1
|
Wang S, Hu Y, Wang Y, Song Y, Liang D, Yin J, Li Y, Yang W, Zhang D. Joint Analysis of Multiple Omics to Describe the Biological Characteristics of Resistant Hypertension. J Clin Hypertens (Greenwich) 2025; 27:e14961. [PMID: 39716980 PMCID: PMC11774085 DOI: 10.1111/jch.14961] [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: 09/06/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/25/2024]
Abstract
Resistant hypertension (RH) may cause severe target organ damage and poses significant challenges in the field of hypertension prevention and treatment. Mining biological characteristics is crucial for exploring the pathogenesis of RH and for early diagnosis and treatment. Although several single-omics studies have been conducted on RH, its complex pathogenesis has only been partially elucidated. In this study, metabolomics, proteomics, and transcriptomics were jointly analyzed in healthy subjects and patients with hypertension and RH. The multi-omics analysis found that differential substances of RH were enriched in the HIF-1 signaling pathway and that differential substances such as ascorbic acid, reduced glutathione (GSH), choline, citric acid, transferrin receptor (TfR), Egl-9 family hypoxia-inducible factor 2 (EGLN2), and glutathione peroxidase 1 (GPX1) were screened out. The results of intergroup comparisons were as follows: RH versus N: ascorbic acid (Fold Change (FC):0.42, p < 0.01), GSH (FC:0.65, p < 0.05), choline (FC:1.32, p < 0.05), citric acid (FC:0.48, p < 0.001), TfR (FC2.32, p < 0.001), GPX1 (FC:16.02, p < 0.001), EGLN2 (FC:0.76, p < 0.001); RH versus EH: ascorbic acid (FC:0.52, p < 0.05), GSH (FC:0.55, p < 0.05), choline (FC:1.28, p < 0.05), citric acid (FC:0.59, p < 0.001), TfR (FC:1.71, p < 0.001), GPX1 (FC:2.11, p < 0.05), EGLN2 (FC:0.76, p < 0.05). These differential substances may reflect the biology of RH. This study provides multi-omics analysis for a deeper understanding of the complex molecular characteristics of RH, providing new insights into the pathogenesis, early diagnosis, and precise treatment of the disease.
Collapse
Affiliation(s)
- Shanshan Wang
- Innovative Institute of Chinese Medicine and PharmacyShandong University of Traditional Chinese MedicineJinanChina
- Department of CardiologyThe Fifth People's Hospital of JinanJinanChina
| | - Yuanlong Hu
- First Faculty of Clinical MedicineShandong University of Traditional Chinese MedicineJinanChina
| | - Yuqi Wang
- First Faculty of Clinical MedicineShandong University of Traditional Chinese MedicineJinanChina
| | - Yueyue Song
- Innovative Institute of Chinese Medicine and PharmacyShandong University of Traditional Chinese MedicineJinanChina
| | - Dan Liang
- Innovative Institute of Chinese Medicine and PharmacyShandong University of Traditional Chinese MedicineJinanChina
| | - Jiufeng Yin
- Innovative Institute of Chinese Medicine and PharmacyShandong University of Traditional Chinese MedicineJinanChina
| | - Yunlun Li
- Innovative Institute of Chinese Medicine and PharmacyShandong University of Traditional Chinese MedicineJinanChina
- Shandong Engineering Laboratory of Traditional Chinese Medicine Precise Therapy for Cardiovascular DiseasesJinanChina
- Department of CardiologyAffiliated Hospital of Shandong University of Traditional Chinese MedicineJinanChina
| | - Wenqing Yang
- Innovative Institute of Chinese Medicine and PharmacyShandong University of Traditional Chinese MedicineJinanChina
- Shandong Engineering Laboratory of Traditional Chinese Medicine Precise Therapy for Cardiovascular DiseasesJinanChina
| | - Dan Zhang
- Experimental CenterShandong University of Traditional Chinese MedicineJinanChina
- Key Laboratory of Traditional Chinese Medicine Classical TheoryMinistry of EducationShandong University of Traditional Chinese MedicineJinanChina
| |
Collapse
|
2
|
Shaydakov ME, Diaz JA, Eklöf B, Lurie F. Venous valve hypoxia as a possible mechanism of deep vein thrombosis: a scoping review. INT ANGIOL 2024; 43:309-322. [PMID: 38864688 DOI: 10.23736/s0392-9590.24.05170-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2024]
Abstract
INTRODUCTION The pathogenesis of deep vein thrombosis (DVT) has been explained by an interplay between a changed blood composition, vein wall alteration, and blood flow abnormalities. A comprehensive investigation of these components of DVT pathogenesis has substantially promoted our understanding of thrombogenesis in the venous system. Meanwhile, the process of DVT initiation remains obscure. This systematic review aims to collect, analyze, and synthesize the published evidence to propose hypoxia as a possible trigger of DVT. EVIDENCE ACQUISITION An exhaustive literature search was conducted across multiple electronic databased including PubMed, EMBASE, Scopus, and Web of Science to identify studies pertinent to the research hypothesis. The search was aimed at exploring the connection between hypoxia, reoxygenation, and the initiation of deep vein thrombosis (DVT). The following key words were used: "deep vein thrombosis," "venous thrombosis," "venous thromboembolism," "hypoxia," "reoxygenation," "venous valve," and "venous endothelium." Reviews, case reports, editorials, and letters were excluded. EVIDENCE SYNTHESIS Based on the systematic search outcome, 156 original papers relevant to the issue were selected for detailed review. These studies encompassed a range of experimental and observational clinical research, focusing on various aspects of DVT, including the anatomical, physiological, and cellular bases of the disease. A number of studies suggested limitations in the traditional understanding of Virchow's triad as an acceptable explanation for DVT initiation. Emerging evidence points to more complex interactions and additional factors that may be critical in the early stages of thrombogenesis. The role of venous valves has been recognized but remains underappreciated, with several studies indicating that these sites may act as primary loci for thrombus formation. A collection of studies describes the effects of hypoxia on venous endothelial cells at the cellular and molecular levels. Hypoxia influences several pathways that regulate endothelial cell permeability, inflammatory response, and procoagulation activity, underpinning the endothelial dysfunction noted in DVT. CONCLUSIONS Hypoxia of the venous valve may serve as an independent hypothesis to outline the DVT triggering process. Further research projects in this field may discover new molecular pathways responsible for the disease and suggest new therapeutic targets.
Collapse
Affiliation(s)
- Maxim E Shaydakov
- Division of Vascular Surgery, University of Pittsburgh Medical Center, Pittsburg, PA, USA -
| | - Jose A Diaz
- Division of Surgical Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Fedor Lurie
- Jobst Vascular Institute, ProMedica Health System, Toledo, OH, USA
| |
Collapse
|
3
|
Forbes S, Bettenay S, Meertens NM, Wildermuth BE, Wildermuth K, Mueller RS. Diascopy and histopathological evaluation of nonblanching erythematous dermatoses in dogs. Vet Dermatol 2024; 35:255-262. [PMID: 38111018 DOI: 10.1111/vde.13230] [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: 02/12/2023] [Revised: 09/19/2023] [Accepted: 12/06/2023] [Indexed: 12/20/2023]
Abstract
BACKGROUND Diascopy is a point-of-care diagnostic test used to differentiate skin erythema due to vascular dilation from haemorrhage. In the veterinary literature, only a handful of diseases have been described to be associated with a negative (nonblanching) diascopy result, and histological investigation of haemorrhage has been inconsistent. OBJECTIVES Retrospective study to undertake a histopathological investigation of canine, nonblanching erythematous dermatoses for the presence or absence of haemorrhage and vascular changes. MATERIALS AND METHODS Skin biopsies from dogs presented with moderate-to-severe nonblanching erythema were evaluated histologically. Additionally, clinical data about each patient were analysed. RESULTS Twenty cases were identified with nonblanching erythema. Diagnoses included vasculopathy (n = 6), canine eosinophilic dermatitis (n = 3), cutaneous epitheliotropic T-cell lymphoma (n = 2), and one case each of sterile granuloma and pyogranuloma syndrome, German shepherd dog pyoderma, multiple mast cell tumours, haemangiosarcoma, exfoliative cutaneous lupus erythematosus, canine leishmaniosis with sebaceous adenitis, sebaceous adenitis with concurrent dermatophytosis, calcinosis cutis and canine atopic dermatitis with insect-bite reaction. One or more vascular changes were present in all 20 cases and included perivascular oedema, endothelial swelling and neutrophilic infiltration of vessel walls. Haemorrhage was identified in 17 of 20 cases (85%). Three cases without dermal haemorrhage were calcinosis cutis, sebaceous adenitis with dermatophytosis and canine atopic dermatitis with insect-bite reaction. CONCLUSIONS AND CLINICAL RELEVANCE Negative diascopy was associated with haemorrhage and vascular pathological findings in the majority of cases, yet not all. Haemorrhage was identified histologically in all diseases previously reported as nonblanching as well as in a few additional diseases.
Collapse
Affiliation(s)
| | - Sonya Bettenay
- Centre for Clinical Veterinary Medicine, University of Munich, Munich, Germany
| | | | | | | | - Ralf S Mueller
- Centre for Clinical Veterinary Medicine, University of Munich, Munich, Germany
| |
Collapse
|
4
|
Li C, Jiang M, Fang Z, Chen Z, Li L, Liu Z, Wang J, Yin X, Wang J, Wu M. Current evidence of synaptic dysfunction after stroke: Cellular and molecular mechanisms. CNS Neurosci Ther 2024; 30:e14744. [PMID: 38727249 PMCID: PMC11084978 DOI: 10.1111/cns.14744] [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] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/07/2024] [Accepted: 04/10/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND Stroke is an acute cerebrovascular disease in which brain tissue is damaged due to sudden obstruction of blood flow to the brain or the rupture of blood vessels in the brain, which can prompt ischemic or hemorrhagic stroke. After stroke onset, ischemia, hypoxia, infiltration of blood components into the brain parenchyma, and lysed cell fragments, among other factors, invariably increase blood-brain barrier (BBB) permeability, the inflammatory response, and brain edema. These changes lead to neuronal cell death and synaptic dysfunction, the latter of which poses a significant challenge to stroke treatment. RESULTS Synaptic dysfunction occurs in various ways after stroke and includes the following: damage to neuronal structures, accumulation of pathologic proteins in the cell body, decreased fluidity and release of synaptic vesicles, disruption of mitochondrial transport in synapses, activation of synaptic phagocytosis by microglia/macrophages and astrocytes, and a reduction in synapse formation. CONCLUSIONS This review summarizes the cellular and molecular mechanisms related to synapses and the protective effects of drugs or compounds and rehabilitation therapy on synapses in stroke according to recent research. Such an exploration will help to elucidate the relationship between stroke and synaptic damage and provide new insights into protecting synapses and restoring neurologic function.
Collapse
Affiliation(s)
- Chuan Li
- Department of Medical LaboratoryAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
| | - Min Jiang
- Jiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| | - Zhi‐Ting Fang
- Department of Pathophysiology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Zhiying Chen
- Department of NeurologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
| | - Li Li
- Department of Intensive Care UnitThe Affiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
| | - Ziying Liu
- Department of Medical LaboratoryAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Xiaoping Yin
- Department of NeurologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
| | - Jian Wang
- Department of Human Anatomy, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Moxin Wu
- Department of Medical LaboratoryAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
- Jiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| |
Collapse
|
5
|
Gwozdzinski L, Pieniazek A, Gwozdzinski K. Factors Influencing Venous Remodeling in the Development of Varicose Veins of the Lower Limbs. Int J Mol Sci 2024; 25:1560. [PMID: 38338837 PMCID: PMC10855638 DOI: 10.3390/ijms25031560] [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: 12/16/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
One of the early symptoms of chronic venous disease (CVD) is varicose veins (VV) of the lower limbs. There are many etiological environmental factors influencing the development of chronic venous insufficiency (CVI), although genetic factors and family history of the disease play a key role. All these factors induce changes in the hemodynamic in the venous system of the lower limbs leading to blood stasis, hypoxia, inflammation, oxidative stress, proteolytic activity of matrix metalloproteinases (MMPs), changes in microcirculation and, consequently, the remodeling of the venous wall. The aim of this review is to present current knowledge on CVD, including the pathophysiology and mechanisms related to vein wall remodeling. Particular emphasis has been placed on describing the role of inflammation and oxidative stress and the involvement of extracellular hemoglobin as pathogenetic factors of VV. Additionally, active substances used in the treatment of VV were discussed.
Collapse
Affiliation(s)
- Lukasz Gwozdzinski
- Department of Pharmacology and Toxicology, Medical University of Lodz, 90-752 Lodz, Poland;
| | - Anna Pieniazek
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland;
| | - Krzysztof Gwozdzinski
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland;
| |
Collapse
|
6
|
Jeong HJ, Nam H, Kim JS, Cho S, Park HH, Cho YS, Jeon H, Jang J, Lee SJ. Dragging 3D printing technique controls pore sizes of tissue engineered blood vessels to induce spontaneous cellular assembly. Bioact Mater 2024; 31:590-602. [PMID: 37876874 PMCID: PMC10593581 DOI: 10.1016/j.bioactmat.2023.07.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 06/23/2023] [Accepted: 07/24/2023] [Indexed: 10/26/2023] Open
Abstract
To date, several off-the-shelf products such as artificial blood vessel grafts have been reported and clinically tested for small diameter vessel (SDV) replacement. However, conventional artificial blood vessel grafts lack endothelium and, thus, are not ideal for SDV transplantation as they can cause thrombosis. In addition, a successful artificial blood vessel graft for SDV must have sufficient mechanical properties to withstand various external stresses. Here, we developed a spontaneous cellular assembly SDV (S-SDV) that develops without additional intervention. By improving the dragging 3D printing technique, SDV constructs with free-form, multilayers and controllable pore size can be fabricated at once. Then, The S-SDV filled in the natural polymer bioink containing human umbilical vein endothelial cells (HUVECs) and human aorta smooth muscle cells (HAoSMCs). The endothelium can be induced by migration and self-assembly of endothelial cells through pores of the SDV construct. The antiplatelet adhesion of the formed endothelium on the luminal surface was also confirmed. In addition, this S-SDV had sufficient mechanical properties (burst pressure, suture retention, leakage test) for transplantation. We believe that the S-SDV could address the challenges of conventional SDVs: notably, endothelial formation and mechanical properties. In particular, the S-SDV can be designed simply as a free-form structure with a desired pore size. Since endothelial formation through the pore is easy even in free-form constructs, it is expected to be useful for endothelial formation in vascular structures with branch or curve shapes, and in other tubular tissues such as the esophagus.
Collapse
Affiliation(s)
- Hun-Jin Jeong
- Department of Mechanical Engineering, Wonkwang University, 54538, Iksan, Republic of Korea
- Regenerative Engineering Laboratory, Columbia University, 630W 168th ST, New York, 10032, USA
| | - Hyoryung Nam
- Department of Convergence IT Engineering, Pohang University of Science and Technology, 37673, Pohang, Gyeongbuk, Republic of Korea
| | - Jae-Seok Kim
- Department of Mechanical Engineering, Wonkwang University, 54538, Iksan, Republic of Korea
| | - Sungkeon Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology, 37673, Pohang, Gyeongbuk, Republic of Korea
| | - Hyun-Ha Park
- Department of Mechanical Engineering, Wonkwang University, 54538, Iksan, Republic of Korea
| | - Young-Sam Cho
- Department of Mechanical and Design Engineering, Wonkwang University, 54538, Iksan, Republic of Korea
| | - Hyungkook Jeon
- Department of Manufacturing Systems and Design Engineering, Seoul National University of Science and Technology, 01811, Seoul, Republic of Korea
| | - Jinah Jang
- Department of Convergence IT Engineering, Pohang University of Science and Technology, 37673, Pohang, Gyeongbuk, Republic of Korea
- Department of Mechanical Engineering, Pohang University of Science and Technology, 37673, Pohang, Gyeongbuk, Republic of Korea
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, 37673, Pohang, Gyeongbuk, Republic of Korea
- Institute of Convergence Science, Yonsei University, 03722, Seoul, Republic of Korea
| | - Seung-Jae Lee
- Department of Mechanical and Design Engineering, Wonkwang University, 54538, Iksan, Republic of Korea
| |
Collapse
|
7
|
Chandel S, Kumaragurubaran R, Giri H, Dixit M. Isolation and Culture of Human Umbilical Vein Endothelial Cells (HUVECs). Methods Mol Biol 2024; 2711:147-162. [PMID: 37776455 DOI: 10.1007/978-1-0716-3429-5_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/02/2023]
Abstract
This protocol describes a simple and an economical method for isolation of endothelial cells from human umbilical vein. Umbilical cord is easily available postpartum following informed consent, and the method for its collection is noninvasive with few ethical concerns. Thus, umbilical vein is an ideal source for isolation of endothelial cells of human origin. Endothelial cells are isolated from umbilical vein by collagenase digestion. This is followed by their culture on extracellular matrix (ECM) protein coated tissue culture flasks in presence of endothelial cell growth medium. In the last few decades, human umbilical vein endothelial cells (HUVECs) have come to be regarded as a standard model system by vascular biologists to understand general principles of endothelial cell biology and dysfunction. Here, we describe isolation, culture, as well as validation of HUVECs.
Collapse
Affiliation(s)
- Shivam Chandel
- Laboratory of Vascular Biology, Centre of Excellence in Molecular Medicine, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras , Chennai, India
| | - Rathnakumar Kumaragurubaran
- Single Cell Genomics, Cincinnati Children's Hospital Medical Centre , University of Cincinnati School of Medicine , Cincinnati, OH, USA
| | - Hemant Giri
- Oklahoma Medical Research Foundation , Oklahoma City, OK, USA
| | - Madhulika Dixit
- Laboratory of Vascular Biology, Centre of Excellence in Molecular Medicine, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras , Chennai, India.
| |
Collapse
|
8
|
Zhou X, Jiang Y, Wang Y, Fan L, Zhu Y, Chen Y, Wang Y, Zhu Y, Wang H, Pan Z, Li Z, Zhu X, Ren R, Ge Z, Lai D, Lai EY, Chen T, Wang K, Liang P, Qin L, Liu C, Qiu C, Simons M, Yu L. Endothelial FIS1 DeSUMOylation Protects Against Hypoxic Pulmonary Hypertension. Circ Res 2023; 133:508-531. [PMID: 37589160 DOI: 10.1161/circresaha.122.321200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/07/2023] [Indexed: 08/18/2023]
Abstract
BACKGROUND Hypoxia is a major cause and promoter of pulmonary hypertension (PH), a representative vascular remodeling disease with poor prognosis and high mortality. However, the mechanism underlying how pulmonary arterial system responds to hypoxic stress during PH remains unclear. Endothelial mitochondria are considered signaling organelles on oxygen tension. Results from previous clinical research and our studies suggested a potential role of posttranslational SUMOylation (small ubiquitin-like modifier modification) in endothelial mitochondria in hypoxia-related vasculopathy. METHODS Chronic hypoxia mouse model and Sugen/hypoxia rat model were employed as PH animal models. Mitochondrial morphology and subcellular structure were determined by transmission electron and immunofluorescent microscopies. Mitochondrial metabolism was determined by mitochondrial oxygen consumption rate and extracellular acidification rate. SUMOylation and protein interaction were determined by immunoprecipitation. RESULTS The involvement of SENP1 (sentrin-specific protease 1)-mediated SUMOylation in mitochondrial remodeling in the pulmonary endothelium was identified in clinical specimens of hypoxia-related PH and was verified in human pulmonary artery endothelial cells under hypoxia. Further analyses in clinical specimens, hypoxic rat and mouse PH models, and human pulmonary artery endothelial cells and human embryonic stem cell-derived endothelial cells revealed that short-term hypoxia-induced SENP1 translocation to endothelial mitochondria to regulate deSUMOylation (the reversible process of SUMOylation) of mitochondrial fission protein FIS1 (mitochondrial fission 1), which facilitated FIS1 assembling with fusion protein MFN2 (mitofusin 2) and mitochondrial gatekeeper VDAC1 (voltage-dependent anion channel 1), and the membrane tethering activity of MFN2 by enhancing its oligomerization. Consequently, FIS1 deSUMOylation maintained the mitochondrial integrity and endoplasmic reticulum-mitochondria calcium communication across mitochondrial-associated membranes, subsequently preserving pulmonary endothelial function and vascular homeostasis. In contrast, prolonged hypoxia disabled the FIS1 deSUMOylation by diminishing the availability of SENP1 in mitochondria via inducing miR (micro RNA)-138 and consequently resulted in mitochondrial dysfunction and metabolic reprogramming in pulmonary endothelium. Functionally, introduction of viral-packaged deSUMOylated FIS1 within pulmonary endothelium in mice improved pulmonary endothelial dysfunction and hypoxic PH development, while knock-in of SUMO (small ubiquitin-like modifier)-conjugated FIS1 in mice exaggerated the diseased cellular and tissue phenotypes. CONCLUSIONS By maintaining endothelial mitochondrial homeostasis, deSUMOylation of FIS1 adaptively preserves pulmonary endothelial function against hypoxic stress and consequently protects against PH. The FIS1 deSUMOylation-SUMOylation transition in pulmonary endothelium is an intrinsic pathogenesis of hypoxic PH.
Collapse
Affiliation(s)
- Xiaofei Zhou
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, X. Zhu, R.R., D.L., C.Q., L.Y.), Hangzhou, China
- MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, R.R., C.Q., L.Y.), Hangzhou, China
| | - Yuanqing Jiang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, X. Zhu, R.R., D.L., C.Q., L.Y.), Hangzhou, China
- MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, R.R., C.Q., L.Y.), Hangzhou, China
| | - Yuewen Wang
- School of Basic Medical Sciences, Shaanxi University of Chinese Medicine, Xianyang, China (Yuewen Wang)
| | - Linge Fan
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, X. Zhu, R.R., D.L., C.Q., L.Y.), Hangzhou, China
- MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, R.R., C.Q., L.Y.), Hangzhou, China
| | - Yunhui Zhu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, X. Zhu, R.R., D.L., C.Q., L.Y.), Hangzhou, China
- Cardiovascular Research Center, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, CT (X. Zhu, L.Q., M.S.)
| | - Yefeng Chen
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, X. Zhu, R.R., D.L., C.Q., L.Y.), Hangzhou, China
- MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, R.R., C.Q., L.Y.), Hangzhou, China
| | - Yiran Wang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, X. Zhu, R.R., D.L., C.Q., L.Y.), Hangzhou, China
- MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, R.R., C.Q., L.Y.), Hangzhou, China
| | - Yingyi Zhu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, X. Zhu, R.R., D.L., C.Q., L.Y.), Hangzhou, China
- MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, R.R., C.Q., L.Y.), Hangzhou, China
| | - Hongkun Wang
- Institute of Translational Medicine (H.W., P.L.), Hangzhou, China
| | - Zihang Pan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China (Z.P., K.W.)
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China (Z.P., K.W.)
| | - Zhoubin Li
- The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China (Z.L., E.Y.-L., T.C.)
| | - Xiaolong Zhu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, X. Zhu, R.R., D.L., C.Q., L.Y.), Hangzhou, China
| | - Ruizhe Ren
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, X. Zhu, R.R., D.L., C.Q., L.Y.), Hangzhou, China
- MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, R.R., C.Q., L.Y.), Hangzhou, China
| | - Zhen Ge
- School of Pharmaceutical Sciences, Hangzhou Medical College, Zhejiang, China (Z.G.)
| | - Dongwu Lai
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, X. Zhu, R.R., D.L., C.Q., L.Y.), Hangzhou, China
| | - En Yin Lai
- The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China (Z.L., E.Y.-L., T.C.)
| | - Ting Chen
- The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China (Z.L., E.Y.-L., T.C.)
| | - Kai Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China (Z.P., K.W.)
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China (Z.P., K.W.)
| | - Ping Liang
- Institute of Translational Medicine (H.W., P.L.), Hangzhou, China
| | - Lingfeng Qin
- Cardiovascular Research Center, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, CT (X. Zhu, L.Q., M.S.)
| | - Cuiqing Liu
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China (C.L.)
| | - Cong Qiu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, X. Zhu, R.R., D.L., C.Q., L.Y.), Hangzhou, China
- MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, R.R., C.Q., L.Y.), Hangzhou, China
- Cancer Center, Zhejiang University (C.Q., L.Y.), Hangzhou, China
| | - Michael Simons
- Cardiovascular Research Center, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, CT (X. Zhu, L.Q., M.S.)
| | - Luyang Yu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, X. Zhu, R.R., D.L., C.Q., L.Y.), Hangzhou, China
- MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, R.R., C.Q., L.Y.), Hangzhou, China
- Cancer Center, Zhejiang University (C.Q., L.Y.), Hangzhou, China
| |
Collapse
|
9
|
Ullah K, Ai L, Humayun Z, Wu R. Targeting Endothelial HIF2α/ARNT Expression for Ischemic Heart Disease Therapy. BIOLOGY 2023; 12:995. [PMID: 37508425 PMCID: PMC10376750 DOI: 10.3390/biology12070995] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 07/07/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023]
Abstract
Ischemic heart disease (IHD) is a major cause of mortality and morbidity worldwide, with novel therapeutic strategies urgently needed. Endothelial dysfunction is a hallmark of IHD, contributing to its development and progression. Hypoxia-inducible factors (HIFs) are transcription factors activated in response to low oxygen levels, playing crucial roles in various pathophysiological processes related to cardiovascular diseases. Among the HIF isoforms, HIF2α is predominantly expressed in cardiac vascular endothelial cells and has a key role in cardiovascular diseases. HIFβ, also known as ARNT, is the obligate binding partner of HIFα subunits and is necessary for HIFα's transcriptional activity. ARNT itself plays an essential role in the development of the cardiovascular system, regulating angiogenesis, limiting inflammatory cytokine production, and protecting against cardiomyopathy. This review provides an overview of the current understanding of HIF2α and ARNT signaling in endothelial cell function and dysfunction and their involvement in IHD pathogenesis. We highlight their roles in inflammation and maintaining the integrity of the endothelial barrier, as well as their potential as therapeutic targets for IHD.
Collapse
Affiliation(s)
- Karim Ullah
- Section of Cardiology, Department of Medicine, Biological Sciences Division, University of Chicago, Chicago, IL 60637, USA
| | - Lizhuo Ai
- Section of Cardiology, Department of Medicine, Biological Sciences Division, University of Chicago, Chicago, IL 60637, USA
- The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Zainab Humayun
- Section of Cardiology, Department of Medicine, Biological Sciences Division, University of Chicago, Chicago, IL 60637, USA
| | - Rongxue Wu
- Section of Cardiology, Department of Medicine, Biological Sciences Division, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
10
|
Chambliss C, Stiles JK, Gee BE. Neuregulin-1 attenuates hemolysis- and ischemia induced-cerebrovascular inflammation associated with sickle cell disease. J Stroke Cerebrovasc Dis 2023; 32:106912. [PMID: 36473396 PMCID: PMC10448832 DOI: 10.1016/j.jstrokecerebrovasdis.2022.106912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 11/21/2022] [Accepted: 11/26/2022] [Indexed: 12/09/2022] Open
Abstract
OBJECTIVES Individuals with sickle cell disease (SCD) are at severely heightened risk for cerebrovascular injury and acute cerebrovascular events, including ischemic and hemorrhagic stroke, potentially leading to impaired development and life-long physical and cognitive disabilities. Cerebrovascular injury specific to SCD includes inflammation caused by underlying conditions of chronic hemolysis and reduced cerebrovascular perfusion. The objectives of this study were to investigate whether expression of neuregulin-1β (NRG-1), an endogenous neuroprotective polypeptide, is increased in SCD or experimental conditions mimicking the hemolysis and ischemic conditions of SCD, and to determine if treatment with exogenous NRG-1 reduces markers of cerebrovascular inflammation. MATERIALS AND METHODS Plasma and brain-specific NRG-1 levels were measured in transgenic SCD mice. Endogenous NRG-1 levels and response to experimental conditions of excess heme and ischemia were measured in cultured human brain microvascular cells and astrocytes. Pre-treatment with NRG-1 was used to determine NRG-1's ability to ameliorate resultant cerebrovascular inflammation. RESULTS Plasma and brain-specific NRG-1 were elevated in transgenic SCD mice compared to healthy controls. Neuregulin-1 expression was significantly increased in cultured human microvascular cells and astrocytes exposed to excess heme and ischemia. Pre-treatment with NRG-1 reduced inflammatory chemokine (CXCL-1 and CXCL-10) and adhesion molecule (ICAM-1 and VCAM-1) expression and increased pro-angiogenic factors (VEGF-A) in microvascular cells and astrocytes exposed to excess heme and ischemia. CONCLUSIONS Elevated NRG-1 in SCD is likely a protective endogenous response to ongoing cerebrovascular insults caused by chronic hemolysis and reduced cerebrovascular perfusion. Administration of NRG-1 to reduce cerebrovascular inflammation may be therapeutically beneficial in SCD and warrants continued investigation.
Collapse
Affiliation(s)
- Christopher Chambliss
- Pediatrics Institute, Emory University School of Medicine, 2015 Uppergate Drive, Atlanta, GA 30322, United States; Aflac Cancer and Blood Disorders Center, 2015 Uppergate Drive, Atlanta, GA 30322, United States; Cardiovascular Research Institute, Morehouse School of Medicine, 720 Westview Drive SW, Atlanta, GA 30310, United States.
| | - Jonathan K Stiles
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, 720 Westview Drive SW, Atlanta, GA 30310, United States.
| | - Beatrice E Gee
- Pediatrics Institute, Emory University School of Medicine, 2015 Uppergate Drive, Atlanta, GA 30322, United States; Aflac Cancer and Blood Disorders Center, 2015 Uppergate Drive, Atlanta, GA 30322, United States; Children's Healthcare of Atlanta, 35 Jesse Hill Jr Drive SE, Atlanta, GA 30303, United States; Department of Pediatrics, Morehouse School of Medicine, 720 Westview Drive SW, Atlanta, GA 30310, United States
| |
Collapse
|
11
|
Mahapatra C, Kumar P, Paul MK, Kumar A. Angiogenic stimulation strategies in bone tissue regeneration. Tissue Cell 2022; 79:101908. [DOI: 10.1016/j.tice.2022.101908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/24/2022] [Accepted: 08/22/2022] [Indexed: 11/28/2022]
|
12
|
Brain Microvascular Endothelial Cell-Derived Exosomes Protect Neurons from Ischemia–Reperfusion Injury in Mice. Pharmaceuticals (Basel) 2022; 15:ph15101287. [PMID: 36297399 PMCID: PMC9608440 DOI: 10.3390/ph15101287] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/14/2022] [Accepted: 10/16/2022] [Indexed: 11/17/2022] Open
Abstract
Stroke often results in neurological and neuropsychiatric sequela. Exosomes derived from brain endothelial cells (EC-Exo) protect neurons from hypoxic injury. However, the biological role of exosomes in apoptosis and synaptic plasticity remains unclear. This research aimed to assess whether cerebral microvascular endothelial cells inhibit apoptosis and promote synaptic remodeling through exosome-mediated cell–cell interaction after the ischemic attack. The effects of EC-Exo on primary neuronal apoptosis and synapses in oxyglucose deprivation reoxygenation (OGD/R) injury were first assessed in vitro. Animal experiments were performed using C57BL/6J mice, divided into three groups: a sham group, a model (middle cerebral artery occlusion/reperfusion, MCAO/R) group, and an EC-Exo group (tail vein injection of EC-Exo, once/2 days for 14 days) to evaluate the neuromotor and exploratory abilities of mice after MCAO/R. Apoptosis and synaptic protein expression levels were detected. The results demonstrated that EC-Exo inhibited neuronal apoptosis and increased synaptic length after OGD/R. In vivo, EC-Exo not only improved neural motor behavior and increased regional cerebral blood flow (rCBF) in MCAO/R-injured mice but also promoted the expression of synaptic regulatory proteins and inhibited apoptosis in the brain. These results suggest that EC-Exo may provide neuroprotection against stroke by promoting synaptic remodeling and inhibiting apoptosis from protecting neurons.
Collapse
|
13
|
Altuntaş C, Alper M, Keleş Y, Sav FN, Köçkar F. Hypoxic regulation of ADAMTS-2 and -3 (a disintegrin and matrix metalloproteinase with thrombospondin motifs 2 and 3) procollagen N proteinases by HIF-1α in endothelial cells. Mol Cell Biochem 2022; 478:1151-1160. [PMID: 36241950 DOI: 10.1007/s11010-022-04549-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 08/26/2022] [Indexed: 10/17/2022]
Abstract
ADAMTS-2 and ADAMTS-3, known as procollagen amino proteases (PNP), are primarily responsible for processing the amino ends of the fibrillar collagen precursors. ADAMTS-2 is a highly expressed gene in type I collagen-rich tissues, such as skin, bones, tendons, and aorta. ADAMTS-3 is mainly expressed in cartilage, where it colocalizes with type II procollagen and in the nervous system. Studies about ADAMTS-2 and ADAMTS-3 enzymes primarily focused on their collagen processing activity. Knowledge about the transcriptional regulations of these genes is rather limited. Here we analyzed the transcriptional regulations of ADAMTS-2 and ADAMTS-3 genes under chemically induced hypoxic conditions in endothelial cell model, HUVECs. We elucidated that hypoxia is the potent positive regulator of ADAMTS-2 and ADAMTS-3 genes. qRT-PCR and western blotting studies revealed that ADAMTS-2 and ADAMTS-3 expressions were increased at mRNA and protein levels under chemically induced hypoxic conditions in HUVECs. In addition, Transient transfection experiments of ADAMTS-2 and ADAMTS-3 promoter-reporter constructs indicated that low oxygen conditions increased ADAMTS-2 and ADAMTS-3 promoter activities. Furthermore, the DNA-protein interaction assay provided evidence of the functional binding of HIF-1α on bioinformatically determined HRE regions on the ADAMTS-2 and ADAMTS-3 promoters.
Collapse
Affiliation(s)
- Candan Altuntaş
- Department of Stem Cell and Tissue Regeneration, Institute of Health Sciences, Kocaeli University, 4138, Kocaeli, Turkey
| | - Meltem Alper
- Department of Translational Oncology, Institute of Oncology, Dokuz Eylül University, İzmir, Turkey
| | - Yasemin Keleş
- Faculty of Science and Literature, Department of Molecular Biology, Balıkesir University, 10145, Balıkesir, Turkey
| | - Feyza Nur Sav
- Faculty of Science and Literature, Department of Molecular Biology, Balıkesir University, 10145, Balıkesir, Turkey
| | - Feray Köçkar
- Faculty of Science and Literature, Department of Molecular Biology, Balıkesir University, 10145, Balıkesir, Turkey.
| |
Collapse
|
14
|
Circulating Microparticles Are Differentially Increased in Lowlanders and Highlanders with High Altitude Induced Pulmonary Hypertension during the Cold Season. Cells 2022; 11:cells11192932. [PMID: 36230894 PMCID: PMC9563667 DOI: 10.3390/cells11192932] [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: 08/18/2022] [Revised: 09/12/2022] [Accepted: 09/15/2022] [Indexed: 11/16/2022] Open
Abstract
The role of microparticles (MPs) and cold in high altitude pulmonary hypertension (HAPH) remains unexplored. We investigated the impact of long-term cold exposure on the pulmonary circulation in lowlanders and high-altitude natives and the role of MPs. Pulmonary hemodynamics were evaluated using Doppler echocardiography at the end of the colder and warmer seasons. We further examined the miRNA content of MPs isolated from the study participants and studied their effects on human pulmonary artery smooth muscle (hPASMCs) and endothelial cells (hPAECs). Long-term exposure to cold environment was associated with an enhanced pulmonary artery pressure in highlanders. Plasma levels of CD62E-positive and CD68-positive MPs increased in response to cold in lowlanders and HAPH highlanders. The miRNA-210 expression contained in MPs differentially changed in response to cold in lowlanders and highlanders. MPs isolated from lowlanders and highlanders increased proliferation and reduced apoptosis of hPASMCs. Further, MPs isolated from warm-exposed HAPH highlanders and cold-exposed highlanders exerted the most pronounced effects on VEGF expression in hPAECs. We demonstrated that prolonged exposure to cold is associated with elevated pulmonary artery pressures, which are most pronounced in high-altitude residents. Further, the numbers of circulating MPs are differentially increased in lowlanders and HAPH highlanders during the colder season.
Collapse
|
15
|
CoCl2-Mimicked Endothelial Cell Hypoxia Induces Nucleotide Depletion and Functional Impairment That Is Reversed by Nucleotide Precursors. Biomedicines 2022; 10:biomedicines10071540. [PMID: 35884844 PMCID: PMC9313011 DOI: 10.3390/biomedicines10071540] [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: 05/26/2022] [Revised: 06/20/2022] [Accepted: 06/24/2022] [Indexed: 11/17/2022] Open
Abstract
Chronic hypoxia drives vascular dysfunction by various mechanisms, including changes in mitochondrial respiration. Although endothelial cells (ECs) rely predominantly on glycolysis, hypoxia is known to alter oxidative phosphorylation, promote oxidative stress and induce dysfunction in ECs. Our work aimed to analyze the effects of prolonged treatment with hypoxia-mimetic agent CoCl2 on intracellular nucleotide concentration, extracellular nucleotide breakdown, mitochondrial function, and nitric oxide (NO) production in microvascular ECs. Moreover, we investigated how nucleotide precursor supplementation and adenosine deaminase inhibition protected against CoCl2-mediated disturbances. Mouse (H5V) and human (HMEC-1) microvascular ECs were exposed to CoCl2-mimicked hypoxia for 24 h in the presence of nucleotide precursors: adenine and ribose, and adenosine deaminase inhibitor, 2′deoxycoformycin. CoCl2 treatment decreased NO production by ECs, depleted intracellular ATP concentration, and increased extracellular nucleotide and adenosine catabolism in both H5V and HMEC-1 cell lines. Diminished intracellular ATP level was the effect of disturbed mitochondrial phosphorylation, while nucleotide precursors effectively restored the ATP pool via the salvage pathway and improved endothelial function under CoCl2 treatment. Endothelial protective effects of adenine and ribose were further enhanced by adenosine deaminase inhibition, that increased adenosine concentration. This work points to a novel strategy for protection of hypoxic ECs by replenishing the adenine nucleotide pool and promoting adenosine signaling.
Collapse
|
16
|
Schmidt A, Fuchs M, Stojanović SD, Liang C, Schmidt K, Jung M, Xiao K, Weusthoff J, Just A, Pfanne A, Distler JHW, Dandekar T, Fiedler J, Thum T, Kunz M. Deciphering Pro-angiogenic Transcription Factor Profiles in Hypoxic Human Endothelial Cells by Combined Bioinformatics and in vitro Modeling. Front Cardiovasc Med 2022; 9:877450. [PMID: 35783871 PMCID: PMC9247153 DOI: 10.3389/fcvm.2022.877450] [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: 02/16/2022] [Accepted: 05/23/2022] [Indexed: 11/29/2022] Open
Abstract
Background Constant supply of oxygen is crucial for multicellular tissue homeostasis and energy metabolism in cardiac tissue. As a first response to acute hypoxia, endothelial cells (ECs) promote recruitment and adherence of immune cells to the dysbalanced EC barrier by releasing inflammatory mediators and growth factors, whereas chronic hypoxia leads to the activation of a transcription factor (TF) battery, that potently induces expression of growth factors and cytokines including platelet-derived growth factor (PDGF) and vascular endothelial growth factor (VEGF). We report a hypoxia-minded, targeted bioinformatics approach aiming to identify and validate TFs that regulate angiogenic signaling. Results A comprehensive RNA-Seq dataset derived from human ECs subjected to normoxic or hypoxic conditions was selected to identify significantly regulated genes based on (i) fold change (normoxia vs. hypoxia) and (ii) relative abundancy. Transcriptional regulation of this gene set was confirmed via qPCR in validation experiments where HUVECs were subjected to hypoxic conditions for 24 h. Screening the promoter and upstream regulatory elements of these genes identified two TFs, KLF5 and SP1, both with a potential binding site within these regions of selected target genes. In vitro, siRNA experiments confirmed SP1- and KLF5-mediated regulation of identified hypoxia-sensitive endothelial genes. Next to angiogenic signaling, we also validated the impact of TFs on inflammatory signaling, both key events in hypoxic sensing. Both TFs impacted on inflammatory signaling since endogenous repression led to increased NF-κB signaling. Additionally, SP1 silencing eventuated decreased angiogenic properties in terms of proliferation and tube formation. Conclusion By detailed in silico analysis of promoter region and upstream regulatory elements for a list of hypoxia-sensitive genes, our bioinformatics approach identified putative binding sites for TFs of SP or KLF family in vitro. This strategy helped to identify TFs functionally involved in human angiogenic signaling and therefore serves as a base for identifying novel RNA-based drug entities in a therapeutic setting of vascularization.
Collapse
Affiliation(s)
- Arne Schmidt
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hanover, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Hanover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hanover, Germany
| | - Maximilian Fuchs
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Hanover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hanover, Germany
| | - Stevan D. Stojanović
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hanover, Germany
- Department of Cardiology and Angiology, Hannover Medical School, Hanover, Germany
| | - Chunguang Liang
- Department of Bioinformatics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Kevin Schmidt
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hanover, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Hanover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hanover, Germany
| | - Mira Jung
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hanover, Germany
| | - Ke Xiao
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hanover, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Hanover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hanover, Germany
| | - Jan Weusthoff
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hanover, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Hanover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hanover, Germany
| | - Annette Just
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hanover, Germany
| | - Angelika Pfanne
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hanover, Germany
| | - Jörg H. W. Distler
- Department of Internal Medicine 3 – Rheumatology and Immunology, Universitätsklinikum Erlangen, Friedrich-Alexander University (FAU) of Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas Dandekar
- Department of Bioinformatics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Jan Fiedler
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hanover, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Hanover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hanover, Germany
- Jan Fiedler,
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hanover, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Hanover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hanover, Germany
- Thomas Thum,
| | - Meik Kunz
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Hanover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hanover, Germany
- Chair of Medical Informatics, Friedrich-Alexander University (FAU) of Erlangen-Nürnberg, Erlangen, Germany
- *Correspondence: Meik Kunz,
| |
Collapse
|
17
|
Schumacher A, Roumans N, Rademakers T, Joris V, Eischen-Loges MJ, van Griensven M, LaPointe VL. Enhanced Microvasculature Formation and Patterning in iPSC–Derived Kidney Organoids Cultured in Physiological Hypoxia. Front Bioeng Biotechnol 2022; 10:860138. [PMID: 35782512 PMCID: PMC9240933 DOI: 10.3389/fbioe.2022.860138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 05/05/2022] [Indexed: 01/10/2023] Open
Abstract
Stem cell–derived kidney organoids have been shown to self-organize from induced pluripotent stem cells into most important renal structures. However, the structures remain immature in culture and contain endothelial networks with low connectivity and limited organoid invasion. Furthermore, the nephrons lose their phenotype after approximately 25 days. To become applicable for future transplantation, further maturation in vitro is essential. Since kidneys in vivo develop in hypoxia, we studied the modulation of oxygen availability in culture. We hypothesized that introducing long-term culture at physiological hypoxia, rather than the normally applied non-physiological, hyperoxic 21% O2, could initiate angiogenesis, lead to enhanced growth factor expression and improve the endothelial patterning. We therefore cultured the kidney organoids at 7% O2 instead of 21% O2 for up to 25 days and evaluated nephrogenesis, growth factor expression such as VEGF-A and vascularization. Whole mount imaging revealed a homogenous morphology of the endothelial network with enhanced sprouting and interconnectivity when the kidney organoids were cultured in hypoxia. Three-dimensional vessel quantification confirmed that the hypoxic culture led to an increased average vessel length, likely due to the observed upregulation of VEGFA-189 and VEGFA-121, and downregulation of the antiangiogenic protein VEGF-A165b measured in hypoxia. This research indicates the importance of optimization of oxygen availability in organoid systems and the potential of hypoxic culture conditions in improving the vascularization of organoids.
Collapse
Affiliation(s)
- Anika Schumacher
- Department of Cell Biology–Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Nadia Roumans
- Department of Cell Biology–Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Timo Rademakers
- Department of Cell Biology–Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Virginie Joris
- Department of Cell Biology–Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Maria José Eischen-Loges
- Department of Cell Biology–Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Martijn van Griensven
- Department of Cell Biology–Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Vanessa L.S. LaPointe
- Department of Cell Biology–Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
- *Correspondence: Vanessa L.S. LaPointe,
| |
Collapse
|
18
|
Yang C, Zhang X, Ge X, He C, Liu S, Yang S, Huang C. N-Acetylcysteine protects against cobalt chloride-induced endothelial dysfunction by enhancing glucose-6-phosphate dehydrogenase activity. FEBS Open Bio 2022; 12:1475-1488. [PMID: 35666067 PMCID: PMC9340863 DOI: 10.1002/2211-5463.13449] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 05/05/2022] [Accepted: 06/03/2022] [Indexed: 11/07/2022] Open
Abstract
Hypoxia‐induced endothelial dysfunction is known to be involved in the pathogenesis of several vascular diseases. However, it remains unclear whether the pentose phosphate pathway (PPP) is involved in regulating the response of endothelial cells to hypoxia. Here, we established an in vitro model by treating EA.hy926 (a hybrid human umbilical vein cell line) with cobalt chloride (CoCl2; a chemical mimic that stabilizes HIF‐1α, thereby leading to the development of hypoxia), and used this to investigate the involvement of PPP by examining expression of its key enzyme, glucose‐6‐phosphate dehydrogenase (G6PD). We report that CoCl2 induces the accumulation of HIF‐1α, leading to endothelial cell dysfunction characterized by reduced cell viability, proliferation, tube formation, and activation of cytokine production, accompanied with a significant decrease in G6PD expression and activity. The addition of 6‐aminonicotinamide (6‐AN) to inhibit PPP directly causes endothelial dysfunction. Additionally, N‐Acetylcysteine (NAC), a precursor of glutathione, was further evaluated for its protective effects; NAC displayed a protective effect against CoCl2‐induced cell damage by enhancing G6PD activity, and this was abrogated by 6‐AN. The effects of CoCl2 and the involvement of G6PD in endothelial dysfunction have been confirmed in primary human aortic endothelial cells. In summary, G6PD was identified as a novel target of CoCl2‐induced damage, which highlighted the involvement of PPP in regulating the response of endothelial cell CoCl2. Treatment with NAC may be a potential strategy to treat hypoxia or ischemia, which are widely observed in vascular diseases.
Collapse
Affiliation(s)
- Chen Yang
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China.,Department of Geriatrics, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Xiaofang Zhang
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
| | - Xilin Ge
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
| | - Chunmei He
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
| | - Suhuan Liu
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China.,Research Center for Translational Medicine, the First Affiliated Hospital of Xiamen University, Xiamen, 361003, China
| | - Shuyu Yang
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China.,Traditional Chinese Medicine research studio, the First Affiliated Hospital of Xiamen University, Xiamen, 361003, China
| | - Caoxin Huang
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
| |
Collapse
|
19
|
Hypoxia and hemorheological properties in older individuals. Ageing Res Rev 2022; 79:101650. [PMID: 35597435 DOI: 10.1016/j.arr.2022.101650] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/20/2022] [Accepted: 05/13/2022] [Indexed: 12/17/2022]
Abstract
Hypoxia is caused by insufficient oxygen availability for the organism leading to reduced oxygen delivery to tissues and cells. It has been regarded as a severe threat to human health and it is indeed implicated in pathophysiological mechanisms involved in the development and progression of many diseases. Nevertheless, the potential of controlled hypoxia interventions (i.e. hypoxia conditioning) for improving cardio-vascular health is gaining increased attention. However, blood rheology is often a forgotten factor for vascular health while aging and hypoxia exposure are both suspected to alter hemorheological properties. These changes in blood rheology may influence the benefits-risks balance of hypoxia exposure in older individuals. The benefits of hypoxia exposure for vascular health are mainly reported for healthy populations and the combined impact of aging and hypoxia on blood rheology could therefore be deleterious in older individuals. This review discusses evidence of hypoxia-related and aging-related changes in blood viscosity and its determinants. It draws upon an extensive literature search on the effects of hypoxia/altitude and aging on blood rheology. Aging increases blood viscosity mainly through a rise in plasma viscosity, red blood cell (RBC) aggregation and a decrease in RBC deformability. Hypoxia also causes an increase in RBC aggregation and plasma viscosity. In addition, hypoxia exposure may increase hematocrit and modulate RBC deformability, depending on the hypoxic dose, i.e, beneficial effect of intermittent hypoxia with moderate dose vs deleterious effect of chronic continuous or intermittent hypoxia or if the hypoxic dose is too high. Special attention is directed toward the risks vs. benefits of hemorheological changes during hypoxia exposure in older individuals, and its clinical relevance for vascular disorders.
Collapse
|
20
|
Hypoxia as a Modulator of Inflammation and Immune Response in Cancer. Cancers (Basel) 2022; 14:cancers14092291. [PMID: 35565420 PMCID: PMC9099524 DOI: 10.3390/cancers14092291] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/25/2022] [Accepted: 04/25/2022] [Indexed: 02/01/2023] Open
Abstract
A clear association between hypoxia and cancer has heretofore been established; however, it has not been completely developed. In this sense, the understanding of the tumoral microenvironment is critical to dissect the complexity of cancer, including the reduction in oxygen distribution inside the tumoral mass, defined as tumoral hypoxia. Moreover, hypoxia not only influences the tumoral cells but also the surrounding cells, including those related to the inflammatory processes. In this review, we analyze the participation of HIF, NF-κB, and STAT signaling pathways as the main components that interconnect hypoxia and immune response and how they modulate tumoral growth. In addition, we closely examine the participation of the immune cells and how they are affected by hypoxia, the effects of the progression of cancer, and some innovative applications that take advantage of this knowledge, to suggest potential therapies. Therefore, we contribute to the understanding of the complexity of cancer to propose innovative therapeutic strategies in the future.
Collapse
|
21
|
Barry M, Pati S. Targeting repair of the vascular endothelium and glycocalyx after traumatic injury with plasma and platelet resuscitation. Matrix Biol Plus 2022; 14:100107. [PMID: 35392184 PMCID: PMC8981767 DOI: 10.1016/j.mbplus.2022.100107] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/10/2022] [Accepted: 03/10/2022] [Indexed: 02/06/2023] Open
Abstract
Endothelial glycocalyx shedding is a key instigator of the endotheliopathy of trauma. Plasma and platelet transfusions preserve vascular integrity in pre-clinical models. However, platelets may be less effective than plasma in preserving the glycocalyx.
Severely injured patients with hemorrhagic shock can develop endothelial dysfunction, systemic inflammation, and coagulation disturbances collectively known as the endotheliopathy of trauma (EOT). Shedding of the endothelial glycocalyx occurs early after injury, contributes to breakdown of the vascular barrier, and plays a critical role in the pathogenesis of multiple organ dysfunction, leading to poor outcomes in trauma patients. In this review we discuss (i) the pathophysiology of endothelial glycocalyx and vascular barrier breakdown following hemorrhagic shock and trauma, and (ii) the role of plasma and platelet transfusion in maintaining the glycocalyx and vascular endothelial integrity.
Collapse
Affiliation(s)
- Mark Barry
- University of California, San Francisco, Department of Surgery. 513 Parnassus Ave., San Francisco, CA 94143, United States
- Corresponding author.
| | - Shibani Pati
- University of California, San Francisco, Department of Surgery. 513 Parnassus Ave., San Francisco, CA 94143, United States
- University of California, San Francisco, Department of Laboratory Medicine. 513 Parnassus Ave., San Francisco, CA 94143, United States
| |
Collapse
|
22
|
Wagner K, Unger L, Salman MM, Kitchen P, Bill RM, Yool AJ. Signaling Mechanisms and Pharmacological Modulators Governing Diverse Aquaporin Functions in Human Health and Disease. Int J Mol Sci 2022; 23:1388. [PMID: 35163313 PMCID: PMC8836214 DOI: 10.3390/ijms23031388] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 02/07/2023] Open
Abstract
The aquaporins (AQPs) are a family of small integral membrane proteins that facilitate the bidirectional transport of water across biological membranes in response to osmotic pressure gradients as well as enable the transmembrane diffusion of small neutral solutes (such as urea, glycerol, and hydrogen peroxide) and ions. AQPs are expressed throughout the human body. Here, we review their key roles in fluid homeostasis, glandular secretions, signal transduction and sensation, barrier function, immunity and inflammation, cell migration, and angiogenesis. Evidence from a wide variety of studies now supports a view of the functions of AQPs being much more complex than simply mediating the passive flow of water across biological membranes. The discovery and development of small-molecule AQP inhibitors for research use and therapeutic development will lead to new insights into the basic biology of and novel treatments for the wide range of AQP-associated disorders.
Collapse
Affiliation(s)
- Kim Wagner
- School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia;
| | - Lucas Unger
- College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK; (L.U.); (P.K.)
| | - Mootaz M. Salman
- Department of Physiology Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK;
- Oxford Parkinson’s Disease Centre, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Philip Kitchen
- College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK; (L.U.); (P.K.)
| | - Roslyn M. Bill
- College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK; (L.U.); (P.K.)
| | - Andrea J. Yool
- School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia;
| |
Collapse
|
23
|
Razi O, Tartibian B, Teixeira AM, Zamani N, Govindasamy K, Suzuki K, Laher I, Zouhal H. Thermal dysregulation in patients with multiple sclerosis during SARS-CoV-2 infection. The potential therapeutic role of exercise. Mult Scler Relat Disord 2022; 59:103557. [PMID: 35092946 PMCID: PMC8785368 DOI: 10.1016/j.msard.2022.103557] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 01/16/2022] [Accepted: 01/22/2022] [Indexed: 12/15/2022]
Abstract
Thermoregulation is a homeostatic mechanism that is disrupted in some neurological diseases. Patients with multiple sclerosis (MS) are susceptible to increases in body temperature, especially with more severe neurological signs. This condition can become intolerable when these patients suffer febrile infections such as coronavirus disease-2019 (COVID-19). We review the mechanisms of hyperthermia in patients with MS, and they may encounter when infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Finally, the thermoregulatory role and relevant adaptation to regular physical exercise are summarized.
Collapse
Affiliation(s)
- Omid Razi
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, Razi University, Kermanshah, Iran
| | - Bakhtyar Tartibian
- Department of Exercise Physiology, Faculty of Physical Education and Sports Sciences, Allameh Tabataba'i University, Tehran, Iran
| | - Ana Maria Teixeira
- University of Coimbra, Research Center for Sport and Physical Activity, Faculty of Sport Sciences and Physical Education, Coimbra, Portugal
| | - Nastaran Zamani
- Department of Biology, Faculty of Science, Payame-Noor University, Tehran, Iran
| | - Karuppasamy Govindasamy
- Department of Physical Education & Sports Science, SRM Institute of Science and Technology, Kattankulathur, Tamilnadu, India
| | - Katsuhiko Suzuki
- Faculty of Sport Sciences, Waseda University, Tokorozawa 359-1192, Japan.
| | - Ismail Laher
- Department of Anesthesiology, Pharmacology and Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Hassane Zouhal
- Univ Rennes, M2S (Laboratoire Mouvement, Sport, Santé) - EA 1274, Rennes F-35000, France; Institut International des Sciences du Sport (2I2S), Irodouer 35850, France.
| |
Collapse
|
24
|
Azzoni E, Frontera V, Anselmi G, Rode C, James C, Deltcheva EM, Demian AS, Brown J, Barone C, Patelli A, Harman JR, Nicholls M, Conway SJ, Morrissey E, Jacobsen SEW, Sparrow DB, Harris AL, Enver T, de Bruijn MFTR. The onset of circulation triggers a metabolic switch required for endothelial to hematopoietic transition. Cell Rep 2021; 37:110103. [PMID: 34910918 PMCID: PMC8692754 DOI: 10.1016/j.celrep.2021.110103] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 10/11/2021] [Accepted: 11/15/2021] [Indexed: 12/14/2022] Open
Abstract
Hematopoietic stem cells (HSCs) emerge during development from the vascular wall of the main embryonic arteries. The onset of circulation triggers several processes that provide critical external factors for HSC generation. Nevertheless, it is not fully understood how and when the onset of circulation affects HSC emergence. Here we show that in Ncx1-/- mouse embryos devoid of circulation the HSC lineage develops until the phenotypic pro-HSC stage. However, these cells reside in an abnormal microenvironment, fail to activate the hematopoietic program downstream of Runx1, and are functionally impaired. Single-cell transcriptomics shows that during the endothelial-to-hematopoietic transition, Ncx1-/- cells fail to undergo a glycolysis to oxidative phosphorylation metabolic switch present in wild-type cells. Interestingly, experimental activation of glycolysis results in decreased intraembryonic hematopoiesis. Our results suggest that the onset of circulation triggers metabolic changes that allow HSC generation to proceed.
Collapse
Affiliation(s)
- Emanuele Azzoni
- MRC Molecular Hematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK.
| | - Vincent Frontera
- MRC Molecular Hematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Giorgio Anselmi
- MRC Molecular Hematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Christina Rode
- MRC Molecular Hematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Chela James
- Department of Cancer Biology, UCL Cancer Institute, University College London, London, WC1E 6DD, UK
| | - Elitza M Deltcheva
- Department of Cancer Biology, UCL Cancer Institute, University College London, London, WC1E 6DD, UK
| | - Atanasiu S Demian
- MRC WIMM Centre for Computational Biology, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - John Brown
- Department of Cancer Biology, UCL Cancer Institute, University College London, London, WC1E 6DD, UK
| | - Cristiana Barone
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, 20900, Italy
| | - Arianna Patelli
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, 20900, Italy
| | - Joe R Harman
- MRC Molecular Hematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Matthew Nicholls
- MRC Molecular Hematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Simon J Conway
- HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, IN 46033, USA
| | - Edward Morrissey
- MRC WIMM Centre for Computational Biology, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Sten Eirik W Jacobsen
- MRC Molecular Hematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK; Hematopoietic Stem Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK; Department of Cell and Molecular Biology, Wallenberg Institute for Regenerative Medicine and Department of Medicine, Center for Hematology and Regenerative Medicine, Karolinska Institutet and Karolinska University Hospital, 171 77 Stockholm, Sweden
| | - Duncan B Sparrow
- Department of Physiology, Anatomy and Genetics, BHF Centre of Research Excellence, University of Oxford, Oxford, OX1 3PT, UK
| | - Adrian L Harris
- Department of Oncology, Molecular Oncology Laboratories, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DS, UK
| | - Tariq Enver
- Department of Cancer Biology, UCL Cancer Institute, University College London, London, WC1E 6DD, UK; Division of Molecular Medicine and Gene Therapy, Lund University, Lund, 22184, Sweden
| | - Marella F T R de Bruijn
- MRC Molecular Hematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK.
| |
Collapse
|
25
|
Hypoxia alters the expression of ACE2 and TMPRSS2 SARS-CoV-2 cell entry mediators in hCMEC/D3 brain endothelial cells. Microvasc Res 2021; 138:104232. [PMID: 34416267 PMCID: PMC8372440 DOI: 10.1016/j.mvr.2021.104232] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 05/30/2021] [Accepted: 08/01/2021] [Indexed: 12/16/2022]
Abstract
The mechanisms by which the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) induces neurological complications remain to be elucidated. We aimed to identify possible effects of hypoxia on the expression of SARS-CoV-2 cell entry mediators, angiotensin-converting enzyme 2 (ACE2) receptor and transmembrane protease serine 2 (TMPRSS2) protein, in human brain endothelial cells, in vitro. hCMEC/D3 cells were exposed to different oxygen tensions: 20% (Control group), 8% or 2% O2 (Hypoxia groups). Cells were harvested 6-, 24- and 48 h following hypoxic challenge for assessment of mRNA and protein, using qPCR and Western Blot. The response of the brain endothelial cells to hypoxia was replicated using modular incubator chambers. We observed an acute increase (6 h, p < 0.05), followed by a longer-term decrease (48 h, p < 0.05) in ACE2 mRNA and protein expression, accompanied by reduced expression of TMPRSS2 protein levels (48 h, p < 0.05) under the more severe hypoxic condition (2% O2). No changes in levels of von Willebrand Factor (vWF - an endothelial cell damage marker) or interleukin 6 (IL-6 - a pro-inflammatory cytokine) mRNA were observed. We conclude that hypoxia regulates brain endothelial cell ACE2 and TMPRSS2 expression in vitro, which may indicate human brain endothelial susceptibility to SARS-CoV-2 infection and subsequent brain sequelae.
Collapse
|
26
|
Jiang S, Chen G, Yang Z, Wang D, Lu Y, Zhu L, Wang X. Testosterone attenuates hypoxia-induced hypertension by affecting NRF1-mediated transcriptional regulation of ET-1 and ACE. Hypertens Res 2021; 44:1395-1405. [PMID: 34257425 DOI: 10.1038/s41440-021-00703-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 05/08/2021] [Accepted: 06/10/2021] [Indexed: 02/07/2023]
Abstract
Hypertension induced by hypoxia at high altitude is one of the typical symptoms of high-altitude reactions (HARs). Emerging evidence indicates that endothelial abnormalities, including increases in angiotensin-2 (Ang-2) and endothelin-1 (ET-1), are closely associated with hypertension. Thus, low blood oxygen-induced endothelial dysfunction through acceleration of Ang-2 and ET-1 synthesis may alleviate HARs. In this study, we investigated the effects of hypoxia on rat blood pressure (BP) and endothelial injury. We found that BP increased by 10 mmHg after treatment with 10% O2 (~5500 m above sea level) for 24 h. Consistently, serum Ang-2 and ET-1 levels were increased along with decreases in NO levels. In endothelial cells, angiotensin-1-converting enzyme (ACE) and ET-1 expression levels were upregulated. Interestingly, nuclear respiratory factor 1 (NRF1) levels were also upregulated, consistent with the changes in ACE and ET-1 levels. We further demonstrated that NRF1 transcriptionally activated ACE and ET-1 by directly binding to their promoter regions, suggesting that the endothelial cell dysfunction induced by hypoxia was due to NRF1-dependent upregulation of ACE and ET-1. Surprisingly, testosterone supplementation showed significant protective effects on BP, while castration induced even higher BPs in rats exposed to hypoxia. We further showed that physiological testosterone repressed NRF1 expression in vivo and in vitro and thereby reduced Ang-2 and ET-1 levels, which was dependent on hypoxia. In summary, we have identified that physiological testosterone protects against hypoxia-induced hypertension through inhibition of NRF1, which transcriptionally regulates ACE and ET-1 expression.
Collapse
Affiliation(s)
- Shan Jiang
- Institute of Special Environmental Medicine, Nantong University, Nantong, 226019, Jiangsu, China.,School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Guijuan Chen
- Institute of Special Environmental Medicine, Nantong University, Nantong, 226019, Jiangsu, China
| | - Zhihui Yang
- Institute of Special Environmental Medicine, Nantong University, Nantong, 226019, Jiangsu, China
| | - Dan Wang
- Institute of Special Environmental Medicine, Nantong University, Nantong, 226019, Jiangsu, China
| | - Yapeng Lu
- Institute of Special Environmental Medicine, Nantong University, Nantong, 226019, Jiangsu, China
| | - Li Zhu
- Institute of Special Environmental Medicine, Nantong University, Nantong, 226019, Jiangsu, China. .,Co-Innovation Center of Neuroregeneration, Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, 226019, Jiangsu, China.
| | - Xueting Wang
- Institute of Special Environmental Medicine, Nantong University, Nantong, 226019, Jiangsu, China.
| |
Collapse
|
27
|
Woo KV, Shen IY, Weinheimer CJ, Kovacs A, Nigro J, Lin CY, Chakinala M, Byers DE, Ornitz DM. Endothelial FGF signaling is protective in hypoxia-induced pulmonary hypertension. J Clin Invest 2021; 131:141467. [PMID: 34623323 DOI: 10.1172/jci141467] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 06/25/2021] [Indexed: 01/08/2023] Open
Abstract
Hypoxia-induced pulmonary hypertension (PH) is one of the most common and deadliest forms of PH. Fibroblast growth factor receptors 1 and 2 (FGFR1/2) are elevated in patients with PH and in mice exposed to chronic hypoxia. Endothelial FGFR1/2 signaling is important for the adaptive response to several injury types and we hypothesized that endothelial FGFR1/2 signaling would protect against hypoxia-induced PH. Mice lacking endothelial FGFR1/2, mice with activated endothelial FGFR signaling, and human pulmonary artery endothelial cells (HPAECs) were challenged with hypoxia. We assessed the effect of FGFR activation and inhibition on right ventricular pressure, vascular remodeling, and endothelial-mesenchymal transition (EndMT), a known pathologic change seen in patients with PH. Hypoxia-exposed mice lacking endothelial FGFRs developed increased PH, while mice overexpressing a constitutively active FGFR in endothelial cells did not develop PH. Mechanistically, lack of endothelial FGFRs or inhibition of FGFRs in HPAECs led to increased TGF-β signaling and increased EndMT in response to hypoxia. These phenotypes were reversed in mice with activated endothelial FGFR signaling, suggesting that FGFR signaling inhibits TGF-β pathway-mediated EndMT during chronic hypoxia. Consistent with these observations, lung tissue from patients with PH showed activation of FGFR and TGF-β signaling. Collectively, these data suggest that activation of endothelial FGFR signaling could be therapeutic for hypoxia-induced PH.
Collapse
Affiliation(s)
- Kel Vin Woo
- Division of Cardiology, Department of Pediatrics.,Department of Developmental Biology
| | | | | | | | | | | | - Murali Chakinala
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Derek E Byers
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | | |
Collapse
|
28
|
Ortega MA, Fraile-Martínez O, García-Montero C, Álvarez-Mon MA, Chaowen C, Ruiz-Grande F, Pekarek L, Monserrat J, Asúnsolo A, García-Honduvilla N, Álvarez-Mon M, Bujan J. Understanding Chronic Venous Disease: A Critical Overview of Its Pathophysiology and Medical Management. J Clin Med 2021; 10:3239. [PMID: 34362022 PMCID: PMC8348673 DOI: 10.3390/jcm10153239] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 01/19/2023] Open
Abstract
Chronic venous disease (CVD) is a multifactorial condition affecting an important percentage of the global population. It ranges from mild clinical signs, such as telangiectasias or reticular veins, to severe manifestations, such as venous ulcerations. However, varicose veins (VVs) are the most common manifestation of CVD. The explicit mechanisms of the disease are not well-understood. It seems that genetics and a plethora of environmental agents play an important role in the development and progression of CVD. The exposure to these factors leads to altered hemodynamics of the venous system, described as ambulatory venous hypertension, therefore promoting microcirculatory changes, inflammatory responses, hypoxia, venous wall remodeling, and epigenetic variations, even with important systemic implications. Thus, a proper clinical management of patients with CVD is essential to prevent potential harms of the disease, which also entails a significant loss of the quality of life in these individuals. Hence, the aim of the present review is to collect the current knowledge of CVD, including its epidemiology, etiology, and risk factors, but emphasizing the pathophysiology and medical care of these patients, including clinical manifestations, diagnosis, and treatments. Furthermore, future directions will also be covered in this work in order to provide potential fields to explore in the context of CVD.
Collapse
Affiliation(s)
- Miguel A. Ortega
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (M.A.O.); (O.F.-M.); (C.G.-M.); (C.C.); (L.P.); (J.M.); (N.G.-H.); (M.Á.-M.); (J.B.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain;
- Cancer Registry and Pathology Department, Hospital Universitario Principe de Asturias, 28806 Alcalá de Henares, Spain
| | - Oscar Fraile-Martínez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (M.A.O.); (O.F.-M.); (C.G.-M.); (C.C.); (L.P.); (J.M.); (N.G.-H.); (M.Á.-M.); (J.B.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain;
| | - Cielo García-Montero
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (M.A.O.); (O.F.-M.); (C.G.-M.); (C.C.); (L.P.); (J.M.); (N.G.-H.); (M.Á.-M.); (J.B.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain;
| | - Miguel A. Álvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (M.A.O.); (O.F.-M.); (C.G.-M.); (C.C.); (L.P.); (J.M.); (N.G.-H.); (M.Á.-M.); (J.B.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain;
| | - Chen Chaowen
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (M.A.O.); (O.F.-M.); (C.G.-M.); (C.C.); (L.P.); (J.M.); (N.G.-H.); (M.Á.-M.); (J.B.)
| | - Fernando Ruiz-Grande
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain;
- Department of Vascular Surgery, Príncipe de Asturias Hospital, 28801 Alcalá de Henares, Spain
| | - Leonel Pekarek
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (M.A.O.); (O.F.-M.); (C.G.-M.); (C.C.); (L.P.); (J.M.); (N.G.-H.); (M.Á.-M.); (J.B.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain;
| | - Jorge Monserrat
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (M.A.O.); (O.F.-M.); (C.G.-M.); (C.C.); (L.P.); (J.M.); (N.G.-H.); (M.Á.-M.); (J.B.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain;
| | - Angel Asúnsolo
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain;
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain;
- Department of Epidemiology and Biostatistics, Graduate School of Public Health and Health Policy, The City University of New York, New York, NY 10027, USA
| | - Natalio García-Honduvilla
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (M.A.O.); (O.F.-M.); (C.G.-M.); (C.C.); (L.P.); (J.M.); (N.G.-H.); (M.Á.-M.); (J.B.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain;
| | - Melchor Álvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (M.A.O.); (O.F.-M.); (C.G.-M.); (C.C.); (L.P.); (J.M.); (N.G.-H.); (M.Á.-M.); (J.B.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain;
- Immune System Diseases—Rheumatology and Internal Medicine Service, University Hospital Príncipe de Asturias, (CIBEREHD), 28806 Alcalá de Henares, Spain
| | - Julia Bujan
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (M.A.O.); (O.F.-M.); (C.G.-M.); (C.C.); (L.P.); (J.M.); (N.G.-H.); (M.Á.-M.); (J.B.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain;
| |
Collapse
|
29
|
Jones DT, Macdonald JH, Sandoo A, Oliver SJ, Rossetti GMK. The deleterious effects of acute hypoxia on microvascular and large vessel endothelial function. Exp Physiol 2021; 106:1699-1709. [PMID: 34036677 DOI: 10.1113/ep089393] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 05/12/2021] [Indexed: 12/14/2022]
Abstract
NEW FINDINGS What is the central question of this study? The aim was primarily to determine the effect of hypoxia on microvascular function and secondarily whether superior cardiorespiratory fitness is protective against hypoxia-induced impairment in vascular function. What is the main finding and its importance? Hypoxia reduced endothelium-dependent but not endothelium-independent microvascular function. The extent of impairment was twofold higher in the microcirculation compared with the large blood vessels. This study suggests that individuals with superior cardiorespiratory fitness might preserve microvascular function in hypoxia. These findings highlight the sensitivity of the microvascular circulation to hypoxia. ABSTRACT Hypoxia is associated with diminished bioavailability of the endothelium-derived vasodilator, nitric oxide (NO). Diminished NO bioavailability can have deleterious effects on endothelial function. The endothelium is a heterogeneous tissue; therefore, a comprehensive assessment of endothelial function is crucial to understand the significance of hypoxia-induced endothelial dysfunction. We hypothesized that acute hypoxia would have a deleterious effect on microvascular and large vessel endothelial function. Twenty-nine healthy adults [24 (SD = 4 ) years of age] completed normoxic and hypoxic [inspired O2 fraction = 0.209] trials in this double-blinded, counterbalanced crossover study. After 30 min, we assessed the laser Doppler imaging-determined perfusion response to iontophoresis of ACh as a measure of endothelium-dependent microvascular function and iontophoresis of sodium nitroprusside as a measure of endothelium-independent microvascular function. After 60 min, we assessed brachial flow-mediated dilatation as a measure of large vessel endothelial function. Thirty minutes of hypoxia reduced endothelium-dependent microvascular function determined by the perfusion response to ACh (median difference (x̃∆) = -109% {interquartile range: 542.7}, P < 0.05), but not endothelium-independent microvascular function determined by the perfusion response to sodium nitroprusside (x̃∆ = 69% {interquartile range: 453.7}, P = 0.6). In addition, 60 min of hypoxia reduced allometrically scaled flow-mediated dilatation compared with normoxia ( x ¯ Δ = - 1.19 [95% CI = -1.80, -0.58 (Confidence Intervals)]%, P < 0.001). The decrease in microvascular endothelial function was associated with cardiorespiratory fitness (r = 0.45, P = 0.02). In conclusion, acute exposure to normobaric hypoxia significantly reduced endothelium-dependent vasodilatory capacity in small and large vessels. Collectively, these findings highlight the sensitivity of the microvascular circulation to hypoxic insult, particularly in those with poor cardiorespiratory fitness.
Collapse
Affiliation(s)
- Danial T Jones
- School of Sport, Health and Exercise Sciences, College of Human Sciences, Bangor University, Bangor, UK
| | - Jamie H Macdonald
- School of Sport, Health and Exercise Sciences, College of Human Sciences, Bangor University, Bangor, UK
| | - Aamer Sandoo
- School of Sport, Health and Exercise Sciences, College of Human Sciences, Bangor University, Bangor, UK
| | - Samuel J Oliver
- School of Sport, Health and Exercise Sciences, College of Human Sciences, Bangor University, Bangor, UK
| | - Gabriella M K Rossetti
- School of Sport, Health and Exercise Sciences, College of Human Sciences, Bangor University, Bangor, UK.,Centre for Integrative Neuroscience and Neurodynamics, School of Psychology and Clinical Language Sciences, University of Reading, Reading, UK
| |
Collapse
|
30
|
Mukherjee D, Konduri GG. Pediatric Pulmonary Hypertension: Definitions, Mechanisms, Diagnosis, and Treatment. Compr Physiol 2021; 11:2135-2190. [PMID: 34190343 PMCID: PMC8289457 DOI: 10.1002/cphy.c200023] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pediatric pulmonary hypertension (PPH) is a multifactorial disease with diverse etiologies and presenting features. Pulmonary hypertension (PH), defined as elevated pulmonary artery pressure, is the presenting feature for several pulmonary vascular diseases. It is often a hidden component of other lung diseases, such as cystic fibrosis and bronchopulmonary dysplasia. Alterations in lung development and genetic conditions are an important contributor to pediatric pulmonary hypertensive disease, which is a distinct entity from adult PH. Many of the causes of pediatric PH have prenatal onset with altered lung development due to maternal and fetal conditions. Since lung growth is altered in several conditions that lead to PPH, therapy for PPH includes both pulmonary vasodilators and strategies to restore lung growth. These strategies include optimal alveolar recruitment, maintaining physiologic blood gas tension, nutritional support, and addressing contributing factors, such as airway disease and gastroesophageal reflux. The outcome for infants and children with PH is highly variable and largely dependent on the underlying cause. The best outcomes are for neonates with persistent pulmonary hypertension (PPHN) and reversible lung diseases, while some genetic conditions such as alveolar capillary dysplasia are lethal. © 2021 American Physiological Society. Compr Physiol 11:2135-2190, 2021.
Collapse
Affiliation(s)
- Devashis Mukherjee
- Division of Neonatology, Department of Pediatrics, Medical College of Wisconsin, Children’s Research Institute, Children’s Wisconsin, Milwaukee, Wisconsin, 53226 USA
| | - Girija G. Konduri
- Division of Neonatology, Department of Pediatrics, Medical College of Wisconsin, Children’s Research Institute, Children’s Wisconsin, Milwaukee, Wisconsin, 53226 USA
| |
Collapse
|
31
|
Yadid M, Lind JU, Ardoña HAM, Sheehy SP, Dickinson LE, Eweje F, Bastings MMC, Pope B, O'Connor BB, Straubhaar JR, Budnik B, Kleber AG, Parker KK. Endothelial extracellular vesicles contain protective proteins and rescue ischemia-reperfusion injury in a human heart-on-chip. Sci Transl Med 2021; 12:12/565/eaax8005. [PMID: 33055246 DOI: 10.1126/scitranslmed.aax8005] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 09/08/2020] [Indexed: 12/17/2022]
Abstract
Extracellular vesicles (EVs) derived from various stem cell sources induce cardioprotective effects during ischemia-reperfusion injury (IRI). These have been attributed mainly to the antiapoptotic, proangiogenic, microRNA (miRNA) cargo within the stem cell-derived EVs. However, the mechanisms of EV-mediated endothelial signaling to cardiomyocytes, as well as their therapeutic potential toward ischemic myocardial injury, are not clear. EV content beyond miRNA that may contribute to cardioprotection has not been fully illuminated. This study characterized the protein cargo of human vascular endothelial EVs (EEVs) to identify lead cardioactive proteins and assessed the effect of EEVs on human laminar cardiac tissues (hlCTs) exposed to IRI. We mapped the protein content of human vascular EEVs and identified proteins that were previously associated with cellular metabolism, redox state, and calcium handling, among other processes. Analysis of the protein landscape of human cardiomyocytes revealed corresponding modifications induced by EEV treatment. To assess their human-specific cardioprotection in vitro, we developed a human heart-on-a-chip IRI assay using human stem cell-derived, engineered cardiac tissues. We found that EEVs alleviated cardiac cell death as well as the loss in contractile capacity during and after simulated IRI in an uptake- and dose-dependent manner. Moreover, we found that EEVs increased the respiratory capacity of normoxic cardiomyocytes. These results suggest that vascular EEVs rescue hlCTs exposed to IRI possibly by supplementing injured myocytes with cargo that supports multiple metabolic and salvage pathways and therefore may serve as a multitargeted therapy for IRI.
Collapse
Affiliation(s)
- Moran Yadid
- Disease Biophysics Group, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Johan U Lind
- Disease Biophysics Group, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA.,Department of Health Technology, Technical University of Denmark, Kgs. Lyngby 2800, Denmark
| | - Herdeline Ann M Ardoña
- Disease Biophysics Group, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| | - Sean P Sheehy
- Disease Biophysics Group, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Lauren E Dickinson
- Disease Biophysics Group, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Feyisayo Eweje
- Disease Biophysics Group, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Maartje M C Bastings
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA.,Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA.,Ecole Polytechnique Federale Lausanne (EPFL), School of Engineering, Institute of Materials, Programmable Biomaterials Laboratory, Station 12, 1015 Lausanne, Switzerland
| | - Benjamin Pope
- Disease Biophysics Group, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Blakely B O'Connor
- Disease Biophysics Group, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| | | | - Bogdan Budnik
- FAS Division of Science, Harvard University, Cambridge, MA 02138, USA
| | - Andre G Kleber
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Kevin Kit Parker
- Disease Biophysics Group, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA. .,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| |
Collapse
|
32
|
Devi S, Alexandre YO, Loi JK, Gillis R, Ghazanfari N, Creed SJ, Holz LE, Shackleford D, Mackay LK, Heath WR, Sloan EK, Mueller SN. Adrenergic regulation of the vasculature impairs leukocyte interstitial migration and suppresses immune responses. Immunity 2021; 54:1219-1230.e7. [PMID: 33915109 DOI: 10.1016/j.immuni.2021.03.025] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/14/2020] [Accepted: 03/29/2021] [Indexed: 12/16/2022]
Abstract
The sympathetic nervous system (SNS) controls various physiological functions via the neurotransmitter noradrenaline. Activation of the SNS in response to psychological or physical stress is frequently associated with weakened immunity. Here, we investigated how adrenoceptor signaling influences leukocyte behavior. Intravital two-photon imaging after injection of noradrenaline revealed transient inhibition of CD8+ and CD4+ T cell locomotion in tissues. Expression of β-adrenergic receptor in hematopoietic cells was not required for NA-mediated inhibition of motility. Rather, chemogenetic activation of the SNS or treatment with adrenergic receptor agonists induced vasoconstriction and decreased local blood flow, resulting in abrupt hypoxia that triggered rapid calcium signaling in leukocytes and halted cell motility. Oxygen supplementation reversed these effects. Treatment with adrenergic receptor agonists impaired T cell responses induced in response to viral and parasitic infections, as well as anti-tumor responses. Thus, stimulation of the SNS impairs leukocyte mobility, providing a mechanistic understanding of the link between adrenergic receptors and compromised immunity.
Collapse
Affiliation(s)
- Sapna Devi
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, 3000, Australia; The Australian Research Council Centre of Excellence in Advanced Molecular Imaging, The University of Melbourne, Melbourne, Victoria, 3000, Australia
| | - Yannick O Alexandre
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, 3000, Australia
| | - Joon Keit Loi
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, 3000, Australia
| | - Ryan Gillis
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052 Australia
| | - Nazanin Ghazanfari
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, 3000, Australia
| | - Sarah J Creed
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052 Australia
| | - Lauren E Holz
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, 3000, Australia; The Australian Research Council Centre of Excellence in Advanced Molecular Imaging, The University of Melbourne, Melbourne, Victoria, 3000, Australia
| | - David Shackleford
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052 Australia
| | - Laura K Mackay
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, 3000, Australia
| | - William R Heath
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, 3000, Australia; The Australian Research Council Centre of Excellence in Advanced Molecular Imaging, The University of Melbourne, Melbourne, Victoria, 3000, Australia
| | - Erica K Sloan
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052 Australia; Division of Surgery, Peter MacCallum Cancer Center, Victoria, 3000, Australia
| | - Scott N Mueller
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, 3000, Australia; The Australian Research Council Centre of Excellence in Advanced Molecular Imaging, The University of Melbourne, Melbourne, Victoria, 3000, Australia.
| |
Collapse
|
33
|
Plasma microparticles of sickle patients during crisis or taking hydroxyurea modify endothelium inflammatory properties. Blood 2021; 136:247-256. [PMID: 32285120 DOI: 10.1182/blood.2020004853] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/05/2020] [Indexed: 12/29/2022] Open
Abstract
Microparticles (MPs) are submicron extracellular vesicles exposing phosphatidylserine (PS), detected at high concentration in the circulation of sickle cell anemia (SS) patients. Several groups studied the biological effects of MPs generated ex vivo. Here, we analyzed for the first time the impact of circulating MPs on endothelial cells (ECs) from 60 sickle cell disease (SCD) patients. MPs were collected from SCD patients and compared with MPs isolated from healthy individuals (AA). Other plasma MPs were purified from SS patients before and 2 years after the onset of hydroxyurea (HU) treatment or during a vaso-occlusive crisis and at steady-state. Compared with AA MPs, SS MPs increased EC ICAM-1 messenger RNA and protein levels, as well as neutrophil adhesion. We showed that ICAM-1 overexpression was primarily caused by MPs derived from erythrocytes, rather than from platelets, and that it was abolished by MP PS capping using annexin V. MPs from SS patients treated with HU were less efficient to induce a proinflammatory phenotype in ECs compared with MPs collected before therapy. In contrast, MPs released during crisis increased ICAM-1 and neutrophil adhesion levels, in a PS-dependent manner, compared with MPs collected at steady-state. Furthermore, neutrophil adhesion was abolished by a blocking anti-ICAM-1 antibody. Our study provides evidence that MPs play a key role in SCD pathophysiology by triggering a proinflammatory phenotype of ECs. We also uncover a new mode of action for HU and identify potential therapeutics: annexin V and anti-ICAM-1 antibodies.
Collapse
|
34
|
A coaxially structured trilayered gallic acid-based antioxidant vascular graft for treating coronary artery disease. Eur Polym J 2021. [DOI: 10.1016/j.eurpolymj.2020.110203] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
35
|
Hypoxia-Mediated Decrease of Ovarian Cancer Cells Reaction to Treatment: Significance for Chemo- and Immunotherapies. Int J Mol Sci 2020; 21:ijms21249492. [PMID: 33327450 PMCID: PMC7764929 DOI: 10.3390/ijms21249492] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 11/30/2020] [Accepted: 12/08/2020] [Indexed: 12/20/2022] Open
Abstract
Hypoxia, a common factor ruling the microenvironment composition, leads to tumor progression. In this hypoxic context, cytokines and cells cooperate to favor cancer development and metastasis. Tumor hypoxia is heterogeneously distributed. Oxygen gradients depend on the vicinity, functionality of blood vessels, and oxygen ability to diffuse into surrounding tissues. Thus, the vasculature state modulates the microenvironment of the tumor cells. Cells sense and react to small variations in oxygen tension, which explains the lack of tumor cells’ unicity in their reaction to drugs. Ovarian cancers are highly hypoxia-dependent, ascites worsening the access to oxygen, in their reactions to both chemotherapy and new immunotherapy. Consequently, hypoxia affects the results of immunotherapy, and is thus, crucial for the design of treatments. Controlling key immunosuppressive factors and receptors, as well as immune checkpoint molecule expression on tumor, immune and stromal cells, hypoxia induces immunosuppression. Consequently, new approaches to alleviate hypoxia in the tumor microenvironment bring promises for ovarian cancer immunotherapeutic strategies. This review focuses on the effects of hypoxia in the microenvironment and its consequences on tumor treatments. This opens the way to innovative combined treatments to the advantage of immunotherapy outcome in ovarian cancers.
Collapse
|
36
|
Yumnamcha T, Guerra M, Singh LP, Ibrahim AS. Metabolic Dysregulation and Neurovascular Dysfunction in Diabetic Retinopathy. Antioxidants (Basel) 2020; 9:E1244. [PMID: 33302369 PMCID: PMC7762582 DOI: 10.3390/antiox9121244] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/29/2020] [Accepted: 12/01/2020] [Indexed: 12/11/2022] Open
Abstract
Diabetic retinopathy is a major cause of ocular complications in patients with type 1 and type 2 diabetes in developed countries. Due to the continued increase in the number of people with obesity and diabetes in the United States of America and globally, the incidence of diabetic retinopathy is expected to increase significantly in the coming years. Diabetic retinopathy is widely accepted as a combination of neurodegenerative and microvascular changes; however, which change occurs first is not yet understood. Although the pathogenesis of diabetic retinopathy is very complex, regulated by numerous signaling pathways and cellular processes, maintaining glucose homeostasis is still an essential component for normal physiological functioning of retinal cells. The maintenance of glucose homeostasis is finely regulated by coordinated interplay between glycolysis, Krebs cycle, and oxidative phosphorylation. Glycolysis is the most conserved metabolic pathway in biology and is tightly regulated to maintain a steady-state concentration of glycolytic intermediates; this regulation is called scheduled or regulated glycolysis. However, an abnormal increase in glycolytic flux generates large amounts of intermediate metabolites that can be shunted into different damaging pathways including the polyol pathway, hexosamine pathway, diacylglycerol-dependent activation of the protein kinase C pathway, and Amadori/advanced glycation end products (AGEs) pathway. In addition, disrupting the balance between glycolysis and oxidative phosphorylation leads to other biochemical and molecular changes observed in diabetic retinopathy including endoplasmic reticulum-mitochondria miscommunication and mitophagy dysregulation. This review will focus on how dysregulation of glycolysis contributes to diabetic retinopathy.
Collapse
Affiliation(s)
- Thangal Yumnamcha
- Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, MI 48201, USA; (M.G.); (L.P.S.)
| | - Michael Guerra
- Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, MI 48201, USA; (M.G.); (L.P.S.)
| | - Lalit Pukhrambam Singh
- Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, MI 48201, USA; (M.G.); (L.P.S.)
| | - Ahmed S. Ibrahim
- Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, MI 48201, USA; (M.G.); (L.P.S.)
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
37
|
Abstract
Most severe cases with COVID-19, especially those with pulmonary failure, are not a consequence of viral burden and/or failure of the 'adaptive' immune response to subdue the pathogen by utilizing an adequate 'adaptive' immune defense. Rather it is a consequence of immunopathology, resulting from imbalanced innate immune response, which may not be linked to pathogen burden at all. In fact, it might be described as an autoinflammatory disease. The Kawasaki-like disease seen in children with SARS-CoV-2 exposure might be another example of similar mechanism.
Collapse
Affiliation(s)
- Chaim Oscar Jacob
- Department of Medicine, Division of Rheumatology and Immunology, Keck School of Medicine, University of Southern California, 2110 Zonal Ave, HMR 705 Los Angeles, CA, USA.
| |
Collapse
|
38
|
Lichota A, Szewczyk EM, Gwozdzinski K. Factors Affecting the Formation and Treatment of Thrombosis by Natural and Synthetic Compounds. Int J Mol Sci 2020; 21:E7975. [PMID: 33121005 PMCID: PMC7663413 DOI: 10.3390/ijms21217975] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 12/13/2022] Open
Abstract
Venous thromboembolism (VTE) refers to deep vein thrombosis (DVT), whose consequence may be a pulmonary embolism (PE). Thrombosis is associated with significant morbidity and mortality and is the third most common cardiovascular disease after myocardial infarction and stroke. DVT is associated with the formation of a blood clot in a deep vein in the body. Thrombosis promotes slowed blood flow, hypoxia, cell activation, and the associated release of many active substances involved in blood clot formation. All thrombi which adhere to endothelium consist of fibrin, platelets, and trapped red and white blood cells. In this review, we summarise the impact of various factors affecting haemostatic disorders leading to blood clot formation. The paper discusses the causes of thrombosis, the mechanism of blood clot formation, and factors such as hypoxia, the involvement of endothelial cells (ECs), and the activation of platelets and neutrophils along with the effects of bacteria and reactive oxygen species (ROS). Mechanisms related to the action of anticoagulants affecting coagulation factors including antiplatelet drugs have also been discussed. However, many aspects related to the pathogenesis of thrombosis still need to be clarified. A review of the drugs used to treat and prevent thrombosis and natural anticoagulants that occur in the plant world and are traditionally used in Far Eastern medicine has also been carried out.
Collapse
Affiliation(s)
- Anna Lichota
- Department of Pharmaceutical Microbiology and Microbiological Diagnostic, Faculty of Pharmacy, Medical University of Lodz, 90-235 Lodz, Poland; (A.L.); (E.M.S.)
| | - Eligia M. Szewczyk
- Department of Pharmaceutical Microbiology and Microbiological Diagnostic, Faculty of Pharmacy, Medical University of Lodz, 90-235 Lodz, Poland; (A.L.); (E.M.S.)
| | - Krzysztof Gwozdzinski
- Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland
| |
Collapse
|
39
|
Xin J, Zhang H, He Y, Duren Z, Bai C, Chen L, Luo X, Yan DS, Zhang C, Zhu X, Yuan Q, Feng Z, Cui C, Qi X, Ouzhuluobu, Wong WH, Wang Y, Su B. Chromatin accessibility landscape and regulatory network of high-altitude hypoxia adaptation. Nat Commun 2020; 11:4928. [PMID: 33004791 PMCID: PMC7529806 DOI: 10.1038/s41467-020-18638-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 09/03/2020] [Indexed: 12/27/2022] Open
Abstract
High-altitude adaptation of Tibetans represents a remarkable case of natural selection during recent human evolution. Previous genome-wide scans found many non-coding variants under selection, suggesting a pressing need to understand the functional role of non-coding regulatory elements (REs). Here, we generate time courses of paired ATAC-seq and RNA-seq data on cultured HUVECs under hypoxic and normoxic conditions. We further develop a variant interpretation methodology (vPECA) to identify active selected REs (ASREs) and associated regulatory network. We discover three causal SNPs of EPAS1, the key adaptive gene for Tibetans. These SNPs decrease the accessibility of ASREs with weakened binding strength of relevant TFs, and cooperatively down-regulate EPAS1 expression. We further construct the downstream network of EPAS1, elucidating its roles in hypoxic response and angiogenesis. Collectively, we provide a systematic approach to interpret phenotype-associated noncoding variants in proper cell types and relevant dynamic conditions, to model their impact on gene regulation.
Collapse
Affiliation(s)
- Jingxue Xin
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, China
- CEMS, NCMIS, MDIS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, 100190, Beijing, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, 650223, Kunming, China
- Bio-X Program, Stanford University, Stanford, CA, 94305, USA
- University of Chinese Academy of Sciences, 100101, Beijing, China
| | - Hui Zhang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, 650223, Kunming, China
| | - Yaoxi He
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, 650223, Kunming, China
- University of Chinese Academy of Sciences, 100101, Beijing, China
| | - Zhana Duren
- Departments of Statistics, Stanford University, Stanford, CA, 94305, USA
- Center for Human Genetics and Department of Genetics and Biochemistry, Clemson University, Greenwood, SC, 29646, USA
| | - Caijuan Bai
- High Altitude Medical Research Center, School of Medicine, Tibetan University, 850000, Lhasa, China
| | - Lang Chen
- CEMS, NCMIS, MDIS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, 100190, Beijing, China
- University of Chinese Academy of Sciences, 100101, Beijing, China
| | - Xin Luo
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, 650223, Kunming, China
- University of Chinese Academy of Sciences, 100101, Beijing, China
| | - Dong-Sheng Yan
- School of Mathematical Science, Inner Mongolia University, 010021, Huhhot, China
| | - Chaoyu Zhang
- CEMS, NCMIS, MDIS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, 100190, Beijing, China
- University of Chinese Academy of Sciences, 100101, Beijing, China
| | - Xiang Zhu
- Departments of Statistics, Stanford University, Stanford, CA, 94305, USA
| | - Qiuyue Yuan
- CEMS, NCMIS, MDIS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, 100190, Beijing, China
- University of Chinese Academy of Sciences, 100101, Beijing, China
| | - Zhanying Feng
- CEMS, NCMIS, MDIS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, 100190, Beijing, China
- University of Chinese Academy of Sciences, 100101, Beijing, China
| | - Chaoying Cui
- High Altitude Medical Research Center, School of Medicine, Tibetan University, 850000, Lhasa, China
| | - Xuebin Qi
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, 650223, Kunming, China
| | - Ouzhuluobu
- High Altitude Medical Research Center, School of Medicine, Tibetan University, 850000, Lhasa, China
| | - Wing Hung Wong
- Bio-X Program, Stanford University, Stanford, CA, 94305, USA.
- Departments of Statistics, Stanford University, Stanford, CA, 94305, USA.
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Yong Wang
- CEMS, NCMIS, MDIS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, 100190, Beijing, China.
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, 650223, Kunming, China.
- University of Chinese Academy of Sciences, 100101, Beijing, China.
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 330106, Hangzhou, China.
| | - Bing Su
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, China.
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, 650223, Kunming, China.
| |
Collapse
|
40
|
Java A, Apicelli AJ, Liszewski MK, Coler-Reilly A, Atkinson JP, Kim AH, Kulkarni HS. The complement system in COVID-19: friend and foe? JCI Insight 2020; 5:140711. [PMID: 32554923 PMCID: PMC7455060 DOI: 10.1172/jci.insight.140711] [Citation(s) in RCA: 246] [Impact Index Per Article: 49.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19), the disease caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has resulted in a global pandemic and a disruptive health crisis. COVID-19-related morbidity and mortality have been attributed to an exaggerated immune response. The role of complement activation and its contribution to illness severity is being increasingly recognized. Here, we summarize current knowledge about the interaction of coronaviruses with the complement system. We posit that (a) coronaviruses activate multiple complement pathways; (b) severe COVID-19 clinical features often resemble complementopathies; (c) the combined effects of complement activation, dysregulated neutrophilia, endothelial injury, and hypercoagulability appear to be intertwined to drive the severe features of COVID-19; (d) a subset of patients with COVID-19 may have a genetic predisposition associated with complement dysregulation; and (e) these observations create a basis for clinical trials of complement inhibitors in life-threatening illness.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Hrishikesh S. Kulkarni
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| |
Collapse
|
41
|
Yolgösteren A, Leba LK, Demir AB. Investigation of chronic venous insufficiency in patients with sleep disorders due to restless legs syndrome. Phlebology 2020; 35:771-776. [PMID: 32664803 DOI: 10.1177/0268355520940921] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND We aimed to investigate of chronic venous insufficiency on patients with sleep disorder due to restless legs syndrome. METHOD Five hundred forty-one cases on whom polysomnography was performed due to sleep disorder were evaluated retrospectively. Forty patients with restless legs syndrome were determined. They were examined by history, physical examination, and duplex ultrasonography in terms of chronic venous insufficiency. The sleep stage rates of both groups were compared (that the rate of total sleep time in polysomnography to sleep stages is expressed as minute and percentage has been defined as sleep stage rate). RESULT Chronic venous insufficiency was identified in 20 out of 40 patients (group 1; female, 90%). In group 2, there were patients with only restless legs syndrome but with no chronic venous insufficiency (female, 80%). The mean ages of both groups were 56.4 ± 11.8 and 54.3 ± 14.7 years. Stage 1 sleep rate in group 1 was 5% ± 2.7 and in group 2 was 8% ± 3.8 (p = 0.006). Periodic limb movement index (polysomnography finding evaluating involuntary leg movements during sleep) was 11.4 ± 17.5 in group 1, and it was 29.4 ± 37.9 in group 2 (p = 0.006). CONCLUSION We recommend that chronic venous insufficiency should be investigated in patients with primary restless legs syndrome diagnosis.
Collapse
Affiliation(s)
- Atıf Yolgösteren
- Faculty of Medicine, Department of Cardiovascular Surgery, Bursa Uludağ University, Bursa, Turkey
| | - Leyla Köse Leba
- Faculty of Medicine, Department of Neurology, Necmettin Erbakan University, Konya, Turkey
| | - Aylin Bican Demir
- Faculty of Medicine, Department of Neurology, Bursa Uludağ University, Bursa, Turkey
| |
Collapse
|
42
|
Hu X, Li Y, Li J, Chen H. Effects of altered blood flow induced by the muscle pump on thrombosis in a microfluidic venous valve model. LAB ON A CHIP 2020; 20:2473-2481. [PMID: 32543635 DOI: 10.1039/d0lc00287a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Deep vein thrombosis (DVT) often occurs in the lower limb veins of bedridden patients and greatly reduces the quality of life. The altered blood flow in venous valves induced by the insufficient efficacy of the muscle pump is commonly considered as a main factor. However, it is still a great challenge to observe the altered blood flow in real time, and its role in the formation of thrombi is poorly understood. Here we make a microfluidic venous valve model with flexible leaflets in a deformable channel that can mimic the motion of valves and the compression of vessels by muscle contraction, and identify the stasis and intermittent reflux in the valve pocket generated by the muscle pump. A thrombus forms in the stasis flow, while the intermittent reflux removes the fibrin and inhibits the growth of the thrombus. A flexible microfluidic device that can mimic the motion of valves and the contraction of vessels would have wide applications in the research on cardiovascular diseases.
Collapse
Affiliation(s)
- Xiangyu Hu
- State Key Laboratory of Tribology, Mechanical Engineering Department, Tsinghua University, Beijing, 100084, China.
| | | | | | | |
Collapse
|
43
|
Gant P, McBride D, Humm K. Abnormal platelet activity in dogs and cats - impact and measurement. J Small Anim Pract 2020; 61:3-18. [PMID: 31919851 DOI: 10.1111/jsap.13092] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 05/06/2019] [Accepted: 10/28/2019] [Indexed: 01/08/2023]
Abstract
Abnormal platelet activity can either lead to bleeding tendencies or inappropriate thrombus formation and can occur secondarily to a wide variety of disease processes, with a range of clinical consequences and severity. This article will discuss the pathophysiology of platelet function abnormalities and consider a logical diagnostic approach applicable to veterinary practice. Recent advances in platelet function testing will then be discussed, with regards to detection of platelet dysfunction and tailoring of pharmacological manipulation. Although many of these tests are still confined to research or academic institutions, techniques for indirectly assessing platelet function are starting to become more widely available. Although we still require further research to develop guidelines for the use of these tests in clinical decision-making, the recent advances in this field are an exciting step forward in being able to detect and manage platelet dysfunction in both primary care and referral practice.
Collapse
Affiliation(s)
- P Gant
- Queen Mother Hospital for Animals (QMHA), The Royal Veterinary College, Hatfield, Hertfordshire, AL9 7TA, UK
| | - D McBride
- Queen Mother Hospital for Animals (QMHA), The Royal Veterinary College, Hatfield, Hertfordshire, AL9 7TA, UK
| | - K Humm
- Queen Mother Hospital for Animals (QMHA), The Royal Veterinary College, Hatfield, Hertfordshire, AL9 7TA, UK
| |
Collapse
|
44
|
Chung JJ, Han J, Wang LL, Arisi MF, Zaman S, Gordon J, Li E, Kim ST, Tran Z, Chen CW, Gaffey AC, Burdick JA, Atluri P. Delayed delivery of endothelial progenitor cell-derived extracellular vesicles via shear thinning gel improves postinfarct hemodynamics. J Thorac Cardiovasc Surg 2020; 159:1825-1835.e2. [PMID: 31353103 PMCID: PMC7077034 DOI: 10.1016/j.jtcvs.2019.06.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 05/04/2019] [Accepted: 06/04/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND Extracellular vesicles (EVs) are promising therapeutics for cardiovascular disease, but poorly-timed delivery might hinder efficacy. We characterized the time-dependent response to endothelial progenitor cell (EPC)-EVs within an injectable shear-thinning hydrogel (STG+EV) post-myocardial infarction (MI) to identify when an optimal response is achieved. METHODS The angiogenic effects of prolonged hypoxia on cell response to EPC-EV therapy and EV uptake affinity were tested in vitro. A rat model of acute MI via left anterior descending artery ligation was created and STG+EV was delivered via intramyocardial injections into the infarct border zone at time points corresponding to phases of post-MI inflammation: 0 hours (immediate), 3 hours (acute inflammation), 4 days (proliferative), and 2 weeks (fibrosis). Hemodynamics 4 weeks post-treatment were compared across treatment and control groups (phosphate buffered saline [PBS], shear-thinning gel). Scar thickness and ventricular diameter were assessed histologically. The primary hemodynamic end point was end systolic elastance. The secondary end point was scar thickness. RESULTS EPC-EVs incubated with chronically versus acutely hypoxic human umbilical vein endothelial cells resulted in a 2.56 ± 0.53 versus 1.65 ± 0.15-fold increase (P = .05) in a number of vascular meshes and higher uptake of EVs over 14 hours. End systolic elastance improved with STG+EV therapy at 4 days (0.54 ± 0.08) versus PBS or shear-thinning gel (0.26 ± 0.03 [P = .02]; 0.23 ± 0.02 [P = .01]). Preservation of ventricular diameter (6.20 ± 0.73 mm vs 8.58 ± 0.38 mm [P = .04]; 9.13 ± 0.25 mm [P = .01]) and scar thickness (0.89 ± 0.05 mm vs 0.62 ± 0.03 mm [P < .0001] and 0.58 ± 0.05 mm [P < .0001]) was significantly greater at 4 days, compared wit PBS and shear-thinning gel controls. CONCLUSIONS Delivery of STG+EV 4 days post-MI improved left ventricular contractility and preserved global ventricular geometry, compared with controls and immediate therapy post-MI. These findings suggest other cell-derived therapies can be optimized by strategic timing of therapeutic intervention.
Collapse
Affiliation(s)
- Jennifer J Chung
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pa; Department of Bioengineering, University of Pennsylvania, Philadelphia, Pa
| | - Jason Han
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pa; Department of Bioengineering, University of Pennsylvania, Philadelphia, Pa
| | - Leo L Wang
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pa; Department of Bioengineering, University of Pennsylvania, Philadelphia, Pa
| | - Maria F Arisi
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pa; Department of Bioengineering, University of Pennsylvania, Philadelphia, Pa
| | - Samir Zaman
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pa; Department of Bioengineering, University of Pennsylvania, Philadelphia, Pa
| | - Jonathan Gordon
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pa; Department of Bioengineering, University of Pennsylvania, Philadelphia, Pa
| | - Elizabeth Li
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pa; Department of Bioengineering, University of Pennsylvania, Philadelphia, Pa
| | - Samuel T Kim
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pa; Department of Bioengineering, University of Pennsylvania, Philadelphia, Pa
| | - Zoe Tran
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pa; Department of Bioengineering, University of Pennsylvania, Philadelphia, Pa
| | - Carol W Chen
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pa; Department of Bioengineering, University of Pennsylvania, Philadelphia, Pa
| | - Ann C Gaffey
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pa; Department of Bioengineering, University of Pennsylvania, Philadelphia, Pa
| | - Jason A Burdick
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pa; Department of Bioengineering, University of Pennsylvania, Philadelphia, Pa
| | - Pavan Atluri
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pa; Department of Bioengineering, University of Pennsylvania, Philadelphia, Pa.
| |
Collapse
|
45
|
Tabata Y, Yoshino D, Funamoto K, Koens R, Kamm RD, Funamoto K. Migration of vascular endothelial cells in monolayers under hypoxic exposure. Integr Biol (Camb) 2020; 11:26-35. [PMID: 31584068 DOI: 10.1093/intbio/zyz002] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 11/26/2018] [Accepted: 01/02/2019] [Indexed: 11/13/2022]
Abstract
The hypoxic microenvironment existing in vivo is known to significantly affect cell morphology and dynamics, and cell group behaviour. Collective migration of vascular endothelial cells is essential for vasculogenesis and angiogenesis, and for maintenance of monolayer integrity. Although hypoxic stress increases vascular endothelial permeability, the changes in collective migration and intracellular junction morphology of vascular endothelial cells remain poorly understood. This study reveals the migration of confluent vascular endothelial cells and changes in their adherens junction, as reflected by changes in the vascular endothelial (VE)-cadherin distribution, under hypoxic exposure. Vascular endothelial monolayers of human umbilical vein endothelial cells (HUVECs) were formed in microfluidic devices with controllability of oxygen tension. The oxygen tension was set to either normoxia (21% O2) or hypoxia (<3% O2) by supplying gas mixtures into separate gas channels. The migration velocity of HUVECs was measured using particle image velocimetry with a time series of phase-contrast microscopic images of the vascular endothelial monolayers. Hypoxia inducible factor-1α (HIF-1α) and VE-cadherin in HUVECs were observed after exposure to normoxic or hypoxic conditions using immunofluorescence staining and quantitative confocal image analysis. Changes in the migration speed of HUVECs were observed in as little as one hour after exposure to hypoxic condition, showing that the migration speed was increased 1.4-fold under hypoxia compared to that under normoxia. Nuclear translocation of HIF-1α peaked after the hypoxic gas mixture was supplied for 2 h. VE-cadherin expression was also found to be reduced. When ethanol was added to the cell culture medium, cell migration increased. By contrast, by strengthening VE-cadherin junctions with forskolin, cell migration decreased gradually in spite the effect of ethanol to stimulate migration. These results indicate that the increase of cell migration by hypoxic exposure was attributable to loosening of intercellular junction resulting from the decrease of VE-cadherin expression.
Collapse
Affiliation(s)
- Yugo Tabata
- Graduate School of Biomedical Engineering, Tohoku University, 6-6-12 Aramaki-aza Aoba, Aoba-ku, Sendai, Miyagi, Japan
| | - Daisuke Yoshino
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, 6-3 Aramaki-aza Aoba, Aoba-ku, Sendai, Miyagi, Japan
| | - Kiyoe Funamoto
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, 6-3 Aramaki-aza Aoba, Aoba-ku, Sendai, Miyagi, Japan
| | - Rei Koens
- Graduate School of Biomedical Engineering, Tohoku University, 6-6-12 Aramaki-aza Aoba, Aoba-ku, Sendai, Miyagi, Japan
| | - Roger D Kamm
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA, USA
| | - Kenichi Funamoto
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, 6-3 Aramaki-aza Aoba, Aoba-ku, Sendai, Miyagi, Japan
| |
Collapse
|
46
|
Li TT, Liu MR, Pei DS. Friend or foe, the role of EGR-1 in cancer. Med Oncol 2019; 37:7. [PMID: 31748910 DOI: 10.1007/s12032-019-1333-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 11/11/2019] [Indexed: 12/18/2022]
Abstract
Early growth response-1 (EGR-1), also termed NEFI-A and Krox-24, as a multi-domain protein is implicated in several vital physiological processes, including development, metabolism, cell growth and proliferation. Previous studies have implied that EGR-1 was producing in response to the tissue injury, immune response and fibrosis. Meanwhile, emerging studies stressed the pronounced correlation of EGR-1 and human cancers. Nevertheless, the intricate mechanisms of cancer-reduce EGR-1 alteration still poorly characterized. In the review, we evaluated the effects of EGR-1 in tumor cell proliferation, apoptosis, migration, invasion and tumor microenvironment, and then, we dwell on the intricate signaling pathways that EGR-1 involved in. The aberrantly expressed of EGR-1 in cancers are expected to provide a new cancer therapy strategy or a new marker for assessing treatment efficacy.
Collapse
Affiliation(s)
- Tong-Tong Li
- Department of Pathology, Xuzhou Medical University, 209 Tong-shan Road, Xuzhou, 221004, Jiangsu, People's Republic of China
| | - Man-Ru Liu
- Department of Pathology, Xuzhou Medical University, 209 Tong-shan Road, Xuzhou, 221004, Jiangsu, People's Republic of China
| | - Dong-Sheng Pei
- Department of Pathology, Xuzhou Medical University, 209 Tong-shan Road, Xuzhou, 221004, Jiangsu, People's Republic of China.
| |
Collapse
|
47
|
Papah MB, Abasht B. Dysregulation of lipid metabolism and appearance of slow myofiber-specific isoforms accompany the development of Wooden Breast myopathy in modern broiler chickens. Sci Rep 2019; 9:17170. [PMID: 31748687 PMCID: PMC6868161 DOI: 10.1038/s41598-019-53728-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 11/05/2019] [Indexed: 01/05/2023] Open
Abstract
Previous transcriptomic studies have hypothesized the occurrence of slow myofiber-phenotype, and dysregulation of lipid metabolism as being associated with the development of Wooden Breast (WB), a meat quality defect in commercial broiler chickens. To gain a deep understanding of the manifestation and implication of these two biological processes in health and disease states in chickens, cellular and global expression of specific genes related to the respective processes were examined in pectoralis major muscles of modern fast-growing and unselected slow-growing chickens. Using RNA in situ hybridization, lipoprotein lipase (LPL) was found to be expressed in endothelial cells of capillaries and small-caliber veins in chickens. RNA-seq analysis revealed upregulation of lipid-related genes in WB-affected chickens at week 3 and downregulation at week 7 of age. On the other hand, cellular localization of slow myofiber-type genes revealed their increased expression in mature myofibers of WB-affected chickens. Similarly, global expression of slow myofiber-type genes showed upregulation in affected chickens at both timepoints. To our knowledge, this is the first study to show the expression of LPL from the vascular endothelium in chickens. This study also confirms the existence of slow myofiber-phenotype and provides mechanistic insights into increased lipid uptake and metabolism in WB disease process.
Collapse
Affiliation(s)
- Michael B Papah
- Department of Animal and Food Sciences, University of Delaware, Delaware, DE, USA
| | - Behnam Abasht
- Department of Animal and Food Sciences, University of Delaware, Delaware, DE, USA.
| |
Collapse
|
48
|
Li Y, Yu Y, VanderBeek BL. Anaemia and the risk of progression from non-proliferative diabetic retinopathy to vision threatening diabetic retinopathy. Eye (Lond) 2019; 34:934-941. [PMID: 31586167 DOI: 10.1038/s41433-019-0617-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 07/13/2019] [Accepted: 07/28/2019] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND/AIMS To determine if anaemia and oxygen delivery-related co-morbidities (ODCs) affect progression from non-proliferative diabetic retinopathy (NPDR) to vision-threatening diabetic retinopathy (VTDR). METHODS This is a retrospective cohort study using medical claims data from a large US insurer. Cohorts were created from all NPDR patients between 2002 and 2016. Primary exclusion criteria consisted of any previous diagnosis of proliferative diabetic retinopathy (PDR), diabetic macular oedema (DME) or treatment used in the care of VTDR. The main outcome was a new diagnosis of VTDR (DME or PDR), PDR, or DME. A time-dependent, multivariate Cox proportional hazard regression was used to determine the association between anaemia and other ODCs with NPDR progression. RESULTS Of the total 69,982 NPDR patients included for analysis, 12,270, 2,162, and 10,322 progressed to VTDR, PDR and DME, respectively. Both mild and moderate/severe (mod/sev) anaemia were associated with an increased hazard for progression to VTDR (mild HR:1.10, 95% CI:1.04-1.16, p < 0.001; mod/sev HR:1.20, 95% CI:1.12-1.29, p < 0.001), PDR (mild HR:1.29, 95% CI:1.13-1.46, p < 0.001; mod/sev HR:1.43, 95% CI:1.21-1.69, p < 0.001), and DME (mild HR:1.06, 95% CI:1.00-1.13, p < 0.001; mod/sev HR:1.14, 95% CI:1.05-1.24, p < 0.001). ODCs such as chronic pulmonary disease and history of blood disorder/cancer were also significantly associated with an increased hazard for NPDR progression (HR > 1.00, p < 0.001 for all comparisons). CONCLUSIONS Anaemia, independent of kidney disease, appears to play a significant role in progression from NPDR to VTDR, PDR, or DME. Concurrently, association of ODCs with NPDR progression lends support to the underlying mechanisms of anaemia in the pathogenesis of diabetic retinopathy.
Collapse
Affiliation(s)
- Yafeng Li
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Yinxi Yu
- Center for Preventive Ophthalmology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Brian L VanderBeek
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA. .,Center for Clinical Epidemiology and Biostatistics, Department of Biostatistics & Epidemiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA. .,Leonard Davis Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
49
|
Binotti WW, Romano AC. Projection-Resolved Optical Coherence Tomography Angiography Parameters to Determine Severity in Diabetic Retinopathy. Invest Ophthalmol Vis Sci 2019; 60:1321-1327. [PMID: 30933259 DOI: 10.1167/iovs.18-24154] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose The purpose of this study was to assess projection-resolved optical coherence tomography angiography (PR-OCTA) vessel density (VD) and foveal avascular zone (FAZ) in determining severity within diabetic retinopathy (DR) and their accuracy in identifying high-risk DR patients. Methods This was a retrospective study with 72 eyes of 52 DR patients, assessing the VD and FAZ area of the superficial capillary plexus (SCP) and deep vascular plexus (DVP), for both 3 × 3-mm and 6 × 6-mm scans between the DR groups (mild to moderate, severe and proliferative DR [PDR]). For accuracy, the severe and PDR groups were merged, representing the high-risk DR group for receiver operator characteristic analysis. VD of OCTA images with and without PR were compared. Results In mild to moderate, severe, and PDR groups, there were 31, 21, and 20 eyes, respectively. PR-OCTA improved VD analysis only in the DVP and particularly in advanced DR stages (P = 0.042). In the 3 × 3-mm PR scans, all superficial and deep parameters were significantly different between severe and PDR groups (P ≤ 0.020), but only the mean VD of SCP and DVP was also significant between the mild to moderate and severe groups (P ≤ 0.007). In the 6 × 6-mm scans, the superficial VD, deep VD, and superficial FAZ were significantly different between the severe and PDR groups (P ≤ 0.029). The superficial VD and deep VD of the 3 × 3-mm scans were good parameters for detecting high-risk patients (area under the curve = 0.829 and 0.895, respectively). Conclusions PR-OCTA improved VD analysis of DVP. The 3 × 3-mm SCP and DVP VD were the most accurate in detecting high-risk DR.
Collapse
|
50
|
Microfluidic Collective Cell Migration Assay for Study of Endothelial Cell Proliferation and Migration under Combinations of Oxygen Gradients, Tensions, and Drug Treatments. Sci Rep 2019; 9:8234. [PMID: 31160651 PMCID: PMC6546762 DOI: 10.1038/s41598-019-44594-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 05/20/2019] [Indexed: 01/28/2023] Open
Abstract
Proliferation and migration of endothelial cells play an important role in many biological activities, and they can be regulated by various microenvironmental factors. In this paper, a novel microfluidic collective cell migration assay is developed to study endothelial cell migration and proliferation under combinations of three oxygen conditions: normoxia, oxygen gradient, and hypoxia and three medium compositions: normal growth medium, the medium with cytochalasin-D for actin polymerization inhibition, and with YC-1 for hypoxia-inducible factor (HIF) inhibition. The microfluidic device designed in the paper allows cell patterns formed with consistent dimensions using laminar flow patterning. In addition, stable oxygen gradients can be generated within the device by a spatially confined chemical reaction method. The device can be operated in conventional cell incubators with minimal chemical reagents and instrumentation for practical applications. The results show directional collective cell migration of the endothelial cells under the oxygen gradients for all the medium compositions. The directional behavior has never been discussed before, and indicates critical roles of oxygen gradients in guiding endothelial cell migration during various biological activities. The developed assay provides a practical yet powerful tool for further in vitro study of endothelial cell behaviors under various physiological microenvironments.
Collapse
|