1
|
Zeng X, Qiu R, Peng W. The protective effects of annexin A1 against oxidized-LDL-induced monocytes adhesion to endothelial cells: implication in atherosclerosis. J Thromb Thrombolysis 2024:10.1007/s11239-024-03050-6. [PMID: 39397189 DOI: 10.1007/s11239-024-03050-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/25/2024] [Indexed: 10/15/2024]
Abstract
Oxidized low-density lipoprotein (ox-LDL)-associated endothelial dysfunction is a critical factor in the initiation and progression of Atherosclerosis (AS). Annexin A1 is an important member of the annexin family. Despite its wide range of biological functions across various tissues and cells, the role of Annexin A1 in AS remains largely unexplored. In this study, we demonstrate that Annexin A1 treatment effectively reduced the expression of LOX-1 at both the mRNA and protein levels in HUVECs exposed to ox-LDL. Annexin A1 also ameliorated oxidative stress (OS) by decreasing mitochondrial ROS levels and restoring reduced GSH levels. Moreover, Annexin A1 decreased the expression of pro-inflammatory cytokines, including IL-6 and MCP-1. Importantly, Annexin A1 inhibited ox-LDL-induced expressions of the endothelial adhesion molecules, such as E-selectin and VCAM-1 in HUVECs, which leads to reduced attachment of THP-1 monocytes to HUVECs. Mechanically, we found that Annexin A1 reversed the expression of KLF2 against ox-LDL mediated by the PI3K/Akt axis. Notably, the silencing of KLF2 abrogated the protective effects of Annexin A1 on E-selectin and VCAM-1 expression and the attachment of THP-1 monocytes to HUVECs. Our findings suggest that Annexin A1 is a potential therapeutic agent for atherosclerosis, offering a novel approach to mitigate endothelial dysfunction and inflammation.
Collapse
Affiliation(s)
- Xiaoling Zeng
- Department of Cardiology, Jingzhou Central Hospital, No. 26, Chu Yuan Road, Jingzhou, Hubei, 434020, China.
| | - Ruhui Qiu
- Department of Cardiology, Jingzhou Central Hospital, No. 26, Chu Yuan Road, Jingzhou, Hubei, 434020, China
| | - Wen Peng
- Department of Cardiology, Jingzhou Central Hospital, No. 26, Chu Yuan Road, Jingzhou, Hubei, 434020, China
| |
Collapse
|
2
|
Li Y, Li Z, Ren Y, Lei Y, Yang S, Shi Y, Peng H, Yang W, Guo T, Yu Y, Xiong Y. Mitochondrial-derived peptides in cardiovascular disease: Novel insights and therapeutic opportunities. J Adv Res 2024; 64:99-115. [PMID: 38008175 PMCID: PMC11464474 DOI: 10.1016/j.jare.2023.11.018] [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: 09/20/2023] [Revised: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 11/28/2023] Open
Abstract
BACKGROUND Mitochondria-derived peptides (MDPs) represent a recently discovered family of peptides encoded by short open reading frames (ORFs) found within mitochondrial genes. This group includes notable members including humanin (HN), mitochondrial ORF of the 12S rDNA type-c (MOTS-c), and small humanin-like peptides 1-6 (SHLP1-6). MDPs assume pivotal roles in the regulation of diverse cellular processes, encompassing apoptosis, inflammation, and oxidative stress, which are all essential for sustaining cellular viability and normal physiological functions. Their emerging significance extends beyond this, prompting a deeper exploration into their multifaceted roles and potential applications. AIM OF REVIEW This review aims to comprehensively explore the biogenesis, various types, and diverse functions of MDPs. It seeks to elucidate the central roles and underlying mechanisms by which MDPs participate in the onset and development of cardiovascular diseases (CVDs), bridging the connections between cell apoptosis, inflammation, and oxidative stress. Furthermore, the review highlights recent advancements in clinical research related to the utilization of MDPs in CVD diagnosis and treatment. KEY SCIENTIFIC CONCEPTS OF REVIEW MDPs levels are diminished with aging and in the presence of CVDs, rendering them potential new indicators for the diagnosis of CVDs. Also, MDPs may represent a novel and promising strategy for CVD therapy. In this review, we delve into the biogenesis, various types, and diverse functions of MDPs. We aim to shed light on the pivotal roles and the underlying mechanisms through which MDPs contribute to the onset and advancement of CVDs connecting cell apoptosis, inflammation, and oxidative stress. We also provide insights into the current advancements in clinical research related to the utilization of MDPs in the treatment of CVDs. This review may provide valuable information with MDPs for CVD diagnosis and treatment.
Collapse
Affiliation(s)
- Yang Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Zhuozhuo Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Yuanyuan Ren
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Ying Lei
- School of Medicine, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Silong Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Yuqi Shi
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Han Peng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Weijie Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Tiantian Guo
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Yi Yu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China; School of Medicine, Northwest University, Xi'an 710069, Shaanxi, PR China.
| | - Yuyan Xiong
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China; Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, 710018 Xi'an, Shaanxi, PR China.
| |
Collapse
|
3
|
Joshi D, Coon BG, Chakraborty R, Deng H, Yang Z, Babar MU, Fernandez-Tussy P, Meredith E, Attanasio J, Joshi N, Traylor JG, Orr AW, Fernandez-Hernando C, Libreros S, Schwartz MA. Endothelial γ-protocadherins inhibit KLF2 and KLF4 to promote atherosclerosis. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1035-1048. [PMID: 39232138 PMCID: PMC11399086 DOI: 10.1038/s44161-024-00522-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 07/18/2024] [Indexed: 09/06/2024]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) is the leading cause of mortality worldwide. Laminar shear stress from blood flow, sensed by vascular endothelial cells, protects from ASCVD by upregulating the transcription factors KLF2 and KLF4, which induces an anti-inflammatory program that promotes vascular resilience. Here we identify clustered γ-protocadherins as therapeutically targetable, potent KLF2 and KLF4 suppressors whose upregulation contributes to ASCVD. Mechanistic studies show that γ-protocadherin cleavage results in translocation of the conserved intracellular domain to the nucleus where it physically associates with and suppresses signaling by the Notch intracellular domain. γ-Protocadherins are elevated in human ASCVD endothelium; their genetic deletion or antibody blockade protects from ASCVD in mice without detectably compromising host defense against bacterial or viral infection. These results elucidate a fundamental mechanism of vascular inflammation and reveal a method to target the endothelium rather than the immune system as a protective strategy in ASCVD.
Collapse
Affiliation(s)
- Divyesh Joshi
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
| | - Brian G Coon
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
| | - Raja Chakraborty
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
| | - Hanqiang Deng
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
| | - Ziyu Yang
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, USA
- Department of Pathology, Yale University, New Haven, CT, USA
| | - Muhammad Usman Babar
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, USA
- Department of Pathology, Yale University, New Haven, CT, USA
| | | | - Emily Meredith
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
| | - John Attanasio
- Department of Immunobiology, Yale University, New Haven, CT, USA
| | - Nikhil Joshi
- Department of Immunobiology, Yale University, New Haven, CT, USA
| | - James G Traylor
- Department of Pathology and Translational Pathobiology, LSU Health Shreveport, Shreveport, LA, USA
| | - Anthony Wayne Orr
- Department of Pathology and Translational Pathobiology, LSU Health Shreveport, Shreveport, LA, USA
| | | | - Stephania Libreros
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, USA
- Department of Pathology, Yale University, New Haven, CT, USA
| | - Martin A Schwartz
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA.
- Department of Cell Biology, Yale University, New Haven, CT, USA.
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA.
| |
Collapse
|
4
|
Min L, Zhong F, Gu L, Lee K, He JC. Krüppel-like factor 2 is an endoprotective transcription factor in diabetic kidney disease. Am J Physiol Cell Physiol 2024; 327:C477-C486. [PMID: 38981608 DOI: 10.1152/ajpcell.00222.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/02/2024] [Accepted: 07/02/2024] [Indexed: 07/11/2024]
Abstract
Diabetic kidney disease (DKD) is a microvascular complication of diabetes, and glomerular endothelial cell (GEC) dysfunction is a key driver of DKD pathogenesis. Krüppel-like factor 2 (KLF2), a shear stress-induced transcription factor, is among the highly regulated genes in early DKD. In the kidney, KLF2 expression is mostly restricted to endothelial cells, but its expression is also found in immune cell subsets. KLF2 expression is upregulated in response to increased shear stress by the activation of mechanosensory receptors but suppressed by inflammatory cytokines, both of which characterize the early diabetic kidney milieu. KLF2 expression is reduced in progressive DKD and hypertensive nephropathy in humans and mice, likely due to high glucose and inflammatory cytokines such as TNF-α. However, KLF2 expression is increased in glomerular hyperfiltration-induced shear stress without metabolic dysregulation, such as in settings of unilateral nephrectomy. Lower KLF2 expression is associated with CKD progression in patients with unilateral nephrectomy, consistent with its endoprotective role. KLF2 confers endoprotection by inhibition of inflammation, thrombotic activation, and angiogenesis, and thus KLF2 is considered a protective factor for cardiovascular disease (CVD). Based on similar mechanisms, KLF2 also exhibits renoprotection, and its reduced expression in endothelial cells worsens glomerular injury and albuminuria in settings of diabetes or unilateral nephrectomy. Thus KLF2 confers endoprotective effects in both CVD and DKD, and its activators could potentially be developed as a novel class of drugs for cardiorenal protection in diabetic patients.
Collapse
Affiliation(s)
- Lulin Min
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Medicine/Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Fang Zhong
- Department of Medicine/Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Leyi Gu
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kyung Lee
- Department of Medicine/Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - John Cijiang He
- Department of Medicine/Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
- Renal Section, James J. Peters Veterans Affair Medical Center, Bronx, New York, United States
| |
Collapse
|
5
|
Abello J, Yin Y, Zhao Y, Maurer J, Lee J, Bodell C, Clevenger AJ, Burton Z, Goeckel ME, Lin M, Grainger S, Halabi CM, Raghavan SA, Sah R, Stratman AN. Endothelial cell Piezo1 promotes vascular smooth muscle cell differentiation on large arteries. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.11.598539. [PMID: 38915529 PMCID: PMC11195244 DOI: 10.1101/2024.06.11.598539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Vascular stabilization is a mechanosensitive process, in part driven by blood flow. Here, we demonstrate the involvement of the mechanosensitive ion channel, Piezo1, in promoting arterial accumulation of vascular smooth muscle cells (vSMCs) during zebrafish development. Using a series of small molecule antagonists or agonists to temporally regulate Piezo1 activity, we identified a role for the Piezo1 channel in regulating klf2a levels and altered targeting of vSMCs between arteries and veins. Increasing Piezo1 activity suppressed klf2a and increased vSMC association with the cardinal vein, while inhibition of Piezo1 activity increased klf2a levels and decreased vSMC association with arteries. We supported the small molecule data with in vivo genetic suppression of piezo1 and 2 in zebrafish, resulting in loss of transgelin+ vSMCs on the dorsal aorta. Further, endothelial cell (EC)-specific Piezo1 knockout in mice was sufficient to decrease vSMC accumulation along the descending dorsal aorta during development, thus phenocopying our zebrafish data, and supporting functional conservation of Piezo1 in mammals. To determine mechanism, we used in vitro modeling assays to demonstrate that differential sensing of pulsatile versus laminar flow forces across endothelial cells changes the expression of mural cell differentiation genes. Together, our findings suggest a crucial role for EC Piezo1 in sensing force within large arteries to mediate mural cell differentiation and stabilization of the arterial vasculature.
Collapse
Affiliation(s)
- Javier Abello
- Department of Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Ying Yin
- Department of Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Yonghui Zhao
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, USA
| | - Josh Maurer
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, USA
| | - Jihui Lee
- Department of Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Cherokee Bodell
- Department of Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Abigail J. Clevenger
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Zarek Burton
- Department of Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Megan E. Goeckel
- Department of Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Michelle Lin
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Stephanie Grainger
- Department of Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | - Carmen M. Halabi
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Shreya A. Raghavan
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Rajan Sah
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, USA
- Center for Cardiovascular Research, Washington University, St Louis, MO, USA
| | - Amber N. Stratman
- Department of Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| |
Collapse
|
6
|
Berg K, Gorham J, Lundt F, Seidman J, Brueckner M. Endocardial primary cilia and blood flow are required for regulation of EndoMT during endocardial cushion development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.15.594405. [PMID: 38798559 PMCID: PMC11118576 DOI: 10.1101/2024.05.15.594405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Blood flow is critical for heart valve formation, and cellular mechanosensors are essential to translate flow into transcriptional regulation of development. Here, we identify a role for primary cilia in vivo in the spatial regulation of cushion formation, the first stage of valve development, by regionally controlling endothelial to mesenchymal transition (EndoMT) via modulation of Kruppel-like Factor 4 (Klf4) . We find that high shear stress intracardiac regions decrease endocardial ciliation over cushion development, correlating with KLF4 downregulation and EndoMT progression. Mouse embryos constitutively lacking cilia exhibit a blood-flow dependent accumulation of KLF4 in these regions, independent of upstream left-right abnormalities, resulting in impaired cushion cellularization. snRNA-seq revealed that cilia KO endocardium fails to progress to late-EndoMT, retains endothelial markers and has reduced EndoMT/mesenchymal genes that KLF4 antagonizes. Together, these data identify a mechanosensory role for endocardial primary cilia in cushion development through regional regulation of KLF4.
Collapse
|
7
|
He J, Blazeski A, Nilanthi U, Menéndez J, Pirani SC, Levic DS, Bagnat M, Singh MK, Raya JG, García-Cardeña G, Torres-Vázquez J. Plxnd1-mediated mechanosensing of blood flow controls the caliber of the Dorsal Aorta via the transcription factor Klf2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.24.576555. [PMID: 38328196 PMCID: PMC10849625 DOI: 10.1101/2024.01.24.576555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
The cardiovascular system generates and responds to mechanical forces. The heartbeat pumps blood through a network of vascular tubes, which adjust their caliber in response to the hemodynamic environment. However, how endothelial cells in the developing vascular system integrate inputs from circulatory forces into signaling pathways to define vessel caliber is poorly understood. Using vertebrate embryos and in vitro-assembled microvascular networks of human endothelial cells as models, flow and genetic manipulations, and custom software, we reveal that Plexin-D1, an endothelial Semaphorin receptor critical for angiogenic guidance, employs its mechanosensing activity to serve as a crucial positive regulator of the Dorsal Aorta's (DA) caliber. We also uncover that the flow-responsive transcription factor KLF2 acts as a paramount mechanosensitive effector of Plexin-D1 that enlarges endothelial cells to widen the vessel. These findings illuminate the molecular and cellular mechanisms orchestrating the interplay between cardiovascular development and hemodynamic forces.
Collapse
Affiliation(s)
- Jia He
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Adriana Blazeski
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA and Harvard Medical School, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Uthayanan Nilanthi
- Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 8 College Road, Singapore, 169857
| | - Javier Menéndez
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Samuel C. Pirani
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Daniel S. Levic
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Michel Bagnat
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Manvendra K. Singh
- Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 8 College Road, Singapore, 169857
- National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, Singapore, 169609
| | - José G Raya
- Department of Radiology, New York University School of Medicine, New York, NY 10016, USA
| | - Guillermo García-Cardeña
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA and Harvard Medical School, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jesús Torres-Vázquez
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
8
|
Joshi D, Coon BG, Chakraborty R, Deng H, Fernandez-Tussy P, Meredith E, Traylor JG, Orr AW, Fernandez-Hernando C, Schwartz MA. Gamma protocadherins in vascular endothelial cells inhibit Klf2/4 to promote atherosclerosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.16.575958. [PMID: 38293157 PMCID: PMC10827163 DOI: 10.1101/2024.01.16.575958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) is the leading cause of mortality worldwide1. Laminar shear stress (LSS) from blood flow in straight regions of arteries protects against ASCVD by upregulating the Klf2/4 anti-inflammatory program in endothelial cells (ECs)2-8. Conversely, disturbed shear stress (DSS) at curves or branches predisposes these regions to plaque formation9,10. We previously reported a whole genome CRISPR knockout screen11 that identified novel inducers of Klf2/4. Here we report suppressors of Klf2/4 and characterize one candidate, protocadherin gamma A9 (Pcdhga9), a member of the clustered protocadherin gene family12. Pcdhg deletion increases Klf2/4 levels in vitro and in vivo and suppresses inflammatory activation of ECs. Pcdhg suppresses Klf2/4 by inhibiting the Notch pathway via physical interaction of cleaved Notch1 intracellular domain (NICD Val1744) with nuclear Pcdhg C-terminal constant domain (CCD). Pcdhg inhibition by EC knockout (KO) or blocking antibody protects from atherosclerosis. Pcdhg is elevated in the arteries of human atherosclerosis. This study identifies a novel fundamental mechanism of EC resilience and therapeutic target for treating inflammatory vascular disease.
Collapse
Affiliation(s)
- Divyesh Joshi
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT 06511, USA
| | - Brian G. Coon
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT 06511, USA
| | - Raja Chakraborty
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT 06511, USA
| | - Hanqiang Deng
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT 06511, USA
| | - Pablo Fernandez-Tussy
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT 06520, USA
| | - Emily Meredith
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT 06511, USA
| | - James G. Traylor
- Department of Pathology and Translational Pathobiology, LSU Health Shreveport, LA 71103, USA
| | - Anthony Wayne Orr
- Department of Pathology and Translational Pathobiology, LSU Health Shreveport, LA 71103, USA
| | | | - Martin A. Schwartz
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT 06511, USA
- Department of Cell Biology, Yale University, New Haven, CT 06510, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT 06510, USA
| |
Collapse
|
9
|
Rojas-González DM, Babendreyer A, Ludwig A, Mela P. Analysis of flow-induced transcriptional response and cell alignment of different sources of endothelial cells used in vascular tissue engineering. Sci Rep 2023; 13:14384. [PMID: 37658092 PMCID: PMC10474151 DOI: 10.1038/s41598-023-41247-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 08/23/2023] [Indexed: 09/03/2023] Open
Abstract
Endothelialization of tissue-engineered vascular grafts has proven crucial for implant functionality and thus clinical outcome, however, the choice of endothelial cells (ECs) is often driven by availability rather than by the type of vessel to be replaced. In this work we studied the response to flow of different human ECs with the aim of examining whether their response in vitro is dictated by their original in vivo conditions. Arterial, venous, and microvascular ECs were cultured under shear stress (SS) of 0, 0.3, 3, 1, 10, and 30 dyne/cm2 for 24 h. Regulation of flow-induced marker KLF2 was similar across the different ECs. Upregulation of anti-thrombotic markers, TM and TPA, was mainly seen at higher SS. Cell elongation and alignment was observed for the different ECs at 10 and 30 dyne/cm2 while at lower SS cells maintained a random orientation. Downregulation of pro-inflammatory factors SELE, IL8, and VCAM1 and up-regulation of anti-oxidant markers NQO1 and HO1 was present even at SS for which cell alignment was not observed. Our results evidenced similarities in the response to flow among the different ECs, suggesting that the maintenance of the resting state in vitro is not dictated by the SS typical of the tissue of origin and that absence of flow-induced cell orientation does not necessarily correlate with a pro-inflammatory state of the ECs. These results support the use of ECs from easily accessible sources for in vitro vascular tissue engineering independently from the target vessel.
Collapse
Affiliation(s)
- Diana M Rojas-González
- Department of Biohybrid & Medical Textiles (BioTex) at Center of Biohybrid Medical Systems (CBMS), AME-Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstr. 55, 52074, Aachen, Germany
- Chair of Medical Materials and Implants, Department of Mechanical Engineering, School of Engineering and Design and Munich Institute of Biomedical Engineering, Technical University of Munich, Boltzmannstr 15, 85748, Garching, Germany
| | - Aaron Babendreyer
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany.
| | - Andreas Ludwig
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Petra Mela
- Department of Biohybrid & Medical Textiles (BioTex) at Center of Biohybrid Medical Systems (CBMS), AME-Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstr. 55, 52074, Aachen, Germany.
- Chair of Medical Materials and Implants, Department of Mechanical Engineering, School of Engineering and Design and Munich Institute of Biomedical Engineering, Technical University of Munich, Boltzmannstr 15, 85748, Garching, Germany.
| |
Collapse
|
10
|
Tsitsikov EN, Phan KP, Liu Y, Tsytsykova AV, Kinter M, Selland L, Garman L, Griffin C, Dunn IF. TRAF7 is an essential regulator of blood vessel integrity during mouse embryonic and neonatal development. iScience 2023; 26:107474. [PMID: 37583551 PMCID: PMC10424150 DOI: 10.1016/j.isci.2023.107474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/19/2023] [Accepted: 07/21/2023] [Indexed: 08/17/2023] Open
Abstract
Targeted deletion of TRAF7 revealed that it is a crucial part of shear stress-responsive MEKK3-MEK5-ERK5 signaling pathway induced in endothelial cells by blood flow. Similar to Mekk3-, Mek5- or Erk5-deficient mice, Traf7-deficient embryos died in utero around midgestation due to impaired endothelium integrity. They displayed significantly lower expression of transcription factor Klf2, an essential regulator of vascular hemodynamic forces downstream of the MEKK3-MEK-ERK5 signaling pathway. In addition, deletion of Traf7 in endothelial cells of postnatal mice was associated with severe cerebral hemorrhage. Here, we show that besides MEKK3 and MEK5, TRAF7 associates with a planar cell polarity protein SCRIB. SCRIB binds with an N-terminal region of TRAF7, while MEKK3 associates with the C-terminal WD40 domain. Downregulation of TRAF7 as well as SCRIB inhibited fluid shear stress-induced phosphorylation of ERK5 in cultured endothelial cells. These findings suggest that TRAF7 and SCRIB may comprise an upstream part of the MEKK3-MEK5-ERK5 signaling pathway.
Collapse
Affiliation(s)
- Erdyni N. Tsitsikov
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Khanh P. Phan
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Yufeng Liu
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Alla V. Tsytsykova
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Mike Kinter
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Lauren Selland
- Histology, Immunohistochemistry, Microscopy Core-COBRE Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Lori Garman
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Courtney Griffin
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Ian F. Dunn
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
11
|
Kotlyarov S, Kotlyarova A. Participation of Krüppel-like Factors in Atherogenesis. Metabolites 2023; 13:metabo13030448. [PMID: 36984888 PMCID: PMC10052737 DOI: 10.3390/metabo13030448] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/17/2023] [Accepted: 03/18/2023] [Indexed: 03/30/2023] Open
Abstract
Atherosclerosis is an important problem in modern medicine, the keys to understanding many aspects of which are still not available to clinicians. Atherosclerosis develops as a result of a complex chain of events in which many cells of the vascular wall and peripheral blood flow are involved. Endothelial cells, which line the vascular wall in a monolayer, play an important role in vascular biology. A growing body of evidence strengthens the understanding of the multifaceted functions of endothelial cells, which not only organize the barrier between blood flow and tissues but also act as regulators of hemodynamics and play an important role in regulating the function of other cells in the vascular wall. Krüppel-like factors (KLFs) perform several biological functions in various cells of the vascular wall. The large family of KLFs in humans includes 18 members, among which KLF2 and KLF4 are at the crossroads between endothelial cell mechanobiology and immunometabolism, which play important roles in both the normal vascular wall and atherosclerosis.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| | - Anna Kotlyarova
- Department of Pharmacy Management and Economics, Ryazan State Medical University, 390026 Ryazan, Russia
| |
Collapse
|
12
|
Yerra VG, Drosatos K. Specificity Proteins (SP) and Krüppel-like Factors (KLF) in Liver Physiology and Pathology. Int J Mol Sci 2023; 24:4682. [PMID: 36902112 PMCID: PMC10003758 DOI: 10.3390/ijms24054682] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 03/04/2023] Open
Abstract
The liver acts as a central hub that controls several essential physiological processes ranging from metabolism to detoxification of xenobiotics. At the cellular level, these pleiotropic functions are facilitated through transcriptional regulation in hepatocytes. Defects in hepatocyte function and its transcriptional regulatory mechanisms have a detrimental influence on liver function leading to the development of hepatic diseases. In recent years, increased intake of alcohol and western diet also resulted in a significantly increasing number of people predisposed to the incidence of hepatic diseases. Liver diseases constitute one of the serious contributors to global deaths, constituting the cause of approximately two million deaths worldwide. Understanding hepatocyte transcriptional mechanisms and gene regulation is essential to delineate pathophysiology during disease progression. The current review summarizes the contribution of a family of zinc finger family transcription factors, named specificity protein (SP) and Krüppel-like factors (KLF), in physiological hepatocyte functions, as well as how they are involved in the onset and development of hepatic diseases.
Collapse
Affiliation(s)
| | - Konstantinos Drosatos
- Metabolic Biology Laboratory, Cardiovascular Center, Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| |
Collapse
|
13
|
Tong T, Zhou Y, Huang Q, Xiao C, Bai Q, Deng B, Chen L. The regulation roles of miRNAs in Helicobacter pylori infection. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2023:10.1007/s12094-023-03094-9. [PMID: 36781601 DOI: 10.1007/s12094-023-03094-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 01/17/2023] [Indexed: 02/15/2023]
Abstract
Helicobacter pylori is a kind of Gram-negative bacteria that parasitizes on human gastric mucosa. Helicobacter pylori infection is very common in human beings, which often causes gastrointestinal diseases, including chronic gastritis, duodenal ulcer and gastric cancer. MicroRNAs are a group of endogenous non-coding single stranded RNAs, which play an important role in cell proliferation, differentiation, autophagy, apoptosis and inflammation. In recent years, relevant studies have found that the expression of microRNA is changed after Helicobacter pylori infection, and then regulate the biological process of host cells. This paper reviews the regulation role of microRNAs on cell biological behavior through different signal pathways after Helicobacter pylori infection.
Collapse
Affiliation(s)
- Ting Tong
- Department of Public Health Laboratory Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.,Hunan Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.,Hengyang Engineering Technology Research Center, Hengyang, 421001, Hunan, China
| | - You Zhou
- Department of Public Health Laboratory Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.,Hunan Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.,Hengyang Engineering Technology Research Center, Hengyang, 421001, Hunan, China
| | - Qiaoling Huang
- Department of Public Health Laboratory Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.,Hunan Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.,Hengyang Engineering Technology Research Center, Hengyang, 421001, Hunan, China
| | - Cui Xiao
- Department of Public Health Laboratory Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.,Hunan Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.,Hengyang Engineering Technology Research Center, Hengyang, 421001, Hunan, China
| | - Qinqin Bai
- Department of Public Health Laboratory Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.,Hunan Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.,Hengyang Engineering Technology Research Center, Hengyang, 421001, Hunan, China
| | - Bo Deng
- Department of Public Health Laboratory Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.,Hunan Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.,Hengyang Engineering Technology Research Center, Hengyang, 421001, Hunan, China
| | - Lili Chen
- Department of Public Health Laboratory Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China. .,Hunan Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China. .,Hengyang Engineering Technology Research Center, Hengyang, 421001, Hunan, China.
| |
Collapse
|
14
|
Li YZ, Xie J, Wang RQ, Gao XQ, Liu PJ, Liu J. KLF2 is a clinical diagnostic and treatment biomarker of breast cancer. Front Cell Dev Biol 2023; 11:1182123. [PMID: 37123417 PMCID: PMC10133575 DOI: 10.3389/fcell.2023.1182123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 04/03/2023] [Indexed: 05/02/2023] Open
Abstract
Background: As a highly prevalent malignancy among women worldwide, breast cancer, remains a critical public health issue necessitating the development of novel therapeutics and biomarkers. Kruppel Like Factor 2 (KLF2), a member of the Kruppel family of transcription factors, has been implicated in various types of cancer due to its diminished expression; however, the potential implications of KLF2 expression in relation to breast cancer progression, prognosis, and therapy remain unclear. Methods: The present study employed the Tumor Immune Estimation Resource (TIMER) and The Human Protein Atlas databases to investigate the expression pattern of KLF2 in pan-cancer. The relationship between KLF2 expression and clinical features or immune infiltration of The Cancer Genome Atlas (TCGA) breast cancer samples was evaluated using Breast Cancer Integrative Platform (BCIP) and TIMER. The expression levels of KLF2 in breast cancer were validated via immunohistochemical staining analysis. Gene Set Enrichment Analysis (GSEA) to study the KLF2-related gene ontology. STRING database was employed to construct a protein-protein interaction (PPI) network of KLF2 in relation to vascular endothelial growth factor A (VEGFA) and hypoxia-inducible factor 1α (HIF1α). The expression of KLF2 following diverse breast cancer therapies was analyzed in the Gene Expression Omnibus (GEO) databases. The expression of KLF2 following treatment with simvastatin was validated via immunofluorescence and western blotting. Results: Our study reveals that KLF2 displays significantly reduced expression in cancerous tissues compared to non-cancerous controls. Patients with low KLF2 expression levels exhibited poor prognosis across multiple cancer types. KLF2 expression levels were found to be reduced in advanced cancer stages and grades, while positively correlated with the expression of estrogen receptor (ER), progesterone receptor (PR), and tumor size in breast cancer. KLF2 expression is associated with diverse immune infiltration cells, and may impact the breast tumor immune microenvironment by regulating dendritic cell activation. Additionally, we observed a negative correlation between KLF2 expression levels and angiogenesis, as well as the expression of VEGFA and HIF1α. Notably, the anticancer drug simvastatin could induce KLF2 expression in both breast cancer. Conclusion: Based on our observations, KLF2 has potential as a diagnostic, prognostic, and therapeutic biomarker for breast cancer.
Collapse
Affiliation(s)
- Ya-Zhao Li
- Center for Translational Medicine, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Juan Xie
- Department of Clinical Laboratory, Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi, China
| | - Rui-Qi Wang
- Center for Translational Medicine, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Xiao-Qian Gao
- Center for Translational Medicine, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Pei-Jun Liu
- Center for Translational Medicine, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- *Correspondence: Pei-Jun Liu, ; Jie Liu,
| | - Jie Liu
- Center for Translational Medicine, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- *Correspondence: Pei-Jun Liu, ; Jie Liu,
| |
Collapse
|
15
|
Wang L, Wang B, Jia L, Yu H, Wang Z, Wei F, Jiang A. Shear stress leads to the dysfunction of endothelial cells through the Cav-1-mediated KLF2/eNOS/ERK signaling pathway under physiological conditions. Open Life Sci 2023; 18:20220587. [PMID: 37077343 PMCID: PMC10106974 DOI: 10.1515/biol-2022-0587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 02/07/2023] [Accepted: 02/28/2023] [Indexed: 04/21/2023] Open
Abstract
To investigate the mechanism of shear stress on endothelial cell dysfunction for providing a theoretical basis for the reduction of arteriovenous fistula dysfunction. The in vitro parallel plate flow chamber was used to form different forces and shear stress to mimic the hemodynamic changes in human umbilical vein endothelial cells, and the expression and distribution of krüppel-like factor 2 (KLF2), caveolin-1 (Cav-1), p-extracellular regulated protein kinase (p-ERK), and endothelial nitric oxide synthase (eNOS) were detected by immunofluorescence and real-time quantitative polymerase chain reaction. With the prolongation of the shear stress action time, the expression of KLF2 and eNOS increased gradually, while the expression of Cav-1 and p-ERK decreased gradually. In addition, after cells were exposed to oscillatory shear stress (OSS) and low shear stress, the expression of KLF2, Cav-1, and eNOS decreased and the expression of p-ERK increased. The expression of KLF2 increased gradually with the prolongation of action time, but it was still obviously lower than that of high shear stress. Following the block of Cav-1 expression by methyl β-cyclodextrin, eNOS expression decreased, and KLF2 and p-ERK expression increased. OSS may lead to endothelial cell dysfunction by Cav-1-mediated KLF2/eNOS/ERK signaling pathway.
Collapse
Affiliation(s)
- Lihua Wang
- Department of Kidney Disease and Blood Purification Centre, 2nd Hospital of Tianjin Medical University, 23rd, Pingjiang Road, Hexi District, Tianjin300211, China
| | - Bingyue Wang
- Blood Purification Center of Tianjin Third Central Hospital, Tianjin300170, China
| | - Lan Jia
- Department of Kidney Disease and Blood Purification Centre, 2nd Hospital of Tianjin Medical University, 23rd, Pingjiang Road, Hexi District, Tianjin300211, China
| | - Haibo Yu
- Department of Kidney Disease and Blood Purification Centre, 2nd Hospital of Tianjin Medical University, 23rd, Pingjiang Road, Hexi District, Tianjin300211, China
| | - Zhe Wang
- Department of Kidney Disease and Blood Purification Centre, 2nd Hospital of Tianjin Medical University, 23rd, Pingjiang Road, Hexi District, Tianjin300211, China
| | - Fang Wei
- Department of Kidney Disease and Blood Purification Centre, 2nd Hospital of Tianjin Medical University, 23rd, Pingjiang Road, Hexi District, Tianjin300211, China
| | - Aili Jiang
- Department of Kidney Disease and Blood Purification Centre, 2nd Hospital of Tianjin Medical University, 23rd, Pingjiang Road, Hexi District, Tianjin300211, China
| |
Collapse
|
16
|
Ma X, Su M, He Q, Zhang Z, Zhang F, Liu Z, Sun L, Weng J, Xu S. PHACTR1, a coronary artery disease risk gene, mediates endothelial dysfunction. Front Immunol 2022; 13:958677. [PMID: 36091033 PMCID: PMC9457086 DOI: 10.3389/fimmu.2022.958677] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/01/2022] [Indexed: 01/13/2023] Open
Abstract
Genome-wide association studies (GWAS) have recently identified phosphatase and actin regulator-1 (PHACTR1) as a critical risk gene associated with polyvascular diseases. However, it remains largely unclear how PHACTR1 is involved in endothelial dysfunction. Here, by mining published datasets of human stable and vulnerable/ruptured plaque tissues, we observed upregulated expression of PHACTR1 in vulnerable/ruptured plaques. Congruent with these data, we demonstrated increased Phactr1 gene expression in aortic endothelium from ApoE-/- mice fed a western type diet compared with that in normal C57BL/6J mice. Relevantly, PHACTR1 gene expression was upregulated by pro-inflammatory and pro-atherogenic stimuli, including TNF-α, IL-1β and oxidized LDL (oxLDL). By employing next-generation RNA sequencing, we demonstrate that PHACTR1 overexpression disrupts pathways associated with endothelial homeostasis. Cell biological studies unravel that PHACTR1 mediates endothelial inflammation and monocyte adhesion by activating NF-κB dependent intercellular adhesion molecule 1 (ICAM1) and vascular cell adhesion molecule 1 (VCAM1) expression. In addition, overexpression of PHACTR1 also reduces the generation of nitric oxide (NO) by inhibiting Akt/eNOS activation. In-house compound screening of vasoprotective drugs identifies several drugs, including lipid-lowering statins, decreases PHACTR1 gene expression. However, PHACTR1 gene expression was not affected by another lipid-lowering drug-fenofibrate. We also performed a proteomic study to reveal PHACTR1 interacting proteins and validated that PHACTR1 can interact with heat shock protein A8 (HSPA8) which was reported to be associated with coronary artery disease and eNOS degradation. Further studies are warranted to confirm the precise mechanism of PHACTR1 in driving endothelial dysfunction. In conclusion, by using systems biology approach and molecular validation, we disclose the deleterious effects of PHACTR1 on endothelial function by inducing endothelial inflammation and reducing NO production, highlighting the potential to prevent endothelial dysfunction and atherosclerosis by targeting PHACTR1 expression. The precise role of endothelial cell PHACTR1 in polyvascular diseases remains to be validated in diseased conditions.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Suowen Xu
- *Correspondence: Suowen Xu, ; Jianping Weng,
| |
Collapse
|
17
|
Hu F, Ren Y, Wang Z, Zhou H, Luo Y, Wang M, Tian F, Zheng J, Du J, Pang G. Bioinformatics analysis of KLF2 as a potential prognostic factor in ccRCC and association with epithelial‑mesenchymal transition. Exp Ther Med 2022; 24:561. [PMID: 35978925 PMCID: PMC9366276 DOI: 10.3892/etm.2022.11498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 06/23/2022] [Indexed: 11/29/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is a primary pathological subtype of RCC and has poor clinical outcome. Krüppel-like factors (KLFs), which are zinc-finger proteins, may be involved in ccRCC development and progression. KLFs belong to the zinc-finger family of DNA-binding transcription factors and regulate transcription of downstream target genes. KLFs are involved in cancer development. The present study aimed to investigate the role of KLFs in ccRCC prognosis. The Cancer Genome Atlas database and multifactorial analysis showed that KLFs were widely expressed in pan-cancers and KLF2 was an independent protective factor for ccRCC prognosis. Patients with low KLF2 expression had a low survival probability and expression of KLF2 was downregulated in patients with ccRCC with high pathological grade (II + III vs. I). In addition, western blot and reverse transcription-quantitative PCR revealed that KLF2 was expressed at low levels in ccRCC cell lines and overexpression of KLF2 inhibited cell migration. In addition, KLF2 expression was negatively correlated with methylation. KLF2 expression was elevated following treatment of ccRCC cells with DNA methyltransferase inhibitor. A prognostic risk index prediction model was constructed based on multiple Cox regression. The receiver operating characteristic curve was 0.780 (area under curve >0.5). Furthermore, Gene Ontology enrichment analysis showed that ‘cell adhesion’ and ‘junction’ were negatively correlated with KLF2 and that high-risk group exhibited significantly activated ‘epithelial-mesenchymal transition’. Western blot analysis showed that overexpression of KLF2 increased expression of E-cadherin, while decreasing levels of N-cadherin and vimentin. The present study highlighted the role of KLFs in ccRCC prognosis prediction and provides a research base for the search of validated prognostic biological markers for ccRCC.
Collapse
Affiliation(s)
- Fangfang Hu
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yan Ren
- Department of Human Anatomy, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Zunyun Wang
- The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, P.R. China
| | - Hui Zhou
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yumei Luo
- The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, P.R. China
| | - Minghua Wang
- The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, P.R. China
| | - Faqing Tian
- The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, P.R. China
| | - Jian Zheng
- The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, P.R. China
| | - Juan Du
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Gang Pang
- Department of Human Anatomy, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
18
|
Sun Y, Chen X, Xie Y, Wang Y, Zhang Q, Lu Y, Li X. TRPM7 promotes lipopolysaccharide-induced inflammatory dysfunction in renal tubular epithelial cells. Immun Inflamm Dis 2022; 10:e641. [PMID: 35759233 PMCID: PMC9208284 DOI: 10.1002/iid3.641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 05/04/2022] [Accepted: 05/07/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Sepsis-associated acute kidney injury (S-AKI) has been reported to affect 30%-50% of all sepsis patients; this condition is associated with a notable fatality rate. Following lipopolysaccharide (LPS) stimulation, the expression of transient receptor potential cation channel subfamily M member 7 (TRPM7), a nonselective cation channel expressed by the renal tubular epithelial cells (RTECs) was found to be upregulated. We aimed to determine how TRPM7 functions in S-AKI. METHODS To establish an in vitro model of S-AKI, RTECs were treated with LPS. The effect of TRPM7 knockdown on cell viability, lactate dehydrogenase (LDH) release, apoptosis, inflammation, and oxidative stress was studied. The binding site between Kruppel-like factor 2 (KLF2) and TRPM7 was predicted using JASPAR. The influence of KLF2 on the regulatory roles of TRPM7 in cells, as well as the effect of their knockdown on the MAPK signaling pathway, was investigated. RESULTS TRPM7 was upregulated in LPS-treated cells, and knocking improved cell viability, reduced LDH levels, and minimized apoptosis, inflammation, and oxidative stress. KLF2 was shown to be associated with TRPM7 and its level decreased in LPS-treated cells. KLF2 knockdown increased TRPM7 expression and reversed the effects of TRPM7 knockdown in LPS-treated cells, including suppression of p38 MAPK, ERK1/2, and JNK activation. CONCLUSION Taken together, our results show that TRPM7 is negatively regulated by KLF2 and promotes LPS-induced inflammatory dysfunction by activating the MAPK pathway in RTECs. The theoretical foundation for the prevention and management of S-AKI is laid out in this article.
Collapse
Affiliation(s)
- Yan Sun
- Department of Emergency MedicineLianyungang Clinical College of Nanjing Medical UniversityLianyungangChina
| | - Xiaobing Chen
- Department of Emergency MedicineLianyungang Clinical College of Nanjing Medical UniversityLianyungangChina
| | - Yongpeng Xie
- Department of Emergency MedicineLianyungang Clinical College of Nanjing Medical UniversityLianyungangChina
| | - Yanli Wang
- Department of Emergency MedicineLianyungang Clinical College of Nanjing Medical UniversityLianyungangChina
| | - Qian Zhang
- Department of Emergency MedicineLianyungang Clinical College of Nanjing Medical UniversityLianyungangChina
| | - Yu Lu
- Department of Emergency MedicineLianyungang Clinical College of Nanjing Medical UniversityLianyungangChina
| | - Xiaomin Li
- Department of Emergency MedicineLianyungang Clinical College of Nanjing Medical UniversityLianyungangChina
| |
Collapse
|
19
|
Tsaryk R, Yucel N, Leonard EV, Diaz N, Bondareva O, Odenthal-Schnittler M, Arany Z, Vaquerizas JM, Schnittler H, Siekmann AF. Shear stress switches the association of endothelial enhancers from ETV/ETS to KLF transcription factor binding sites. Sci Rep 2022; 12:4795. [PMID: 35314737 PMCID: PMC8938417 DOI: 10.1038/s41598-022-08645-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 03/10/2022] [Indexed: 02/06/2023] Open
Abstract
Endothelial cells (ECs) lining blood vessels are exposed to mechanical forces, such as shear stress. These forces control many aspects of EC biology, including vascular tone, cell migration and proliferation. Despite a good understanding of the genes responding to shear stress, our insight into the transcriptional regulation of these genes is much more limited. Here, we set out to study alterations in the chromatin landscape of human umbilical vein endothelial cells (HUVEC) exposed to laminar shear stress. To do so, we performed ChIP-Seq for H3K27 acetylation, indicative of active enhancer elements and ATAC-Seq to mark regions of open chromatin in addition to RNA-Seq on HUVEC exposed to 6 h of laminar shear stress. Our results show a correlation of gained and lost enhancers with up and downregulated genes, respectively. DNA motif analysis revealed an over-representation of KLF transcription factor (TF) binding sites in gained enhancers, while lost enhancers contained more ETV/ETS motifs. We validated a subset of flow responsive enhancers using luciferase-based reporter constructs and CRISPR-Cas9 mediated genome editing. Lastly, we characterized the shear stress response in ECs of zebrafish embryos using RNA-Seq. Our results lay the groundwork for the exploration of shear stress responsive elements in controlling EC biology.
Collapse
Affiliation(s)
- Roman Tsaryk
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149, Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
- Department of Cell and Developmental Biology and Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Nora Yucel
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Elvin V Leonard
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149, Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
- Department of Cell and Developmental Biology and Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Noelia Diaz
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149, Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
| | - Olga Bondareva
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
- Institute of Anatomy and Vascular Biology, Faculty of Medicine, Westfälische Wilhelms-Universität Münster, Vesaliusweg 2-4, 48149, Münster, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Philipp-Rosenthal-Str. 27, 04103, Leipzig, Germany
| | - Maria Odenthal-Schnittler
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149, Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
- Institute of Anatomy and Vascular Biology, Faculty of Medicine, Westfälische Wilhelms-Universität Münster, Vesaliusweg 2-4, 48149, Münster, Germany
- Institute of Neuropathology, Westfälische Wilhelms-Universität Münster, Pottkamp 2, 48149, Münster, Germany
| | - Zoltan Arany
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Juan M Vaquerizas
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149, Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
| | - Hans Schnittler
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149, Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
- Institute of Anatomy and Vascular Biology, Faculty of Medicine, Westfälische Wilhelms-Universität Münster, Vesaliusweg 2-4, 48149, Münster, Germany
- Institute of Neuropathology, Westfälische Wilhelms-Universität Münster, Pottkamp 2, 48149, Münster, Germany
| | - Arndt F Siekmann
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149, Münster, Germany.
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany.
- Department of Cell and Developmental Biology and Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
20
|
Siddiqui HB, Dogru S, Lashkarinia SS, Pekkan K. Soft-Tissue Material Properties and Mechanogenetics during Cardiovascular Development. J Cardiovasc Dev Dis 2022; 9:jcdd9020064. [PMID: 35200717 PMCID: PMC8876703 DOI: 10.3390/jcdd9020064] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 01/22/2022] [Accepted: 01/28/2022] [Indexed: 12/17/2022] Open
Abstract
During embryonic development, changes in the cardiovascular microstructure and material properties are essential for an integrated biomechanical understanding. This knowledge also enables realistic predictive computational tools, specifically targeting the formation of congenital heart defects. Material characterization of cardiovascular embryonic tissue at consequent embryonic stages is critical to understand growth, remodeling, and hemodynamic functions. Two biomechanical loading modes, which are wall shear stress and blood pressure, are associated with distinct molecular pathways and govern vascular morphology through microstructural remodeling. Dynamic embryonic tissues have complex signaling networks integrated with mechanical factors such as stress, strain, and stiffness. While the multiscale interplay between the mechanical loading modes and microstructural changes has been studied in animal models, mechanical characterization of early embryonic cardiovascular tissue is challenging due to the miniature sample sizes and active/passive vascular components. Accordingly, this comparative review focuses on the embryonic material characterization of developing cardiovascular systems and attempts to classify it for different species and embryonic timepoints. Key cardiovascular components including the great vessels, ventricles, heart valves, and the umbilical cord arteries are covered. A state-of-the-art review of experimental techniques for embryonic material characterization is provided along with the two novel methods developed to measure the residual and von Mises stress distributions in avian embryonic vessels noninvasively, for the first time in the literature. As attempted in this review, the compilation of embryonic mechanical properties will also contribute to our understanding of the mature cardiovascular system and possibly lead to new microstructural and genetic interventions to correct abnormal development.
Collapse
Affiliation(s)
- Hummaira Banu Siddiqui
- Department of Mechanical Engineering, Koc University, Istanbul 34450, Turkey; (H.B.S.); (S.D.); (S.S.L.)
| | - Sedat Dogru
- Department of Mechanical Engineering, Koc University, Istanbul 34450, Turkey; (H.B.S.); (S.D.); (S.S.L.)
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Seyedeh Samaneh Lashkarinia
- Department of Mechanical Engineering, Koc University, Istanbul 34450, Turkey; (H.B.S.); (S.D.); (S.S.L.)
- Department of Bioengineering, Imperial College London, London SW7 2BX, UK
| | - Kerem Pekkan
- Department of Mechanical Engineering, Koc University, Istanbul 34450, Turkey; (H.B.S.); (S.D.); (S.S.L.)
- Correspondence: ; Tel.: +90-(533)-356-3595
| |
Collapse
|
21
|
Ale-Ebrahim M, Rahmani R, Faryabi K, Mohammadifar N, Mortazavi P, Karkhaneh L. Atheroprotective and hepatoprotective effects of trans-chalcone through modification of eNOS/AMPK/KLF-2 pathway and regulation of COX-2, Ang-II, and PDGF mRNA expression in NMRI mice fed HCD. Mol Biol Rep 2022; 49:3433-3443. [PMID: 35190927 DOI: 10.1007/s11033-022-07174-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 01/19/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND The effects of trans-chalcone on atherosclerosis and NAFLD have been investigated. However, the underlying molecular mechanisms of these effects are not completely understood. This study aimed to deduce the impacts of trans-chalcone on the eNOS/AMPK/KLF-2 pathway in the heart tissues and the expression of Ang-II, PDFG, and COX-2 genes in liver sections of NMRI mice fed HCD. METHODS AND RESULTS Thirty-two male mice were divided into four groups (n = 8): control group; fed normal diet. HCD group; fed HCD (consisted of 2% cholesterol) (12 weeks). TCh groups; received HCD (12 weeks) besides co-treated with trans-chalcone (20 mg/kg and 40 mg/kg b.w. dosages respectively) for 4 weeks. Finally, the blood samples were collected to evaluate the biochemical parameters. Histopathological observations of aorta and liver sections were performed by H&E staining. The real-time PCR method was used for assessing the expression of the aforementioned genes. Histopathological examination demonstrated atheroma plaque formation and fatty liver in mice fed HCD which were accomplished with alteration in biochemical factors and Real-time PCR outcomes. Administration of trans-chalcone significantly modulated the serum of biochemical parameters. These effects were accompanied by significant increasing the expression of eNOS, AMPK, KLF-2 genes in heart sections and significant decrease in COX-2, Ang-II, and PDGF mRNA expression in liver sections. CONCLUSION Our findings propose that the atheroprotective and hepatoprotective effects of trans-chalcone may be attributed to the activation of the eNOS/AMPK/KLF-2 pathway and down-regulation of Ang-II, PDFG, and COX-2 genes, respectively.
Collapse
Affiliation(s)
- Mahsa Ale-Ebrahim
- Department of Physiology, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Raziyeh Rahmani
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Kousar Faryabi
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Niloofar Mohammadifar
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Pejman Mortazavi
- Department of Veterinary Pathology, Faculty of Specialized Veterinary, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Leyla Karkhaneh
- Department of Physiology, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
22
|
Brandt MM, Cheng C, Merkus D, Duncker DJ, Sorop O. Mechanobiology of Microvascular Function and Structure in Health and Disease: Focus on the Coronary Circulation. Front Physiol 2022; 12:771960. [PMID: 35002759 PMCID: PMC8733629 DOI: 10.3389/fphys.2021.771960] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/11/2021] [Indexed: 12/19/2022] Open
Abstract
The coronary microvasculature plays a key role in regulating the tight coupling between myocardial perfusion and myocardial oxygen demand across a wide range of cardiac activity. Short-term regulation of coronary blood flow in response to metabolic stimuli is achieved via adjustment of vascular diameter in different segments of the microvasculature in conjunction with mechanical forces eliciting myogenic and flow-mediated vasodilation. In contrast, chronic adjustments in flow regulation also involve microvascular structural modifications, termed remodeling. Vascular remodeling encompasses changes in microvascular diameter and/or density being largely modulated by mechanical forces acting on the endothelium and vascular smooth muscle cells. Whereas in recent years, substantial knowledge has been gathered regarding the molecular mechanisms controlling microvascular tone and how these are altered in various diseases, the structural adaptations in response to pathologic situations are less well understood. In this article, we review the factors involved in coronary microvascular functional and structural alterations in obstructive and non-obstructive coronary artery disease and the molecular mechanisms involved therein with a focus on mechanobiology. Cardiovascular risk factors including metabolic dysregulation, hypercholesterolemia, hypertension and aging have been shown to induce microvascular (endothelial) dysfunction and vascular remodeling. Additionally, alterations in biomechanical forces produced by a coronary artery stenosis are associated with microvascular functional and structural alterations. Future studies should be directed at further unraveling the mechanisms underlying the coronary microvascular functional and structural alterations in disease; a deeper understanding of these mechanisms is critical for the identification of potential new targets for the treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Maarten M Brandt
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Caroline Cheng
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Division of Internal Medicine and Dermatology, Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, Netherlands
| | - Daphne Merkus
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Walter Brendel Center of Experimental Medicine (WBex), LMU Munich, Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany
| | - Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Oana Sorop
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
23
|
Mauersberger C, Hinterdobler J, Schunkert H, Kessler T, Sager HB. Where the Action Is-Leukocyte Recruitment in Atherosclerosis. Front Cardiovasc Med 2022; 8:813984. [PMID: 35087886 PMCID: PMC8787128 DOI: 10.3389/fcvm.2021.813984] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/15/2021] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis is the leading cause of death worldwide and leukocyte recruitment is a key element of this phenomenon, thus allowing immune cells to enter the arterial wall. There, in concert with accumulating lipids, the invading leukocytes trigger a plethora of inflammatory responses which promote the influx of additional leukocytes and lead to the continued growth of atherosclerotic plaques. The recruitment process follows a precise scheme of tethering, rolling, firm arrest, crawling and transmigration and involves multiple cellular and subcellular players. This review aims to provide a comprehensive up-to-date insight into the process of leukocyte recruitment relevant to atherosclerosis, each from the perspective of endothelial cells, monocytes and macrophages, neutrophils, T lymphocytes and platelets. In addition, therapeutic options targeting leukocyte recruitment into atherosclerotic lesions-or potentially arising from the growing body of insights into its precise mechanisms-are highlighted.
Collapse
Affiliation(s)
- Carina Mauersberger
- Department of Cardiology, German Heart Center Munich, Technical University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Julia Hinterdobler
- Department of Cardiology, German Heart Center Munich, Technical University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Heribert Schunkert
- Department of Cardiology, German Heart Center Munich, Technical University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Thorsten Kessler
- Department of Cardiology, German Heart Center Munich, Technical University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Hendrik B. Sager
- Department of Cardiology, German Heart Center Munich, Technical University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
24
|
Fraxinellone ameliorates intracerebral hemorrhage-induced secondary brain injury by regulating Krüppel-like transcription factor 2 expression in rats. Brain Res Bull 2021; 177:340-351. [PMID: 34717966 DOI: 10.1016/j.brainresbull.2021.10.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 10/11/2021] [Accepted: 10/26/2021] [Indexed: 11/24/2022]
Abstract
Damage to the blood-brain barrier (BBB) is an important factor leading to intracerebral hemorrhage (ICH)-induced secondary brain injury (SBI). Krüppel-like transcription factor 2 (KLF2) plays an important role in the maintenance of the BBB. This study aims to detect the changes of KLF2 after ICH and evaluate the potential effects of fraxinellone on ICH-induced SBI and its correlation with KLF2. An ICH model was established by injecting autologous blood into the right basal ganglia of Sprague-Dawley (SD) rats. First, after ICH induction, the protein levels of KLF2 were reduced. Then, we found that the decrease of KLF2 protein levels induced by ICH could be effectively reversed with the treatment of fraxinellone in vascular endothelial cells. Furthermore, fraxinellone treatment effectively alleviated brain edema, decreased the levels of TNF-α and IL-1β, and improved neuronal cell degeneration induced by ICH. Meanwhile, fraxinellone ameliorated neurobehavioral disorders, motor and sensory impairments, and neurobehavioral disorders and memory loss caused by ICH. Collectively, these findings reveal that KLF2 may be a potential target for fraxinellone to exert neuroprotective effects after ICH, and fraxinellone could be a potential therapeutic agent for SBI after ICH.
Collapse
|
25
|
Liu J, Li Z, Yu G, Wang T, Qu G, Wang Y. LINC01232 Promotes Gastric Cancer Proliferation through Interacting with EZH2 to Inhibit the Transcription of KLF2. J Microbiol Biotechnol 2021; 31:1358-1365. [PMID: 34409953 PMCID: PMC9705925 DOI: 10.4014/jmb.2106.06041] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/10/2021] [Accepted: 08/17/2021] [Indexed: 12/15/2022]
Abstract
To clarify the role of long intergenic nonprotein-coding RNA 1232 (LINC01232) in the progression of gastric cancer and the potential mechanism, we analyzed the expression of LINC01232 in TCGA database using the GEPIA online tool, and the LINC01232 level in gastric cancer cell lines was detected by quantitative real time-polymerase chain reaction (qRT-PCR) as well. Cell proliferation assay, colony formation assay, transwell assay and tumor formation experiment in nude mice were conducted to observe the biological behavior changes of gastric cancer cells through the influence of LINC01232 knockdown. LncATLAS database and subcellular isolation assay were used for subcellular distribution of LINC01232 in gastric cancer cells. The interaction among LINC01232, zeste homolog 2 (EZH2) and kruppel-like factor 2 (KLF2) was clarified by RNA-protein interaction prediction (RPISeq), RNA immunoprecipitation (RIP), qRT-PCR and chromatin immunoprecipitation (ChIP) assay. Rescue experiments were further conducted to elucidate the biological function of LINC01232/KLF2 axis in the progression of gastric cancer. LINC01232 was upregulated in stomach adenocarcinoma (STAD) tissues and gastric cancer lines. LINC01232 knockdown inhibited the proliferative capacities of gastric cancer cells in vitro, and impaired in vivo tumorigenicity. LINC01232 was mainly distributed in the cell nucleus where it epigenetically repressed KLF2 expression via binding to the enhancer of EZH2, which was capable of binding to promoter regions of KLF2 to induce histone H3 lysine 27 trimethylation (H3K27me3). LINC01232 exerts oncogenic activities in gastric cancer via inhibition of KLF2, and therefore, the knockdown of KLF2 could reverse the regulatory effect of LINC01232 in the proliferative ability of gastric cancer cells.
Collapse
Affiliation(s)
- Jing Liu
- Department of Pathology, Yantai Yuhuangding Hospital, Yantai 264000, P.R. China
| | - Zhen Li
- Department of General and Pediatric Surgery, Yantai Yuhuangding Hospital, Yantai 264000, P.R. China
| | - Guohua Yu
- Department of Pathology, Yantai Yuhuangding Hospital, Yantai 264000, P.R. China
| | - Ting Wang
- Department of Pathology, Yantai Yuhuangding Hospital, Yantai 264000, P.R. China
| | - Guimei Qu
- Department of Pathology, Yantai Yuhuangding Hospital, Yantai 264000, P.R. China
| | - Yunhui Wang
- Department of General and Pediatric Surgery, Yantai Yuhuangding Hospital, Yantai 264000, P.R. China
| |
Collapse
|
26
|
Li Z, Wang J, Ma Y. Montelukast attenuates interleukin IL-1β-induced oxidative stress and apoptosis in chondrocytes by inhibiting CYSLTR1 (Cysteinyl Leukotriene Receptor 1) and activating KLF2 (Kruppel Like Factor 2). Bioengineered 2021; 12:8476-8484. [PMID: 34565285 PMCID: PMC8806840 DOI: 10.1080/21655979.2021.1984003] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Montelukast is a cysteinyl leukotriene receptor 1 (CysLTR1) antagonist widely used to suppress the inflammatory response in asthma and allergic rhinitis. This study aimed to investigate the potential impacts of montelukast on osteoarthritis (OA) progression. To determine the role of montelukast in OA, the expression of CysLTR1 was first examined by quantitative reverse transcription PCR (RT-qPCR) and western blot in IL-1β-induced ATDC5 cells treated with or without montelukast. Subsequently, the impacts of montelukast on cell viability and oxidative stress were measured by Cell-Counting-Kit-8 (CCK-8), commercial kits and western blot. Oxidative stress-related protein expressions were determined by western blot analysis in Il-1β-induced ATDC5 cells. Cell apoptosis and cartilage degradation were examined by TdT-mediated dUTP Nick-End Labeling (TUNEL) assay, western blot and RT-qPCR. KLF2 expression was measured in IL-1β-induced ATDC5 cells treated with montelukast. After interference with small interfering RNA (siRNA)-KLF2 in ATDC5 cells, the loss-of-function assays were also performed in same ways. CysLTR1 expression was elevated in IL-1β-induced ATDC5 cells but inhibited significantly by montelukast. Montelukast attenuated the oxidative stress and apoptosis, improved cell viability. Moreover, montelukast enhanced KLF2 expression. After transfected with siRNA-KLF2, montelukast attenuated cell injury, oxidative stress, apoptosis and cartilage degradation in IL-1β-induced ATDC5 cells by activating KLF2.In summary, this work elaborates the evidence that montelukast could attenuate oxidative stress and apoptosis in IL-1β-induced chondrocytes by inhibiting CysLTR1 and activating KLF2, which can guide the therapeutic strategies of montelukast for OA development in the future.
Collapse
Affiliation(s)
- Zongwei Li
- School of Pharmaceutical Engineering, Guangdong Food and Drug Vocational College, Guangzhou City, Guangdong Province, China
| | - Jianming Wang
- School of Pharmaceutical Engineering, Guangdong Food and Drug Vocational College, Guangzhou City, Guangdong Province, China
| | - Yumin Ma
- Department of Pharmaceutical Machinery, Maternal and Child Health and Family Planning Technical Service Center, Wuwei City, Gansu Province, China
| |
Collapse
|
27
|
New mechanism-based approaches to treating and evaluating the vasculopathy of scleroderma. Curr Opin Rheumatol 2021; 33:471-479. [PMID: 34402454 DOI: 10.1097/bor.0000000000000830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
PURPOSE OF REVIEW Utilizing recent insight into the vasculopathy of scleroderma (SSc), the review will highlight new opportunities for evaluating and treating the disease by promoting stabilization and protection of the microvasculature. RECENT FINDINGS Endothelial junctional signaling initiated by vascular endothelial-cadherin (VE-cadherin) and Tie2 receptors, which are fundamental to promoting vascular health and stability, are disrupted in SSc. This would be expected to not only diminish their protective activity, but also increase pathological processes that are normally restrained by these signaling mediators, resulting in pathological changes in vascular function and structure. Indeed, key features of SSc vasculopathy, from the earliest signs of edema and puffy fingers to pathological disruption of hemodynamics, nutritional blood flow, capillary structure and angiogenesis are all consistent with this altered endothelial signaling. It also likely contributes to further progression of the disease including tissue fibrosis, and organ and tissue injury. SUMMARY Restoring protective endothelial junctional signaling should combat the vasculopathy of SSc and prevent further deterioration in vascular and organ function. Indeed, this type of targeted approach has achieved remarkable results in preclinical models for other diseases. Furthermore, tracking this endothelial junctional signaling, for example by assessing vascular permeability, should facilitate insight into disease progression and its response to therapy.
Collapse
|
28
|
Kugler E, Snodgrass R, Bowley G, Plant K, Serbanovic-Canic J, Hamilton N, Evans PC, Chico T, Armitage P. The effect of absent blood flow on the zebrafish cerebral and trunk vasculature. VASCULAR BIOLOGY (BRISTOL, ENGLAND) 2021; 3:1-16. [PMID: 34522840 PMCID: PMC8428019 DOI: 10.1530/vb-21-0009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 07/29/2021] [Indexed: 12/18/2022]
Abstract
The role of blood flow in vascular development is complex and context-dependent. In this study, we quantify the effect of the lack of blood flow on embryonic vascular development on two vascular beds, namely the cerebral and trunk vasculature in zebrafish. We perform this by analysing vascular topology, endothelial cell (EC) number, EC distribution, apoptosis, and inflammatory response in animals with normal blood flow or absent blood flow. We find that absent blood flow reduced vascular area and EC number significantly in both examined vascular beds, but the effect is more severe in the cerebral vasculature, and severity increases over time. Absent blood flow leads to an increase in non-EC-specific apoptosis without increasing tissue inflammation, as quantified by cerebral immune cell numbers and nitric oxide. Similarly, while stereotypic vascular patterning in the trunk is maintained, intra-cerebral vessels show altered patterning, which is likely to be due to vessels failing to initiate effective fusion and anastomosis rather than sprouting or path-seeking. In conclusion, blood flow is essential for cellular survival in both the trunk and cerebral vasculature, but particularly intra-cerebral vessels are affected by the lack of blood flow, suggesting that responses to blood flow differ between these two vascular beds.
Collapse
Affiliation(s)
- Elisabeth Kugler
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Medical School, Sheffield, UK
- The Bateson Centre, Firth Court, University of Sheffield, Western Bank, Sheffield, UK
- Insigneo Institute for in silico Medicine, Sheffield, UK
- Institute of Ophthalmology, Faculty of Brain Sciences, University College London, London, UK
| | - Ryan Snodgrass
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Medical School, Sheffield, UK
- The Bateson Centre, Firth Court, University of Sheffield, Western Bank, Sheffield, UK
| | - George Bowley
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Medical School, Sheffield, UK
- The Bateson Centre, Firth Court, University of Sheffield, Western Bank, Sheffield, UK
| | - Karen Plant
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Medical School, Sheffield, UK
- The Bateson Centre, Firth Court, University of Sheffield, Western Bank, Sheffield, UK
| | - Jovana Serbanovic-Canic
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Medical School, Sheffield, UK
- The Bateson Centre, Firth Court, University of Sheffield, Western Bank, Sheffield, UK
| | - Noémie Hamilton
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Medical School, Sheffield, UK
- The Bateson Centre, Firth Court, University of Sheffield, Western Bank, Sheffield, UK
| | - Paul C Evans
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Medical School, Sheffield, UK
- The Bateson Centre, Firth Court, University of Sheffield, Western Bank, Sheffield, UK
- Insigneo Institute for in silico Medicine, Sheffield, UK
| | - Timothy Chico
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Medical School, Sheffield, UK
- The Bateson Centre, Firth Court, University of Sheffield, Western Bank, Sheffield, UK
| | - Paul Armitage
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Medical School, Sheffield, UK
- Insigneo Institute for in silico Medicine, Sheffield, UK
| |
Collapse
|
29
|
Jiang Y, Shen Q. IRF2BP2 prevents ox-LDL-induced inflammation and EMT in endothelial cells via regulation of KLF2. Exp Ther Med 2021; 21:481. [PMID: 33767776 PMCID: PMC7976449 DOI: 10.3892/etm.2021.9912] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 02/15/2021] [Indexed: 12/13/2022] Open
Abstract
Oxidized low-density lipoprotein (ox-LDL)-induced endothelial dysfunction contributes to the progression of atherosclerosis. Interferon regulatory factor 2-binding protein 2 (IRF2BP2) attenuates macrophage-mediated inflammation and susceptibility to atherosclerosis. However, the effects of IRF2BP2 on vascular endothelial cells in atherosclerosis have not been fully elucidated. In the present study, the effects of IRF2BP2 on cell viability, inflammation and endothelial-to-mesenchymal transition (EMT) of human umbilical vein endothelial cells (HUVECs) were assessed using Cell Counting Kit-8 (CCK-8) assays, ELISA kits and western blot analysis, respectively. In addition, the expression levels of Krüppel-like factor 2 (KLF2) were determined by reverse transcription-quantitative PCR and immunofluorescence assays. A Nitrate/Nitrite assay kit was utilized to detect the production of nitric oxide (NO). The results demonstrated that ox-LDL induced inflammation and EMT of HUVECs, and decreased the NO levels. Furthermore, IRF2BP2 overexpression protected HUVECs against ox-LDL-induced inflammation, EMT and endothelial dysfunction, and resulted in upregulated expression of KLF2. Additionally, IRF2BP2 was shown to bind to KLF2, and KLF2 knockdown reversed the protective effects of IRF2BP2 on ox-LDL-exposed HUVECs. These findings indicated that IRF2BP2 may prevent ox-LDL-induced endothelial damage via upregulating KLF2 expression.
Collapse
Affiliation(s)
- Yongri Jiang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Qiuling Shen
- Department of Laboratory Diagnosis, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
30
|
Chemokine mediated signalling within arteries promotes vascular smooth muscle cell recruitment. Commun Biol 2020; 3:734. [PMID: 33277595 PMCID: PMC7719186 DOI: 10.1038/s42003-020-01462-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 11/05/2020] [Indexed: 01/13/2023] Open
Abstract
The preferential accumulation of vascular smooth muscle cells (vSMCs) on arteries versus veins during early development is a well-described phenomenon, but the molecular pathways underlying this polarization are not well understood. In zebrafish, the cxcr4a receptor (mammalian CXCR4) and its ligand cxcl12b (mammalian CXCL12) are both preferentially expressed on arteries at time points consistent with the arrival and differentiation of the first vSMCs during vascular development. We show that autocrine cxcl12b/cxcr4 activity leads to increased production of the vSMC chemoattractant ligand pdgfb by endothelial cells in vitro and increased expression of pdgfb by arteries of zebrafish and mice in vivo. Additionally, we demonstrate that expression of the blood flow-regulated transcription factor klf2a in primitive veins negatively regulates cxcr4/cxcl12 and pdgfb expression, restricting vSMC recruitment to the arterial vasculature. Together, this signalling axis leads to the differential acquisition of vSMCs at sites where klf2a expression is low and both cxcr4a and pdgfb are co-expressed, i.e. arteries during early development. Stratman et al. provide evidence linking the cxcl12b/cxcr4a signaling axis in endothelial cells to an increased release of platelet-derived growth factor b, leading to the recruitment of smooth muscle cells to developing arteries. This signalling axis is suppressed in the venous endothelium during early development by the high expression of blood flow-regulated transcription factor klf2a.
Collapse
|
31
|
Taniguchi R, Ono S, Isaji T, Gorecka J, Lee SR, Matsubara Y, Yatsula B, Koizumi J, Nishibe T, Hoshina K, Dardik A. A mouse model of stenosis distal to an arteriovenous fistula recapitulates human central venous stenosis. JVS Vasc Sci 2020; 1:109-122. [PMID: 33543148 PMCID: PMC7857464 DOI: 10.1016/j.jvssci.2020.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Objective Central venous stenosis (CVS) is a major cause of arteriovenous fistula (AVF) failure. However, central veins are relatively inaccessible to study with conventional Doppler ultrasound methods. To understand mechanisms underlying AVF failure owing to CVS, an animal model was established that creates a stenosis distal to an AVF. We hypothesized that this mouse model will show comparable morphology and physiology to human CVS. Methods An aortocaval fistula was created between the distal aorta and inferior vena cava (IVC); a stenosis was then created distal to the fistula by partial IVC ligation. Sham-operated animals, AVF without venous stenosis, and venous stenosis without AVF were used as controls. Physiologic properties of the IVC, both upstream and downstream of the stenosis, or the corresponding sites in models without stenosis, were assessed with ultrasound examination on days 0 to 21. The spectral broadening index was measured to assess the degree of disturbed shear stress. The IVC was harvested at day 21 and specimens were analyzed with immunofluorescence. Results The IVC diameter of mice with an AVF and stenosis showed increased upstream (P = .013), but decreased downstream diameter (P = .001) compared with mice with an AVF but without a stenosis, at all postoperative times (days 3-21). IVC wall thickness increased in mice with an AVF, compared with IVC without an AVF (upstream of stenosis: 13.9 μm vs 11.0 μm vs 4.5 μm vs 3.9 μm; P = .020; downstream of stenosis: 6.0 μm vs 6.6 μm vs μm 4.5 μm vs 3.8 μm; P = .002; AVF with stenosis, AVF, stenosis, sham, respectively). AVF patency significantly decreased in mice with an AVF and stenosis by day 21 (50% vs 90%; P = .048). The IVC of mice with AVF and stenosis showed a venous waveform with pulsatility as well as enhanced velocity at and downstream of the stenosis; similar waveforms were observed in a human case of CVS. Downstream to the stenosis, the spectral broadening index was significantly higher compared with mice with AVF alone (1.06 vs 0.78; P = .011; day 21), and there was a trend towards less immunoreactivity of both Krüppel-like factor 2 and phosphorylated-endothelial nitric oxide synthase compared with mice with an AVF alone. Conclusions Partial IVC ligation distal to a mouse aortocaval fistula alters the fistula diameter and wall thickness, decreases patency, and increases distal disturbed flow compared with fistulae without a distal stenosis. Our mouse model of stenosis distal to an AVF may be a faithful representation of human CVS that shows similar morphology and physiology, including disturbed shear stress. A mouse model of venous stenosis distal to an arteriovenous fistula shows similar Doppler waveforms as those observed in a human case of central venous stenosis. These mice retain disturbed shear stress in the vein distal to the fistula, characterized by a sustained increase of the spectral broadening index and diminished expression of proteins upregulated by laminar shear stress. This novel mouse model will enable investigation of the physiology and downstream molecular pathways involved in central venous stenosis in humans.
Collapse
Affiliation(s)
- Ryosuke Taniguchi
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven.,Division of Vascular Surgery, The University of Tokyo, Bunkyo-ku, Tokyo
| | - Shun Ono
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven.,Department of Diagnostic Radiology, Tokai University School of Medicine, Isehara, Kanagawa
| | - Toshihiko Isaji
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven.,Division of Vascular Surgery, The University of Tokyo, Bunkyo-ku, Tokyo
| | - Jolanta Gorecka
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven
| | - Shin-Rong Lee
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven
| | - Yutaka Matsubara
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven.,Department of Surgery and Sciences, Kyushu University, Fukuoka
| | - Bogdan Yatsula
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven
| | - Jun Koizumi
- Department of Diagnostic Radiology, Tokai University School of Medicine, Isehara, Kanagawa
| | - Toshiya Nishibe
- Department of Cardiovascular Surgery, Tokyo Medical University, Shinjuku-ku, Tokyo
| | - Katsuyuki Hoshina
- Division of Vascular Surgery, The University of Tokyo, Bunkyo-ku, Tokyo
| | - Alan Dardik
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven.,Division of Vascular and Endovascular Surgery, Department of Surgery, Yale School of Medicine, New Haven.,Department of Surgery, VA Connecticut Healthcare Systems, West Haven
| |
Collapse
|
32
|
Abstract
Endothelial cell (EC) metabolism is important for health and disease. Metabolic pathways, such as glycolysis, fatty acid oxidation, and amino acid metabolism, determine vasculature formation. These metabolic pathways have different roles in securing the production of energy and biomass and the maintenance of redox homeostasis in vascular migratory tip cells, proliferating stalk cells, and quiescent phalanx cells, respectively. Emerging evidence demonstrates that perturbation of EC metabolism results in EC dysfunction and vascular pathologies. Here, we summarize recent insights into EC metabolic pathways and their deregulation in vascular diseases. We further discuss the therapeutic implications of targeting EC metabolism in various pathologies.
Collapse
Affiliation(s)
- Xuri Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China; ,
| | - Anil Kumar
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China; ,
| | - Peter Carmeliet
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China; , .,Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven B-3000, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven B-3000, Belgium
| |
Collapse
|
33
|
Dheer P, Rautela I, Sharma V, Dhiman M, Sharma A, Sharma N, Sharma MD. Evolution in crop improvement approaches and future prospects of molecular markers to CRISPR/Cas9 system. Gene 2020; 753:144795. [PMID: 32450202 DOI: 10.1016/j.gene.2020.144795] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/07/2020] [Accepted: 05/19/2020] [Indexed: 01/03/2023]
Abstract
The advent of genetic selection and genome modification method assure about a real novel reformation in biotechnology and genetic engineering. With the extensive capabilities of molecular markers of them being stable, cost-effective and easy to use, they ultimately become a potent tool for variety of applications such a gene targeting, selection, editing, functional genomics; mainly for the improvisation of commercially important crops. Three main benefits of molecular marker in the field of agriculture and crop improvement programmes first, reduction of the duration of breeding programmes, second, they allow creation of new genetic variation and genetic diversity of plants and third most promising benefit is help in production of engineered plant for disease resistance, or resistance from pathogen and herbicides. This review is anticipated to present an outline how the techniques have been evolved from the simple conventional applications of DNA based molecular markers to highly throughput CRISPR technology and geared the crop yield. Techniques like using Zinc Finger Nucleases (ZFNs), Transcription Activator-Like Effector Nucleases (TALENs) and Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR/Cas9) systems have revolutionised in the field of genome editing. These have been promptly accepted in both the research and commercial industry. On the whole, the widespread use of molecular markers with their types, their appliance in plant breeding along with the advances in genetic selection and genome editing together being a novel strategy to boost crop yield has been reviewed.
Collapse
Affiliation(s)
- Pallavi Dheer
- Department of Life Sciences, Shri Guru Ram Rai Institute of Technology & Science, Patel Nagar, Dehradun, Uttarakhand, India
| | - Indra Rautela
- Department of Biotechnology, SALS, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Vandana Sharma
- Department of Botany, K.L.DAV (PG) College, Roorkee,Uttarakhand, India
| | - Manjul Dhiman
- Department of Botany, K.L.DAV (PG) College, Roorkee,Uttarakhand, India
| | - Aditi Sharma
- Department of Biotechnology, Graphic Era University, Dehradun, Uttarakhand, India
| | - Nishesh Sharma
- Department of Biotechnology, SALS, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Manish Dev Sharma
- Department of Biotechnology, School of Basic and Applied Sciences, Shri Guru Ram Rai University, Patel Nagar, Dehradun, Uttarakhand, India.
| |
Collapse
|
34
|
Li Q, Xuan W, Jia Z, Li H, Li M, Liang X, Su D. HRD1 prevents atherosclerosis-mediated endothelial cell apoptosis by promoting LOX-1 degradation. Cell Cycle 2020; 19:1466-1477. [PMID: 32308114 DOI: 10.1080/15384101.2020.1754561] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The 3-hydroxy-3-methylglutaryl reductase degradation (HRD1) is an E3 ubiquitin ligase that can preserve heart structure and function, but its role in endothelial dysfunction and atherosclerosis (AS) is unclear. The aim of this study was to explore the role and biological function of HRD1 in AS. HRD1 expression was significantly decreased in atherosclerotic intima and ox-LDL led to a decrease of HRD1 level in endothelial cells (ECs). Forced expression of HRD1 inhibited the endothelial apoptosis induced by ox-LDL. The transcription factor KLF2 specifically bound to the HRD1 promoter and positively regulated HRD1 expression. KLF2 up-regulation could reverse the decrease of HRD1 level in ECs treated with ox-LDL. Further analysis showed that HRD1 interacted with LOX-1 and promoted ubiquitination and degradation of LOX-1 by the proteasome. Deletion of LOX-1 attenuated the ECs apoptosis induced by HRD1 downregulation. Pravastatin, which protected EC from damage via a KLF2-dependent mechanism, could dose-dependently enhanced HRD1 expression in EC exposed to ox-LDL. Interestingly, interference of HRD1 abolished the cytoprotective effect of pravastatin. Collectively, our data indicate that decreased HRD1 expression leads to apoptosis of ECs and restoration of HRD1 expression could represent a novel strategy for human AS therapy.
Collapse
Affiliation(s)
- Qingguo Li
- Department of Cardiovascular Surgery, 2nd Affiliated Hospital of Nanjing Medical University , Nanjing, China
| | - Wenying Xuan
- Department of Stomatology, Xuanwu Hospital , Nanjing, China
| | - Zhijun Jia
- Department of Nuclear Medicine, The Affiliated Drum Tower Hospital of Nanjing University , Nanjing, China
| | - Hongyan Li
- Department of Pathology, Nanjing Medical University , Nanjing, China
| | - Min Li
- Department of Pathology, Nanjing Medical University , Nanjing, China
| | - Xiubin Liang
- Center of Pathology and Clinical Laboratory, Sir Runrun Hospital of Nanjing Medical University , Nanjing, China
| | - Dongming Su
- Department of Pathology, Nanjing Medical University , Nanjing, China.,Center of Pathology and Clinical Laboratory, Sir Runrun Hospital of Nanjing Medical University , Nanjing, China
| |
Collapse
|
35
|
Turpaev KT. Transcription Factor KLF2 and Its Role in the Regulation of Inflammatory Processes. BIOCHEMISTRY (MOSCOW) 2020; 85:54-67. [PMID: 32079517 DOI: 10.1134/s0006297920010058] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
KLF2 is a member of the Krüppel-like transcription factor family of proteins containing highly conserved DNA-binding zinc finger domains. KLF2 participates in the differentiation and regulation of the functional activity of monocytes, T lymphocytes, adipocytes, and vascular endothelial cells. The activity of KLF2 is controlled by several regulatory systems, including the MEKK2,3/MEK5/ERK5/MEF2 MAP kinase cascade, Rho family G-proteins, histone acetyltransferases CBP and p300, and histone deacetylases HDAC4 and HDAC5. Activation of KLF2 in endothelial cells induces eNOS expression and provides vasodilatory effect. Many KLF2-dependent genes participate in the suppression of blood coagulation and aggregation of T cells and macrophages with the vascular endothelium, thereby preventing atherosclerosis progression. KLF2 can have a dual effect on the gene transcription. Thus, it induces expression of multiple genes, but suppresses transcription of NF-κB-dependent genes. Transcription factors KLF2 and NF-κB are reciprocal antagonists. KLF2 inhibits induction of NF-κB-dependent genes, whereas NF-κB downregulates KLF2 expression. KLF2-mediated inhibition of NF-κB signaling leads to the suppression of cell response to the pro-inflammatory cytokines IL-1β and TNFα and results in the attenuation of inflammatory processes.
Collapse
Affiliation(s)
- K T Turpaev
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, Moscow, 119991, Russia.
| |
Collapse
|
36
|
Xia Y, Feng H, Li ZW, Tang KX, Gao HQ, Wang WL, Cui XP, Li XL. Low-dose phloretin alleviates diabetic atherosclerosis through endothelial KLF2 restoration. Biosci Biotechnol Biochem 2020; 84:815-823. [PMID: 31791197 DOI: 10.1080/09168451.2019.1699396] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
ABSTRACT
We investigated whether low-dose phloretin served as daily dietary supplements could ameliorate diabetic atherosclerosis and the role of kruppel-like factor 2 (KLF2). HUVECs cultured in high glucose medium were treated with different concentrations of phloretin and KLF2 mRNA, and protein level was detected. Diabetes was induced using streptozotocin in Apoe−/- mice after which they were fed a high-cholesterol diet for 8 weeks. Diabetic mice injected with KLF2 shRNA-lentivirus or control virus were treated with 20 mg/kg phloretin. Glucose, lipid profile, aortic atheroma, and endothelial nitric oxide synthase (eNOS) expression were detected. Phloretin retained endothelial function by KLF2-eNOS activation under hyperglycemia. Low-dose phloretin helped with lipid metabolism, and blocked the acceleration of atherosclerosis in STZ-induced diabetic mice since the early stage, which was diminished by KLF2 knockdown. Low-dose phloretin exhibited athero-protective effect in diabetic Apoe−/- mice dependent on KLF2 activation. This finding makes phloretin for diabetic atherosclerosis.
Collapse
Affiliation(s)
- Yong Xia
- Department of Geriatric Medicine, Qi-lu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Hua Feng
- Department of Digestive Disease, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Zhen-Wei Li
- Department of Gastroenterology, Mengyin People’s Hospital, Mengyin, China
| | - Kuan-Xiao Tang
- Department of Geriatric Medicine, Qi-lu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Hai-Qing Gao
- Department of Geriatric Medicine, Qi-lu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Wei-Ling Wang
- Department of Geriatric Medicine, Qi-lu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Xiao-Pei Cui
- Department of Geriatric Medicine, Qi-lu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Xiao-Li Li
- Department of Drug Purchase and Supply, Qi-Lu Hospital of Shandong University, Jinan, China
| |
Collapse
|
37
|
KLF2 Protects against Osteoarthritis by Repressing Oxidative Response through Activation of Nrf2/ARE Signaling In Vitro and In Vivo. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8564681. [PMID: 31827706 PMCID: PMC6885785 DOI: 10.1155/2019/8564681] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 10/17/2019] [Indexed: 12/21/2022]
Abstract
Osteoarthritis (OA) is a multifactorial and inflammatory disease characterized by cartilage destruction that can cause disability among aging patients. There is currently no effective treatment that can arrest or reverse OA progression. Kruppel-like factor 2 (KLF2), a member of the zinc finger family, has emerged as a transcription factor involved in a wide variety of inflammatory diseases. Here, we identified that KLF2 expression is downregulated in IL-1β-treated human chondrocytes and OA cartilage. Genetic and pharmacological overexpression of KLF2 suppressed IL-1β-induced apoptosis and matrix degradation through the suppression of reactive oxygen species (ROS) production. In addition, KLF2 overexpression resulted in increased expression of heme oxygenase-1 (HO-1) and NAD(P)H dehydrogenase quinone 1 (NQO1) through the enhanced nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2). Further, Nrf2 inhibition abrogated the chondroprotective effects of KLF2. Safranin O/fast green and TUNEL staining demonstrated that adenovirus-mediated overexpression of KLF2 in joint cartilage protects rats against experimental OA by inhibiting cartilage degradation and chondrocyte apoptosis. Immunohistochemical staining revealed that KLF2 overexpression significantly decreases MMP13 expression caused by OA progression in vivo. This in vitro and in vivo study is the first to investigate the antioxidative effect and mechanisms of KLF2 in OA pathogenesis. Our results collectively provide new insights into OA pathogenesis regulated by KLF2 and a rationale for the development of effective OA intervention strategies.
Collapse
|
38
|
Malik N, Dunn KM, Cassels J, Hay J, Estell C, Sansom OJ, Michie AM. mTORC1 activity is essential for erythropoiesis and B cell lineage commitment. Sci Rep 2019; 9:16917. [PMID: 31729420 PMCID: PMC6858379 DOI: 10.1038/s41598-019-53141-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 10/26/2019] [Indexed: 12/17/2022] Open
Abstract
Mechanistic target of rapamycin (mTOR) is a serine/threonine protein kinase that mediates phosphoinositide-3-kinase (PI3K)/AKT signalling. This pathway is involved in a plethora of cellular functions including protein and lipid synthesis, cell migration, cell proliferation and apoptosis. In this study, we proposed to delineate the role of mTORC1 in haemopoietic lineage commitment using knock out (KO) mouse and cell line models. Mx1-cre and Vav-cre expression systems were used to specifically target Raptorfl/fl (mTORC1), either in all tissues upon poly(I:C) inoculation, or specifically in haemopoietic stem cells, respectively. Assessment of the role of mTORC1 during the early stages of development in Vav-cre+Raptorfl/fl mice, revealed that these mice do not survive post birth due to aberrations in erythropoiesis resulting from an arrest in development at the megakaryocyte-erythrocyte progenitor stage. Furthermore, Raptor-deficient mice exhibited a block in B cell lineage commitment. The essential role of Raptor (mTORC1) in erythrocyte and B lineage commitment was confirmed in adult Mx1-cre+Raptorfl/fl mice upon cre-recombinase induction. These studies were supported by results showing that the expression of key lineage commitment regulators, GATA1, GATA2 and PAX5 were dysregulated in the absence of mTORC1-mediated signals. The regulatory role of mTOR during erythropoiesis was confirmed in vitro by demonstrating a reduction of K562 cell differentiation towards RBCs in the presence of established mTOR inhibitors. While mTORC1 plays a fundamental role in promoting RBC development, we showed that mTORC2 has an opposing role, as Rictor-deficient progenitor cells exhibited an elevation in RBC colony formation ex vivo. Collectively, our data demonstrate a critical role played by mTORC1 in regulating the haemopoietic cell lineage commitment.
Collapse
Affiliation(s)
- Natasha Malik
- Institute of Cancer Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Karen M Dunn
- Institute of Cancer Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Jennifer Cassels
- Institute of Cancer Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Jodie Hay
- Institute of Cancer Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Christopher Estell
- Institute of Cancer Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Owen J Sansom
- Institute of Cancer Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, UK
| | - Alison M Michie
- Institute of Cancer Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
39
|
Li N, Liu SF, Dong K, Zhang GC, Huang J, Wang ZH, Wang TJ. Exosome-Transmitted miR-25 Induced by H. pylori Promotes Vascular Endothelial Cell Injury by Targeting KLF2. Front Cell Infect Microbiol 2019; 9:366. [PMID: 31750260 PMCID: PMC6842922 DOI: 10.3389/fcimb.2019.00366] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 10/08/2019] [Indexed: 02/06/2023] Open
Abstract
Background: Increasing evidence has shown that Helicobacter pylori is associated with coronary heart disease (CHD); however, the underlying mechanism remains unclear. Methods: The expression of miR-25 and mRNAs was measured using qRT-PCR. Protein levels were detected using western blotting and exosomes were assessed with an electron microscope. The target gene of miR-25 was identified using the luciferase report system. Results: H. pylori infection increased the expression of miR-25 in gastric epithelial cells and was associated with increased levels of exosome-transmitted miR-25 in human peripheral blood. Mechanistic investigation showed the Kruppel-like factor 2 (KLF2) was a direct target of exosome-transmitted miR-25 in vascular endothelial cells. In addition, the miR-25/KLF2 axis regulated the NF-κB signaling pathway, resulting in increased expression of interleukin 6 (IL6), monocyte chemoattractant protein-1 (MCP-1), vascular cell adhesion molecule-1 (VCAM-1), and intercellular adhesion molecule-1 (ICAM-1). Conclusion: Our findings suggest that the miR-25/KLF2 axis may be a potential therapeutic target for H. pylori-associated CHD. Furthermore, high levels of exosome-transmitted miR-25 in peripheral blood may pose a potential risk for CHD.
Collapse
Affiliation(s)
- Na Li
- Department of Pediatric Cardiac Surgery, Institute of Cardiovascular Disease, The 960th Hospital of Chinese PLA, Jinan, China
| | - Shi-Feng Liu
- Department of Pediatric Cardiac Surgery, Institute of Cardiovascular Disease, The 960th Hospital of Chinese PLA, Jinan, China
| | - Kai Dong
- Department of Pediatric Cardiac Surgery, Institute of Cardiovascular Disease, The 960th Hospital of Chinese PLA, Jinan, China
| | - Gui-Chun Zhang
- Department of Pediatric Cardiac Surgery, Institute of Cardiovascular Disease, The 960th Hospital of Chinese PLA, Jinan, China
| | - Jing Huang
- Department of Pediatric Cardiac Surgery, Institute of Cardiovascular Disease, The 960th Hospital of Chinese PLA, Jinan, China
| | - Zhi-Heng Wang
- Department of Pediatric Cardiac Surgery, Institute of Cardiovascular Disease, The 960th Hospital of Chinese PLA, Jinan, China
| | - Tong-Jian Wang
- Department of Pediatric Cardiac Surgery, Institute of Cardiovascular Disease, The 960th Hospital of Chinese PLA, Jinan, China
| |
Collapse
|
40
|
Liu C, Chen Y, Deng Y, Dong Y, Jiang J, Chen S, Kang W, Deng J, Sun H. Survival-based bioinformatics analysis to identify hub genes and key pathways in non-small cell lung cancer. Transl Cancer Res 2019; 8:1188-1198. [PMID: 35116861 PMCID: PMC8797769 DOI: 10.21037/tcr.2019.06.35] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 05/21/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND Lung cancer is one of the leading causes of cancer mortality worldwide. Here, we performed an integrative bioinformatics analysis to screen hub genes and critical pathways in non-small cell lung cancer (NSCLC) based on the overall survival rate of differentially expressed genes (DEGs). METHODS Four datasets from the gene expression omnibus (GEO) were used to identify the DEGs. To obtain robust DEGs in NSCLC, only the DEGs that co-existed in the four datasets were selected for subsequent analysis. To identify the genes correlated with overall survival, the overall survival of these genes was then analyzed using the Kaplan-Meier plotter database. The genes significantly correlated with survival were used to perform gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) analysis; next, these genes were used to construct a protein-protein interaction network. MCODE and CytoHubba were used to identify the clusters and hub genes. Finally, the hub genes were validated in the Cancer Genome Atlas (TCGA) and the Human Protein Atlas (HPA). RESULTS We found 522 up-regulated DEGs, and 989 down-regulated DEGs between the NSCLC and normal lung tissue, and 895 of them were correlated with a higher overall survival. GO analysis showed that the DEGs that were associated with a higher overall survival were enriched in cell division, cell cycle, DNA replication, angiogenesis, and cell migration. KEGG analysis was consistent with GO analysis and showed that p53 signaling pathway, pyrimidine metabolism, cGMP-PKG signaling pathway and renin secretion pathway were associated with overall survival in NSCLC. In the protein-protein analysis, we identified seven clusters and six hub genes which were BUB1B, CCNB1, CENPE, KIF18A, NDC10, and MAD2L1. Of these genes, CENPE and KIF18A had not been reported until now. Finally, the dysregulated expression of the six hub genes was validated by the data from the TCGA and HPA. CONCLUSIONS We identified the hub genes and potential mechanisms of NSCLC based on multiple-microarray analysis and overall survival; then, validated the hub genes in the TCGA and HPA database. These hub genes may serve as potential therapeutic targets.
Collapse
Affiliation(s)
- Chunliang Liu
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yu Chen
- Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yuqi Deng
- Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yu Dong
- Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jixuan Jiang
- Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Si Chen
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wenfeng Kang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jiong Deng
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Haipeng Sun
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
41
|
Morin-Brureau M, Milior G, Royer J, Chali F, Le Duigou C, Savary E, Blugeon C, Jourdren L, Akbar D, Dupont S, Navarro V, Baulac M, Bielle F, Mathon B, Clemenceau S, Miles R. Microglial phenotypes in the human epileptic temporal lobe. Brain 2019; 141:3343-3360. [PMID: 30462183 DOI: 10.1093/brain/awy276] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 09/12/2018] [Indexed: 12/11/2022] Open
Abstract
Microglia, the immune cells of the brain, are highly plastic and possess multiple functional phenotypes. Differences in phenotype in different regions and different states of epileptic human brain have been little studied. Here we use transcriptomics, anatomy, imaging of living cells and ELISA measurements of cytokine release to examine microglia from patients with temporal lobe epilepsies. Two distinct microglial phenotypes were explored. First we asked how microglial phenotype differs between regions of high and low neuronal loss in the same brain. Second, we asked how microglial phenotype is changed by a recent seizure. In sclerotic areas with few neurons, microglia have an amoeboid rather than ramified shape, express activation markers and respond faster to purinergic stimuli. The repairing interleukin, IL-10, regulates the basal phenotype of microglia in the CA1 and CA3 regions with neuronal loss and gliosis. To understand changes in phenotype induced by a seizure, we estimated the delay from the last seizure until tissue collection from changes in reads for immediate early gene transcripts. Pseudotime ordering of these data was validated by comparison with results from kainate-treated mice. It revealed a local and transient phenotype in which microglia secrete the human interleukin CXCL8, IL-1B and other cytokines. This secretory response is mediated in part via the NRLP3 inflammasome.
Collapse
Affiliation(s)
- Mélanie Morin-Brureau
- Inserm U1127, CNRS UMR7225, Sorbonne Universités, UPMC Univ Paris 6 UMR S1127, Institut du Cerveau et de la Moelle épinière, Paris, France
| | - Giampaolo Milior
- Inserm U1127, CNRS UMR7225, Sorbonne Universités, UPMC Univ Paris 6 UMR S1127, Institut du Cerveau et de la Moelle épinière, Paris, France
| | - Juliette Royer
- Inserm U1127, CNRS UMR7225, Sorbonne Universités, UPMC Univ Paris 6 UMR S1127, Institut du Cerveau et de la Moelle épinière, Paris, France
| | - Farah Chali
- Inserm U1127, CNRS UMR7225, Sorbonne Universités, UPMC Univ Paris 6 UMR S1127, Institut du Cerveau et de la Moelle épinière, Paris, France
| | - Caroline Le Duigou
- Inserm U1127, CNRS UMR7225, Sorbonne Universités, UPMC Univ Paris 6 UMR S1127, Institut du Cerveau et de la Moelle épinière, Paris, France
| | - Etienne Savary
- Inserm U1127, CNRS UMR7225, Sorbonne Universités, UPMC Univ Paris 6 UMR S1127, Institut du Cerveau et de la Moelle épinière, Paris, France
| | - Corinne Blugeon
- École normale supérieure, PSL Research University, CNRS, Inserm, Institut de Biologie de l'École normale supérieure (IBENS), Plateforme Génomique, Paris, France
| | - Laurent Jourdren
- École normale supérieure, PSL Research University, CNRS, Inserm, Institut de Biologie de l'École normale supérieure (IBENS), Plateforme Génomique, Paris, France
| | - David Akbar
- Inserm U1127, CNRS UMR7225, Sorbonne Universités, UPMC Univ Paris 6 UMR S1127, Institut du Cerveau et de la Moelle épinière, Paris, France
| | - Sophie Dupont
- AP-HP, GH Pitie-Salpêtrière-Charles Foix, Epilepsy Unit (VN, MB, SD), Neuropathologie (FB), Neurochirurgie (BM, SC), Paris, France
| | - Vincent Navarro
- AP-HP, GH Pitie-Salpêtrière-Charles Foix, Epilepsy Unit (VN, MB, SD), Neuropathologie (FB), Neurochirurgie (BM, SC), Paris, France
| | - Michel Baulac
- AP-HP, GH Pitie-Salpêtrière-Charles Foix, Epilepsy Unit (VN, MB, SD), Neuropathologie (FB), Neurochirurgie (BM, SC), Paris, France
| | - Franck Bielle
- AP-HP, GH Pitie-Salpêtrière-Charles Foix, Epilepsy Unit (VN, MB, SD), Neuropathologie (FB), Neurochirurgie (BM, SC), Paris, France
| | - Bertrand Mathon
- AP-HP, GH Pitie-Salpêtrière-Charles Foix, Epilepsy Unit (VN, MB, SD), Neuropathologie (FB), Neurochirurgie (BM, SC), Paris, France
| | - Stéphane Clemenceau
- AP-HP, GH Pitie-Salpêtrière-Charles Foix, Epilepsy Unit (VN, MB, SD), Neuropathologie (FB), Neurochirurgie (BM, SC), Paris, France
| | - Richard Miles
- Inserm U1127, CNRS UMR7225, Sorbonne Universités, UPMC Univ Paris 6 UMR S1127, Institut du Cerveau et de la Moelle épinière, Paris, France
| |
Collapse
|
42
|
Venturini G, Malagrino PA, Padilha K, Tanaka LY, Laurindo FR, Dariolli R, Carvalho VM, Cardozo KHM, Krieger JE, Pereira ADC. Integrated proteomics and metabolomics analysis reveals differential lipid metabolism in human umbilical vein endothelial cells under high and low shear stress. Am J Physiol Cell Physiol 2019; 317:C326-C338. [PMID: 31067084 DOI: 10.1152/ajpcell.00128.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Atherosclerotic plaque development is closely associated with the hemodynamic forces applied to endothelial cells (ECs). Among these, shear stress (SS) plays a key role in disease development since changes in flow intensity and direction could stimulate an atheroprone or atheroprotective phenotype. ECs under low or oscillatory SS (LSS) show upregulation of inflammatory, adhesion, and cellular permeability molecules. On the contrary, cells under high or laminar SS (HSS) increase their expression of protective and anti-inflammatory factors. The mechanism behind SS regulation of an atheroprotective phenotype is not completely elucidated. Here we used proteomics and metabolomics to better understand the changes in endothelial cells (human umbilical vein endothelial cells) under in vitro LSS and HSS that promote an atheroprone or atheroprotective profile and how these modifications can be connected to atherosclerosis development. Our data showed that lipid metabolism, in special cholesterol metabolism, was downregulated in cells under LSS. The low-density lipoprotein receptor (LDLR) showed significant alterations both at the quantitative expression level as well as regarding posttranslational modifications. Under LSS, LDLR was seen at lower concentrations and with a different glycosylation profile. Finally, modulating LDLR with atorvastatin led to the recapitulation of a HSS metabolic phenotype in EC under LSS. Altogether, our data suggest that there is significant modulation of lipid metabolism in endothelial cells under different SS intensities and that this could contribute to the atheroprone phenotype of LSS. Statin treatment was able to partially recover the protective profile of these cells.
Collapse
Affiliation(s)
- Gabriela Venturini
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Pamella Araujo Malagrino
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Kallyandra Padilha
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Leonardo Yuji Tanaka
- Vascular Biology Laboratory, Heart Institute (InCor), Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Francisco Rafael Laurindo
- Vascular Biology Laboratory, Heart Institute (InCor), Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Rafael Dariolli
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | | | | | - Jose Eduardo Krieger
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Alexandre da Costa Pereira
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
43
|
Lopes-da-Silva M, McCormack JJ, Burden JJ, Harrison-Lavoie KJ, Ferraro F, Cutler DF. A GBF1-Dependent Mechanism for Environmentally Responsive Regulation of ER-Golgi Transport. Dev Cell 2019; 49:786-801.e6. [PMID: 31056345 PMCID: PMC6764485 DOI: 10.1016/j.devcel.2019.04.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 02/19/2019] [Accepted: 04/04/2019] [Indexed: 12/17/2022]
Abstract
How can anterograde membrane trafficking be modulated by physiological cues? A screen of Golgi-associated proteins revealed that the ARF-GEF GBF1 can selectively modulate the ER-Golgi trafficking of prohaemostatic von Willebrand factor (VWF) and extracellular matrix (ECM) proteins in human endothelial cells and in mouse fibroblasts. The relationship between levels of GBF1 and the trafficking of VWF into forming secretory granules confirmed GBF1 is a limiting factor in this process. Further, GBF1 activation by AMPK couples its control of anterograde trafficking to physiological cues; levels of glucose control GBF1 activation in turn modulating VWF trafficking into secretory granules. GBF1 modulates both ER and TGN exit, the latter dramatically affecting the size of the VWF storage organelles, thereby influencing the hemostatic capacity of the endothelium. The role of AMPK as a central integrating element of cellular pathways with intra- and extra-cellular cues can now be extended to modulation of the anterograde secretory pathway. The Arf-GEF GBF1 modulates anterograde trafficking of VWF and ECM proteins Loss of GBF1 slows ER and TGN exit, producing swollen ER and giant WPBs Activation of GBF1 via AMPK reduces endothelial WPB size and secretion Metabolic change alters anterograde trafficking and cargo secretion via AMPK-GBF1
Collapse
Affiliation(s)
- Mafalda Lopes-da-Silva
- Endothelial Cell Biology Laboratory, MRC Laboratory for Molecular Cell Biology, University College London, London, UK.
| | - Jessica J McCormack
- Endothelial Cell Biology Laboratory, MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Jemima J Burden
- Electron Microscopy Laboratory, MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Kimberly J Harrison-Lavoie
- Endothelial Cell Biology Laboratory, MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Francesco Ferraro
- Endothelial Cell Biology Laboratory, MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Daniel F Cutler
- Endothelial Cell Biology Laboratory, MRC Laboratory for Molecular Cell Biology, University College London, London, UK.
| |
Collapse
|
44
|
Montalva L, Antounians L, Zani A. Pulmonary hypertension secondary to congenital diaphragmatic hernia: factors and pathways involved in pulmonary vascular remodeling. Pediatr Res 2019; 85:754-768. [PMID: 30780153 DOI: 10.1038/s41390-019-0345-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 02/10/2019] [Indexed: 02/06/2023]
Abstract
Congenital diaphragmatic hernia (CDH) is a severe birth defect that is characterized by pulmonary hypoplasia and pulmonary hypertension (PHTN). PHTN secondary to CDH is a result of vascular remodeling, a structural alteration in the pulmonary vessel wall that occurs in the fetus. Factors involved in vascular remodeling have been reported in several studies, but their interactions remain unclear. To help understand PHTN pathophysiology and design novel preventative and treatment strategies, we have conducted a systematic review of the literature and comprehensively analyzed all factors and pathways involved in the pathogenesis of pulmonary vascular remodeling secondary to CDH in the nitrofen model. Moreover, we have linked the dysregulated factors with pathways involved in human CDH. Of the 358 full-text articles screened, 75 studies reported factors that play a critical role in vascular remodeling secondary to CDH. Overall, the impairment of epithelial homeostasis present in pulmonary hypoplasia results in altered signaling to endothelial cells, leading to endothelial dysfunction. This causes an impairment of the crosstalk between endothelial cells and pulmonary artery smooth muscle cells, resulting in increased smooth muscle cell proliferation, resistance to apoptosis, and vasoconstriction, which clinically translate into PHTN.
Collapse
Affiliation(s)
- Louise Montalva
- Division of General and Thoracic Surgery, Department of Surgery, The Hospital for Sick Children, Toronto, Canada.,Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
| | - Lina Antounians
- Division of General and Thoracic Surgery, Department of Surgery, The Hospital for Sick Children, Toronto, Canada.,Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
| | - Augusto Zani
- Division of General and Thoracic Surgery, Department of Surgery, The Hospital for Sick Children, Toronto, Canada. .,Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Canada.
| |
Collapse
|
45
|
Hou Z, Wang Z, Tao Y, Bai J, Yu B, Shen J, Sun H, Xiao L, Xu Y, Zhou J, Wang Z, Geng D. KLF2 regulates osteoblast differentiation by targeting of Runx2. J Transl Med 2019; 99:271-280. [PMID: 30429507 DOI: 10.1038/s41374-018-0149-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 08/26/2018] [Accepted: 09/14/2018] [Indexed: 01/15/2023] Open
Abstract
Osteoblast differentiation plays a critical role in bone formation and maintaining balance in bone remodeling. Runt-related transcription factor 2 (Runx2) is a central transcription factor regulating osteoblast differentiation and promoting bone mineralization. Until now, the molecular regulatory basis and especially the gene regulatory network of osteogenic differentiation have been unclear. Krüppel-like factor 2 (KLF2) is a zinc finger structure and DNA-binding transcription factor. The current study aimed to investigate the physiological function of KLF2 in osteoblast differentiation. Our results indicate that KLF2 is expressed in pre-osteoblast MC3T3-E1 cells and primary osteoblasts. Interestingly, KLF2 expression is increased in osteoblasts during the osteoblastic differentiation process. Overexpression of KLF2 in MC3T3-E1 cells promoted the expression of the osteoblastic differentiation marker genes Alp, Osx, and Ocn, and stimulated mineralization by increasing Runx2 expression at both the mRNA and protein levels. In contrast, knockdown of KLF2 produced the opposite effects. Importantly, we found that KLF2 could physically interact with Runx2. KLF2 promoted osteoblast differentiation by regulating Runx2 and physically interacting with Runx2. Taken together, the findings of this study identify KLF2 as a novel regulator of osteoblast differentiation. Our findings suggest that KLF2 might be a new therapeutic target for bone disease.
Collapse
Affiliation(s)
- Zhenyang Hou
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China.,Department of Orthopaedics, Tengzhou Central People's Hospital, Tengzhou, Shandong, 277500, China
| | - Zhen Wang
- Department of Orthopaedics, Suzhou Kowloon Hospital Shanghai Jiao Tong University School of Medicine, Suzhou, Jiangsu, 215006, China
| | - Yunxia Tao
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Jiaxiang Bai
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Binqing Yu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Jining Shen
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Houyi Sun
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Long Xiao
- Department of Orthopaedics, Zhangjiagang Hospital of Traditional Chinese Medicine, Zhangjiagang, Suzhou, Jiangsu, 215600, China
| | - Yaozeng Xu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Jun Zhou
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Zhirong Wang
- Department of Orthopaedics, Zhangjiagang Hospital of Traditional Chinese Medicine, Zhangjiagang, Suzhou, Jiangsu, 215600, China.
| | - Dechun Geng
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China.
| |
Collapse
|
46
|
Paolini A, Abdelilah-Seyfried S. The mechanobiology of zebrafish cardiac valve leaflet formation. Curr Opin Cell Biol 2018; 55:52-58. [DOI: 10.1016/j.ceb.2018.05.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 05/10/2018] [Indexed: 12/31/2022]
|
47
|
Poletto V, Rosti V, Biggiogera M, Guerra G, Moccia F, Porta C. The role of endothelial colony forming cells in kidney cancer's pathogenesis, and in resistance to anti-VEGFR agents and mTOR inhibitors: A speculative review. Crit Rev Oncol Hematol 2018; 132:89-99. [PMID: 30447930 DOI: 10.1016/j.critrevonc.2018.09.005] [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] [Received: 02/04/2018] [Revised: 08/07/2018] [Accepted: 09/08/2018] [Indexed: 12/22/2022] Open
Abstract
Renal cell carcinoma (RCC) is highly dependent on angiogenesis, due to the overactivation of the VHL/HIF/VEGF/VEGFRs axis; this justifies the marked sensitivity of this neoplasm to antiangiogenic agents which, however, ultimately fail to control tumor growth. RCC also frequently shows alterations in the mTOR signaling pathway, and mTOR inhibitors have shown a similar pattern of initial activity/late failure as pure antiangiogenic agents. Understanding mechanisms of resistance to these agents would be key to improve the outcome of our patients. Circulating endothelial cells are a family of mainly bone marrow-derived progenitors, which have been postulated to be responsible of the reactivation of angiogenesis in different tumors. In this review, we shall discuss the complex nature and function of these cells, the evidence pro and contra their contribution to tumor vascularization, especially as far as RCC is concerned, and their possible role in determining resistance to presently available treatments.
Collapse
Affiliation(s)
- Valentina Poletto
- Center for the Study of Myelofibrosis, Research and Experimental Biotechnology Laboratory Area, Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Policlinico San Matteo Foundation, Piazzale Golgi 19, 27100, Pavia, Italy.
| | - Vittorio Rosti
- Center for the Study of Myelofibrosis, Research and Experimental Biotechnology Laboratory Area, Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Policlinico San Matteo Foundation, Piazzale Golgi 19, 27100, Pavia, Italy.
| | - Marco Biggiogera
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Italy.
| | - Germano Guerra
- Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, Campobasso, Italy.
| | - Francesco Moccia
- Laboratory of Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, via Forlanini 6, 27100, Pavia, Italy.
| | - Camillo Porta
- Medical Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Policlinico San Matteo Foundation, Piazzale Golgi 19, 27100, Pavia, Italy; present address: Department of Internal Medicine, University of Pavia, and Division of Translational Oncology, IRCCS Istituti Clinici Scientifici Maugeri, via S. Maugeri 10, 27100 Pavia, Italy.
| |
Collapse
|
48
|
Wang X, Wu Z, He Y, Zhang H, Tian L, Zheng C, Shang T, Zhu Q, Li D, He Y. Humanin prevents high glucose-induced monocyte adhesion to endothelial cells by targeting KLF2. Mol Immunol 2018; 101:245-250. [DOI: 10.1016/j.molimm.2018.07.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 06/22/2018] [Accepted: 07/02/2018] [Indexed: 01/10/2023]
|
49
|
Colunga T, Dalton S. Building Blood Vessels with Vascular Progenitor Cells. Trends Mol Med 2018; 24:630-641. [PMID: 29802036 PMCID: PMC6050017 DOI: 10.1016/j.molmed.2018.05.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 05/02/2018] [Accepted: 05/03/2018] [Indexed: 12/20/2022]
Abstract
Vascular progenitor cells have been identified from perivascular cell fractions and peripheral blood and bone marrow mononuclear fractions. These vascular progenitors share the ability to generate some of the vascular lineages, including endothelial cells, smooth muscle cells, and pericytes. The potential therapeutic uses for vascular progenitor cells are broad and relate to stroke, ischemic disease, and to the engineering of whole organs and tissues that require a vascular component. This review summarizes the best-characterized sources of vascular progenitor cells and discusses advances in 3D printing and electrospinning using blended polymers for the creation of biomimetic vascular grafts. These advances are pushing the field of regenerative medicine closer to the creation of small-diameter vascular grafts with long-term clinical utility.
Collapse
Affiliation(s)
- Thomas Colunga
- Center for Molecular Medicine, University of Georgia, 325 Riverbend Road, Athens, GA 30605, USA; Department of Biochemistry and Molecular Biology, University of Georgia, 325 Riverbend Road, Athens, GA 30605, USA
| | - Stephen Dalton
- Center for Molecular Medicine, University of Georgia, 325 Riverbend Road, Athens, GA 30605, USA; Department of Biochemistry and Molecular Biology, University of Georgia, 325 Riverbend Road, Athens, GA 30605, USA.
| |
Collapse
|
50
|
Donat S, Lourenço M, Paolini A, Otten C, Renz M, Abdelilah-Seyfried S. Heg1 and Ccm1/2 proteins control endocardial mechanosensitivity during zebrafish valvulogenesis. eLife 2018; 7:28939. [PMID: 29364115 PMCID: PMC5794256 DOI: 10.7554/elife.28939] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 01/24/2018] [Indexed: 12/18/2022] Open
Abstract
Endothelial cells respond to different levels of fluid shear stress through adaptations of their mechanosensitivity. Currently, we lack a good understanding of how this contributes to sculpting of the cardiovascular system. Cerebral cavernous malformation (CCM) is an inherited vascular disease that occurs when a second somatic mutation causes a loss of CCM1/KRIT1, CCM2, or CCM3 proteins. Here, we demonstrate that zebrafish Krit1 regulates the formation of cardiac valves. Expression of heg1, which encodes a binding partner of Krit1, is positively regulated by blood-flow. In turn, Heg1 stabilizes levels of Krit1 protein, and both Heg1 and Krit1 dampen expression levels of klf2a, a major mechanosensitive gene. Conversely, loss of Krit1 results in increased expression of klf2a and notch1b throughout the endocardium and prevents cardiac valve leaflet formation. Hence, the correct balance of blood-flow-dependent induction and Krit1 protein-mediated repression of klf2a and notch1b ultimately shapes cardiac valve leaflet morphology.
Collapse
Affiliation(s)
- Stefan Donat
- Institute of Biochemistry and Biology, Potsdam University, Potsdam, Germany.,Institute of Molecular Biology, Hannover Medical School, Hannover, Germany
| | - Marta Lourenço
- Institute of Biochemistry and Biology, Potsdam University, Potsdam, Germany
| | - Alessio Paolini
- Institute of Biochemistry and Biology, Potsdam University, Potsdam, Germany
| | - Cécile Otten
- Institute of Biochemistry and Biology, Potsdam University, Potsdam, Germany
| | - Marc Renz
- Institute of Biochemistry and Biology, Potsdam University, Potsdam, Germany
| | - Salim Abdelilah-Seyfried
- Institute of Biochemistry and Biology, Potsdam University, Potsdam, Germany.,Institute of Molecular Biology, Hannover Medical School, Hannover, Germany
| |
Collapse
|