1
|
Liu D, Zhang Y, Guo L, Fang R, Guo J, Li P, Qian T, Li W, Zhao L, Luo X, Zhang S, Shao J, Sun S. Single-cell atlas of healthy vocal folds and cellular function in the endothelial-to-mesenchymal transition. Cell Prolif 2024; 57:e13723. [PMID: 39245637 PMCID: PMC11628749 DOI: 10.1111/cpr.13723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/06/2024] [Accepted: 07/13/2024] [Indexed: 09/10/2024] Open
Abstract
The vocal fold is an architecturally complex organ comprising a heterogeneous mixture of various layers of individual epithelial and mesenchymal cell lineages. Here we performed single-cell RNA sequencing profiling of 5836 cells from the vocal folds of adult Sprague-Dawley rats. Combined with immunostaining, we generated a spatial and transcriptional map of the vocal fold cells and characterized the subpopulations of epithelial cells, mesenchymal cells, endothelial cells, and immune cells. We also identified a novel epithelial-to-mesenchymal transition-associated epithelial cell subset that was mainly found in the basal epithelial layers. We further confirmed that this subset acts as intermediate cells with similar genetic features to epithelial-to-mesenchymal transition in head and neck squamous cell carcinoma. Finally, we present the complex intracellular communication network involved homeostasis using CellChat analysis. These studies define the cellular and molecular framework of the biology and pathology of the VF mucosa and reveal the functional importance of developmental pathways in pathological states in cancer.
Collapse
Affiliation(s)
- Danling Liu
- Department of Otorhinolaryngology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangdong Cardiovascular InstituteSouthern Medical UniversityGuangzhouChina
- ENT Institute and Otorhinolaryngology, Innovation Center, Affiliated Eye and ENT Hospital, Key Laboratory of Hearing Medicine of NHFPC, State Key Laboratory of Medical NeurobiologyFudan UniversityShanghaiChina
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Institute of Microscale Optoelectronics and Otolaryngology Department and Biobank of the First Affiliated Hospital, Shenzhen Second People's Hospital, Health Science CenterShenzhen UniversityShenzhenChina
| | - Yunzhong Zhang
- ENT Institute and Otorhinolaryngology, Innovation Center, Affiliated Eye and ENT Hospital, Key Laboratory of Hearing Medicine of NHFPC, State Key Laboratory of Medical NeurobiologyFudan UniversityShanghaiChina
| | - Luo Guo
- ENT Institute and Otorhinolaryngology, Innovation Center, Affiliated Eye and ENT Hospital, Key Laboratory of Hearing Medicine of NHFPC, State Key Laboratory of Medical NeurobiologyFudan UniversityShanghaiChina
| | - Rui Fang
- ENT Institute and Otorhinolaryngology, Innovation Center, Affiliated Eye and ENT Hospital, Key Laboratory of Hearing Medicine of NHFPC, State Key Laboratory of Medical NeurobiologyFudan UniversityShanghaiChina
| | - Jin Guo
- ENT Institute and Otorhinolaryngology, Innovation Center, Affiliated Eye and ENT Hospital, Key Laboratory of Hearing Medicine of NHFPC, State Key Laboratory of Medical NeurobiologyFudan UniversityShanghaiChina
| | - Peifang Li
- ENT Institute and Otorhinolaryngology, Innovation Center, Affiliated Eye and ENT Hospital, Key Laboratory of Hearing Medicine of NHFPC, State Key Laboratory of Medical NeurobiologyFudan UniversityShanghaiChina
| | - Tingting Qian
- ENT Institute and Otorhinolaryngology, Innovation Center, Affiliated Eye and ENT Hospital, Key Laboratory of Hearing Medicine of NHFPC, State Key Laboratory of Medical NeurobiologyFudan UniversityShanghaiChina
| | - Wen Li
- ENT Institute and Otorhinolaryngology, Innovation Center, Affiliated Eye and ENT Hospital, Key Laboratory of Hearing Medicine of NHFPC, State Key Laboratory of Medical NeurobiologyFudan UniversityShanghaiChina
| | - Liping Zhao
- ENT Institute and Otorhinolaryngology, Innovation Center, Affiliated Eye and ENT Hospital, Key Laboratory of Hearing Medicine of NHFPC, State Key Laboratory of Medical NeurobiologyFudan UniversityShanghaiChina
| | - Xiaoning Luo
- Department of Otorhinolaryngology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangdong Cardiovascular InstituteSouthern Medical UniversityGuangzhouChina
| | - Siyi Zhang
- Department of Otorhinolaryngology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangdong Cardiovascular InstituteSouthern Medical UniversityGuangzhouChina
| | - Jun Shao
- ENT Institute and Otorhinolaryngology, Innovation Center, Affiliated Eye and ENT Hospital, Key Laboratory of Hearing Medicine of NHFPC, State Key Laboratory of Medical NeurobiologyFudan UniversityShanghaiChina
| | - Shan Sun
- ENT Institute and Otorhinolaryngology, Innovation Center, Affiliated Eye and ENT Hospital, Key Laboratory of Hearing Medicine of NHFPC, State Key Laboratory of Medical NeurobiologyFudan UniversityShanghaiChina
| |
Collapse
|
2
|
Park G, Hwang DY, Kim DY, Han JY, Lee E, Hwang H, Park JS, Kim DW, Hong S, Yim SV, Hong HS, Son Y. Identification of CD141 +vasculogenic precursor cells from human bone marrow and their endothelial engagement in the arteriogenesis by co-transplantation with mesenchymal stem cells. Stem Cell Res Ther 2024; 15:388. [PMID: 39482744 PMCID: PMC11526567 DOI: 10.1186/s13287-024-03994-9] [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: 07/09/2024] [Accepted: 10/10/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND Critical limb ischemia (CLI) is a condition characterized by insufficient blood flow to the lower limbs, resulting in severe ischemia and potentially leading to amputation. This study aims to identify novel vasculogenic precursor cells (VPCs) in human bone marrow and evaluate their efficacy in combination with bone marrow-derived mesenchymal stem cells (BM-MSCs) for the treatment of CLI. METHODS Ex vivo cultured VPCs and BM-MSCs from bone marrow were characterized and their effects on neovascularization and long-term tissue regeneration were tested in a mouse CLI model. RESULTS VPCs, expressing high levels of hepatocyte growth factor and c-MET, were identified from human bone marrow aspirates. These cells exhibited strong vasculogenic capacity in vitro but possessed a cellular phenotype distinct from those of previously reported endothelial precursor cells in circulation or cord blood. They also expressed most surface markers of BM-MSCs and demonstrated multipotent differentiation ability. Screening of 376 surface markers revealed that VPCs uniquely display CD141 (thrombomodulin). CD141+VPCs are present in BM aspirates as a rare population and can be expanded ex vivo with a population doubling time of approximately 20 h, generating an elaborate vascular network even under angiogenic factor-deficient conditions and recruiting BM-MSCs to the network as pericyte-like cells. Intramuscular transplantation of a combination of human CD141+VPCs and BM-MSCs at a ratio of 2:1 resulted in limb salvage, blood flow recovery, and regeneration of large vessels in the femoral artery-removed CLI model, with an efficacy superior to that of singular transplantation. Importantly, large arteries and arterioles in dual cell transplantation expressed human CD31 in the intima and human α-smooth muscle actin in media layer at 4 and 12 weeks, likely indicating their lineage commitment to endothelial cells and vascular smooth muscle, respectively, in vivo. CONCLUSION Dual-cell therapy using BM-derived CD141+ VPCs and BM-MSCs holds potential for further development in clinical trials to treat peripheral artery disease and diabetic ulcers.
Collapse
Affiliation(s)
- Gabee Park
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea
| | - Dae Yeon Hwang
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea
| | - Do Young Kim
- Department of Biomedical Science and Technology, Graduated School, Kyung Hee University, Seoul, Korea
| | - Ji Young Han
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea
| | - Euiseon Lee
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea
| | - Hwakyung Hwang
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea
| | - Jeong Seop Park
- Department of Biomedical Science and Technology, Graduated School, Kyung Hee University, Seoul, Korea
| | - Dae Wook Kim
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea
- Department of Genetic Engineering, Graduate School of Biotechnology, Kyung Hee University, Yong In, Korea
| | - Seonmin Hong
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea
| | - Sung Vin Yim
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea
- Department of Clinical Pharmacology and Therapeutics, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Hyun Sook Hong
- Department of Biomedical Science and Technology, Graduated School, Kyung Hee University, Seoul, Korea.
- East-West Medical Research Institute, Kyung Hee University, Seoul, Korea.
| | - Youngsook Son
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea.
- Department of Genetic Engineering, Graduate School of Biotechnology, Kyung Hee University, Yong In, Korea.
| |
Collapse
|
3
|
Stanworth M, Zhang SD. Elucidating the roles of SOD3 correlated genes and reactive oxygen species in rare human diseases using a bioinformatic-ontology approach. PLoS One 2024; 19:e0313139. [PMID: 39480826 PMCID: PMC11527182 DOI: 10.1371/journal.pone.0313139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 10/18/2024] [Indexed: 11/02/2024] Open
Abstract
Superoxide Dismutase 3 (SOD3) scavenges extracellular superoxide giving a hydrogen peroxide metabolite. Both Reactive Oxygen Species diffuse through aquaporins causing oxidative stress and biomolecular damage. SOD3 is differentially expressed in cancer and this research utilises Gene Expression Omnibus data series GSE2109 with 2,158 cancer samples. Genome-wide expression correlation analysis was conducted with SOD3 as the seed gene. Categorical SOD3 Pearson Correlation gene lists incrementing in correlation strength by 0.01 from ρ≥|0.34| to ρ≥|0.41| were extracted from the data. Positively and negatively SOD3 correlated genes were separated for each list and checked for significance against disease overlapping genes in the ClinVar and Orphanet databases via Enrichr. Disease causal genes were added to the relevant gene list and checked against Gene Ontology, Phenotype Ontology, and Elsevier Pathways via Enrichr before the significant ontologies containing causal and non-overlapping genes were reviewed with a literature search for possible disease and oxidative stress associations. 12 significant individually discriminated disorders were identified: Autosomal Dominant Cutis Laxa (p = 6.05x10-7), Renal Tubular Dysgenesis of Genetic Origin (p = 6.05x10-7), Lethal Arteriopathy Syndrome due to Fibulin-4 Deficiency (p = 6.54x10-9), EMILIN-1-related Connective Tissue Disease (p = 6.54x10-9), Holt-Oram Syndrome (p = 7.72x10-10), Multisystemic Smooth Muscle Dysfunction Syndrome (p = 9.95x10-15), Distal Hereditary Motor Neuropathy type 2 (p = 4.48x10-7), Congenital Glaucoma (p = 5.24x210-9), Megacystis-Microcolon-Intestinal Hypoperistalsis Syndrome (p = 3.77x10-16), Classical-like Ehlers-Danlos Syndrome type 1 (p = 3.77x10-16), Retinoblastoma (p = 1.9x10-8), and Lynch Syndrome (p = 5.04x10-9). 35 novel (21 unique) genes across 12 disorders were identified: ADNP, AOC3, CDC42EP2, CHTOP, CNN1, DES, FOXF1, FXR1, HLTF, KCNMB1, MTF2, MYH11, PLN, PNPLA2, REST, SGCA, SORBS1, SYNPO2, TAGLN, WAPL, and ZMYM4. These genes are proffered as potential biomarkers or therapeutic targets for the corresponding rare diseases discussed.
Collapse
Affiliation(s)
- Mark Stanworth
- Personalised Medicine Centre, School of Medicine, Ulster University, C-TRIC Building, Altnagelvin Hospital, Derry, Londonderry, Northern Ireland
| | - Shu-Dong Zhang
- Personalised Medicine Centre, School of Medicine, Ulster University, C-TRIC Building, Altnagelvin Hospital, Derry, Londonderry, Northern Ireland
| |
Collapse
|
4
|
Shrestha B, Stern NB, Zhou A, Dunn A, Porter T. Current trends in the characterization and monitoring of vascular response to cancer therapy. Cancer Imaging 2024; 24:143. [PMID: 39438891 PMCID: PMC11515715 DOI: 10.1186/s40644-024-00767-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 08/26/2024] [Indexed: 10/25/2024] Open
Abstract
Tumor vascular physiology is an important determinant of disease progression as well as the therapeutic outcome of cancer treatment. Angiogenesis or the lack of it provides crucial information about the tumor's blood supply and therefore can be used as an index for cancer growth and progression. While standalone anti-angiogenic therapy demonstrated limited therapeutic benefits, its combination with chemotherapeutic agents improved the overall survival of cancer patients. This could be attributed to the effect of vascular normalization, a dynamic process that temporarily reverts abnormal vasculature to the normal phenotype maximizing the delivery and intratumor distribution of chemotherapeutic agents. Longitudinal monitoring of vascular changes following antiangiogenic therapy can indicate an optimal window for drug administration and estimate the potential outcome of treatment. This review primarily focuses on the status of various imaging modalities used for the longitudinal characterization of vascular changes before and after anti-angiogenic therapies and their clinical prospects.
Collapse
Affiliation(s)
- Binita Shrestha
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA.
| | - Noah B Stern
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Annie Zhou
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Andrew Dunn
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Tyrone Porter
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| |
Collapse
|
5
|
Gomez GT, Shi L, Fohner AE, Chen J, Yang Y, Fornage M, Duggan MR, Peng Z, Daya GN, Tin A, Schlosser P, Longstreth WT, Kalani R, Sharma M, Psaty BM, Nevado-Holgado AJ, Buckley NJ, Gottesman RF, Lutsey PL, Jack CR, Sullivan KJ, Mosley T, Hughes TM, Coresh J, Walker KA. Plasma proteome-wide analysis of cerebral small vessel disease identifies novel biomarkers and disease pathways. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.07.24314972. [PMID: 39417098 PMCID: PMC11483013 DOI: 10.1101/2024.10.07.24314972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Cerebral small vessel disease (SVD), as defined by neuroimaging characteristics such as white matter hyperintensities (WMHs), cerebral microhemorrhages (CMHs), and lacunar infarcts, is highly prevalent and has been associated with dementia risk and other clinical sequelae. Although conditions such as hypertension are known to contribute to SVD, little is known about the diverse set of subclinical biological processes and molecular mediators that may also influence the development and progression of SVD. To better understand the mechanisms underlying SVD and to identify novel SVD biomarkers, we used a large-scale proteomic platform to relate 4,877 plasma proteins to MRI-defined SVD characteristics within 1,508 participants of the Atherosclerosis Risk in Communities (ARIC) Study cohort. Our proteome-wide analysis of older adults (mean age: 76) identified 13 WMH-associated plasma proteins involved in synaptic function, endothelial integrity, and angiogenesis, two of which remained associated with late-life WMH volume when measured nearly 20 years earlier, during midlife. We replicated the relationship between 9 candidate proteins and WMH volume in one or more external cohorts; we found that 11 of the 13 proteins were associated with risk for future dementia; and we leveraged publicly available proteomic data from brain tissue to demonstrate that a subset of WMH-associated proteins was differentially expressed in the context of cerebral atherosclerosis, pathologically-defined Alzheimer's disease, and cognitive decline. Bidirectional two-sample Mendelian randomization analyses examined the causal relationships between candidate proteins and WMH volume, while pathway and network analyses identified discrete biological processes (lipid/cholesterol metabolism, NF-kB signaling, hemostasis) associated with distinct forms of SVD. Finally, we synthesized these findings to identify two plasma proteins, oligodendrocyte myelin glycoprotein (OMG) and neuronal pentraxin receptor (NPTXR), as top candidate biomarkers for elevated WMH volume and its clinical manifestations.
Collapse
|
6
|
Advani D, Farid N, Tariq MH, Kohli N. A systematic review of mesenchymal stem cell secretome: Functional annotations, gene clusters and proteomics analyses for bone formation. Bone 2024; 190:117269. [PMID: 39368726 DOI: 10.1016/j.bone.2024.117269] [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] [Received: 06/19/2024] [Revised: 08/15/2024] [Accepted: 10/02/2024] [Indexed: 10/07/2024]
Abstract
The regenerative capacity of mesenchymal stem cells (MSCs) is now attributed to their ability to release paracrine factors into the extracellular matrix that boost tissue regeneration, reduce inflammation and encourage healing. Understanding the MSC secretome is crucial for shifting the prototypic conventional cell-based therapies to cell-free regenerative treatments. This systematic review aimed to analyse the functional annotations of the secretome of human adult adipose tissue and bone marrow MSCs and unveil the gene clusters responsible for bone formation. Bioinformatics tools were used to identify the biological processes, molecular functions, hallmarks and KEGG pathways of adipose and bone marrow MSC secretome proteins. We found a substantial overlap in the functional annotations and protein compositions of both adipose and bone marrow MSC secretome indicating that MSC source may be noninfluencial with regards to tissue regeneration. Additionally, a novel network pharmacology-based analysis of the secreted proteins revealed that the commonly secreted proteins within a single source interact with multiple drugable targets of bone diseases and regulate various KEGG pathway. This study unravels the secretome profile of human adult adipose and bone marrow MSCs based on the current literature and provides valuable insights into the therapeutic use of the MSC secretome for cell-free therapies.
Collapse
Affiliation(s)
- Dia Advani
- Department of Biomedical Engineering and Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates.
| | - Nouran Farid
- Department of Biomedical Engineering and Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates.
| | - Muhammad Hamza Tariq
- Department of Biomedical Engineering and Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates.
| | - Nupur Kohli
- Department of Biomedical Engineering and Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates; Healthcare Engineering Innovation Center, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates.
| |
Collapse
|
7
|
Dib MJ, Azzo JD, Zhao L, Salman O, Gan S, De Buyzere ML, De Meyer T, Ebert C, Gunawardhana K, Liu L, Gordon D, Seiffert D, Ching-Pin C, Zamani P, Cohen JB, Pourmussa B, Kun S, Gill D, Burgess S, van Empel V, Richards AM, Dennis J, Javaheri A, Mann DL, Cappola TP, Rietzschel E, Chirinos JA. Proteome-Wide Genetic Investigation of Large Artery Stiffness. JACC Basic Transl Sci 2024; 9:1178-1191. [PMID: 39534640 PMCID: PMC11551872 DOI: 10.1016/j.jacbts.2024.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 11/16/2024]
Abstract
The molecular mechanisms contributing to large artery stiffness (LAS) are not fully understood. The aim of this study was to investigate the association between circulating plasma proteins and LAS using complementary proteomic and genomic analyses. A total of 106 proteins associated with carotid-femoral pulse-wave velocity, a noninvasive measure of LAS, were identified in 1,178 individuals from the Asklepios study cohort. Mendelian randomization analyses revealed causal effects of 13 genetically predicted plasma proteins on pulse pressure, including cartilage intermediate layer protein-2, high-temperature requirement A serine peptidase-1, and neuronal growth factor-1. These findings suggest potential novel therapeutic targets to reduce LAS and its related diseases.
Collapse
Affiliation(s)
- Marie-Joe Dib
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Joe David Azzo
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lei Zhao
- Bristol Myers Squibb, Lawrenceville, New Jersey, USA
| | - Oday Salman
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sushrima Gan
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Marc L. De Buyzere
- Department of Cardiovascular Diseases, Ghent University Hospital, Ghent, Belgium
| | - Tim De Meyer
- Department of Mathematical Modelling, Statistics and Bioinformatics, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | | | | | - Laura Liu
- Bristol Myers Squibb, Lawrenceville, New Jersey, USA
| | - David Gordon
- Bristol Myers Squibb, Lawrenceville, New Jersey, USA
| | | | | | - Payman Zamani
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jordana B. Cohen
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Renal-Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Bianca Pourmussa
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Seavmeiyin Kun
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, United Kingdom
| | - Stephen Burgess
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Vanessa van Empel
- Department of Cardiology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - A. Mark Richards
- Cardiovascular Research Institute, National University of Singapore, Singapore, Singapore
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
| | | | - Ali Javaheri
- Washington University School of Medicine, St. Louis, Missouri, USA
- John J. Cochran Veterans Hospital, St. Louis, Missouri, USA
| | - Douglas L. Mann
- Washington University School of Medicine, St. Louis, Missouri, USA
| | - Thomas P. Cappola
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ernst Rietzschel
- Department of Cardiovascular Diseases, Ghent University Hospital, Ghent, Belgium
| | - Julio A. Chirinos
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
8
|
Zhang W, Xu Y, Zheng X, Shen J, Li Y. Identifying cell types by lasso-constraint regularized Gaussian graphical model based on weighted distance penalty. Brief Bioinform 2024; 25:bbae572. [PMID: 39541187 PMCID: PMC11562834 DOI: 10.1093/bib/bbae572] [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: 08/11/2024] [Revised: 10/10/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) technology is one of the most cost-effective and efficacious methods for revealing cellular heterogeneity and diversity. Precise identification of cell types is essential for establishing a robust foundation for downstream analyses and is a prerequisite for understanding heterogeneous mechanisms. However, the accuracy of existing methods warrants improvement, and highly accurate methods often impose stringent equipment requirements. Moreover, most unsupervised learning-based approaches are constrained by the need to input the number of cell types a prior, which limits their widespread application. In this paper, we propose a novel algorithm framework named WLGG. Initially, to capture the underlying nonlinear information, we introduce a weighted distance penalty term utilizing the Gaussian kernel function, which maps data from a low-dimensional nonlinear space to a high-dimensional linear space. We subsequently impose a Lasso constraint on the regularized Gaussian graphical model to enhance its ability to capture linear data characteristics. Additionally, we utilize the Eigengap strategy to predict the number of cell types and obtain predicted labels via spectral clustering. The experimental results on 14 test datasets demonstrate the superior clustering accuracy of the WLGG algorithm over 16 alternative methods. Furthermore, downstream analysis, including marker gene identification, pseudotime inference, and functional enrichment analysis based on the similarity matrix and predicted labels from the WLGG algorithm, substantiates the reliability of WLGG and offers valuable insights into biological dynamic biological processes and regulatory mechanisms.
Collapse
Affiliation(s)
- Wei Zhang
- School of Mathematics and Physics, Wuhan Institute of Technology, Wuhan 430205, China
| | - Yaxin Xu
- Peng Cheng Laboratory, and School of Microelectronics, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xiaoying Zheng
- School of Mathematics and Physics, Wuhan Institute of Technology, Wuhan 430205, China
| | - Juan Shen
- School of Mathematics and Physics, Wuhan Institute of Technology, Wuhan 430205, China
| | - Yuanyuan Li
- School of Mathematics and Physics, Wuhan Institute of Technology, Wuhan 430205, China
| |
Collapse
|
9
|
Tsuji-Tamura K, Sato M, Tamura M. Pharmacological control of angiogenesis by regulating phosphorylation of myosin light chain 2. Cell Signal 2024; 120:111223. [PMID: 38729320 DOI: 10.1016/j.cellsig.2024.111223] [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: 11/15/2023] [Revised: 04/25/2024] [Accepted: 05/07/2024] [Indexed: 05/12/2024]
Abstract
BACKGROUND Control of angiogenesis is widely considered a therapeutic strategy, but reliable control methods are still under development. Phosphorylation of myosin light chain 2 (MLC2), which regulates actin-myosin interaction, is critical to the behavior of vascular endothelial cells (ECs) during angiogenesis. MLC2 is phosphorylated by MLC kinase (MLCK) and dephosphorylated by MLC phosphatase (MLCP) containing a catalytic subunit PP1. We investigated the potential role of MLC2 in the pharmacological control of angiogenesis. METHODS AND RESULTS We exposed transgenic zebrafish Tg(fli1a:Myr-mCherry)ncv1 embryos to chemical inhibitors and observed vascular development. PP1 inhibition by tautomycetin increased length of intersegmental vessels (ISVs), whereas MLCK inhibition by ML7 decreased it; these effects were not accompanied by structural dysplasia. ROCK inhibition by Y-27632 also decreased vessel length. An in vitro angiogenesis model of human umbilical vein endothelial cells (HUVECs) showed that tautomycetin increased vascular cord formation, whereas ML7 and Y-27632 decreased it. These effects appear to be influenced by regulation of cell morphology rather than cell viability or motility. Actin co-localized with phosphorylated MLC2 (pMLC2) was abundant in vascular-like elongated-shaped ECs, but poor in non-elongated ECs. pMLC2 was associated with tightly arranged actin, but not with loosely arranged actin. Moreover, knockdown of MYL9 gene encoding MLC2 reduced total MLC2 and pMLC2 protein and inhibited angiogenesis in HUVECs. CONCLUSION The present study found that MLC2 is a pivotal regulator of angiogenesis. MLC2 phosphorylation may be involved in the regulation of of cell morphogenesis and cell elongation. The functionally opposite inhibitors positively or negatively control angiogenesis, probably through the regulating EC morphology. These findings may provide a unique therapeutic target for angiogenesis.
Collapse
Affiliation(s)
- Kiyomi Tsuji-Tamura
- Oral Biochemistry and Molecular Biology, Department of Oral Health Science, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Kita 13, Nishi 7, Kita-Ku, Sapporo 060-8586, Japan.
| | - Mari Sato
- Oral Biochemistry and Molecular Biology, Department of Oral Health Science, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Kita 13, Nishi 7, Kita-Ku, Sapporo 060-8586, Japan
| | - Masato Tamura
- Oral Biochemistry and Molecular Biology, Department of Oral Health Science, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Kita 13, Nishi 7, Kita-Ku, Sapporo 060-8586, Japan
| |
Collapse
|
10
|
He Y, Yang X, Wu N. TGF β1, SNAIL2, and PAPP-A Expression in Placenta of Gestational Diabetes Mellitus Patients. J Diabetes Res 2024; 2024:1386469. [PMID: 39109165 PMCID: PMC11303042 DOI: 10.1155/2024/1386469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/26/2024] [Accepted: 06/26/2024] [Indexed: 09/17/2024] Open
Abstract
Background: Gestational diabetes mellitus (GDM) is a pregnancy-related diabetic condition that may cause serious complications. However, its pathogenesis remains unclear. Placental damage due to GDM may lead to several health issues that cannot be ignored. Thus, we aimed to identify the mechanisms underlying GDM by screening differentially expressed genes (DEGs) related to vascular endothelial cells in the GDM databases and verify the expression of these DEGs in the placentas of women afflicted by GDM. Methods: We used GDM microarray datasets integrated from the Gene Expression Omnibus (GEO) database. Functional annotation and protein-protein interaction (PPI) analyses were used to screen DEGs. Placental tissues from 20 pregnant women with GDM and 20 healthy pregnant women were collected, and differential gene expression in the placental tissues was verified via qRT-PCR, western blotting, and immunofluorescence. Results: Bioinformatics analysis revealed three significant DEGs: SNAIL2, PAPP-A, and TGFβ1. These genes were all predicted to be underexpressed in patients with GDM. The results of qRT-PCR, western blot, and immunofluorescence analyses indicated that SNAIL2 and PAPP-A in the placenta tissue of patients with GDM were significantly underexpressed. However, TGFβ1 in the placenta tissues of GDM was significantly overexpressed. Conclusion: SNAIL2, TGFβ1, and PAPP-A may affect the placentas of pregnant women with GDM, warranting further investigation.
Collapse
Affiliation(s)
- Yujing He
- Department of EndocrinologyShengjing Hospital of China Medical University, Shenyang 110004, China
- School of Life ScienceLiaoning University, 66 Chongshan Middle Road, Shenyang 110036, China
| | - Xiyao Yang
- Department of EndocrinologyShengjing Hospital of China Medical University, Shenyang 110004, China
| | - Na Wu
- Department of EndocrinologyShengjing Hospital of China Medical University, Shenyang 110004, China
- Department of PediatricsShengjing Hospital of China Medical University, Shenyang 110004, China
| |
Collapse
|
11
|
Gendreizig S, Martínez-Ruiz L, López-Rodríguez A, Pabla H, Hose L, Brasch F, Busche T, Escames G, Sudhoff H, Scholtz LU, Todt I, Oppel F. Human papillomavirus-associated head and neck squamous cell carcinoma cells lose viability during triggered myocyte lineage differentiation. Cell Death Dis 2024; 15:517. [PMID: 39030166 PMCID: PMC11271587 DOI: 10.1038/s41419-024-06867-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 06/25/2024] [Accepted: 06/25/2024] [Indexed: 07/21/2024]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a highly malignant disease, and death rates have remained at approximately 50% for decades. New tumor-targeting strategies are desperately needed, and a previous report indicated the triggered differentiation of HPV-negative HNSCC cells to confer therapeutic benefits. Using patient-derived tumor cells, we created a similar HNSCC differentiation model of HPV+ tumor cells from two patients. We observed a loss of malignant characteristics in differentiating cell culture conditions, including irregularly enlarged cell morphology, cell cycle arrest with downregulation of Ki67, and reduced cell viability. RNA-Seq showed myocyte-like differentiation with upregulation of markers of myofibril assembly. Immunofluorescence staining of differentiated and undifferentiated primary HPV+ HNSCC cells confirmed an upregulation of these markers and the formation of parallel actin fibers reminiscent of myoblast-lineage cells. Moreover, immunofluorescence of HPV+ tumor tissue revealed areas of cells co-expressing the identified markers of myofibril assembly, HPV surrogate marker p16, and stress-associated basal keratinocyte marker KRT17, indicating that the observed myocyte-like in vitro differentiation occurs in human tissue. We are the first to report that carcinoma cells can undergo a triggered myocyte-like differentiation, and our study suggests that the targeted differentiation of HPV+ HNSCCs might be therapeutically valuable.
Collapse
Affiliation(s)
- Sarah Gendreizig
- Department of Otolaryngology, Head and Neck Surgery, Campus Klinikum Bielefeld Mitte, University Hospital OWL of Bielefeld University, Teutoburger Str. 50, 33604, Bielefeld, Germany
| | - Laura Martínez-Ruiz
- Institute of Biotechnology, Biomedical Research Center, Health Sciences Technology Park, University of Granada, Granada, Spain
- Department of Physiology, Faculty of Medicine, University of Granada, 18016, Granada, Spain
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Investigación Biosanitaria (Ibs), Granada, San Cecilio University Hospital, Granada, Spain
| | - Alba López-Rodríguez
- Institute of Biotechnology, Biomedical Research Center, Health Sciences Technology Park, University of Granada, Granada, Spain
- Department of Physiology, Faculty of Medicine, University of Granada, 18016, Granada, Spain
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Investigación Biosanitaria (Ibs), Granada, San Cecilio University Hospital, Granada, Spain
| | - Harkiren Pabla
- Department of Otolaryngology, Head and Neck Surgery, Campus Klinikum Bielefeld Mitte, University Hospital OWL of Bielefeld University, Teutoburger Str. 50, 33604, Bielefeld, Germany
| | - Leonie Hose
- Department of Otolaryngology, Head and Neck Surgery, Campus Klinikum Bielefeld Mitte, University Hospital OWL of Bielefeld University, Teutoburger Str. 50, 33604, Bielefeld, Germany
| | - Frank Brasch
- Department of Pathology, Klinikum Bielefeld, Teutoburger Str. 50, 33604, Bielefeld, Germany
| | - Tobias Busche
- Center for Biotechnology (CeBiTec), University Hospital OWL of Bielefeld University, Bielefeld, Germany
| | - Germaine Escames
- Institute of Biotechnology, Biomedical Research Center, Health Sciences Technology Park, University of Granada, Granada, Spain
- Department of Physiology, Faculty of Medicine, University of Granada, 18016, Granada, Spain
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Investigación Biosanitaria (Ibs), Granada, San Cecilio University Hospital, Granada, Spain
| | - Holger Sudhoff
- Department of Otolaryngology, Head and Neck Surgery, Campus Klinikum Bielefeld Mitte, University Hospital OWL of Bielefeld University, Teutoburger Str. 50, 33604, Bielefeld, Germany
| | - Lars Uwe Scholtz
- Department of Otolaryngology, Head and Neck Surgery, Campus Klinikum Bielefeld Mitte, University Hospital OWL of Bielefeld University, Teutoburger Str. 50, 33604, Bielefeld, Germany
| | - Ingo Todt
- Department of Otolaryngology, Head and Neck Surgery, Campus Klinikum Bielefeld Mitte, University Hospital OWL of Bielefeld University, Teutoburger Str. 50, 33604, Bielefeld, Germany
| | - Felix Oppel
- Department of Otolaryngology, Head and Neck Surgery, Campus Klinikum Bielefeld Mitte, University Hospital OWL of Bielefeld University, Teutoburger Str. 50, 33604, Bielefeld, Germany.
| |
Collapse
|
12
|
Liu Y, Lyons CJ, Ayu C, O’Brien T. Enhancing endothelial colony-forming cells for treating diabetic vascular complications: challenges and clinical prospects. Front Endocrinol (Lausanne) 2024; 15:1396794. [PMID: 39076517 PMCID: PMC11284052 DOI: 10.3389/fendo.2024.1396794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 06/14/2024] [Indexed: 07/31/2024] Open
Abstract
Diabetes mellitus (DM) is a metabolic disease characterized by hyperglycemia, leading to various vascular complications. Accumulating evidence indicates that endothelial colony-forming cells (ECFCs) have attractive prospects for repairing and restoring blood vessels. Thus, ECFCs may be a novel therapeutic option for diabetic patients with vascular complications who require revascularization therapy. However, it has been reported that the function of ECFCs is impaired in DM, which poses challenges for the autologous transplantation of ECFCs. In this review, we summarize the molecular mechanisms that may be responsible for ECFC dysfunction and discuss potential strategies for improving the therapeutic efficacy of ECFCs derived from patients with DM. Finally, we discuss barriers to the use of ECFCs in human studies in light of the fact that there are no published reports using these cells in humans.
Collapse
Affiliation(s)
| | | | | | - Timothy O’Brien
- Regenerative Medicine Institute (REMEDI), Biomedical Sciences Building, University of Galway, Galway, Ireland
| |
Collapse
|
13
|
Wu J, Li W, Su J, Zheng J, Liang Y, Lin J, Xu B, Liu Y. Integration of single-cell sequencing and bulk RNA-seq to identify and develop a prognostic signature related to colorectal cancer stem cells. Sci Rep 2024; 14:12270. [PMID: 38806611 PMCID: PMC11133358 DOI: 10.1038/s41598-024-62913-3] [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: 01/08/2024] [Accepted: 05/22/2024] [Indexed: 05/30/2024] Open
Abstract
The prognosis for patients with colorectal cancer (CRC) remains worse than expected due to metastasis, recurrence, and resistance to chemotherapy. Colorectal cancer stem cells (CRCSCs) play a vital role in tumor metastasis, recurrence, and chemotherapy resistance. However, there are currently no prognostic markers based on CRCSCs-related genes available for clinical use. In this study, single-cell transcriptome sequencing was employed to distinguish cancer stem cells (CSCs) in the CRC microenvironment and analyze their properties at the single-cell level. Subsequently, data from TCGA and GEO databases were utilized to develop a prognostic risk model for CRCSCs-related genes and validate its diagnostic performance. Additionally, functional enrichment, immune response, and chemotherapeutic drug sensitivity of the relevant genes in the risk model were investigated. Lastly, the key gene RPS17 in the risk model was identified as a potential prognostic marker and therapeutic target for further comprehensive studies. Our findings provide new insights into the prognostic treatment of CRC and offer novel perspectives for a systematic and comprehensive understanding of CRC development.
Collapse
Affiliation(s)
- Jiale Wu
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Wanyu Li
- Well Lead Medical Co., Ltd., Guangzhou, 511434, Guangdong, China
| | - Junyu Su
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Jiamin Zheng
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Yanwen Liang
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Jiansuo Lin
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Bilian Xu
- School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China.
| | - Yi Liu
- School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China.
| |
Collapse
|
14
|
Colijn S, Nambara M, Malin G, Sacchetti EA, Stratman AN. Identification of distinct vascular mural cell populations during zebrafish embryonic development. Dev Dyn 2024; 253:519-541. [PMID: 38112237 PMCID: PMC11065631 DOI: 10.1002/dvdy.681] [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: 04/06/2023] [Revised: 11/14/2023] [Accepted: 11/29/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND Mural cells are an essential perivascular cell population that associate with blood vessels and contribute to vascular stabilization and tone. In the embryonic zebrafish vasculature, pdgfrb and tagln are commonly used as markers for identifying pericytes and vascular smooth muscle cells. However, the overlapping and distinct expression patterns of these markers in tandem have not been fully described. RESULTS Here, we used the Tg(pdgfrb:Gal4FF; UAS:RFP) and Tg(tagln:NLS-EGFP) transgenic lines to identify single- and double-positive perivascular cell populations on the cranial, axial, and intersegmental vessels between 1 and 5 days postfertilization. From this comparative analysis, we discovered two novel regions of tagln-positive cell populations that have the potential to function as mural cell precursors. Specifically, we found that the hypochord-a reportedly transient structure-contributes to tagln-positive cells along the dorsal aorta. We also identified a unique mural cell progenitor population that resides along the midline between the neural tube and notochord and contributes to intersegmental vessel mural cell coverage. CONCLUSION Together, our findings highlight the variability and versatility of tracking both pdgfrb and tagln expression in mural cells of the developing zebrafish embryo and reveal unexpected embryonic cell populations that express pdgfrb and tagln.
Collapse
Affiliation(s)
- Sarah Colijn
- Department of Cell Biology and Physiology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110
| | - Miku Nambara
- Department of Cell Biology and Physiology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110
| | - Gracie Malin
- Department of Cell Biology and Physiology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110
| | - Elena A. Sacchetti
- Department of Cell Biology and Physiology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110
| | - Amber N. Stratman
- Department of Cell Biology and Physiology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110
| |
Collapse
|
15
|
Sakamuri SSVP, Sure VN, Oruganti L, Wisen W, Chandra PK, Liu N, Fonseca VA, Wang X, Klein J, Katakam PVG. Acute severe hypoglycemia alters mouse brain microvascular proteome. J Cereb Blood Flow Metab 2024; 44:556-572. [PMID: 37944245 PMCID: PMC10981402 DOI: 10.1177/0271678x231212961] [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: 05/01/2023] [Revised: 09/12/2023] [Accepted: 10/05/2023] [Indexed: 11/12/2023]
Abstract
Hypoglycemia increases the risk related to stroke and neurodegenerative diseases, however, the underlying mechanisms are unclear. For the first time, we studied the effect of a single episode (acute) of severe (ASH) and mild (AMH) hypoglycemia on mouse brain microvascular proteome. After four-hour fasting, insulin was administered (i.p) to lower mean blood glucose in mice and induce ∼30 minutes of ASH (∼30 mg/dL) or AMH (∼75 mg/dL), whereas a similar volume of saline was given to control mice (∼130 mg/dL). Blood glucose was allowed to recover over 60 minutes either spontaneously or by 20% dextrose administration (i.p). Twenty-four hours later, the brain microvessels (BMVs) were isolated, and tandem mass tag (TMT)-based quantitative proteomics was performed using liquid chromatography-mass spectrometry (LC/MS). When compared to control, ASH significantly downregulated 13 proteins (p ≤ 0.05) whereas 23 proteins showed a strong trend toward decrease (p ≤ 0.10). When compared to AMH, ASH significantly induced the expression of 35 proteins with 13 proteins showing an increasing trend. AMH downregulated only 3 proteins. ASH-induced downregulated proteins are involved in actin cytoskeleton maintenance needed for cell shape and migration which are critical for blood-brain barrier maintenance and angiogenesis. In contrast, ASH-induced upregulated proteins are RNA-binding proteins involved in RNA splicing, transport, and stability. Thus, ASH alters BMV proteomics to impair cytoskeletal integrity and RNA processing which are critical for cerebrovascular function.
Collapse
Affiliation(s)
- Siva SVP Sakamuri
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Venkata N Sure
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Lokanatha Oruganti
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - William Wisen
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Partha K Chandra
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
- Neuroscience Program, Tulane Brain Institute, Tulane University, New Orleans, LA, USA
| | - Ning Liu
- Neuroscience Program, Tulane Brain Institute, Tulane University, New Orleans, LA, USA
- Clinical Neuroscience Research Center, New Orleans, LA, USA
- Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Vivian A Fonseca
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Xiaoying Wang
- Neuroscience Program, Tulane Brain Institute, Tulane University, New Orleans, LA, USA
- Clinical Neuroscience Research Center, New Orleans, LA, USA
- Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Jennifer Klein
- Department of Biochemistry & Molecular Biology, Louisiana State University School of Medicine, New Orleans, LA, USA
| | - Prasad VG Katakam
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
- Neuroscience Program, Tulane Brain Institute, Tulane University, New Orleans, LA, USA
- Clinical Neuroscience Research Center, New Orleans, LA, USA
| |
Collapse
|
16
|
Yuan P, Chen Y, Sun T, Cui L, Wei Y, Li T, Meng Q. Exploring potential genes and mechanisms linking erectile dysfunction and depression. Front Endocrinol (Lausanne) 2023; 14:1221043. [PMID: 38111702 PMCID: PMC10726033 DOI: 10.3389/fendo.2023.1221043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 11/20/2023] [Indexed: 12/20/2023] Open
Abstract
Background The clinical correlation between erectile dysfunction (ED) and depression has been revealed in cumulative studies. However, the evidence of shared mechanisms between them was insufficient. This study aimed to explore common transcriptomic alterations associated with ED and depression. Materials and methods The gene sets associated with ED and depression were collected from the Gene Expression Omnibus (GEO) database. Comparative analysis was conducted to obtain common genes. Using R software and other appropriate tools, we conducted a range of analyses, including function enrichment, interactive network creation, gene cluster analysis, and transcriptional and post-transcriptional signature profiling. Candidate hub crosslinks between ED and depression were selected after external validation and molecular experiments. Furthermore, subpopulation location and disease association of hub genes were explored. Results A total of 85 common genes were identified between ED and depression. These genes strongly correlate with cell adhesion, redox homeostasis, reactive oxygen species metabolic process, and neuronal cell body. An interactive network consisting of 80 proteins and 216 interactions was thereby developed. Analysis of the proteomic signature of common genes highlighted eight major shared genes: CLDN5, COL7A1, LDHA, MAP2K2, RETSAT, SEMA3A, TAGLN, and TBC1D1. These genes were involved in blood vessel morphogenesis and muscle cell activity. A subsequent transcription factor (TF)-miRNA network showed 47 TFs and 88 miRNAs relevant to shared genes. Finally, CLDN5 and TBC1D1 were well-validated and identified as the hub crosslinks between ED and depression. These genes had specific subpopulation locations in the corpus cavernosum and brain tissue, respectively. Conclusion Our study is the first to investigate common transcriptomic alterations and the shared biological roles of ED and depression. The findings of this study provide insights into the referential molecular mechanisms underlying the co-existence between depression and ED.
Collapse
Affiliation(s)
- Penghui Yuan
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Urology Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yinwei Chen
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Taotao Sun
- Department of Urology Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lingang Cui
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yinsheng Wei
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Teng Li
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qingjun Meng
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
17
|
Jonmundsson T, Steindorsdottir AE, Austin TR, Frick EA, Axelsson GT, Launer L, Psaty BM, Loureiro J, Orth AP, Aspelund T, Emilsson V, Floyd JS, Jennings L, Gudnason V, Gudmundsdottir V. A proteomic analysis of atrial fibrillation in a prospective longitudinal cohort (AGES-Reykjavik study). Europace 2023; 25:euad320. [PMID: 37967346 PMCID: PMC10685397 DOI: 10.1093/europace/euad320] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/01/2023] [Accepted: 10/06/2023] [Indexed: 11/17/2023] Open
Abstract
AIMS Atrial fibrillation (AF) is associated with high risk of comorbidities and mortality. Our aim was to examine causal and predictive relationships between 4137 serum proteins and incident AF in the prospective population-based Age, Gene/Environment Susceptibility-Reykjavik (AGES-Reykjavik) study. METHODS AND RESULTS The study included 4765 participants, of whom 1172 developed AF. Cox proportional hazards regression models were fitted for 4137 baseline protein measurements adjusting for known risk factors. Protein associations were tested for replication in the Cardiovascular Health Study (CHS). Causal relationships were examined in a bidirectional, two-sample Mendelian randomization analysis. The time-dependent area under the receiver operating characteristic curve (AUC)-statistic was examined as protein levels and an AF-polygenic risk score (PRS) were added to clinical risk models. The proteomic signature of incident AF consisted of 76 proteins, of which 63 (83%) were novel and 29 (38%) were replicated in CHS. The signature included both N-terminal prohormone of brain natriuretic peptide (NT-proBNP)-dependent (e.g. CHST15, ATP1B1, and SVEP1) and independent components (e.g. ASPN, AKR1B, and LAMA1/LAMB1/LAMC1). Nine causal candidates were identified (TAGLN, WARS, CHST15, CHMP3, COL15A1, DUSP13, MANBA, QSOX2, and SRL). The reverse causal analysis suggested that most AF-associated proteins were affected by the genetic liability to AF. N-terminal prohormone of brain natriuretic peptide improved the prediction of incident AF events close to baseline with further improvements gained by the AF-PRS at all time points. CONCLUSION The AF proteomic signature includes biologically relevant proteins, some of which may be causal. It mainly reflects an NT-proBNP-dependent consequence of the genetic liability to AF. N-terminal prohormone of brain natriuretic peptide is a promising marker for incident AF in the short term, but risk assessment incorporating a PRS may improve long-term risk assessment.
Collapse
Affiliation(s)
- Thorarinn Jonmundsson
- Icelandic Heart Association, Holtasmari 1, Kopavogur 201, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik 101, Iceland
| | | | - Thomas R Austin
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Elisabet A Frick
- Icelandic Heart Association, Holtasmari 1, Kopavogur 201, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik 101, Iceland
| | - Gisli T Axelsson
- Icelandic Heart Association, Holtasmari 1, Kopavogur 201, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik 101, Iceland
| | - Lenore Launer
- Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, Bethesda, MD, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Health Systems and Population Health, University of Washington, Seattle, WA, USA
| | | | | | - Thor Aspelund
- Icelandic Heart Association, Holtasmari 1, Kopavogur 201, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik 101, Iceland
| | - Valur Emilsson
- Icelandic Heart Association, Holtasmari 1, Kopavogur 201, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik 101, Iceland
| | - James S Floyd
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | | | - Vilmundur Gudnason
- Icelandic Heart Association, Holtasmari 1, Kopavogur 201, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik 101, Iceland
| | - Valborg Gudmundsdottir
- Icelandic Heart Association, Holtasmari 1, Kopavogur 201, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik 101, Iceland
| |
Collapse
|
18
|
Tsuji-Tamura K, Ogawa M. FOXO1 promotes endothelial cell elongation and angiogenesis by up-regulating the phosphorylation of myosin light chain 2. Angiogenesis 2023; 26:523-545. [PMID: 37488325 DOI: 10.1007/s10456-023-09884-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 07/11/2023] [Indexed: 07/26/2023]
Abstract
The forkhead box O1 (FOXO1) is an important transcription factor related to proliferation, metabolism, and homeostasis, while the major phenotype of FOXO1-null mice is abnormal vascular morphology, such as vessel enlargement and dilation. In in vitro mouse embryonic stem cell (ESC)-differentiation system, Foxo1-/- vascular endothelial cells (ECs) fail to elongate, and mimic the abnormalities of FOXO1-deficiency in vivo. Here, we identified the PPP1R14C gene as the FOXO1 target genes responsible for elongating using transcriptome analyses in ESC-derived ECs (ESC-ECs), and found that the FOXO1-PPP1R14C-myosin light chain 2 (MLC2) axis is required for EC elongation during angiogenesis. MLC2 is phosphorylated by MLC kinase (MLCK) and dephosphorylated by MLC phosphatase (MLCP). PPP1R14C is an inhibitor of PP1, the catalytic subunit of MLCP. The abnormal morphology of Foxo1-/- ESC-ECs was associated with low level of PPP1R14C and loss of MLC2 phosphorylation, which were reversed by PPP1R14C-introduction. Knockdown of either FOXO1 or PPP1R14C suppressed vascular cord formation and reduced MLC2 phosphorylation in human ECs (HUVECs). The mouse and human PPP1R14C locus possesses an enhancer element containing conserved FOXO1-binding motifs. In vivo chemical inhibition of MLC2 phosphorylation caused dilated vascular structures in mouse embryos. Furthermore, foxo1 or ppp1r14c-knockdown zebrafish exhibited vascular malformations, which were also restored by PPP1R14C-introduction. Mechanistically, FOXO1 suppressed MLCP activity by up-regulating PPP1R14C expression, thereby promoting MLC2 phosphorylation and EC elongation, which are necessary for vascular development. Given the importance of MLC2 phosphorylation in cell morphogenesis, this study may provide novel insights into the role of FOXO1 in control of angiogenesis.
Collapse
Affiliation(s)
- Kiyomi Tsuji-Tamura
- Oral Biochemistry and Molecular Biology, Department of Oral Health Science, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Kita 13, Nishi 7, Kita-Ku, Sapporo, 060-8586, Japan.
| | - Minetaro Ogawa
- Department of Cell Differentiation, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-Ku, Kumamoto, 860-0811, Japan
| |
Collapse
|
19
|
Rai V, Le H, Agrawal DK. Novel mediators regulating angiogenesis in diabetic foot ulcer healing. Can J Physiol Pharmacol 2023; 101:488-501. [PMID: 37459652 DOI: 10.1139/cjpp-2023-0193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
A non-healing diabetic foot ulcer (DFU) is a debilitating clinical problem amounting to socioeconomic and psychosocial burdens. DFUs increase morbidity due to prolonged treatment and mortality in the case of non-treatable ulcers resulting in gangrene and septicemia. The overall amputation rate of the lower extremity with DFU ranges from 3.34% to 42.83%. Wound debridement, antibiotics, applying growth factors, negative pressure wound therapy, hyperbaric oxygen therapy, topical oxygen, and skin grafts are common therapies for DFU. However, recurrence and nonhealing ulcers are still major issues. Chronicity of inflammation, hypoxic environment, poor angiogenesis, and decreased formation of the extracellular matrix (ECM) are common impediments leading to nonhealing patterns of DFUs. Angiogenesis is crucial for wound healing since proper vessel formation facilitates nutrients, oxygen, and immune cells to the ulcer tissue to help in clearing out debris and facilitate healing. However, poor angiogenesis due to decreased expression of angiogenic mediators and matrix formation results in nonhealing and ultimately amputation. Multiple proangiogenic mediators and vascular endothelial growth factor (VEGF) therapy exist to enhance angiogenesis, but the results are not satisfactory. Thus, there is a need to investigate novel pro-angiogenic mediators that can either alone or in combination enhance the angiogenesis and healing of DFUs. In this article, we critically reviewed the existing pro-angiogenic mediators followed by potentially novel factors that might play a regulatory role in promoting angiogenesis and wound healing in DFUs.
Collapse
Affiliation(s)
- Vikrant Rai
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Hoangvi Le
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| |
Collapse
|
20
|
Dylag AM, Misra RS, Bandyopadhyay G, Poole C, Huyck HL, Jehrio MG, Haak J, Deutsch GH, Dvorak C, Olson HM, Paurus V, Katzman PJ, Woo J, Purkerson JM, Adkins JN, Mariani TJ, Clair GC, Pryhuber GS. New insights into the natural history of bronchopulmonary dysplasia from proteomics and multiplexed immunohistochemistry. Am J Physiol Lung Cell Mol Physiol 2023; 325:L419-L433. [PMID: 37489262 PMCID: PMC10642360 DOI: 10.1152/ajplung.00130.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/02/2023] [Accepted: 07/04/2023] [Indexed: 07/26/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a disease of prematurity related to the arrest of normal lung development. The objective of this study was to better understand how proteome modulation and cell-type shifts are noted in BPD pathology. Pediatric human donors aged 1-3 yr were classified based on history of prematurity and histopathology consistent with "healed" BPD (hBPD, n = 3) and "established" BPD (eBPD, n = 3) compared with respective full-term born (n = 6) age-matched term controls. Proteins were quantified by tandem mass spectroscopy with selected Western blot validations. Multiplexed immunofluorescence (MxIF) microscopy was performed on lung sections to enumerate cell types. Protein abundances and MxIF cell frequencies were compared among groups using ANOVA. Cell type and ontology enrichment were performed using an in-house tool and/or EnrichR. Proteomics detected 5,746 unique proteins, 186 upregulated and 534 downregulated, in eBPD versus control with fewer proteins differentially abundant in hBPD as compared with age-matched term controls. Cell-type enrichment suggested a loss of alveolar type I, alveolar type II, endothelial/capillary, and lymphatics, and an increase in smooth muscle and fibroblasts consistent with MxIF. Histochemistry and Western analysis also supported predictions of upregulated ferroptosis in eBPD versus control. Finally, several extracellular matrix components mapping to angiogenesis signaling pathways were altered in eBPD. Despite clear parsing by protein abundance, comparative MxIF analysis confirms phenotypic variability in BPD. This work provides the first demonstration of tandem mass spectrometry and multiplexed molecular analysis of human lung tissue for critical elucidation of BPD trajectory-defining factors into early childhood.NEW & NOTEWORTHY We provide new insights into the natural history of bronchopulmonary dysplasia in donor human lungs after the neonatal intensive care unit hospitalization. This study provides new insights into how the proteome and histopathology of BPD changes in early childhood, uncovering novel pathways for future study.
Collapse
Affiliation(s)
- Andrew M Dylag
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
| | - Ravi S Misra
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
| | - Gautam Bandyopadhyay
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
| | - Cory Poole
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
| | - Heidie L Huyck
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
| | - Matthew G Jehrio
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
| | - Jeannie Haak
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
| | - Gail H Deutsch
- Department of Laboratory Medicine and Pathology, University of Washington, University of Washington, Seattle, Washington, United States
| | - Carly Dvorak
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
| | - Heather M Olson
- Pacific Northwest National Laboratories, Richland, Washington, United States
| | - Vanessa Paurus
- Pacific Northwest National Laboratories, Richland, Washington, United States
| | - Philip J Katzman
- Department of Pathology, University of Rochester Medical Center, Rochester, New York, United States
| | - Jongmin Woo
- Pacific Northwest National Laboratories, Richland, Washington, United States
| | - Jeffrey M Purkerson
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
| | - Joshua N Adkins
- Pacific Northwest National Laboratories, Richland, Washington, United States
| | - Thomas J Mariani
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
| | - Geremy C Clair
- Pacific Northwest National Laboratories, Richland, Washington, United States
| | - Gloria S Pryhuber
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
| |
Collapse
|
21
|
Shen J, Liu Y, Teng X, Jin L, Feng L, Sun X, Zhao F, Huang B, Zhong J, Chen Y, Wang L. Spatial Transcriptomics of Aging Rat Ovaries Reveals Unexplored Cell Subpopulations with Reduced Antioxidative Defense. Gerontology 2023; 69:1315-1329. [PMID: 37717573 DOI: 10.1159/000533922] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 08/29/2023] [Indexed: 09/19/2023] Open
Abstract
INTRODUCTION Ovarian aging is characterized by a gradual decline in quantity and quality of oocytes and lower chance of fertility. Better understanding the genetic modulation during ovarian aging can further address available treatment options for aging-related ovarian diseases and fertility preservation. METHODS A novel technique spatial transcriptomics (ST) was used to investigate the spatial transcriptome features of rat ovaries. Transcriptomes from ST spots in the young and aged ovaries were clustered using differentially expressed genes. These data were analyzed to determine the spatial organization of age-induced heterogeneity and potential mechanisms underlying ovarian aging. RESULTS In this study, ST technology was applied to profile the comprehensive spatial imaging in young and aged rat ovary. Fifteen ovarian cell clusters with distinct gene-expression signatures were identified. The gene expression dynamics of granulosa cell clusters revealed three sub-types with sequential developmental stages. Aged ovary showed a significant decrease in the number of granulosa cells from the antral follicle. Besides, a remarkable rearrangement of interstitial gland cells was detected in aging ovary. Further analysis of aging-associated transcriptional changes revealed that the disturbance of oxidative pathway was a crucial factor in ovarian aging. CONCLUSIONS This study firstly described an aging-related spatial transcriptome changes in ovary and identified the potential targets for prevention of ovarian aging. These data may provide the basis for further investigations of the diagnosis and treatment of aging-related ovarian disorders.
Collapse
Affiliation(s)
- Jiayu Shen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China,
| | - Yuanyuan Liu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xinyuan Teng
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Ligui Jin
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Lin Feng
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiwen Sun
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Fengdong Zhao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Bao Huang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinjie Zhong
- Department of Basic Medicine Sciences, and Department of Obstetrics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yingying Chen
- Department of Basic Medicine Sciences, and Department of Obstetrics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liquan Wang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
22
|
Arolkar G, Kumar SK, Wang H, Gonzalez KM, Kumar S, Bishnoi B, Rios Coronado PE, Woo YJ, Red-Horse K, Das S. Dedifferentiation and Proliferation of Artery Endothelial Cells Drive Coronary Collateral Development in Mice. Arterioscler Thromb Vasc Biol 2023; 43:1455-1477. [PMID: 37345524 PMCID: PMC10364966 DOI: 10.1161/atvbaha.123.319319] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 06/08/2023] [Indexed: 06/23/2023]
Abstract
BACKGROUND Collateral arteries act as natural bypasses which reroute blood flow to ischemic regions and facilitate tissue regeneration. In an injured heart, neonatal artery endothelial cells orchestrate a systematic series of cellular events, which includes their outward migration, proliferation, and coalescence into fully functional collateral arteries. This process, called artery reassembly, aids complete cardiac regeneration in neonatal hearts but is absent in adults. The reason for this age-dependent disparity in artery cell response is completely unknown. In this study, we investigated if regenerative potential of coronary arteries is dictated by their ability to dedifferentiate. METHODS Single-cell RNA sequencing of coronary endothelial cells was performed to identify differences in molecular profiles of neonatal and adult endothelial cells in mice. Findings from this in silico analyses were confirmed with in vivo experiments using genetic lineage tracing, whole organ immunostaining, confocal imaging, and cardiac functional assays in mice. RESULTS Upon coronary occlusion, neonates showed a significant increase in actively cycling artery cells and expressed prominent dedifferentiation markers. Data from in silico pathway analyses and in vivo experiments suggested that upon myocardial infarction, cell cycle reentry of preexisting neonatal artery cells, the subsequent collateral artery formation, and recovery of cardiac function are dependent on arterial VegfR2 (vascular endothelial growth factor receptor-2). This subpopulation of dedifferentiated and proliferating artery cells was absent in nonregenerative postnatal day 7 or adult hearts. CONCLUSIONS These data indicate that adult artery endothelial cells fail to drive collateral artery development due to their limited ability to dedifferentiate and proliferate.
Collapse
Affiliation(s)
- Gauri Arolkar
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, India (G.A., S.K.K., S.K., B.B., S.D.)
| | - Sneha K. Kumar
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, India (G.A., S.K.K., S.K., B.B., S.D.)
| | - Hanjay Wang
- Department of Cardiothoracic Surgery (H.W., Y.J.W.), Stanford University School of Medicine, CA
| | - Karen M. Gonzalez
- Institute for Stem Cell Biology and Regenerative Medicine (K.M.G., K.R.-H.), Stanford University School of Medicine, CA
- Department of Biology (K.M.G., K.R.-H.), Stanford University, CA
| | - Suraj Kumar
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, India (G.A., S.K.K., S.K., B.B., S.D.)
| | - Bhavnesh Bishnoi
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, India (G.A., S.K.K., S.K., B.B., S.D.)
| | | | - Y. Joseph Woo
- Department of Cardiothoracic Surgery (H.W., Y.J.W.), Stanford University School of Medicine, CA
| | - Kristy Red-Horse
- Institute for Stem Cell Biology and Regenerative Medicine (K.M.G., K.R.-H.), Stanford University School of Medicine, CA
- Department of Biology (K.M.G., K.R.-H.), Stanford University, CA
- Howard Hughes Medical Institute, Chevy Chase, MD (K.R.-H.)
| | - Soumyashree Das
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, India (G.A., S.K.K., S.K., B.B., S.D.)
| |
Collapse
|
23
|
Asciak L, Gilmour L, Williams JA, Foster E, Díaz-García L, McCormick C, Windmill JFC, Mulvana HE, Jackson-Camargo JC, Domingo-Roca R. Investigating multi-material hydrogel three-dimensional printing for in vitro representation of the neo-vasculature of solid tumours: a comprehensive mechanical analysis and assessment of nitric oxide release from human umbilical vein endothelial cells. ROYAL SOCIETY OPEN SCIENCE 2023; 10:230929. [PMID: 37593713 PMCID: PMC10427827 DOI: 10.1098/rsos.230929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 07/25/2023] [Indexed: 08/19/2023]
Abstract
Many solid tumours (e.g. sarcoma, carcinoma and lymphoma) form a disorganized neo-vasculature that initiates uncontrolled vessel formation to support tumour growth. The complexity of these environments poses a significant challenge for tumour medicine research. While animal models are commonly used to address some of these challenges, they are time-consuming and raise ethical concerns. In vitro microphysiological systems have been explored as an alternative, but their production typically requires multi-step lithographic processes that limit their production. In this work, a novel approach to rapidly develop multi-material tissue-mimicking, cell-compatible platforms able to represent the complexity of a solid tumour's neo-vasculature is investigated via stereolithography three-dimensional printing. To do so, a series of acrylate resins that yield covalently photo-cross-linked hydrogels with healthy and diseased mechano-acoustic tissue-mimicking properties are designed and characterized. The potential viability of these materials to displace animal testing in preclinical research is assessed by studying the morphology, actin expression, focal adhesions and nitric oxide release of human umbilical vein endothelial cells. These materials are exploited to produce a simplified multi-material three-dimensional printed model of the neo-vasculature of a solid tumour, demonstrating the potential of our approach to replicate the complexity of solid tumours in vitro without the need for animal testing.
Collapse
Affiliation(s)
- Lisa Asciak
- Department of Electronic and Electrical Engineering, University of Strathclyde, Glasgow, UK
| | - Lauren Gilmour
- Department of Biomedical Engineering, University of Strathclyde, Glasgow, UK
| | | | - Euan Foster
- Department of Electronic and Electrical Engineering, University of Strathclyde, Glasgow, UK
| | - Lara Díaz-García
- Department of Electronic and Electrical Engineering, University of Strathclyde, Glasgow, UK
| | | | - James F. C. Windmill
- Department of Electronic and Electrical Engineering, University of Strathclyde, Glasgow, UK
| | - Helen E. Mulvana
- Department of Biomedical Engineering, University of Strathclyde, Glasgow, UK
| | | | - Roger Domingo-Roca
- Department of Electronic and Electrical Engineering, University of Strathclyde, Glasgow, UK
| |
Collapse
|
24
|
Trinh LT, Osipovich AB, Liu B, Shrestha S, Cartailler JP, Wright CVE, Magnuson MA. Single-Cell RNA Sequencing of Sox17-Expressing Lineages Reveals Distinct Gene Regulatory Networks and Dynamic Developmental Trajectories. Stem Cells 2023; 41:643-657. [PMID: 37085274 PMCID: PMC10465087 DOI: 10.1093/stmcls/sxad030] [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: 01/24/2023] [Accepted: 04/04/2023] [Indexed: 04/23/2023]
Abstract
During early embryogenesis, the transcription factor SOX17 contributes to hepato-pancreato-biliary system formation and vascular-hematopoietic emergence. To better understand Sox17 function in the developing endoderm and endothelium, we developed a dual-color temporal lineage-tracing strategy in mice combined with single-cell RNA sequencing to analyze 6934 cells from Sox17-expressing lineages at embryonic days 9.0-9.5. Our analyses showed 19 distinct cellular clusters combined from all 3 germ layers. Differential gene expression, trajectory and RNA-velocity analyses of endothelial cells revealed a heterogenous population of uncommitted and specialized endothelial subtypes, including 2 hemogenic populations that arise from different origins. Similarly, analyses of posterior foregut endoderm revealed subsets of hepatic, pancreatic, and biliary progenitors with overlapping developmental potency. Calculated gene-regulatory networks predict gene regulons that are dominated by cell type-specific transcription factors unique to each lineage. Vastly different Sox17 regulons found in endoderm versus endothelial cells support the differential interactions of SOX17 with other regulatory factors thereby enabling lineage-specific regulatory actions.
Collapse
Affiliation(s)
- Linh T Trinh
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
- Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, USA
- Program in Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Anna B Osipovich
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
- Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, USA
| | - Bryan Liu
- College of Arts and Sciences, Vanderbilt University, Nashville, TN, USA
| | - Shristi Shrestha
- Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, USA
| | | | - Christopher V E Wright
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
- Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, USA
- Program in Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Mark A Magnuson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
- Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, USA
- Program in Developmental Biology, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
25
|
Colijn S, Nambara M, Stratman AN. Identification of overlapping and distinct mural cell populations during early embryonic development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.03.535476. [PMID: 37066365 PMCID: PMC10104062 DOI: 10.1101/2023.04.03.535476] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Mural cells are an essential perivascular cell population that associate with blood vessels and contribute to vascular stabilization and tone. In the embryonic zebrafish vasculature, pdgfrb and tagln are commonly used as markers for identifying pericytes and vascular smooth muscle cells (vSMCs). However, the expression patterns of these markers used in tandem have not been fully described. Here, we used the Tg(pdgfrb:Gal4FF; UAS:RFP) and Tg(tagln:NLS-EGFP) transgenic lines to identify single- and double-positive perivascular populations in the cranial, axial, and intersegmental vessels between 1 and 5 days post-fertilization. From this comparative analysis, we discovered two novel regions of tagln-positive cell populations that have the potential to function as mural cell precursors. Specifically, we found that the hypochord- a reportedly transient structure-contributes to tagln-positive cells along the dorsal aorta. We also identified a unique sclerotome-derived mural cell progenitor population that resides along the midline between the neural tube and notochord and contributes to intersegmental vessel mural cell coverage. Together, our findings highlight the variability and versatility of tracking pdgfrb and tagln expression in mural cells of the developing zebrafish embryo.
Collapse
Affiliation(s)
- Sarah Colijn
- Department of Cell Biology and Physiology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110
| | - Miku Nambara
- Department of Cell Biology and Physiology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110
| | - Amber N. Stratman
- Department of Cell Biology and Physiology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110
| |
Collapse
|
26
|
Elhadad S, Redmond D, Tan A, Huang J, Rodriguez BL, Racine-Brzostek SE, Subrahmanian S, Ahamed J, Laurence J. Defibrotide mitigates endothelial cell injury induced by plasmas from patients with COVID-19 and related vasculopathies. Thromb Res 2023; 225:47-56. [PMID: 37001283 PMCID: PMC10033153 DOI: 10.1016/j.thromres.2023.03.009] [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: 01/04/2023] [Revised: 03/14/2023] [Accepted: 03/20/2023] [Indexed: 03/24/2023]
Abstract
Background and objectives COVID-19 progression is characterized by systemic small vessel arterial and venous thrombosis. Microvascular endothelial cell (MVEC) activation and injury, platelet activation, and histopathologic features characteristic of acute COVID-19 also describe certain thrombotic microangiopathies, including atypical hemolytic-uremic syndrome (aHUS), thrombotic thrombocytopenic purpura (TTP), and hematopoietic stem cell transplant (HSCT)-associated veno-occlusive disease (VOD). We explored the effect of clinically relevant doses of defibrotide, approved for HSCT-associated VOD, on MVEC activation/injury. Methods Human dermal MVEC were exposed to plasmas from patients with acute TMAs or acute COVID-19 in the presence and absence of defibrotide (5 μg/ml) and caspase 8, a marker of EC activation and apoptosis, was assessed. RNAseq was used to explore potential mechanisms of defibrotide activity. Results Defibrotide suppressed TMA plasma-induced caspase 8 activation in MVEC (mean 60.2 % inhibition for COVID-19; p = 0.0008). RNAseq identified six major cellular pathways associated with defibrotide's alteration of COVID-19-associated MVEC changes: TNF-α signaling; IL-17 signaling; extracellular matrix (ECM)-EC receptor and platelet receptor interactions; ECM formation; endothelin activity; and fibrosis. Communications across these pathways were revealed by STRING analyses. Forty transcripts showing the greatest changes induced by defibrotide in COVID-19 plasma/MVEC cultures included: claudin 14 and F11R (JAM), important in maintaining EC tight junctions; SOCS3 and TNFRSF18, involved in suppression of inflammation; RAMP3 and transgelin, which promote angiogenesis; and RGS5, which regulates caspase activation and apoptosis. Conclusion Our data, in the context of a recent clinical trial in severe COVID-19, suggest benefits to further exploration of defibrotide and these pathways in COVID-19 and related endotheliopathies.
Collapse
Affiliation(s)
- Sonia Elhadad
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, United States of America
| | - David Redmond
- Division of Regenerative Medicine, Hartman Institute fort Therapeutic Organ Regeneration, Ansary Stem Cell Institute, United States of America
| | - Adrian Tan
- Genomics Resources Core Facility, Weill Cornell Medicine, United States of America
| | - Jenny Huang
- Division of Regenerative Medicine, Hartman Institute fort Therapeutic Organ Regeneration, Ansary Stem Cell Institute, United States of America
| | - Beatriz Lorenzo Rodriguez
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, United States of America
| | | | - Sandeep Subrahmanian
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States of America
| | - Jasimuddin Ahamed
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States of America
| | - Jeffrey Laurence
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, United States of America.
| |
Collapse
|
27
|
Du TY, Gao YX, Zheng YS. Identification of key genes related to immune infiltration in cirrhosis via bioinformatics analysis. Sci Rep 2023; 13:1876. [PMID: 36725885 PMCID: PMC9892033 DOI: 10.1038/s41598-022-26794-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 12/20/2022] [Indexed: 02/03/2023] Open
Abstract
Cirrhosis is the most common subclass of liver disease worldwide and correlated to immune infiltration. However, the immune-related molecular mechanism underlying cirrhosis remains obscure. Two gene expression profiles GSE89377 and GSE139602 were investigated to identify differentially expressed genes (DEGs) related to cirrhosis. Enrichment analysis for DEGs was conducted. Next, the immune infiltration of DEGs was evaluated using CIBERSORT algorithm. The hub DEGs with tight connectivity were identified using the String and Cytoscape databases, and the expression difference of these hub genes between normal liver and cirrhosis samples was determined. Moreover, in order to evaluate the discriminatory ability of hub genes and obtained the area under the receiver operating characteristic curve values in the GSE89377 and GSE139602 datasets. Finally, the association between hub DEGs and immune cell infiltration was explored by Spearman method. Among the 299 DEGs attained, 136 were up-regulated and 163 were down-regulated. Then the enrichment function analysis of DEGs and CIBERSORT algorithm showed significant enrichment in immune and inflammatory responses. And four hub DEGs (ACTB, TAGLN, VIM, SOX9) were identified, which also showed a diagnostic value in the GSE89377 and GSE 139,602 datasets. Finally, the immune infiltration analysis indicated that, these hub DEGs were highly related to immune cells. This study revealed key DEGs involved in inflammatory immune responses of cirrhosis, which could be used as biomarkers for diagnosis or therapeutic targets of cirrhosis.
Collapse
Affiliation(s)
- Tong-Yue Du
- Department of Critical Care Medicine, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, No.-1-1, Zhongfu Road, Nanjing, 210003, China
| | - Ya-Xian Gao
- Department of Critical Care Medicine, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, No.-1-1, Zhongfu Road, Nanjing, 210003, China
| | - Yi-Shan Zheng
- Department of Critical Care Medicine, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, No.-1-1, Zhongfu Road, Nanjing, 210003, China.
| |
Collapse
|
28
|
Dual genome-wide coding and lncRNA screens in neural induction of induced pluripotent stem cells. CELL GENOMICS 2022; 2:100177. [PMID: 36381608 PMCID: PMC9648144 DOI: 10.1016/j.xgen.2022.100177] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Human chromosomes are pervasively transcribed, but systematic understanding of coding and lncRNA genome function in cell differentiation is lacking. Using CRISPR interference (CRISPRi) in human induced pluripotent stem cells, we performed dual genome-wide screens - assessing 18,905 protein-coding and 10,678 lncRNA loci - and identified 419 coding and 201 lncRNA genes that regulate neural induction. Integrative analyses revealed distinct properties of coding and lncRNA genome function, including a 10-fold enrichment of lncRNA genes for roles in differentiation compared to proliferation. Further, we applied Perturb-seq to obtain granular insights into neural induction phenotypes. While most coding hits stalled or aborted differentiation, lncRNA hits were enriched for the genesis of diverse cellular states, including those outside the neural lineage. In addition to providing a rich resource (danlimlab.shinyapps.io/dualgenomewide) for understanding coding and lncRNA gene function in development, these results indicate that the lncRNA genome regulates lineage commitment in a manner fundamentally distinct from coding genes.
Collapse
|
29
|
Physiology and Proteomic Basis of Lung Adaptation to High-Altitude Hypoxia in Tibetan Sheep. Animals (Basel) 2022; 12:ani12162134. [PMID: 36009723 PMCID: PMC9405401 DOI: 10.3390/ani12162134] [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: 06/30/2022] [Revised: 08/09/2022] [Accepted: 08/16/2022] [Indexed: 11/25/2022] Open
Abstract
Simple Summary As an indigenous animal living in the Tibetan plateau, the Tibetan sheep is well adapted to high-altitude hypoxia, and the lungs play an important role in overcoming the hypoxic environment. To reveal the physiological and proteomic basis of Tibetan sheep lungs during their adaptation to hypoxia, we studied the lungs of Tibetan sheep at different altitudes using light and electron microscopy and proteome sequencing. The results showed that in the lungs of Tibetan sheep occurred a series of physiological changes with increasing altitude, and some important proteins and pathways identified by proteome sequencing further support these physiology findings. These changes at the physiological and molecular levels may facilitate the adaptation of Tibetan sheep to high-altitude hypoxia. In conclusion, these findings may provide a reference for the prevention of altitude sickness in humans. Abstract The Tibetan sheep is an indigenous animal of the Tibetan plateau, and after a long period of adaptation have adapted to high-altitude hypoxia. Many physiological changes occur in Tibetan sheep as they adapt to high-altitude hypoxia, especially in the lungs. To reveal the physiological changes and their molecular mechanisms in the lungs of Tibetan sheep during adaptation to high altitudes, we selected Tibetan sheep from three altitudes (2500 m, 3500 m, and 4500 m) and measured blood-gas indicators, observed lung structures, and compared lung proteome changes. The results showed that the Tibetan sheep increased their O2-carrying capacity by increasing the hemoglobin (Hb) concentration and Hematocrit (Hct) at an altitude of 3500 m. While at altitude of 4500 m, Tibetan sheep decreased their Hb concentration and Hct to avoid pulmonary hypertension and increased the efficiency of air-blood exchange and O2 transfer by increasing the surface area of gas exchange and half-saturation oxygen partial pressure. Besides these, some important proteins and pathways related to gas transport, oxidative stress, and angiogenesis identified by proteome sequencing further support these physiology findings, including HBB, PRDX2, GPX1, GSTA1, COL14A1, and LTBP4, etc. In conclusion, the lungs of Tibetan sheep are adapted to different altitudes by different strategies; these findings are valuable for understanding the basis of hypoxic adaptation in Tibetan sheep.
Collapse
|
30
|
Tsuji-Tamura K, Tamura M. Basic fibroblast growth factor uniquely stimulates quiescent vascular smooth muscle cells and induces proliferation and dedifferentiation. FEBS Lett 2022; 596:1686-1699. [PMID: 35363891 DOI: 10.1002/1873-3468.14345] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 03/23/2022] [Accepted: 03/23/2022] [Indexed: 11/11/2022]
Abstract
Blood vessels normally remain stable over the long-term. However, in atherosclerosis, vascular cells leave the quiescent state and enter an activated state. Here, we investigated the factors that trigger breakage of the quiescent state by screening growth factors and cytokines using a vascular smooth muscle cell (SMC) line and an endothelial cell (EC) line. Despite known functions of the tested factors, only basic fibroblast growth factor (bFGF) was identified as a potent trigger of quiescence breakage in SMCs, but not ECs. bFGF disrupted tight SMC-monolayers, and caused morphological changes, proliferation and dedifferentiation. Human primary SMCs, but not ECs, also showed similar results. Aberrant SMC-proliferation is a critical histological event in atherosclerosis. We thus provide further insights into the role of bFGF in vascular pathobiology.
Collapse
Affiliation(s)
- Kiyomi Tsuji-Tamura
- Oral Biochemistry and Molecular Biology, Department of Oral Health Science, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Kita 13, Nishi 7, Kita-ku, Sapporo, 060-8586, Japan
| | - Masato Tamura
- Oral Biochemistry and Molecular Biology, Department of Oral Health Science, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Kita 13, Nishi 7, Kita-ku, Sapporo, 060-8586, Japan
| |
Collapse
|