1
|
Ogunbawo AR, Hidalgo J, Mulim HA, Carrara ER, Ventura HT, Souza NO, Lourenco D, Oliveira HR. Applying the algorithm for Proven and young in GWAS Reveals high polygenicity for key traits in Nellore cattle. Front Genet 2025; 16:1549284. [PMID: 40370699 PMCID: PMC12075139 DOI: 10.3389/fgene.2025.1549284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 04/07/2025] [Indexed: 05/16/2025] Open
Abstract
Background Identifying genomic regions associated with traits of interest and their biological processes provides valuable insights into the phenotypic variability of these traits. This study aimed to identify candidate genes and genomic regions associated with 16 traits currently evaluated by the Brazilian Association of Zebu Breeders (ABCZ). These traits include reproductive traits such as age at first calving (AFC), stayability (STAY), and scrotal circumference at 365 (SC365) and 450 days (SC450). Growth traits include birthweight (BW), expected progeny difference for weight at 120days of age (EPD120), as well as weight at 120 (W120), 210 (W210), 365 (W365), and 450 days of age (W450). Carcass traits include body conformation (BC), finishing score (FS), marbling (MARB), muscularity (MUSC), finishing precocity (FP), and ribeye area (REA). Methods A dataset containing 304,782 Nellore cattle genotyped with 437,650 SNPs (after quality control) was used for this study. The Algorithm for Proven and Young (APY), implemented in the PREGSF90 software, was used to compute theG A P Y - 1 matrix using 36,000 core animals (which explained 98% of the variance in the genomic matrix). Subsequently, the SNP solutions were estimated by back-solving the Genomic Estimated Breeding Values (GEBVs) predicted by ABCZ using the single-step GBLUP method. Genomic regions were identified using sliding windows of 175 consecutive SNPs, and the top 1% genomic windows, ranked based on their proportion of the additive genetic variance, were used to annotate positional candidate genes and genomic regions associated with each of the 16 traits. Results The top 1% windows for all traits explained between 2.779% (STAY) to 3.158% (FP) of the additive genetic variance, highlighting the polygenic nature of these traits. Functional analysis of the candidate genes and genomic regions provided valuable insights into the genetic architecture underlying these traits in Nellore cattle. For instance, our results revealed genes with important functions for each trait, such as SERPINA14 (plays a key role for the endometrial epithelium) identified for AFC, HSPG2 (associated with morphological development and tissue differentiation) identified for BW, among others. Conclusion We identified genomic regions and candidate genes, some of which have been previously reported in the literature, while others are novel discoveries that warrant further investigation. These findings contribute to gene prioritization efforts, facilitating the identification of functional candidate genes that can enhance genomic selection strategies for economically important traits in Nellore cattle.
Collapse
Affiliation(s)
- Adebisi R. Ogunbawo
- Department of Animal Sciences, Purdue University, West Lafayette, IN, United States
| | - Jorge Hidalgo
- Department of Animal and Dairy Sciences, University of Georgia, Athens, GA, United States
| | - Henrique A. Mulim
- Department of Animal Sciences, Purdue University, West Lafayette, IN, United States
| | - Eula R. Carrara
- Department of Animal and Dairy Sciences, University of Georgia, Athens, GA, United States
| | | | - Nadson O. Souza
- Brazilian Association of Zebu Breeders, Uberaba, Minas Gerais, Brazil
| | - Daniela Lourenco
- Department of Animal and Dairy Sciences, University of Georgia, Athens, GA, United States
| | - Hinayah R. Oliveira
- Department of Animal Sciences, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
2
|
Murray EC, Hodge GM, Lee LS, Mitchell CAR, Lombardo AT. The Rho effector ARHGAP18 coordinates a Hippo pathway feedback loop through YAP and Merlin to regulate the cytoskeleton and epithelial cell polarity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.26.625473. [PMID: 39651219 PMCID: PMC11623603 DOI: 10.1101/2024.11.26.625473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
The organization of the cell's cytoskeletal filaments is coordinated through a complex symphony of signaling cascades originating from internal and external cues. Two major actin regulatory pathways are signal transduction through Rho family GTPases and growth and proliferation signaling through the Hippo pathway. These two pathways act to define the actin cytoskeleton, controlling foundational cellular attributes such as morphology and polarity. In this study, we use human epithelial cells to investigate the interplay between the Hippo and Rho Family signaling pathways, which have predominantly been characterized as independent actin regulatory mechanisms. We identify that the RhoA effector, ARHGAP18, forms a complex with the Hippo pathway transcription factor YAP to address a long-standing enigma in the field. Using super resolution STORM microscopy, we characterize the changes in the actin cytoskeleton, on the single filament level, that arise from CRISPR/Cas9 knockout of ARHGAP18. We report that the loss of ARHGAP18 results in alterations of the cell that derive from both aberrant RhoA signaling and inappropriate nuclear localization of YAP. These findings indicate that the Hippo and Rho family GTPase signaling cascades are coordinated in their temporal and spatial control of the actin cytoskeleton.
Collapse
|
3
|
Chandurkar MK, Mittal N, Royer-Weeden SP, Lehmann SD, Michels EB, Haarman SE, Severance SA, Rho Y, Han SJ. Transient low shear-stress preconditioning influences long-term endothelial traction and alignment under high shear flow. Am J Physiol Heart Circ Physiol 2024; 326:H1180-H1192. [PMID: 38457352 PMCID: PMC11649189 DOI: 10.1152/ajpheart.00067.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/01/2024] [Accepted: 03/03/2024] [Indexed: 03/10/2024]
Abstract
Endothelial cells (ECs) within the vascular system encounter fluid shear stress (FSS). High, laminar FSS promotes vasodilation and anti-inflammatory responses, whereas low or disturbed FSS induces dysfunction and inflammation. However, the adaptation of endothelial cells (ECs) to dynamically changing FSS patterns remains underexplored. Here, by combining traction force microscopy with a custom flow chamber, we examined human umbilical vein endothelial cells adapting their traction during transitions from short-term low shear to long-term high shear stress. We discovered that the initial low FSS elevates the traction by only half of the amount in response to direct high FSS even after flow changes to high FSS. However, in the long term under high FSS, the flow started with low FSS triggers a substantial second rise in traction for over 10 h. In contrast, the flow started directly with high FSS results in a quick traction surge followed by a huge reduction below the baseline traction in <30 min. Importantly, we find that the orientation of traction vectors is steered by initial shear exposure. Using Granger causality analysis, we show that the traction that aligns in the flow direction under direct high FSS functionally causes cell alignment toward the flow direction. However, EC traction that orients perpendicular to the flow that starts with temporary low FSS functionally causes cell orientation perpendicular to the flow. Taken together, our findings elucidate the significant influence of initial short-term low FSS on lasting changes in endothelial traction that induces EC alignment.NEW & NOTEWORTHY In our study, we uncover that preconditioning with low shear stress yields enduring impacts on endothelial cell traction and orientation, persisting even after transitioning to high-shear conditions. Using Granger causality analysis, we demonstrate a functional link between the direction of cell traction and subsequent cellular alignment across varying shear environments.
Collapse
Affiliation(s)
- Mohanish K Chandurkar
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, United States
- Health Research Institute, Michigan Technological University, Houghton, Michigan, United States
| | - Nikhil Mittal
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, United States
- Health Research Institute, Michigan Technological University, Houghton, Michigan, United States
| | - Shaina P Royer-Weeden
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, United States
- Health Research Institute, Michigan Technological University, Houghton, Michigan, United States
| | - Steven D Lehmann
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, United States
| | - Etienne B Michels
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, United States
| | - Samuel E Haarman
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, United States
- Health Research Institute, Michigan Technological University, Houghton, Michigan, United States
| | - Scott A Severance
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, United States
| | - Yeonwoo Rho
- Department of Mathematical Sciences, Michigan Technological University, Houghton, Michigan, United States
| | - Sangyoon J Han
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, United States
- Health Research Institute, Michigan Technological University, Houghton, Michigan, United States
- Department of Mechanical Engineering and Engineering Mechanics, Michigan Technological University, Houghton, Michigan, United States
| |
Collapse
|
4
|
Chandurkar MK, Mittal N, Royer-Weeden SP, Lehmann SD, Rho Y, Han SJ. Low Shear in Short-Term Impacts Endothelial Cell Traction and Alignment in Long-Term. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.20.558732. [PMID: 37790318 PMCID: PMC10542130 DOI: 10.1101/2023.09.20.558732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Within the vascular system, endothelial cells (ECs) are exposed to fluid shear stress (FSS), a mechanical force exerted by blood flow that is critical for regulating cellular tension and maintaining vascular homeostasis. The way ECs react to FSS varies significantly; while high, laminar FSS supports vasodilation and suppresses inflammation, low or disturbed FSS can lead to endothelial dysfunction and increase the risk of cardiovascular diseases. Yet, the adaptation of ECs to dynamically varying FSS remains poorly understood. This study focuses on the dynamic responses of ECs to brief periods of low FSS, examining its impact on endothelial traction-a measure of cellular tension that plays a crucial role in how endothelial cells respond to mechanical stimuli. By integrating traction force microscopy (TFM) with a custom-built flow chamber, we analyzed how human umbilical vein endothelial cells (HUVECs) adjust their traction in response to shifts from low to high shear stress. We discovered that initial exposure to low FSS prompts a marked increase in traction force, which continues to rise over 10 hours before slowly decreasing. In contrast, immediate exposure to high FSS causes a quick spike in traction followed by a swift reduction, revealing distinct patterns of traction behavior under different shear conditions. Importantly, the direction of traction forces and the resulting cellular alignment under these conditions indicate that the initial shear experience dictates long-term endothelial behavior. Our findings shed light on the critical influence of short-lived low-shear stress experiences in shaping endothelial function, indicating that early exposure to low FSS results in enduring changes in endothelial contractility and alignment, with significant consequences for vascular health and the development of cardiovascular diseases.
Collapse
Affiliation(s)
- Mohanish K. Chandurkar
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931
- Health Research Institute, Michigan Technological University, Houghton, MI 49931
| | - Nikhil Mittal
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931
- Health Research Institute, Michigan Technological University, Houghton, MI 49931
| | - Shaina P. Royer-Weeden
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931
- Health Research Institute, Michigan Technological University, Houghton, MI 49931
| | - Steven D. Lehmann
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931
| | - Yeonwoo Rho
- Department of Mathematical Sciences, Michigan Technological University, Houghton, MI 49931
| | - Sangyoon J. Han
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931
- Health Research Institute, Michigan Technological University, Houghton, MI 49931
- Department of Mechanical Engineering and Engineering Mechanics, Michigan Technological University, Houghton, MI 49931
| |
Collapse
|
5
|
Choi JS, Doo HM, Kim B, Lee SH, Sung S, Go G, Suarez A, Kim Y, Weon BM, Choi B, Kim HJ, Kim D. NanoIEA: A Nanopatterned Interdigitated Electrode Array-Based Impedance Assay for Real-Time Measurement of Aligned Endothelial Cell Barrier Functions. Adv Healthc Mater 2024; 13:e2301124. [PMID: 37820720 PMCID: PMC10841753 DOI: 10.1002/adhm.202301124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 09/18/2023] [Indexed: 10/13/2023]
Abstract
A nanopatterned interdigitated electrode array (nanoIEA)-based impedance assay is developed for quantitative real-time measurement of aligned endothelial cell (EC) barrier functions in vitro. A bioinspired poly(3,4-dihydroxy-L-phenylalanine) (poly (l-DOPA)) coating is applied to improve the human brain EC adhesion onto the Nafion nanopatterned surfaces. It is found that a poly (l-DOPA)-coated Nafion grooved nanopattern makes the human brain ECs orient along the nanopattern direction. Aligned human brain ECs on Nafion nanopatterns exhibit increased expression of genes encoding tight and adherens junction proteins. Aligned human brain ECs also have enhanced impedance and resistance versus unaligned ones. Treatment with a glycogen synthase kinase-3 inhibitor (GSK3i) further increases impedance and resistance, suggesting synergistic effects occur on the cell-cell tightness of in vitro human brain ECs via a combination of anisotropic matrix nanotopography and GSK3i treatment. It is found that this enhanced cell-cell tightness of the combined approach is accompanied by increased expression of claudin protein. These data demonstrate that the proposed nanoIEA assay integrated with poly (l-DOPA)-coated Nafion nanopatterns and interdigitated electrode arrays can make not only biomimetic aligned ECs, but also enable real-time measurement of the enhanced barrier functions of aligned ECs via tighter cell-cell junctions.
Collapse
Affiliation(s)
- Jong Seob Choi
- Department of Biomedical Engineering, Center for Microphysiological SystemsJohns Hopkins UniversityBaltimoreMD21205USA
- Division of Advanced Materials EngineeringKongju National UniversityCheonanChungnam31080South Korea
| | - Hyun Myung Doo
- Department of Health Sciences and TechnologySAIHSTSungkyunkwan UniversitySeoul06351South Korea
- Department of Biomedical Research CenterKorea University Guro HospitalSeoul08308South Korea
- Division of Medical Oncology, Department of Internal MedicineKorea University Guro Hospital, Korea University College of MedicineSeoul08308South Korea
| | - Byunggik Kim
- Department of Mechanical EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
| | - Su Han Lee
- Digital Health Care Research CenterGumi Electronics and Information Technology Research Institute (GERI)GumiGyeongbuk39253South Korea
| | - Sang‐keun Sung
- Digital Health Care Research CenterGumi Electronics and Information Technology Research Institute (GERI)GumiGyeongbuk39253South Korea
| | - Gwangjun Go
- Department of Biomedical Engineering, Center for Microphysiological SystemsJohns Hopkins UniversityBaltimoreMD21205USA
- Department of Mechanical EngineeringChosun UniversityGwangju61452South Korea
| | - Allister Suarez
- Department of Biomedical Engineering, Center for Microphysiological SystemsJohns Hopkins UniversityBaltimoreMD21205USA
| | - Yeseul Kim
- SKKU Advanced Institute of Nanotechnology (SAINT)School of Advanced Materials Science and EngineeringSungkyunkwan UniversitySuwon16419South Korea
| | - Byung Mook Weon
- SKKU Advanced Institute of Nanotechnology (SAINT)School of Advanced Materials Science and EngineeringSungkyunkwan UniversitySuwon16419South Korea
| | - Byung‐Ok Choi
- Department of Health Sciences and TechnologySAIHSTSungkyunkwan UniversitySeoul06351South Korea
- Department of NeurologySamsung Medical CenterSungkyunkwan University School of MedicineSeoul06351South Korea
| | - Hyung Jin Kim
- School of Electrical and Electronic EngineeringUlsan CollegeUlsan44610South Korea
| | - Deok‐Ho Kim
- Department of Biomedical Engineering, Center for Microphysiological SystemsJohns Hopkins UniversityBaltimoreMD21205USA
- Department of Mechanical EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMD21205USA
- Institute for NanobiotechnologyJohns Hopkins UniversityBaltimoreMD21218USA
| |
Collapse
|
6
|
A current overview of RhoA, RhoB, and RhoC functions in vascular biology and pathology. Biochem Pharmacol 2022; 206:115321. [DOI: 10.1016/j.bcp.2022.115321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/17/2022] [Accepted: 10/18/2022] [Indexed: 11/24/2022]
|
7
|
Salvador J, Iruela-Arispe ML. Nuclear Mechanosensation and Mechanotransduction in Vascular Cells. Front Cell Dev Biol 2022; 10:905927. [PMID: 35784481 PMCID: PMC9247619 DOI: 10.3389/fcell.2022.905927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/31/2022] [Indexed: 11/24/2022] Open
Abstract
Vascular cells are constantly subjected to physical forces associated with the rhythmic activities of the heart, which combined with the individual geometry of vessels further imposes oscillatory, turbulent, or laminar shear stresses on vascular cells. These hemodynamic forces play an important role in regulating the transcriptional program and phenotype of endothelial and smooth muscle cells in different regions of the vascular tree. Within the aorta, the lesser curvature of the arch is characterized by disturbed, oscillatory flow. There, endothelial cells become activated, adopting pro-inflammatory and athero-prone phenotypes. This contrasts the descending aorta where flow is laminar and endothelial cells maintain a quiescent and atheroprotective phenotype. While still unclear, the specific mechanisms involved in mechanosensing flow patterns and their molecular mechanotransduction directly impact the nucleus with consequences to transcriptional and epigenetic states. The linker of nucleoskeleton and cytoskeleton (LINC) protein complex transmits both internal and external forces, including shear stress, through the cytoskeleton to the nucleus. These forces can ultimately lead to changes in nuclear integrity, chromatin organization, and gene expression that significantly impact emergence of pathology such as the high incidence of atherosclerosis in progeria. Therefore, there is strong motivation to understand how endothelial nuclei can sense and respond to physical signals and how abnormal responses to mechanical cues can lead to disease. Here, we review the evidence for a critical role of the nucleus as a mechanosensor and the importance of maintaining nuclear integrity in response to continuous biophysical forces, specifically shear stress, for proper vascular function and stability.
Collapse
Affiliation(s)
| | - M. Luisa Iruela-Arispe
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
8
|
Polk T, Schmitt S, Aldrich JL, Long DS. Human dermal microvascular endothelial cell morphological response to fluid shear stress. Microvasc Res 2022; 143:104377. [PMID: 35561754 DOI: 10.1016/j.mvr.2022.104377] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 04/15/2022] [Accepted: 05/05/2022] [Indexed: 10/18/2022]
Abstract
As the cells that line the vasculature, endothelial cells are continually exposed to fluid shear stress by blood flow. Recent studies suggest that the morphological response of endothelial cells to fluid shear stress depends on the endothelial cell type. Thus, the present study characterizes the morphological response of human dermal microvascular endothelial cells (HMEC-1) and nuclei to steady, laminar, and unidirectional fluid shear stress. Cultured HMEC-1 monolayers were exposed to shear stress of 0.3 dyn/cm2, 16 dyn/cm2, or 32 dyn/cm2 for 72 h with hourly live-cell imaging capturing both the nuclear and cellular morphology. Despite changes in elongation and alignment occurring with increasing fluid shear stress, there was a lack of elongation and alignment over time under each fluid shear stress condition. Conversely, changes in cellular and nuclear area exhibited dependence on both time and fluid shear stress magnitude. The trends in cellular morphology differed at shear stress levels above and below 16 dyn/cm2, whereas the nuclear orientation was independent of fluid shear stress magnitude. These findings show the complex morphological response of HMEC-1 to fluid shear stress.
Collapse
Affiliation(s)
- Tabatha Polk
- Mechanobiology and Biomedicine Lab, Department of Biomedical Engineering, Wichita State University, Wichita, KS, USA
| | - Sarah Schmitt
- Mechanobiology and Biomedicine Lab, Department of Biomedical Engineering, Wichita State University, Wichita, KS, USA
| | - Jessica L Aldrich
- Mechanobiology and Biomedicine Lab, Department of Biomedical Engineering, Wichita State University, Wichita, KS, USA
| | - David S Long
- Mechanobiology and Biomedicine Lab, Department of Biomedical Engineering, Wichita State University, Wichita, KS, USA.
| |
Collapse
|
9
|
Williams D, Mahmoud M, Liu R, Andueza A, Kumar S, Kang DW, Zhang J, Tamargo I, Villa-Roel N, Baek KI, Lee H, An Y, Zhang L, Tate EW, Bagchi P, Pohl J, Mosnier LO, Diamandis EP, Mihara K, Hollenberg MD, Dai Z, Jo H. Stable flow-induced expression of KLK10 inhibits endothelial inflammation and atherosclerosis. eLife 2022; 11:e72579. [PMID: 35014606 PMCID: PMC8806187 DOI: 10.7554/elife.72579] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 01/08/2022] [Indexed: 12/15/2022] Open
Abstract
Atherosclerosis preferentially occurs in arterial regions exposed to disturbed blood flow (d-flow), while regions exposed to stable flow (s-flow) are protected. The proatherogenic and atheroprotective effects of d-flow and s-flow are mediated in part by the global changes in endothelial cell (EC) gene expression, which regulates endothelial dysfunction, inflammation, and atherosclerosis. Previously, we identified kallikrein-related peptidase 10 (Klk10, a secreted serine protease) as a flow-sensitive gene in mouse arterial ECs, but its role in endothelial biology and atherosclerosis was unknown. Here, we show that KLK10 is upregulated under s-flow conditions and downregulated under d-flow conditions using in vivo mouse models and in vitro studies with cultured ECs. Single-cell RNA sequencing (scRNAseq) and scATAC sequencing (scATACseq) study using the partial carotid ligation mouse model showed flow-regulated Klk10 expression at the epigenomic and transcription levels. Functionally, KLK10 protected against d-flow-induced permeability dysfunction and inflammation in human artery ECs, as determined by NFκB activation, expression of vascular cell adhesion molecule 1 and intracellular adhesion molecule 1, and monocyte adhesion. Furthermore, treatment of mice in vivo with rKLK10 decreased arterial endothelial inflammation in d-flow regions. Additionally, rKLK10 injection or ultrasound-mediated transfection of Klk10-expressing plasmids inhibited atherosclerosis in Apoe-/- mice. Moreover, KLK10 expression was significantly reduced in human coronary arteries with advanced atherosclerotic plaques compared to those with less severe plaques. KLK10 is a flow-sensitive endothelial protein that serves as an anti-inflammatory, barrier-protective, and anti-atherogenic factor.
Collapse
Affiliation(s)
- Darian Williams
- Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of TechnologyAtlantaUnited States
- Molecular and Systems Pharmacology Program, Emory UniversityAtlantaUnited States
| | - Marwa Mahmoud
- Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of TechnologyAtlantaUnited States
| | - Renfa Liu
- Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of TechnologyAtlantaUnited States
- Department of Biomedical Engineering, Peking UniversityBeijingChina
| | - Aitor Andueza
- Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of TechnologyAtlantaUnited States
| | - Sandeep Kumar
- Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of TechnologyAtlantaUnited States
| | - Dong-Won Kang
- Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of TechnologyAtlantaUnited States
| | - Jiahui Zhang
- Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of TechnologyAtlantaUnited States
| | - Ian Tamargo
- Molecular and Systems Pharmacology Program, Emory UniversityAtlantaUnited States
| | - Nicolas Villa-Roel
- Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of TechnologyAtlantaUnited States
| | - Kyung-In Baek
- Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of TechnologyAtlantaUnited States
| | | | | | - Leran Zhang
- Department of Chemistry, Imperial College LondonLondonUnited Kingdom
| | - Edward W Tate
- Department of Chemistry, Imperial College LondonLondonUnited Kingdom
| | - Pritha Bagchi
- Emory Integrated Proteomics Core, Emory UniversityAtlantaUnited States
| | - Jan Pohl
- Biotechnology Core Facility Branch, Centers for Disease Control and PreventionAtlantaUnited States
| | - Laurent O Mosnier
- Department of Molecular Medicine, Scripps Research InstituteSan DiegoUnited States
| | | | - Koichiro Mihara
- Department of Physiology and Pharmacology, University of CalgaryCalgaryCanada
| | - Morley D Hollenberg
- Department of Physiology and Pharmacology, University of CalgaryCalgaryCanada
| | - Zhifei Dai
- Department of Biomedical Engineering, Peking UniversityBeijingChina
| | - Hanjoong Jo
- Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of TechnologyAtlantaUnited States
- Molecular and Systems Pharmacology Program, Emory UniversityAtlantaUnited States
- Department of Medicine, Emory UniversityAtlantaUnited States
| |
Collapse
|
10
|
Xu Z, Hu Z, Xu H, Zhang L, Li L, Wang Y, Zhu Y, Yang L, Hu D. Liquiritigenin alleviates doxorubicin-induced chronic heart failure via promoting ARHGAP18 and suppressing RhoA/ROCK1 pathway. Exp Cell Res 2022; 411:113008. [PMID: 34990617 DOI: 10.1016/j.yexcr.2022.113008] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 12/30/2021] [Accepted: 01/01/2022] [Indexed: 12/01/2022]
Abstract
Chronic heart failure (CHF) is one of the most common chronic diseases with increasing incidence and mortality. Liquiritigenin (LQG) is shown to protect mice from cardiotoxicity. However, its underlying mechanism remains unclear. Our study aimed to reveal the role of ARHGAP18 in LQG-mediated cardioprotective effects in CHF. In the current study, CHF cell model and rat model were established by the application of doxorubicin (DOX). The reactive oxygen species (ROS) level and cell apoptosis were determined by flow cytometry. The cardiac function of rats was evaluated by measuring left ventricular systolic pressure, left ventricular end diastolic pressure, and serum level of lactate dehydrogenase and brain natriuretic peptide. The expression of active RhoA was elevated and that of ARHGAP18 was decreased in DOX-induced CHF cell model. ARHGAP18 could reduce DOX-induced RhoA activation, ROS elevation, and cell apoptosis. Meanwhile, the knockdown of ARHGAP18 could promote the activation of RhoA, the level of ROS, and the rate of cell apoptosis, which could be reversed by the application of RhoA inhibitor. LQG promoted the expression of ARHGAP18 and exerted similar effects of ARHGAP18 in CHF cell model. The application of LQG could also reverse the effects mediated by ARHGAP18 knockdown. Moreover, LQG significantly improved cardiac function and ameliorated DOX-induced cardiotoxicity of CHF rats. In conclusion, LQG could alleviate DOX-induced CHF via promoting ARHGAP18 and suppressing RhoA/ROCK1 pathway. LQG was a potential agent for CHF treatment.
Collapse
Affiliation(s)
- Zhibing Xu
- Department of Emergency, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, China
| | - Zongde Hu
- Department of Traditional Chinese Medicine, Shanghai Pudong New Area Hospital of Traditional Chinese Medicine, China
| | - Hanchen Xu
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, China
| | - Lifen Zhang
- Department of Emergency, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, China
| | - Liang Li
- Department of Emergency, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, China
| | - Yi Wang
- Department of Emergency, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, China
| | - Yuanqing Zhu
- Department of Emergency, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, China
| | - Limeng Yang
- Department of Traditional Chinese Medicine, Shanghai Pudong New Area Hospital of Traditional Chinese Medicine, China.
| | - Dan Hu
- Department of Traditional Chinese Medicine, Shanghai Pudong New Area Hospital of Traditional Chinese Medicine, China.
| |
Collapse
|
11
|
Sunderland K, Jiang J, Zhao F. Disturbed flow's impact on cellular changes indicative of vascular aneurysm initiation, expansion, and rupture: A pathological and methodological review. J Cell Physiol 2022; 237:278-300. [PMID: 34486114 PMCID: PMC8810685 DOI: 10.1002/jcp.30569] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 08/06/2021] [Accepted: 08/16/2021] [Indexed: 01/03/2023]
Abstract
Aneurysms are malformations within the arterial vasculature brought on by the structural breakdown of the microarchitecture of the vessel wall, with aneurysms posing serious health risks in the event of their rupture. Blood flow within vessels is generally laminar with high, unidirectional wall shear stressors that modulate vascular endothelial cell functionality and regulate vascular smooth muscle cells. However, altered vascular geometry induced by bifurcations, significant curvature, stenosis, or clinical interventions can alter the flow, generating low stressor disturbed flow patterns. Disturbed flow is associated with altered cellular morphology, upregulated expression of proteins modulating inflammation, decreased regulation of vascular permeability, degraded extracellular matrix, and heightened cellular apoptosis. The understanding of the effects disturbed flow has on the cellular cascades which initiate aneurysms and promote their subsequent growth can further elucidate the nature of this complex pathology. This review summarizes the current knowledge about the disturbed flow and its relation to aneurysm pathology, the methods used to investigate these relations, as well as how such knowledge has impacted clinical treatment methodologies. This information can contribute to the understanding of the development, growth, and rupture of aneurysms and help develop novel research and aneurysmal treatment techniques.
Collapse
Affiliation(s)
- Kevin Sunderland
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931
| | - Jingfeng Jiang
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931,Corresponding Authors: Feng Zhao, 101 Bizzell Street, College Station, TX 77843-312, Tel : 979-458-1239, , Jingfeng Jiang, 1400 Townsend Dr., Houghton, MI 49931, Tel: 906-487-1943
| | - Feng Zhao
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843,Corresponding Authors: Feng Zhao, 101 Bizzell Street, College Station, TX 77843-312, Tel : 979-458-1239, , Jingfeng Jiang, 1400 Townsend Dr., Houghton, MI 49931, Tel: 906-487-1943
| |
Collapse
|
12
|
Botts SR, Fish JE, Howe KL. Dysfunctional Vascular Endothelium as a Driver of Atherosclerosis: Emerging Insights Into Pathogenesis and Treatment. Front Pharmacol 2021; 12:787541. [PMID: 35002720 PMCID: PMC8727904 DOI: 10.3389/fphar.2021.787541] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/06/2021] [Indexed: 12/28/2022] Open
Abstract
Atherosclerosis, the chronic accumulation of cholesterol-rich plaque within arteries, is associated with a broad spectrum of cardiovascular diseases including myocardial infarction, aortic aneurysm, peripheral vascular disease, and stroke. Atherosclerotic cardiovascular disease remains a leading cause of mortality in high-income countries and recent years have witnessed a notable increase in prevalence within low- and middle-income regions of the world. Considering this prominent and evolving global burden, there is a need to identify the cellular mechanisms that underlie the pathogenesis of atherosclerosis to discover novel therapeutic targets for preventing or mitigating its clinical sequelae. Despite decades of research, we still do not fully understand the complex cell-cell interactions that drive atherosclerosis, but new investigative approaches are rapidly shedding light on these essential mechanisms. The vascular endothelium resides at the interface of systemic circulation and the underlying vessel wall and plays an essential role in governing pathophysiological processes during atherogenesis. In this review, we present emerging evidence that implicates the activated endothelium as a driver of atherosclerosis by directing site-specificity of plaque formation and by promoting plaque development through intracellular processes, which regulate endothelial cell proliferation and turnover, metabolism, permeability, and plasticity. Moreover, we highlight novel mechanisms of intercellular communication by which endothelial cells modulate the activity of key vascular cell populations involved in atherogenesis, and discuss how endothelial cells contribute to resolution biology - a process that is dysregulated in advanced plaques. Finally, we describe important future directions for preclinical atherosclerosis research, including epigenetic and targeted therapies, to limit the progression of atherosclerosis in at-risk or affected patients.
Collapse
Affiliation(s)
- Steven R. Botts
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Jason E. Fish
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
| | - Kathryn L. Howe
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
- Division of Vascular Surgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
13
|
Dawson A, Wang Y, Li Y, LeMaire SA, Shen YH. New Technologies With Increased Precision Improve Understanding of Endothelial Cell Heterogeneity in Cardiovascular Health and Disease. Front Cell Dev Biol 2021; 9:679995. [PMID: 34513826 PMCID: PMC8430032 DOI: 10.3389/fcell.2021.679995] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 06/17/2021] [Indexed: 01/08/2023] Open
Abstract
Endothelial cells (ECs) are vital for blood vessel integrity and have roles in maintaining normal vascular function, healing after injury, and vascular dysfunction. Extensive phenotypic heterogeneity has been observed among ECs of different types of blood vessels in the normal and diseased vascular wall. Although ECs with different phenotypes can share common functions, each has unique features that may dictate a fine-tuned role in vascular health and disease. Recent studies performed with single-cell technology have generated powerful information that has significantly improved our understanding of EC biology. Here, we summarize a variety of EC types, states, and phenotypes recently identified by using new, increasingly precise techniques in transcriptome analysis.
Collapse
Affiliation(s)
- Ashley Dawson
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Yidan Wang
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Yanming Li
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Scott A. LeMaire
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
- Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX, United States
| | - Ying H. Shen
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
- Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX, United States
| |
Collapse
|
14
|
Li H, Luo Q, Shan W, Cai S, Tie R, Xu Y, Lin Y, Qian P, Huang H. Biomechanical cues as master regulators of hematopoietic stem cell fate. Cell Mol Life Sci 2021; 78:5881-5902. [PMID: 34232331 PMCID: PMC8316214 DOI: 10.1007/s00018-021-03882-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 06/02/2021] [Accepted: 06/15/2021] [Indexed: 01/09/2023]
Abstract
Hematopoietic stem cells (HSCs) perceive both soluble signals and biomechanical inputs from their microenvironment and cells themselves. Emerging as critical regulators of the blood program, biomechanical cues such as extracellular matrix stiffness, fluid mechanical stress, confined adhesiveness, and cell-intrinsic forces modulate multiple capacities of HSCs through mechanotransduction. In recent years, research has furthered the scientific community's perception of mechano-based signaling networks in the regulation of several cellular processes. However, the underlying molecular details of the biomechanical regulatory paradigm in HSCs remain poorly elucidated and researchers are still lacking in the ability to produce bona fide HSCs ex vivo for clinical use. This review presents an overview of the mechanical control of both embryonic and adult HSCs, discusses some recent insights into the mechanisms of mechanosensing and mechanotransduction, and highlights the application of mechanical cues aiming at HSC expansion or differentiation.
Collapse
Affiliation(s)
- Honghu Li
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China
- Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310012, Zhejiang, People's Republic of China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, 310012, Zhejiang, People's Republic of China
| | - Qian Luo
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China
- Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310012, Zhejiang, People's Republic of China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, 310012, Zhejiang, People's Republic of China
| | - Wei Shan
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China
- Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310012, Zhejiang, People's Republic of China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, 310012, Zhejiang, People's Republic of China
| | - Shuyang Cai
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China
- Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310012, Zhejiang, People's Republic of China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, 310012, Zhejiang, People's Republic of China
| | - Ruxiu Tie
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China
- Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310012, Zhejiang, People's Republic of China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, 310012, Zhejiang, People's Republic of China
| | - Yulin Xu
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China
- Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310012, Zhejiang, People's Republic of China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, 310012, Zhejiang, People's Republic of China
| | - Yu Lin
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China
- Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310012, Zhejiang, People's Republic of China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, 310012, Zhejiang, People's Republic of China
| | - Pengxu Qian
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China.
- Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310012, Zhejiang, People's Republic of China.
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, 310012, Zhejiang, People's Republic of China.
- Center of Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, 310012, China.
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China.
| | - He Huang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China.
- Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310012, Zhejiang, People's Republic of China.
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, 310012, Zhejiang, People's Republic of China.
- Center of Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, 310012, China.
| |
Collapse
|
15
|
Chen M, Chen S, Yang D, Zhou J, Liu B, Chen Y, Ye W, Zhang H, Ji L, Zheng Y. Weighted Gene Co-expression Network Analysis Identifies Crucial Genes Mediating Progression of Carotid Plaque. Front Physiol 2021; 12:601952. [PMID: 33613306 PMCID: PMC7894049 DOI: 10.3389/fphys.2021.601952] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 01/12/2021] [Indexed: 12/28/2022] Open
Abstract
Background Surface rupture of carotid plaque can cause severe cerebrovascular disease, including transient ischemic attack and stroke. The aim of this study was to elucidate the molecular mechanism governing carotid plaque progression and to provide candidate treatment targets for carotid atherosclerosis. Methods The microarray dataset GSE28829 and the RNA-seq dataset GSE104140, which contain advanced plaque and early plaque samples, were utilized in our analysis. Differentially expressed genes (DEGs) were screened using the “limma” R package. Gene modules for both early and advanced plaques were identified based on co-expression networks constructed by weighted gene co-expression network analysis (WGCNA). Gene Ontology (GO) and Kyoto Encyclopedia of Genes Genomes (KEGG) analyses were employed in each module. In addition, hub genes for each module were identified. Crucial genes were identified by molecular complex detection (MCODE) based on the DEG co-expression network and were validated by the GSE43292 dataset. Gene set enrichment analysis (GSEA) for crucial genes was performed. Sensitivity analysis was performed to evaluate the robustness of the networks that we constructed. Results A total of 436 DEGs were screened, of which 335 were up-regulated and 81 were down-regulated. The pathways related to inflammation and immune response were determined to be concentrated in the black module of the advanced plaques. The hub gene of the black module was ARHGAP18 (Rho GTPase activating protein 18). NCF2 (neutrophil cytosolic factor 2), IQGAP2 (IQ motif containing GTPase activating protein 2) and CD86 (CD86 molecule) had the highest connectivity among the crucial genes. All crucial genes were validated successfully, and sensitivity analysis demonstrated that our results were reliable. Conclusion To the best of our knowledge, this study is the first to combine DEGs and WGCNA to establish a DEG co-expression network in carotid plaques, and it proposes potential therapeutic targets for carotid atherosclerosis.
Collapse
Affiliation(s)
- Mengyin Chen
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Siliang Chen
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dan Yang
- Department of Computational Biology and Bioinformatics, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiawei Zhou
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bao Liu
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuexin Chen
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Ye
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hui Zhang
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei Ji
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuehong Zheng
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
16
|
Dibus M, Brábek J, Rösel D. A Screen for PKN3 Substrates Reveals an Activating Phosphorylation of ARHGAP18. Int J Mol Sci 2020; 21:ijms21207769. [PMID: 33092266 PMCID: PMC7594087 DOI: 10.3390/ijms21207769] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/16/2020] [Accepted: 10/17/2020] [Indexed: 12/13/2022] Open
Abstract
Protein kinase N3 (PKN3) is a serine/threonine kinase implicated in tumor progression of multiple cancer types, however, its substrates and effector proteins still remain largely understudied. In the present work we aimed to identify novel PKN3 substrates in a phosphoproteomic screen using analog sensitive PKN3. Among the identified putative substrates we selected ARHGAP18, a protein from RhoGAP family, for validation of the screen and further study. We confirmed that PKN3 can phosphorylate ARHGAP18 in vitro and we also characterized the interaction of the two proteins, which is mediated via the N-terminal part of ARHGAP18. We present strong evidence that PKN3-ARHGAP18 interaction is increased upon ARHGAP18 phosphorylation and that the phosphorylation of ARHGAP18 by PKN3 enhances its GAP domain activity and contributes to negative regulation of active RhoA. Taken together, we identified new set of potential PKN3 substrates and revealed a new negative feedback regulatory mechanism of Rho signaling mediated by PKN3-induced ARHGAP18 activation.
Collapse
Affiliation(s)
- Michal Dibus
- Department of Cell Biology, Charles University, Viničná 7, 12800 Prague, Czech Republic; (M.D.); (J.B.)
- Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242 Vestec u Prahy, Czech Republic
| | - Jan Brábek
- Department of Cell Biology, Charles University, Viničná 7, 12800 Prague, Czech Republic; (M.D.); (J.B.)
- Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242 Vestec u Prahy, Czech Republic
| | - Daniel Rösel
- Department of Cell Biology, Charles University, Viničná 7, 12800 Prague, Czech Republic; (M.D.); (J.B.)
- Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242 Vestec u Prahy, Czech Republic
- Correspondence:
| |
Collapse
|
17
|
Witjes JJ, Smits LP, Pekmez CT, Prodan A, Meijnikman AS, Troelstra MA, Bouter KEC, Herrema H, Levin E, Holleboom AG, Winkelmeijer M, Beuers UH, van Lienden K, Aron-Wisnewky J, Mannisto V, Bergman JJ, Runge JH, Nederveen AJ, Dragsted LO, Konstanti P, Zoetendal EG, de Vos W, Verheij J, Groen AK, Nieuwdorp M. Donor Fecal Microbiota Transplantation Alters Gut Microbiota and Metabolites in Obese Individuals With Steatohepatitis. Hepatol Commun 2020; 4:1578-1590. [PMID: 33163830 PMCID: PMC7603524 DOI: 10.1002/hep4.1601] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/27/2020] [Accepted: 07/31/2020] [Indexed: 12/11/2022] Open
Abstract
The intestinal microbiota has been linked to the development and prevalence of steatohepatitis in humans. Interestingly, steatohepatitis is significantly lower in individuals taking a plant-based, low-animal-protein diet, which is thought to be mediated by gut microbiota. However, data on causality between these observations in humans is scarce. In this regard, fecal microbiota transplantation (FMT) using healthy donors is safe and is capable of changing microbial composition in human disease. We therefore performed a double-blind randomized controlled proof-of-principle study in which individuals with hepatic steatosis on ultrasound were randomized to two study arms: lean vegan donor (allogenic n = 10) or own (autologous n = 11) FMT. Both were performed three times at 8-week intervals. A liver biopsy was performed at baseline and after 24 weeks in every subject to determine histopathology (Nonalcoholic Steatohepatitis Clinical Research Network) classification and changes in hepatic gene expression based on RNA sequencing. Secondary outcome parameters were changes in intestinal microbiota composition and fasting plasma metabolomics. We observed a trend toward improved necro-inflammatory histology, and found significant changes in expression of hepatic genes involved in inflammation and lipid metabolism following allogenic FMT. Intestinal microbial community structure changed following allogenic FMT, which was associated with changes in plasma metabolites as well as markers of . Conclusion: Allogenic FMT using lean vegan donors in individuals with hepatic steatosis shows an effect on intestinal microbiota composition, which is associated with beneficial changes in plasma metabolites and markers of steatohepatitis.
Collapse
Affiliation(s)
- Julia J Witjes
- Department of Internal and Vascular Medicine Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Loek P Smits
- Department of Internal and Vascular Medicine Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Ceyda T Pekmez
- Department of Nutrition, Exercise and Sports University of Copenhagen Copenhagen Denmark
| | - Andrei Prodan
- Department of Internal and Vascular Medicine Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Abraham S Meijnikman
- Department of Internal and Vascular Medicine Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Marian A Troelstra
- Department of Radiology & Nuclear Medicine Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Kristien E C Bouter
- Department of Internal and Vascular Medicine Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Hilde Herrema
- Department of Internal and Vascular Medicine Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Evgeni Levin
- Department of Internal and Vascular Medicine Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Adriaan G Holleboom
- Department of Internal and Vascular Medicine Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Maaike Winkelmeijer
- Department of Internal and Vascular Medicine Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Ulrich H Beuers
- Department of Gastroenterology and Hepatology Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Krijn van Lienden
- Department of Radiology & Nuclear Medicine Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Judith Aron-Wisnewky
- Department of Internal and Vascular Medicine Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Ville Mannisto
- Department of Internal and Vascular Medicine Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Jacques J Bergman
- Department of Gastroenterology and Hepatology Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Jurgen H Runge
- Department of Radiology & Nuclear Medicine Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Aart J Nederveen
- Department of Radiology & Nuclear Medicine Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Lars O Dragsted
- Department of Nutrition, Exercise and Sports University of Copenhagen Copenhagen Denmark
| | - Prokopis Konstanti
- Laboratory of Microbiology Wageningen University Wageningen the Netherlands
| | - Erwin G Zoetendal
- Laboratory of Microbiology Wageningen University Wageningen the Netherlands
| | - Willem de Vos
- Laboratory of Microbiology Wageningen University Wageningen the Netherlands.,Faculty of Medicine Human Microbiome Research Program University of Helsinki Finland
| | - Joanne Verheij
- Department of Pathology Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Albert K Groen
- Department of Internal and Vascular Medicine Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands.,Department of Laboratory Medicine University of Groningen University Medical Center Groningen the Netherlands
| | - Max Nieuwdorp
- Department of Internal and Vascular Medicine Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| |
Collapse
|
18
|
Coleman PR, Lay AJ, Ting KK, Zhao Y, Li J, Jarrah S, Vadas MA, Gamble JR. YAP and the RhoC regulator ARHGAP18, are required to mediate flow-dependent endothelial cell alignment. Cell Commun Signal 2020; 18:18. [PMID: 32013974 PMCID: PMC6998144 DOI: 10.1186/s12964-020-0511-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 01/04/2020] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Vascular endothelial cell alignment in the direction of flow is an adaptive response that protects against aortic diseases such as atherosclerosis. The RhoGTPases are known to regulate this alignment. We have shown previously that ARHGAP18 in endothelial cells is a negative regulator of RhoC and its expression is essential in flow-mediated alignment. Depletion of ARHGAP18 inhibits alignment and results in the induction of a pro-inflammatory phenotype. In embryogenesis, ARHGAP18 was identified as a downstream effector of the Yes-associated protein, YAP, which regulates cell shape and size. METHODS We have used siRNA technology to deplete either ARHGAP18 or YAP in human endothelial cells. The in vitro studies were performed under athero-protective, laminar flow conditions. The analysis of YAP activity was also investigated, using high performance confocal imaging, in our ARHGAP18 knockout mutant mice. RESULTS We show here that loss of ARHGAP18, although decreasing the expression of YAP results in its nuclear localisation consistent with activation. We further show that depletion of YAP itself results in its activation as defined by an in increase in its nuclear localisation and an increase in the YAP target gene, CyR61. Depletion of YAP, similar to that observed for ARHGAP18 depletion, results in loss of endothelial cell alignment under high shear stress mediated flow and also in the activation of NFkB, as determined by p65 nuclear localisation. In contrast, ARHGAP18 overexpression results in upregulation of YAP, its phosphorylation, and a decrease in the YAP target gene Cyr61, consistent with YAP inactivation. Finally, in ARHGAP18 deleted mice, in regions where there is a loss of endothelial cell alignment, a situation associated with a priming of the cells to a pro-inflammatory phenotype, YAP shows nuclear localisation. CONCLUSION Our results show that YAP is downstream of ARHGAP18 in mature endothelial cells and that this pathway is involved in the athero-protective alignment of endothelial cells under laminar shear stress. ARHGAP18 depletion leads to a disruption of the junctions as seen by loss of VE-Cadherin localisation to these regions and a concomitant localisation of YAP to the nucleus.
Collapse
Affiliation(s)
- Paul R Coleman
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, The University of Sydney, Locked Bag 6, Newtown, Sydney, 2042, Australia
| | - Angelina J Lay
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, The University of Sydney, Locked Bag 6, Newtown, Sydney, 2042, Australia
| | - Ka Ka Ting
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, The University of Sydney, Locked Bag 6, Newtown, Sydney, 2042, Australia
| | - Yang Zhao
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, The University of Sydney, Locked Bag 6, Newtown, Sydney, 2042, Australia
| | - Jia Li
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, The University of Sydney, Locked Bag 6, Newtown, Sydney, 2042, Australia
| | - Sorour Jarrah
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, The University of Sydney, Locked Bag 6, Newtown, Sydney, 2042, Australia
| | - Mathew A Vadas
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, The University of Sydney, Locked Bag 6, Newtown, Sydney, 2042, Australia
| | - Jennifer R Gamble
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, The University of Sydney, Locked Bag 6, Newtown, Sydney, 2042, Australia.
| |
Collapse
|
19
|
Lay AJ, Coleman PR, Formaz-Preston A, Ting KK, Roediger B, Weninger W, Schwartz MA, Vadas MA, Gamble JR. ARHGAP18: A Flow-Responsive Gene That Regulates Endothelial Cell Alignment and Protects Against Atherosclerosis. J Am Heart Assoc 2020; 8:e010057. [PMID: 30630384 PMCID: PMC6497359 DOI: 10.1161/jaha.118.010057] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background Vascular endothelial cell (EC) alignment in the direction of flow is an adaptive response that protects against aortic diseases, such as atherosclerosis. The Rho GTPases are known to regulate this alignment. Herein, we analyze the effect of ARHGAP18 on the regulation of EC alignment and examine the effect of ARHGAP18 deficiency on the development of atherosclerosis in mice. Methods and Results We used in vitro analysis of ECs under flow conditions together with apolipoprotein E−/−Arhgap18−/− double‐mutant mice to study the function of ARHGAP18 in a high‐fat diet–induced model of atherosclerosis. Depletion of ARHGAP18 inhibited the alignment of ECs in the direction of flow and promoted inflammatory phenotype, as evidenced by disrupted junctions and increased expression of nuclear factor‐κB and intercellular adhesion molecule‐1 and decreased endothelial nitric oxide synthase. Mice with double deletion in ARHGAP18 and apolipoprotein E and fed a high‐fat diet show early onset of atherosclerosis, with lesions developing in atheroprotective regions. Conclusions ARHGAP18 is a protective gene that maintains EC alignments in the direction of flow. Deletion of ARHGAP18 led to loss of EC ability to align and promoted atherosclerosis development.
Collapse
Affiliation(s)
- Angelina J Lay
- 1 Vascular Biology Program Centre for the Endothelium Centenary Institute The University of Sydney Newtown Australia
| | - Paul R Coleman
- 1 Vascular Biology Program Centre for the Endothelium Centenary Institute The University of Sydney Newtown Australia
| | - Ann Formaz-Preston
- 1 Vascular Biology Program Centre for the Endothelium Centenary Institute The University of Sydney Newtown Australia
| | - Ka Ka Ting
- 1 Vascular Biology Program Centre for the Endothelium Centenary Institute The University of Sydney Newtown Australia
| | - Ben Roediger
- 2 Immune Imaging Program, Centenary Institute The University of Sydney Newtown Australia
| | - Wolfgang Weninger
- 2 Immune Imaging Program, Centenary Institute The University of Sydney Newtown Australia
| | - Martin A Schwartz
- 3 Department of Internal Medicine Yale Cardiovascular Research Center Yale University New Haven CT
| | - Mathew A Vadas
- 1 Vascular Biology Program Centre for the Endothelium Centenary Institute The University of Sydney Newtown Australia
| | - Jennifer R Gamble
- 1 Vascular Biology Program Centre for the Endothelium Centenary Institute The University of Sydney Newtown Australia
| |
Collapse
|
20
|
Strassheim D, Gerasimovskaya E, Irwin D, Dempsey EC, Stenmark K, Karoor V. RhoGTPase in Vascular Disease. Cells 2019; 8:E551. [PMID: 31174369 PMCID: PMC6627336 DOI: 10.3390/cells8060551] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/24/2019] [Accepted: 05/27/2019] [Indexed: 12/24/2022] Open
Abstract
Ras-homologous (Rho)A/Rho-kinase pathway plays an essential role in many cellular functions, including contraction, motility, proliferation, and apoptosis, inflammation, and its excessive activity induces oxidative stress and promotes the development of cardiovascular diseases. Given its role in many physiological and pathological functions, targeting can result in adverse effects and limit its use for therapy. In this review, we have summarized the role of RhoGTPases with an emphasis on RhoA in vascular disease and its impact on endothelial, smooth muscle, and heart and lung fibroblasts. It is clear from the various studies that understanding the regulation of RhoGTPases and their regulators in physiology and pathological conditions is required for effective targeting of Rho.
Collapse
Affiliation(s)
- Derek Strassheim
- Cardiovascular and Pulmonary Research Lab, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
| | - Evgenia Gerasimovskaya
- Cardiovascular and Pulmonary Research Lab, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
- Department of Pediatrics, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
| | - David Irwin
- Cardiovascular and Pulmonary Research Lab, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
| | - Edward C Dempsey
- Cardiovascular and Pulmonary Research Lab, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
- Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045, USA.
| | - Kurt Stenmark
- Cardiovascular and Pulmonary Research Lab, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
- Department of Pediatrics, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
| | - Vijaya Karoor
- Cardiovascular and Pulmonary Research Lab, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
- Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
| |
Collapse
|