute, Timisoara, Romania
| | - Gabriela Radu
- Department of Medical Semiotics, Faculty of Medicine, "Victor Babeş" University of Medicine and Pharmacy, Timisoara, Romania
| | - Dorina Coricovac
- Department of Toxicology, Faculty of Pharmacy, "Victor Babeş" University of Medicine and Pharmacy, Timisoara, Romania
| | - Corina Danciu
- Department of Pharmacognosy, Faculty of Pharmacy, "Victor Babeş" University of Medicine and Pharmacy, Timisoara, Romania
| | - Ionut Ledeti
- Department of Physical Chemistry, Faculty of Pharmacy, "Victor Babeş" University of Medicine and Pharmacy, Timisoara, Romania
| | - Adelina Cheveresan
- Department of Pharmacology, Faculty of Medicine, "Victor Babeş" University of Medicine and Pharmacy, Timisoara, Romania
| | - Cristian Nica
- Department of Surgery, Faculty of Medicine, "Victor Babeş" University of Medicine and Pharmacy, Timisoara, Romania
| | - Paul Tuduce
- Department of Balneophysiotherapy, Rheumatology and Rehabilitation, Faculty of Medicine, Vasile Goldis Western University, Arad, Romania
| | - Mirela Cleopatra Tomescu
- Department of Medical Semiotics, Faculty of Medicine, "Victor Babeş" University of Medicine and Pharmacy, Timisoara, Romania
| |
Collapse
|
29
|
Vicen M, Vitverova B, Havelek R, Blazickova K, Machacek M, Rathouska J, Najmanová I, Dolezelova E, Prasnicka A, Sternak M, Bernabeu C, Nachtigal P. Regulation and role of endoglin in cholesterol-induced endothelial and vascular dysfunction in vivo and in vitro. FASEB J 2019; 33:6099-6114. [PMID: 30753095 DOI: 10.1096/fj.201802245r] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Our objective was to investigate the effect of cholesterol [hypercholesterolemia and 7-ketocholesterol (7K)] on endoglin (Eng) expression and regulation with respect to endothelial or vascular dysfunction in vivo and in vitro. In vivo experiments were performed in 2-mo-old atherosclerosis-prone apolipoprotein E-deficient/LDL receptor-deficient (ApoE-/-/LDLR-/-) female mice and their wild-type C57BL/6J littermates. In in vitro experiments, human aortic endothelial cells (HAECs) were treated with 7K. ApoE-/-/LDLR-/- mice developed hypercholesterolemia accompanied by increased circulating levels of P-selectin and Eng and a disruption of NO metabolism. Functional analysis of the aorta demonstrated impaired vascular reactivity, and Western blot analysis revealed down-regulation of membrane Eng/Smad2/3/eNOS signaling in ApoE-/-/LDLR-/- mice. 7K increased Eng expression via Krüppel-like factor 6 (KLF6), liver X nuclear receptor, and NF-κB in HAECs. 7K-induced Eng expression was prevented by the treatment with 2-hydroxypropyl-β-cyclodextrin; 8-{[5-chloro-2-(4-methylpiperazin-1-yl) pyridine-4-carbonyl] amino}-1-(4-fluorophenyl)-4, 5-dihydrobenzo[g]indazole-3-carboxamide; or by KLF6 silencing. 7K induced increased adhesion and transmigration of monocytic human leukemia promonocytic cell line cells and was prevented by Eng silencing. We concluded that hypercholesterolemia altered Eng expression and signaling, followed by endothelial or vascular dysfunction before formation of atherosclerotic lesions in ApoE-/-/LDLR-/- mice. By contrast, 7K increased Eng expression and induced inflammation in HAECs, which was followed by an increased adhesion and transmigration of monocytes via endothelium, which was prevented by Eng inhibition. Thus, we propose a relevant role for Eng in endothelial or vascular dysfunction or inflammation when exposed to cholesterol.-Vicen, M., Vitverova, B., Havelek, R., Blazickova, K., Machacek, M., Rathouska, J., Najmanová, I., Dolezelova, E., Prasnicka, A., Sternak, M., Bernabeu, C., Nachtigal, P. Regulation and role of endoglin in cholesterol-induced endothelial and vascular dysfunction in vivo and in vitro.
Collapse
Affiliation(s)
- Matej Vicen
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Barbora Vitverova
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Radim Havelek
- Department of Medical Biochemistry, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Katerina Blazickova
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Miloslav Machacek
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Jana Rathouska
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Iveta Najmanová
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Eva Dolezelova
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Alena Prasnicka
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Magdalena Sternak
- Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego, Krakow, Poland
| | - Carmelo Bernabeu
- Center for Biological Research, Spanish National Research Council (CSIC), Madrid, Spain.,Biomedical Research Networking Center on Rare Diseases (CIBERER), Madrid, Spain
| | - Petr Nachtigal
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| |
Collapse
|
30
|
Rane MJ, Zhao Y, Cai L. Krϋppel-like factors (KLFs) in renal physiology and disease. EBioMedicine 2019; 40:743-750. [PMID: 30662001 PMCID: PMC6414320 DOI: 10.1016/j.ebiom.2019.01.021] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 01/08/2019] [Accepted: 01/08/2019] [Indexed: 12/20/2022] Open
Abstract
Dysregulated Krϋppel-like factor (KLF) gene expression appears in many disease-associated pathologies. In this review, we discuss physiological functions of KLFs in the kidney with a focus on potential pharmacological modulation/therapeutic applications of these KLF proteins. KLF2 is critical to maintaining endothelial barrier integrity and preventing gap formations and in prevention of glomerular endothelial cell and podocyte damage in diabetic mice. KLF4 is renoprotective in the setting of AKI and is a critical regulator of proteinuria in mice and humans. KLF6 expression in podocytes preserves mitochondrial function and prevents podocyte apoptosis, while KLF5 expression prevents podocyte apoptosis by blockade of ERK/p38 MAPK pathways. KLF15 is a critical regulator of podocyte differentiation and is protective against podocyte injury. Loss of KLF4 and KLF15 promotes renal fibrosis, while fibrotic kidneys have increased KLF5 and KLF6 expression. For therapeutic modulation of KLFs, continued screening of small molecules will promote drug discoveries targeting KLF proteins.
Collapse
Affiliation(s)
- Madhavi J Rane
- Department of Medicine, Division Nephrology, Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, KY 40292, USA.
| | - Yuguang Zhao
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Lu Cai
- Pediatric Research Institute, Department of Pediatrics, Radiation Oncology, Pharmacology and Toxicology, University of Louisville, Louisville, KY 40292, USA.
| |
Collapse
|
31
|
Abstract
Supplemental Digital Content is available in the text. Kruppel like factor 6 (KLF6), a member of KLF family, which has classic zinc finger structure, is broadly considered to have anticancer activity. The role of SV2 variant, one of KLF6 alternative splicing isoforms has not yet been definite in the colorectal cancer. This study aimed to detect the expression of the KLF6-SV2 in colorectal cancer and investigate its impact on cell proliferation and apoptosis. qRT-PCR was used to quantitatively determine KLF6-SV2 mRNA expression in colorectal cancer samples, corresponding normal tissue, normal colonic mucosal cell line FHC and seven colorectal cancer cell lines. SW480 and SW620 cell models with over-expressing KLF6-SV2 were constructed. Cell proliferation, cell cycle and apoptosis were measured respectively using MTT assay, DNA ploidy detection and Annexin V flow cytometry. Meanwhile, expression of p53, p21 and Bax were detected by qRT-PCR and western blot. The mRNA expression level of KLF6-SV2 in colorectal cancer tissues (0.783±0.409) was decreased than in corresponding normal tissues (1.086±0.449) (P<0.01), and expression in SW480 and SW620 were lower than in FHC, HCT116, LoVo, HT29, Caco-2 and RKO. In cell lines over-expressing KLF6-SV2, cell proliferation was markedly suppressed, cell cycle was blocked and cell apoptosis was significantly induced. Simultaneously, expression of p21 and Bax were remarkably up-regulated, while p53 remained unchanged. Decreased expression of KLF6-SV2 may be associated with the occurrence and development of colorectal cancer. KLF6-SV2 plays a role as tumor suppressor by efficiently blocking cell proliferation, arresting cell cycle and inducing apoptosis in colorectal cancer, which may be related to increased expression of p21 and Bax.
Collapse
|
32
|
Chi Y, Xu Y, Luo F, Lin Y, Li Z. Molecular cloning, expression profiles and associations of KLF6 gene with intramuscular fat in Tibetan chicken. Anim Biotechnol 2018; 31:67-75. [PMID: 30501383 DOI: 10.1080/10495398.2018.1540428] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The aim of this study was to clone KLF6 gene of Tibetan chicken, clarify its temporal-spatial expression characteristics, and build the correlation between the expression level of KLF6 gene and IMF content in different developmental stages. RT-PCR was used to clone Tibetan chicken KLF6 gene, qPCR was used to detect the expression level of KLF6 gene in different tissues and developmental stages. The sequence of KLF6 gene was 919 bp including a complete 852 bp CDS region. The gene was highest expression in lung tissues, which was significantly higher than in other tissues (p < 0.01). In male Tibetan chicken breast muscle the levels of KLF6 mRNA were negatively related to IMF content (r=-0.097, p > 0.05), while in females they were positively correlated (r = 0.077, p > 0.05). At the age of 119-210 days, the expression of KLF6 mRNA in the male chicken leg muscles was highly positively correlated (r = 0.506, p < 0.01), but negatively correlated in the female chicken leg muscles (r=-0.198, p > 0.05). The expression level of KLF6 in breast muscle decreased gradually with the increasing age, while in leg muscle the expression level increased firstly and then descended with the increasing age.
Collapse
Affiliation(s)
- Yongdong Chi
- College of Life Science and Technology, Southwest Minzu University, Chengdu, China.,Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu, China
| | - Yaou Xu
- College of Life Science and Technology, Southwest Minzu University, Chengdu, China.,Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu, China
| | - Fan Luo
- College of Life Science and Technology, Southwest Minzu University, Chengdu, China.,Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu, China
| | - Yaqiu Lin
- College of Life Science and Technology, Southwest Minzu University, Chengdu, China.,Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu, China
| | - Zhixiong Li
- College of Life Science and Technology, Southwest Minzu University, Chengdu, China.,Key Laboratory of Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation, Southwest Minzu University, Chengdu, China
| |
Collapse
|
33
|
Wang R, Lin J, Bagchi RA. Novel molecular therapeutic targets in cardiac fibrosis: a brief overview 1. Can J Physiol Pharmacol 2018; 97:246-256. [PMID: 30388374 DOI: 10.1139/cjpp-2018-0430] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cardiac fibrosis, characterized by excessive accumulation of extracellular matrix, abolishes cardiac contractility, impairs cardiac function, and ultimately leads to heart failure. In recent years, significant evidence has emerged that supports the highly dynamic and responsive nature of the cardiac extracellular matrix. Although our knowledge of cardiac fibrosis has advanced tremendously over the past decade, there is still a lack of specific therapies owing to an incomplete understanding of the disease etiology and process. In this review, we attempt to highlight some of the recently investigated molecular determinants of ischemic and non-ischemic fibrotic remodeling of the myocardium that present as promising avenues for development of anti-fibrotic therapies.
Collapse
Affiliation(s)
- Ryan Wang
- a Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Justin Lin
- b Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Rushita A Bagchi
- c Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
34
|
Hong J, Wang X, Mei C, Zan L. Competitive regulation by transcription factors and DNA methylation in the bovine SIRT5 promoter: Roles of E2F4 and KLF6. Gene 2018; 684:39-46. [PMID: 30359737 DOI: 10.1016/j.gene.2018.10.052] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 10/15/2018] [Accepted: 10/19/2018] [Indexed: 11/19/2022]
Abstract
Sirtuin 5 (SIRT5) belongs to the mitochondrial sirtuin family, which constitutes a highly conserved family of nicotinamide adenine dinucleotide NAD+-dependent deacetylases and ADP-ribosyltransferases that play important regulatory roles in stress resistance and metabolic homeostasis. SIRT5 was shown to have deacetylase, desuccinylase, and demalonylase activities. However, the mechanisms regulating SIRT5 transcription remain unclear. To explore the molecular regulation of bovine SIRT5 expression, we obtained a 500-base pair fragment of the 5'-regulatory region of bovine SIRT5 by molecular cloning, which contained a region with 3 CpG islands. Electrophoretic mobility shift assays and luciferase reporter assays revealed the E2F transcription factor 4 (E2F4) and Kruppel-like factor 6 (KLF6) binding sites as transcriptional activators or repressors in the promoter region of SIRT5. We further verified that E2F4 and KLF6 bind to the SIRT5 promoter by chromatin immunoprecipitation assays. Additionally, methylation and luciferase report assays showed that SIRT5 promoter activity was enhanced by demethylation, and transcriptional activation by E2F4 and transcriptional inhibition by KLF6 of SIRT5 expression was strengthened by demethylation during adipocytes differentiation. This study focused on the mechanism underlying the methylation and transcriptional regulation of SIRT5 expression in bovine adipocytes.
Collapse
Affiliation(s)
- Jieyun Hong
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xiaoyu Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Chugang Mei
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China; National Beef Cattle Improvement Center of Northwest A&F University, Yangling 712100, China
| | - Linsen Zan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China; National Beef Cattle Improvement Center of Northwest A&F University, Yangling 712100, China.
| |
Collapse
|
35
|
Bus P, Gerrits T, Heemskerk SAC, Zandbergen M, Wolterbeek R, Bruijn JA, Baelde HJ, Scharpfenecker M. Endoglin Mediates Vascular Endothelial Growth Factor-A-Induced Endothelial Cell Activation by Regulating Akt Signaling. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:2924-2935. [PMID: 30248336 DOI: 10.1016/j.ajpath.2018.08.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 07/26/2018] [Accepted: 08/02/2018] [Indexed: 12/24/2022]
Abstract
In diabetic nephropathy, differential expression of growth factors leads to vascular changes, including endothelial cell activation, monocyte infiltration, and inflammation. Endoglin plays an important role in endothelial function and is also associated with inflammation. In the kidney, vascular endoglin expression is increased in animal models of renal injury, where it contributes to disease severity, possibly by promoting endothelial cell activation and inflammation. Herein, we investigated whether endoglin expression is associated with diabetic nephropathy. In addition, we examined whether reducing endothelial endoglin expression in vitro affects endothelial cell activation and monocyte adhesion and, if so, which intracellular pathways are involved. Finally, we analyzed whether glomerular endoglin expression is correlated with endothelial cell activation in patients with diabetic nephropathy. Endoglin levels were significantly increased in mice with type 1 diabetes compared with control mice. Reducing endoglin expression in cultured endothelial cells significantly impaired the vascular endothelial growth factor-A-induced up-regulation of activation markers and monocyte adhesion. This was mediated by increased phosphorylation of Akt, thereby inhibiting activating transcription factor 2 phosphorylation, which regulates vascular cell adhesion molecule-1 (VCAM1) gene transcription in these cells. Last, endoglin colocalized with VCAM-1 in the glomeruli of diabetic patients, glomerular VCAM-1 expression was significantly increased in these patients, and this increase in VCAM-1 expression was correlated with increased glomerular endoglin expression. Thus, targeting endoglin function may have therapeutic value in patients at risk for diabetic nephropathy.
Collapse
Affiliation(s)
- Pascal Bus
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands.
| | - Tessa Gerrits
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Sharon A C Heemskerk
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Malu Zandbergen
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ron Wolterbeek
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, Leiden, the Netherlands
| | - Jan A Bruijn
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Hans J Baelde
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | | |
Collapse
|
36
|
Fan Y, Lu H, Liang W, Hu W, Zhang J, Chen YE. Krüppel-like factors and vascular wall homeostasis. J Mol Cell Biol 2018; 9:352-363. [PMID: 28992202 DOI: 10.1093/jmcb/mjx037] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 08/22/2017] [Indexed: 12/19/2022] Open
Abstract
Cardiovascular diseases (CVDs) are major causes of death worldwide. Identification of promising targets for prevention and treatment of CVDs is paramount in the cardiovascular field. Numerous transcription factors regulate cellular function through modulation of specific genes and thereby are involved in the physiological and pathophysiological processes of CVDs. Although Krüppel-like factors (KLFs) have a similar protein structure with a conserved zinc finger domain, they possess distinct tissue and cell distribution patterns as well as biological functions. In the vascular system, KLF activities are regulated at both transcriptional and posttranscriptional levels. Growing in vitro, in vivo, and genetic epidemiology studies suggest that specific KLFs play important roles in vascular wall biology, which further affect vascular diseases. KLFs regulate various functional aspects such as cell growth, differentiation, activation, and development through controlling a whole cluster of functionally related genes and modulating various signaling pathways in response to pathological conditions. Therapeutic targeting of selective KLF family members may be desirable to achieve distinct treatment effects in the context of various vascular diseases. Further elucidation of the association of KLFs with human CVDs, their underlying molecular mechanisms, and precise protein structure studies will be essential to define KLFs as promising targets for therapeutic interventions in CVDs.
Collapse
Affiliation(s)
- Yanbo Fan
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Haocheng Lu
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Wenying Liang
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Wenting Hu
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Jifeng Zhang
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Y Eugene Chen
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| |
Collapse
|
37
|
Plumitallo S, Ruiz-Llorente L, Langa C, Morini J, Babini G, Cappelletti D, Scelsi L, Greco A, Danesino C, Bernabeu C, Olivieri C. Functional analysis of a novel ENG variant in a patient with hereditary hemorrhagic telangiectasia (HHT) identifies a new Sp1 binding-site. Gene 2018; 647:85-92. [PMID: 29305977 DOI: 10.1016/j.gene.2018.01.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 12/01/2017] [Accepted: 01/02/2018] [Indexed: 10/18/2022]
Abstract
Hereditary Hemorrhagic Telangiectasia (HHT) is a rare disease, with an autosomal dominant inheritance and a worldwide incidence of about 1: 5000 individuals. In >80% of patients, HHT is caused by mutations in either ENG or ACVRL1, which code for ENDOGLIN and Activin A Receptor Type II-Like Kinase 1 (ALK1), belonging to the TGF-β/BMP signalling pathway. Typical HHT clinical features are mucocutaneous telangiectases, arteriovenous malformations, spontaneous and recurrent epistaxis, as well as gastrointestinal bleedings. An additional, but less frequent, clinical manifestation in some HHT patients is the presence of Pulmonary Arterial Hypertension (PAH). The aim of this work is to describe the functional role of a novel ENG intronic variant found in a patient affected by both HHT and PAH, in order to assess whether it has a pathogenic role. We proved that the variant lies in a novel binding-site for the transcription factor Sp1, known to be involved in the regulation of ENG and ACVRL1 transcription. We confirmed a pathogenic role for this intronic variant, as it significantly reduces ENG transcription by affecting this novel Sp1 binding-site.
Collapse
Affiliation(s)
- Sara Plumitallo
- Molecular Medicine Department, General Biology and Medical Genetics Unit, University of Pavia, Via Forlanini 14, 27100 Pavia, Italy.
| | - Lidia Ruiz-Llorente
- Centro de Investigaciones Biológicas - Consejo Superior de Investigaciones Científicas and Centro de Investigación Biomédica en Red de Enfermedades Raras, Calle Ramiro de Maeztu 9, 28040 Madrid, Spain.
| | - Carmen Langa
- Centro de Investigaciones Biológicas - Consejo Superior de Investigaciones Científicas and Centro de Investigación Biomédica en Red de Enfermedades Raras, Calle Ramiro de Maeztu 9, 28040 Madrid, Spain.
| | - Jacopo Morini
- Physics Department, Radiation Biophysics and Radiobiology Lab, University of Pavia, Via Bassi 6, 27100 Pavia, Italy.
| | - Gabriele Babini
- Physics Department, Radiation Biophysics and Radiobiology Lab, University of Pavia, Via Bassi 6, 27100 Pavia, Italy.
| | - Donata Cappelletti
- Coronary Care Unit and Laboratory of Clinical and Experimental Cardiology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.
| | - Laura Scelsi
- Cardiothoracic-Vascular Department, Cardiology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.
| | - Alessandra Greco
- Cardiothoracic-Vascular Department, Cardiology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.
| | - Cesare Danesino
- Molecular Medicine Department, General Biology and Medical Genetics Unit, University of Pavia, Via Forlanini 14, 27100 Pavia, Italy; Genetic Counselling Service, IRCCS Fondazione Policlinico San Matteo, Piazzale Golgi 2, 27100 Pavia, Italy.
| | - Carmelo Bernabeu
- Centro de Investigaciones Biológicas - Consejo Superior de Investigaciones Científicas and Centro de Investigación Biomédica en Red de Enfermedades Raras, Calle Ramiro de Maeztu 9, 28040 Madrid, Spain.
| | - Carla Olivieri
- Molecular Medicine Department, General Biology and Medical Genetics Unit, University of Pavia, Via Forlanini 14, 27100 Pavia, Italy.
| |
Collapse
|
38
|
Sweet DR, Fan L, Hsieh PN, Jain MK. Krüppel-Like Factors in Vascular Inflammation: Mechanistic Insights and Therapeutic Potential. Front Cardiovasc Med 2018; 5:6. [PMID: 29459900 PMCID: PMC5807683 DOI: 10.3389/fcvm.2018.00006] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 01/17/2018] [Indexed: 12/19/2022] Open
Abstract
The role of inflammation in vascular disease is well recognized, involving dysregulation of both circulating immune cells as well as the cells of the vessel wall itself. Unrestrained vascular inflammation leads to pathological remodeling that eventually contributes to atherothrombotic disease and its associated sequelae (e.g., myocardial/cerebral infarction, embolism, and critical limb ischemia). Signaling events during vascular inflammation orchestrate widespread transcriptional programs that affect the functions of vascular and circulating inflammatory cells. The Krüppel-like factors (KLFs) are a family of transcription factors central in regulating vascular biology in states of homeostasis and disease. Given their abundance and diversity of function in cells associated with vascular inflammation, understanding the transcriptional networks regulated by KLFs will further our understanding of the pathogenesis underlying several pervasive health concerns (e.g., atherosclerosis, stroke, etc.) and consequently inform the treatment of cardiovascular disease. Within this review, we will discuss the role of KLFs in coordinating protective and deleterious responses during vascular inflammation, while addressing the potential targeting of these critical transcription factors in future therapies.
Collapse
Affiliation(s)
- David R Sweet
- Case Cardiovascular Research Institute, Case Western Reserve University, Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, United States.,Department of Pathology, Case Western Reserve University, Cleveland, OH, United States
| | - Liyan Fan
- Case Cardiovascular Research Institute, Case Western Reserve University, Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, United States.,Department of Pathology, Case Western Reserve University, Cleveland, OH, United States
| | - Paishiun N Hsieh
- Case Cardiovascular Research Institute, Case Western Reserve University, Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, United States.,Department of Pathology, Case Western Reserve University, Cleveland, OH, United States
| | - Mukesh K Jain
- Case Cardiovascular Research Institute, Case Western Reserve University, Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| |
Collapse
|
39
|
Ruiz-Llorente L, Gallardo-Vara E, Rossi E, Smadja DM, Botella LM, Bernabeu C. Endoglin and alk1 as therapeutic targets for hereditary hemorrhagic telangiectasia. Expert Opin Ther Targets 2017; 21:933-947. [PMID: 28796572 DOI: 10.1080/14728222.2017.1365839] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
INTRODUCTION Hereditary Haemorrhagic Telangiectasia (HHT) is as an autosomal dominant trait characterized by frequent nose bleeds, mucocutaneous telangiectases, arteriovenous malformations (AVMs) of the lung, liver and brain, and gastrointestinal bleedings due to telangiectases. HHT is originated by mutations in genes whose encoded proteins are involved in the transforming growth factor β (TGF-β) family signalling of vascular endothelial cells. In spite of the great advances in the diagnosis as well as in the molecular, cellular and animal models of HHT, the current treatments remain just at the palliative level. Areas covered: Pathogenic mutations in genes coding for the TGF-β receptors endoglin (ENG) (HHT1) or the activin receptor-like kinase-1 (ACVRL1 or ALK1) (HHT2), are responsible for more than 80% of patients with HHT. Therefore, ENG and ALK1 are the main potential therapeutic targets for HHT and the focus of this review. The current status of the preclinical and clinical studies, including the anti-angiogenic strategy, have been addressed. Expert opinion: Endoglin and ALK1 are attractive therapeutic targets in HHT. Because haploinsufficiency is the pathogenic mechanism in HHT, several therapeutic approaches able to enhance protein expression and/or function of endoglin and ALK1 are keys to find novel and efficient treatments for the disease.
Collapse
Affiliation(s)
- Lidia Ruiz-Llorente
- a Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) , Madrid , Spain
| | - Eunate Gallardo-Vara
- a Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) , Madrid , Spain
| | - Elisa Rossi
- b Faculté de Pharmacie , Paris Descartes University, Sorbonne Paris Cité and Inserm UMR-S1140 , Paris , France
| | - David M Smadja
- b Faculté de Pharmacie , Paris Descartes University, Sorbonne Paris Cité and Inserm UMR-S1140 , Paris , France
| | - Luisa M Botella
- a Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) , Madrid , Spain
| | - Carmelo Bernabeu
- a Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) , Madrid , Spain
| |
Collapse
|
40
|
Harbi S, Park H, Gregory M, Lopez P, Chiriboga L, Mignatti P. Arrested Development: Infantile Hemangioma and the Stem Cell Teratogenic Hypothesis. Lymphat Res Biol 2017; 15:153-165. [PMID: 28520518 DOI: 10.1089/lrb.2016.0030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Early-life programming is defined by the adaptive changes made by the fetus in response to an adverse in utero environment. Infantile hemangioma (IH), a vascular anomaly, is the most common tumor of infancy. Here we take IH as the tumor model to propose the stem cell teratogenic hypothesis of tumorigenesis and the potential involvement of the immune system. OBJECTIVES Teratogenic agents include chemicals, heavy metals, pathogens, and ionizing radiation. To investigate the etiology and pathogenesis of IH, we hypothesized that they result from a teratogenic mechanism. Immature, incompletely differentiated, dysregulated progenitor cells (multipotential stem cells) are arrested in development with vasculogenic, angiogenic, and tumorigenic potential due to exposure to teratogenic agents such as extrinsic factors that disrupt intrinsic factors via molecular mimicry. During the critical period of immunological tolerance, environmental exposure to immunotoxic agents may harness the teratogenic potential in the developing embryo or fetus and modify the early-life programming algorithm by altering normal fetal development, causing malformations, and inducing tumorigenesis. Specifically, exposure to environmental agents may interfere with physiological signaling pathways and contribute to the generation of IH, by several mechanisms. DISCUSSION An adverse in utero environment no longer serves as a sustainable environment for proper embryogenesis and normal development. Targeted disruption of stem cells by extrinsic factors can alter the genetic program. CONCLUSIONS This article offers new perspectives to stimulate discussion, explore novel experimental approaches (such as immunotoxicity/vasculotoxicity assays and novel isogenic models), and to address the questions raised to convert the hypotheses into nontoxic, noninvasive treatments.
Collapse
Affiliation(s)
| | - Hannah Park
- 2 Department of Epidemiology, University of California , Irvine, School of Medicine, Irvine, California
| | - Michael Gregory
- 3 Department of Pathology, New York University School of Medicine , New York, New York
| | - Peter Lopez
- 3 Department of Pathology, New York University School of Medicine , New York, New York
| | - Luis Chiriboga
- 3 Department of Pathology, New York University School of Medicine , New York, New York
| | - Paolo Mignatti
- 4 Department of Medicine, New York University School of Medicine , New York, New York.,5 Department of Cell Biology, New York University School of Medicine , New York, New York
| |
Collapse
|
41
|
|
42
|
Plasma Soluble Endoglin Levels Are Inversely Associated With the Severity of Coronary Atherosclerosis—Brief Report. Arterioscler Thromb Vasc Biol 2017; 37:49-52. [DOI: 10.1161/atvbaha.116.308494] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Accepted: 10/06/2016] [Indexed: 11/16/2022]
Abstract
Objective—
Transforming growth factor-β inhibits migration and proliferation of endothelial and smooth muscle cells. Endoglin is a transmembrane receptor for transforming growth factor-β1 and transforming growth factor-β3. Endoglin is released into blood as a soluble form (soluble endoglin [sEng]), but plasma sEng levels in patients with coronary artery disease (CAD) have not been elucidated.
Approach and Results—
We measured plasma sEng levels in 244 patients undergoing coronary angiography. The severity of coronary atherosclerosis was evaluated as the numbers of >50% stenotic vessels and segments. CAD was found in 147 patients, of whom 55 had 1-vessel, 42 had 2-vessel, and 50 had 3-vessel disease. Compared with 97 patients without CAD, 147 with CAD had lower sEng levels (median 4.04 versus 4.37 ng/mL;
P
<0.005). A stepwise decrease in sEng levels was found based on the number of stenotic vessels: 4.37 in CAD(−), 4.23 in 1-vessel, 4.13 in 2-vessel, and 3.74 ng/mL in 3-vessel disease (
P
<0.005). sEng levels inversely correlated with the number of stenotic segments (
r
=−0.25;
P
<0.001). In multivariate analysis, sEng was an independent factor for 3-vessel disease and CAD. Odds ratios for CAD and 3-vessel disease were 0.97 (95% confidence interval, 0.95–0.99;
P
<0.02) and 0.96 (95% confidence interval, 0.93–0.99;
P
<0.01) for a 0.1 ng/mL increase in sEng levels, respectively.
Conclusions—
Plasma sEng levels were low in patients with CAD, especially 3-vessel disease, and were inversely associated with the severity of coronary atherosclerosis.
Collapse
|
43
|
Harbi S, Wang R, Gregory M, Hanson N, Kobylarz K, Ryan K, Deng Y, Lopez P, Chiriboga L, Mignatti P. Infantile Hemangioma Originates From A Dysregulated But Not Fully Transformed Multipotent Stem Cell. Sci Rep 2016; 6:35811. [PMID: 27786256 PMCID: PMC5081534 DOI: 10.1038/srep35811] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 10/05/2016] [Indexed: 12/11/2022] Open
Abstract
Infantile hemangioma (IH) is the most common tumor of infancy. Its cellular origin and biological signals for uncontrolled growth are poorly understood, and specific pharmacological treatment is unavailable. To understand the process of hemangioma-genesis we characterized the progenitor hemangioma-derived stem cell (HemSC) and its lineage and non-lineage derivatives. For this purpose we performed a high-throughput (HT) phenotypic and gene expression analysis of HemSCs, and analyzed HemSC-derived tumorspheres. We found that IH is characterized by high expression of genes involved in vasculogenesis, angiogenesis, tumorigenesis and associated signaling pathways. These results show that IH derives from a dysregulated stem cell that remains in an immature, arrested stage of development. The potential biomarkers we identified can afford the development of diagnostic tools and precision-medicine therapies to "rewire" or redirect cellular transitions at an early stage, such as signaling pathways or immune response modifiers.
Collapse
Affiliation(s)
- Shaghayegh Harbi
- Department of Medicine, New York University School of Medicine, 550 First Avenue New York, NY 10016, USA
- VasculoTox Inc., New York, NY 10001, USA
| | - Rong Wang
- Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Michael Gregory
- Department of Pathology, New York University School of Medicine, 550 First Avenue New York, NY 10016, USA
| | - Nicole Hanson
- Department of Pathology, New York University School of Medicine, 550 First Avenue New York, NY 10016, USA
| | - Keith Kobylarz
- Department of Pathology, New York University School of Medicine, 550 First Avenue New York, NY 10016, USA
- Pfizer Inc., Pearl River, NY 10965, USA
| | - Kamilah Ryan
- Department of Pathology, New York University School of Medicine, 550 First Avenue New York, NY 10016, USA
| | - Yan Deng
- Department of Pathology, New York University School of Medicine, 550 First Avenue New York, NY 10016, USA
| | - Peter Lopez
- Department of Pathology, New York University School of Medicine, 550 First Avenue New York, NY 10016, USA
| | - Luis Chiriboga
- Department of Pathology, New York University School of Medicine, 550 First Avenue New York, NY 10016, USA
| | - Paolo Mignatti
- Department of Medicine, New York University School of Medicine, 550 First Avenue New York, NY 10016, USA
- Department of Cell Biology, New York University School of Medicine, 550 First Avenue New York, NY 10016, USA
| |
Collapse
|
44
|
Racca AC, Ridano ME, Bandeira CL, Bevilacqua E, Avvad Portari E, Genti-Raimondi S, Graham CH, Panzetta-Dutari GM. Low oxygen tension induces Krüppel-Like Factor 6 expression in trophoblast cells. Placenta 2016; 45:50-7. [PMID: 27577710 DOI: 10.1016/j.placenta.2016.07.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 07/06/2016] [Accepted: 07/25/2016] [Indexed: 12/11/2022]
Abstract
The transcription factor Krüppel-Like Factor 6 (KLF6) has important roles in cell differentiation, angiogenesis, apoptosis, and proliferation. Furthermore, there is evidence that KLF6 is required for proper placental development. While oxygen is a critical mediator of trophoblast differentiation and function, the involvement of oxygen in the regulation of KLF6 expression remains unexplored. In the present study we examined the expression of KLF6 in placental tissue from uncomplicated and preeclamptic pregnancies, the latter often characterized by an inadequately perfused placenta. We also determined the effect of hypoxia and the involvement of Hypoxia-Inducible Factor 1α (HIF-1α) on the expression of KLF6 in cultured trophoblast cells and placental tissues. Results revealed that villous, interstitial and endovascular extravillous cytotrophoblasts from placentas from normal and preeclamptic pregnancies express KLF6. In addition, KLF6 immunoreactivity was higher in the placental bed of preeclamptic pregnancies than in those of uncomplicated pregnancies. We demonstrated that hypoxia induced an early and transient increase in KLF6 protein levels in HTR8/SVneo extravillous cytotrophoblast cells and in placental explants. Reoxygenation returned KLF6 protein to basal levels. Moreover, hypoxia-induced up-regulation of KLF6 expression was dependent on HIF-1α as revealed by siRNA knockdown in HTR8/SVneo cells. These results indicate that KLF6 may mediate some of the effects of hypoxia in placental development. The regulation of KLF6 protein levels by oxygen has significant implications for understanding its putative role in diseases affected by tissue hypoxia.
Collapse
Affiliation(s)
- A C Racca
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - M E Ridano
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - C L Bandeira
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, Brazil
| | - E Bevilacqua
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, Brazil
| | - E Avvad Portari
- Department of Pathology at Medical Sciences School, State University of Rio de Janeiro, Brazil
| | - S Genti-Raimondi
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - C H Graham
- Departments of Biomedical and Molecular Sciences and Urology, Queen's University, Kingston, Ontario, Canada
| | - G M Panzetta-Dutari
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.
| |
Collapse
|
45
|
Kong LM, Yao L, Lu N, Dong YL, Zhang J, Wang YQ, Liu L, Zhang HL, Huang JG, Liao CG. Interaction of KLF6 and Sp1 regulates basigin-2 expression mediated proliferation, invasion and metastasis in hepatocellular carcinoma. Oncotarget 2016; 7:27975-87. [PMID: 27057625 PMCID: PMC5053703 DOI: 10.18632/oncotarget.8564] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 03/26/2016] [Indexed: 02/07/2023] Open
Abstract
Accumulating evidence suggests that the tumor suppressor gene Krüppel-like factor 6 (KLF6) plays important roles in both development and progression of cancer. However, the role of KLF6 in hepatocellular carcinoma (HCC) remains unclear. Cancer-related molecule basigin-2 plays an important role in HCC progression and metastasis. Sp1, one of Sp/KLFs family members, regulates basigin-2 expression in HCC. The involvement of KLFs in basigin-2 regulation and HCC progression and metastasis has not been investigated. We first measured KLF6 expression levels in 50 pairs of HCC and adjacent normal tissues (ANTs) by immunohistochemistry. Specifically, low KLF6 expression but high Sp1 and basigin-2 expression were found in HCC tissues. By contrast, the ANTs showed high KLF6 expression but low Sp1 and basigin-2 expression. Kaplan-Meier analysis showed that higher expression of KLF6 was associated with better overall survival. The survival rate of KLF6-negative patients was lower than that of KLF6-positive patients (P = 0.015). We also found that KLF6 binds to the basigin-2 and Sp1 promoters and decreases their expression. Thus, we identified a microcircuitry mechanism in which KLF6 can repress basigin-2 expression directly by binding to its promoter or indirectly by inhibiting the expression of the transcription factor Sp1 to block gene expression. Additionally, overexpression of KLF6 suppressed the invasion, metastasis and proliferation of HCC cells in vitro and in vivo by targeting basigin-2. Our study provides new evidence that interaction of KLF6 and Sp1 regulates basigin-2 expression in HCC and that KLF6 represses the invasive and metastatic capacities of HCC through basigin-2.
Collapse
Affiliation(s)
- Ling-Min Kong
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Li Yao
- Department of Pathology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, P. R. China
| | - Ning Lu
- Department of Oncology, Urumqi General Hospital of Lanzhou Military Command of PLA, Urumqi, 830000, P. R. China
| | - Ya-Lu Dong
- Department of Oncology, Urumqi General Hospital of Lanzhou Military Command of PLA, Urumqi, 830000, P. R. China
| | - Jing Zhang
- Department of Oncology, Urumqi General Hospital of Lanzhou Military Command of PLA, Urumqi, 830000, P. R. China
| | - Yong-Qiang Wang
- Department of Oncology, Urumqi General Hospital of Lanzhou Military Command of PLA, Urumqi, 830000, P. R. China
| | - Lili Liu
- Department of Oncology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, P. R. China
- Cancer Institute, Fourth Military Medical University, Xi'an, 710038, P. R. China
| | - He-Long Zhang
- Department of Oncology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, P. R. China
- Cancer Institute, Fourth Military Medical University, Xi'an, 710038, P. R. China
| | - Jian-Guo Huang
- Department of Oncology, Urumqi General Hospital of Lanzhou Military Command of PLA, Urumqi, 830000, P. R. China
| | - Cheng-Gong Liao
- Department of Oncology, Urumqi General Hospital of Lanzhou Military Command of PLA, Urumqi, 830000, P. R. China
- Department of Oncology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, P. R. China
- Cancer Institute, Fourth Military Medical University, Xi'an, 710038, P. R. China
| |
Collapse
|
46
|
Rossi E, Smadja DM, Boscolo E, Langa C, Arevalo MA, Pericacho M, Gamella-Pozuelo L, Kauskot A, Botella LM, Gaussem P, Bischoff J, Lopez-Novoa JM, Bernabeu C. Endoglin regulates mural cell adhesion in the circulatory system. Cell Mol Life Sci 2016; 73:1715-39. [PMID: 26646071 PMCID: PMC4805714 DOI: 10.1007/s00018-015-2099-4] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 11/19/2015] [Accepted: 11/23/2015] [Indexed: 02/06/2023]
Abstract
The circulatory system is walled off by different cell types, including vascular mural cells and podocytes. The interaction and interplay between endothelial cells (ECs) and mural cells, such as vascular smooth muscle cells or pericytes, play a pivotal role in vascular biology. Endoglin is an RGD-containing counter-receptor for β1 integrins and is highly expressed by ECs during angiogenesis. We find that the adhesion between vascular ECs and mural cells is enhanced by integrin activators and inhibited upon suppression of membrane endoglin or β1-integrin, as well as by addition of soluble endoglin (SolEng), anti-integrin α5β1 antibody or an RGD peptide. Analysis of different endoglin mutants, allowed the mapping of the endoglin RGD motif as involved in the adhesion process. In Eng (+/-) mice, a model for hereditary hemorrhagic telangectasia type 1, endoglin haploinsufficiency induces a pericyte-dependent increase in vascular permeability. Also, transgenic mice overexpressing SolEng, an animal model for preeclampsia, show podocyturia, suggesting that SolEng is responsible for podocytes detachment from glomerular capillaries. These results suggest a critical role for endoglin in integrin-mediated adhesion of mural cells and provide a better understanding on the mechanisms of vessel maturation in normal physiology as well as in pathologies such as preeclampsia or hereditary hemorrhagic telangiectasia.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Cell Adhesion/physiology
- Cell Line, Tumor
- Disease Models, Animal
- Endoglin
- Endothelium, Vascular/metabolism
- Female
- Human Umbilical Vein Endothelial Cells/metabolism
- Humans
- Integrin beta1/genetics
- Jurkat Cells
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Nude
- Mice, Transgenic
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/metabolism
- Neovascularization, Pathologic/metabolism
- Pericytes/metabolism
- Podocytes/metabolism
- Pre-Eclampsia/genetics
- Pre-Eclampsia/pathology
- Pregnancy
- Protein Binding
- RNA Interference
- RNA, Small Interfering
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Retina/metabolism
- Telangiectasia, Hereditary Hemorrhagic/genetics
- Telangiectasia, Hereditary Hemorrhagic/pathology
Collapse
Affiliation(s)
- Elisa Rossi
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), c/Ramiro de Maeztu 9, 28040, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28040, Madrid, Spain
- Paris Descartes University, Sorbonne Paris Cite, Paris, France
- Hematology Department, AP-HP, Hôpital Européen Georges Pompidou, Paris, France
| | - David M Smadja
- Hematology Department, AP-HP, Hôpital Européen Georges Pompidou, Paris, France
- Faculté de Pharmacie, Inserm UMR-S1140, Paris, France
| | - Elisa Boscolo
- Department of Surgery, Harvard Medical School, Children's Hospital, Boston, MA, 02115, USA
| | - Carmen Langa
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), c/Ramiro de Maeztu 9, 28040, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28040, Madrid, Spain
| | - Miguel A Arevalo
- Departamento de Anatomía e Histología Humanas, Facultad de Medicina, Universidad de Salamanca, 37007, Salamanca, Spain
- Instituto de Investigaciones Biomédicas de Salamanca (IBSAL), 37007, Salamanca, Spain
| | - Miguel Pericacho
- Instituto de Investigaciones Biomédicas de Salamanca (IBSAL), 37007, Salamanca, Spain
- Departamento de Fisiología y Farmacología, Unidad de Fisiopatología Renal y Cardiovascular, Universidad de Salamanca, 37007, Salamanca, Spain
| | - Luis Gamella-Pozuelo
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), c/Ramiro de Maeztu 9, 28040, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28040, Madrid, Spain
- Departamento de Fisiología y Farmacología, Unidad de Fisiopatología Renal y Cardiovascular, Universidad de Salamanca, 37007, Salamanca, Spain
| | - Alexandre Kauskot
- Inserm UMR-S1176, Le Kremlin Bicêtre, Paris, France
- Université Paris Sud, Le Kremlin Bicêtre, Paris, France
| | - Luisa M Botella
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), c/Ramiro de Maeztu 9, 28040, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28040, Madrid, Spain
| | - Pascale Gaussem
- Hematology Department, AP-HP, Hôpital Européen Georges Pompidou, Paris, France
- Faculté de Pharmacie, Inserm UMR-S1140, Paris, France
| | - Joyce Bischoff
- Department of Surgery, Harvard Medical School, Children's Hospital, Boston, MA, 02115, USA
| | - José M Lopez-Novoa
- Instituto de Investigaciones Biomédicas de Salamanca (IBSAL), 37007, Salamanca, Spain
- Departamento de Fisiología y Farmacología, Unidad de Fisiopatología Renal y Cardiovascular, Universidad de Salamanca, 37007, Salamanca, Spain
| | - Carmelo Bernabeu
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), c/Ramiro de Maeztu 9, 28040, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28040, Madrid, Spain.
| |
Collapse
|
47
|
Transcription factor KLF6 upregulates expression of metalloprotease MMP14 and subsequent release of soluble endoglin during vascular injury. Angiogenesis 2016; 19:155-71. [PMID: 26850053 PMCID: PMC4819519 DOI: 10.1007/s10456-016-9495-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Accepted: 01/23/2016] [Indexed: 12/31/2022]
Abstract
After endothelial injury, the transcription factor Krüppel-like factor 6 (KLF6) translocates into the cell nucleus to regulate a variety of target genes involved in angiogenesis, vascular repair and remodeling, including components of the membrane transforming growth factor beta (TGF-β) receptor complex such as endoglin and activin receptor-like kinase 1. The membrane metalloproteinase 14 (MMP14 or MT1-MMP) targets endoglin to release soluble endoglin and is involved in vascular inflammation and endothelial tubulogenesis. However, little is known about the regulation of MMP14 expression during vascular wounding. In vitro denudation of monolayers of human endothelial cell monolayers leads to an increase in the KLF6 gene transcriptional rate, followed by an upregulation of MMP14 and release of soluble endoglin. Concomitant with this process, MMP14 co-localizes with endoglin in the sprouting endothelial cells surrounding the wound border. MMP14 expression at mRNA and protein levels is increased by ectopic KLF6 and downregulated by KLF6 suppression in cultured endothelial cells. Moreover, after wire-induced endothelial denudation, Klf6+/− mice show lower levels of MMP14 in their vasculature compared with their wild-type siblings. Ectopic cellular expression of KLF6 results in an increased transcription rate of MMP14, and chromatin immunoprecipitation assays show that KLF6 interacts with MMP14 promoter in ECs, this interaction being enhanced during wound healing. Furthermore, KLF6 markedly increases the transcriptional activity of different reporter constructs of MMP14 gene promoter. These results suggest that KLF6 regulates MMP14 transcription and is a critical player of the gene expression network triggered during endothelial repair.
Collapse
|
48
|
Hu X, Fang Y, Li YK, Liu WK, Li H, Ma L, You C. Role of Endoglin Insertion and rs1800956 Polymorphisms in Intracranial Aneurysm Susceptibility: A Meta-Analysis. Medicine (Baltimore) 2015; 94:e1847. [PMID: 26559253 PMCID: PMC4912247 DOI: 10.1097/md.0000000000001847] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Endoglin is an essential molecule during angiogenesis, vascular development, and integrity. Till now, many studies have investigated the association between endoglin polymorphisms and intracranial aneurysm (IA) risk, with the results remained inconclusive. Therefore, we performed a meta-analysis to summarize the possible association.We searched PubMed and Embase until June 2015 to identify studies addressing the association between endoglin polymorphisms and IA risk. The summary odds ratios (ORs) and their corresponding 95% confidence interval (CI) were calculated to assess the strength of the association.Eleven studies with a total of 1501 cases and 2012 controls were finally included in this meta-analysis, with 10 studies investigating endoglin 6-bp insertion (6bINS) polymorphism and 4 studies investigating 1800956 polymorphism. No significant association between endoglin 6bINS polymorphism and IA risk was detected in overall estimation (I/I vs wt/I + wt/wt: OR = 1.21, 95% CI = 0.87-1.69) or in the subgroup analysis by ethnicity, control source, or ruptured status. However, we observed an association with borderline significance of 6bINS with IA occurrence (I/I vs wt/I + wt/wt: OR = 1.49, 95% CI = 0.99-2.25, P = 0.058) in studies applying matched controls. Furthermore, we detected a significant association for 6bINS polymorphism of endoglin with increased risk of familial IA (I vs wt, OR = 1.64, 95% CI = 1.10-2.42) but not sporadic IA (I vs wt, OR = 1.09, 95% CI = 0.68-1.45). With regard to rs1800956, our pooled results indicated a significantly decreased IA risk in individuals carrying C allele (C/C vs G/C + G/G: OR = 0.65; 95% CI = 0.45-0.94).This meta-analysis provided no evidence for the association between 6bINS polymorphism with overall IA risk. However, we detected a significant association of 6bINS allele with increased risk of familial IA. Also, we found that rs1800956 was significantly related to IA occurrence. Further, well-designed studies with large sample size are warranted and updated meta-analysis is needed to verify our findings.
Collapse
Affiliation(s)
- Xin Hu
- From the Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | | | | | | | | | | | | |
Collapse
|
49
|
Conroy AL, Gélvez M, Hawkes M, Rajwans N, Tran V, Liles WC, Villar-Centeno LA, Kain KC. Host biomarkers are associated with progression to dengue haemorrhagic fever: a nested case-control study. Int J Infect Dis 2015; 40:45-53. [PMID: 26255888 DOI: 10.1016/j.ijid.2015.07.027] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Revised: 07/28/2015] [Accepted: 07/30/2015] [Indexed: 01/19/2023] Open
Abstract
OBJECTIVES Dengue represents the most important arboviral infection worldwide. Onset of circulatory collapse can be unpredictable. Biomarkers that can identify individuals at risk of plasma leakage may facilitate better triage and clinical management. DESIGN Using a nested case-control design, we randomly selected subjects from a prospective cohort study of dengue in Colombia (n=1582). Using serum collected within 96 hours of fever onset, we tested 19 biomarkers by ELISA in cases (developed dengue hemorrhagic fever or dengue shock syndrome (DHF/DSS); n=46), and controls (uncomplicated dengue fever (DF); n=65) and healthy controls (HC); n=15. RESULTS Ang-1 levels were lower and angptl3, sKDR, sEng, sICAM-1, CRP, CXCL10/IP-10, IL-18 binding protein, CHI3L1, C5a and Factor D levels were increased in dengue compared to HC. sICAM-1, sEng and CXCL10/IP-10 were further elevated in subjects who subsequently developed DHF/DSS (p=0.008, p=0.028 and p=0.025, respectively). In a logistic regression model, age (odds ratio (OR) (95% CI): 0.95 (0.92-0.98), p=0.001), hyperesthesia/hyperalgesia (OR; 3.8 (1.4-10.4), p=0.008) and elevated sICAM-1 (>298ng/mL: OR; 6.3 (1.5-25.7), p=0.011) at presentation were independently associated with progression to DHF/DSS. CONCLUSIONS These results suggest that inflammation and endothelial activation are important pathways in the pathogenesis of dengue and sICAM-1 levels may identify individuals at risk of plasma leakage.
Collapse
Affiliation(s)
- Andrea L Conroy
- Sandra A. Rotman Laboratories, Sandra Rotman Centre, University Health Network-Toronto General Hospital, University of Toronto, Toronto, ON, M5G 1L7, Canada.
| | - Margarita Gélvez
- Centro de Investigaciones Epidemiológicas, Facultad de Salud, Universidad Industrial de Santander, Bucaramanga, Colombia.
| | - Michael Hawkes
- Department of Pediatrics, University of Alberta, Edmonton, AB, T6G 2E1, Canada.
| | - Nimerta Rajwans
- Sandra A. Rotman Laboratories, Sandra Rotman Centre, University Health Network-Toronto General Hospital, University of Toronto, Toronto, ON, M5G 1L7, Canada.
| | - Vanessa Tran
- Sandra A. Rotman Laboratories, Sandra Rotman Centre, University Health Network-Toronto General Hospital, University of Toronto, Toronto, ON, M5G 1L7, Canada.
| | - W Conrad Liles
- University of Washington, Department of Medicine, Seattle, WA, 98195, USA.
| | - Luis Angel Villar-Centeno
- Centro de Investigaciones Epidemiológicas, Facultad de Salud, Universidad Industrial de Santander, Bucaramanga, Colombia.
| | - Kevin C Kain
- Sandra A. Rotman Laboratories, Sandra Rotman Centre, University Health Network-Toronto General Hospital, University of Toronto, Toronto, ON, M5G 1L7, Canada; Tropical Disease Unit, Division of Infectious Diseases, Department of Medicine, University of Toronto, Toronto, ON, M5G 2C4, Canada.
| |
Collapse
|
50
|
Functional interplay of SP family members and nuclear factor Y is essential for transcriptional activation of the human Calreticulin gene. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2015; 1849:1188-97. [PMID: 26162987 DOI: 10.1016/j.bbagrm.2015.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 06/18/2015] [Accepted: 07/06/2015] [Indexed: 11/21/2022]
Abstract
Calreticulin (CALR) is a highly conserved, multifunctional protein involved in a variety of cellular processes including the maintenance of intracellular calcium homeostasis, proper protein folding, differentiation and immunogenic cell death. More recently, a crucial role for CALR in the pathogenesis of certain hematologic malignancies was discovered: in clinical subgroups of acute myeloid leukemia, CALR overexpression mediates a block in differentiation, while somatic mutations have been found in the majority of patients with myeloproliferative neoplasms with nonmutated Janus kinase 2 gene (JAK2) or thrombopoietin receptor gene (MPL). However, the mechanisms underlying CALR promoter activation have insufficiently been investigated so far. By dissecting the core promoter region, we could identify a functional TATA-box relevant for transcriptional activation. In addition, we characterized two evolutionary highly conserved cis-regulatory modules (CRMs) within the proximal promoter each composed of one binding site for the transcription factors SP1 and SP3 as well as for the nuclear transcription factor Y (NFY) and we verified binding of these factors to their cognate sites in vitro and in vivo.
Collapse
|