1
|
Vandal M, Institoris A, Reveret L, Korin B, Gunn C, Hirai S, Jiang Y, Lee S, Lee J, Bourassa P, Mishra RC, Peringod G, Arellano F, Belzil C, Tremblay C, Hashem M, Gorzo K, Elias E, Yao J, Meilandt B, Foreman O, Roose-Girma M, Shin S, Muruve D, Nicola W, Körbelin J, Dunn JF, Chen W, Park SK, Braun AP, Bennett DA, Gordon GRJ, Calon F, Shaw AS, Nguyen MD. Loss of endothelial CD2AP causes sex-dependent cerebrovascular dysfunction. Neuron 2025:S0896-6273(25)00010-8. [PMID: 39892386 DOI: 10.1016/j.neuron.2025.01.006] [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: 09/06/2022] [Revised: 08/27/2024] [Accepted: 01/09/2025] [Indexed: 02/03/2025]
Abstract
Polymorphisms in CD2-associated protein (CD2AP) predispose to Alzheimer's disease (AD), but the underlying mechanisms remain unknown. Here, we show that loss of CD2AP in cerebral blood vessels is associated with cognitive decline in AD subjects and that genetic downregulation of CD2AP in brain vascular endothelial cells impairs memory function in male mice. Animals with reduced brain endothelial CD2AP display altered blood flow regulation at rest and during neurovascular coupling, defects in mural cell activity, and an abnormal vascular sex-dependent response to Aβ. Antagonizing endothelin-1 receptor A signaling partly rescues the vascular impairments, but only in male mice. Treatment of CD2AP mutant mice with reelin glycoprotein that mitigates the effects of CD2AP loss function via ApoER2 increases resting cerebral blood flow and even protects male mice against the noxious effect of Aβ. Thus, endothelial CD2AP plays critical roles in cerebrovascular functions and represents a novel target for sex-specific treatment in AD.
Collapse
Affiliation(s)
- Milène Vandal
- Departments of Clinical Neurosciences, Cell Biology and Anatomy, and Biochemistry and Molecular Biology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1 Canada
| | - Adam Institoris
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary AB T2N 4N1, Canada
| | - Louise Reveret
- Faculté de pharmacie, Université Laval, Québec, QC G1V 0A6, Canada; Centre de Hospitalier Universitaire de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada
| | - Ben Korin
- Department of Research Biology, Genentech, South San Francisco, CA 94080, USA
| | - Colin Gunn
- Departments of Clinical Neurosciences, Cell Biology and Anatomy, and Biochemistry and Molecular Biology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1 Canada
| | - Sotaro Hirai
- Departments of Clinical Neurosciences, Cell Biology and Anatomy, and Biochemistry and Molecular Biology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1 Canada
| | - Yulan Jiang
- Departments of Clinical Neurosciences, Cell Biology and Anatomy, and Biochemistry and Molecular Biology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1 Canada
| | - Sukyoung Lee
- Departments of Clinical Neurosciences, Cell Biology and Anatomy, and Biochemistry and Molecular Biology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1 Canada
| | - Jiyeon Lee
- Department of Research Biology, Genentech, South San Francisco, CA 94080, USA
| | - Philippe Bourassa
- Faculté de pharmacie, Université Laval, Québec, QC G1V 0A6, Canada; Centre de Hospitalier Universitaire de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada
| | - Ramesh C Mishra
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary AB T2N 4N1, Canada
| | - Govind Peringod
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary AB T2N 4N1, Canada
| | - Faye Arellano
- Departments of Clinical Neurosciences, Cell Biology and Anatomy, and Biochemistry and Molecular Biology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1 Canada
| | - Camille Belzil
- Departments of Clinical Neurosciences, Cell Biology and Anatomy, and Biochemistry and Molecular Biology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1 Canada
| | - Cyntia Tremblay
- Centre de Hospitalier Universitaire de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada
| | - Mada Hashem
- Department of Radiology, Hotchkiss Brain Institute, University of Calgary, Calgary AB T2N 4N1, Canada
| | - Kelsea Gorzo
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary AB T2N 4N1, Canada
| | - Esteban Elias
- Department of Medicine, Snyder Institute for Chronic Diseases, University of Calgary, Calgary AB T2N 4N1, Canada
| | - Jinjing Yao
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary AB T2N 4N1, Canada
| | - Bill Meilandt
- Department of Research Biology, Genentech, South San Francisco, CA 94080, USA
| | - Oded Foreman
- Department of Research Biology, Genentech, South San Francisco, CA 94080, USA
| | - Meron Roose-Girma
- Department of Research Biology, Genentech, South San Francisco, CA 94080, USA
| | - Steven Shin
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary AB T2N 4N1, Canada
| | - Daniel Muruve
- Department of Medicine, Snyder Institute for Chronic Diseases, University of Calgary, Calgary AB T2N 4N1, Canada
| | - Wilten Nicola
- Departments of Cell Biology and Anatomy, Hotchkiss Brain Institute, University of Calgary, Calgary AB T2N 4N1, Canada
| | - Jakob Körbelin
- Department of Oncology, Hematology and Bone Marrow Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany
| | - Jeff F Dunn
- Departments of Clinical Neurosciences, Cell Biology and Anatomy, and Biochemistry and Molecular Biology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1 Canada; Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary AB T2N 4N1, Canada; Department of Radiology, Hotchkiss Brain Institute, University of Calgary, Calgary AB T2N 4N1, Canada
| | - Wayne Chen
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary AB T2N 4N1, Canada
| | - Sang-Ki Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Andrew P Braun
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary AB T2N 4N1, Canada
| | - David A Bennett
- Rush Alzheimer's disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Grant R J Gordon
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary AB T2N 4N1, Canada
| | - Frédéric Calon
- Faculté de pharmacie, Université Laval, Québec, QC G1V 0A6, Canada; Centre de Hospitalier Universitaire de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada.
| | - Andrey S Shaw
- Department of Research Biology, Genentech, South San Francisco, CA 94080, USA.
| | - Minh Dang Nguyen
- Departments of Clinical Neurosciences, Cell Biology and Anatomy, and Biochemistry and Molecular Biology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1 Canada.
| |
Collapse
|
2
|
Fakhari S, Campolina‐Silva G, Asayesh F, Girardet L, Scott‐Boyer M, Droit A, Soulet D, Greener J, Belleannée C. Shear stress effects on epididymal epithelial cell via primary cilia mechanosensory signaling. J Cell Physiol 2025; 240:e31475. [PMID: 39508588 PMCID: PMC11733861 DOI: 10.1002/jcp.31475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/24/2024] [Accepted: 10/17/2024] [Indexed: 11/15/2024]
Abstract
Shear stress, resulting from fluid flow, is a fundamental mechanical stimulus affecting various cellular functions. The epididymis, essential for sperm maturation, offers a compelling model to study the effects of shear stress on cellular behavior. This organ undergoes extensive proliferation and differentiation until puberty, achieving full functionality as spermatozoa commence their post-testicular maturation. Although the mechanical tension exerted by testicular fluid is hypothesized to drive epithelial proliferation and differentiation, the precise mechanisms remain unclear. Here we assessed whether the responsiveness of the epididymal cells to shear stress depends on functional primary cilia by combining microfluidic strategies on immortalized epididymal cells, calcium signaling assays, and high-throughput gene expression analysis. We identified 97 genes overexpressed in response to shear stress, including early growth response (Egr) 2/3, cellular communication network factor (Ccn) 1/2, and Fos proto-oncogene (Fos). While shear stress triggered a rapid increase of intracellular Ca2+, this response was abrogated following the impairment of primary ciliogenesis through pharmacological and siRNA approaches. Overall, our findings provide valuable insights into how mechanical forces influence the development of the male reproductive system, a requisite to sperm maturation.
Collapse
Affiliation(s)
- Sepideh Fakhari
- Department of Obstetrics, Gynecology, and Reproduction, Centre de recherche en Reproduction, Développement et Santé IntergénérationnelleFaculty of MedicineQuébec CityQuebecCanada
- Centre de recherche du centre hospitalier universitaire de Québec ‐ Université LavalQuébec CityQuebecCanada
- Department of ChemistryFaculty of Science and EngineeringQuébec CityQuebecCanada
| | - Gabriel Campolina‐Silva
- Department of Obstetrics, Gynecology, and Reproduction, Centre de recherche en Reproduction, Développement et Santé IntergénérationnelleFaculty of MedicineQuébec CityQuebecCanada
- Centre de recherche du centre hospitalier universitaire de Québec ‐ Université LavalQuébec CityQuebecCanada
| | - Farnaz Asayesh
- Department of Obstetrics, Gynecology, and Reproduction, Centre de recherche en Reproduction, Développement et Santé IntergénérationnelleFaculty of MedicineQuébec CityQuebecCanada
| | - Laura Girardet
- Department of Obstetrics, Gynecology, and Reproduction, Centre de recherche en Reproduction, Développement et Santé IntergénérationnelleFaculty of MedicineQuébec CityQuebecCanada
- Centre de recherche du centre hospitalier universitaire de Québec ‐ Université LavalQuébec CityQuebecCanada
| | - Marie‐Pier Scott‐Boyer
- Proteomics Platform, Québec Genomic Center, Université Laval, CHU de Québec Research Center (CHUL)Québec CityQuebecCanada
| | - Arnaud Droit
- Proteomics Platform, Québec Genomic Center, Université Laval, CHU de Québec Research Center (CHUL)Québec CityQuebecCanada
| | - Denis Soulet
- Centre de recherche du centre hospitalier universitaire de Québec ‐ Université LavalQuébec CityQuebecCanada
- Faculté de pharmacieUniversité LavalQuébec CityQuebecCanada
| | - Jesse Greener
- Centre de recherche du centre hospitalier universitaire de Québec ‐ Université LavalQuébec CityQuebecCanada
- Department of ChemistryFaculty of Science and EngineeringQuébec CityQuebecCanada
| | - Clémence Belleannée
- Department of Obstetrics, Gynecology, and Reproduction, Centre de recherche en Reproduction, Développement et Santé IntergénérationnelleFaculty of MedicineQuébec CityQuebecCanada
- Centre de recherche du centre hospitalier universitaire de Québec ‐ Université LavalQuébec CityQuebecCanada
| |
Collapse
|
3
|
Mierke CT. Mechanosensory entities and functionality of endothelial cells. Front Cell Dev Biol 2024; 12:1446452. [PMID: 39507419 PMCID: PMC11538060 DOI: 10.3389/fcell.2024.1446452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 10/04/2024] [Indexed: 11/08/2024] Open
Abstract
The endothelial cells of the blood circulation are exposed to hemodynamic forces, such as cyclic strain, hydrostatic forces, and shear stress caused by the blood fluid's frictional force. Endothelial cells perceive mechanical forces via mechanosensors and thus elicit physiological reactions such as alterations in vessel width. The mechanosensors considered comprise ion channels, structures linked to the plasma membrane, cytoskeletal spectrin scaffold, mechanoreceptors, and junctional proteins. This review focuses on endothelial mechanosensors and how they alter the vascular functions of endothelial cells. The current state of knowledge on the dysregulation of endothelial mechanosensitivity in disease is briefly presented. The interplay in mechanical perception between endothelial cells and vascular smooth muscle cells is briefly outlined. Finally, future research avenues are highlighted, which are necessary to overcome existing limitations.
Collapse
|
4
|
Bhattacharyya A, Barbee KA. Vascular endothelial cell morphology and alignment regulate VEGF-induced endothelial nitric oxide synthase activation. Cytoskeleton (Hoboken) 2024; 81:473-487. [PMID: 38775643 PMCID: PMC11496009 DOI: 10.1002/cm.21872] [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: 11/29/2023] [Revised: 03/12/2024] [Accepted: 03/18/2024] [Indexed: 06/13/2024]
Abstract
Nitric oxide (NO) production by endothelial nitric oxide synthase (eNOS) inhibits platelet and leukocyte adhesion while promoting vasorelaxation in smooth muscle cells. Dysfunctional regulation of eNOS is a hallmark of various vascular pathologies, notably atherosclerosis, often associated with areas of low shear stress on endothelial cells (ECs). While the link between EC morphology and local hemodynamics is acknowledged, the specific impact of EC morphology on eNOS regulation remains unclear. Morphological differences between elongated, aligned ECs and polygonal, randomly oriented ECs correspond to variations in focal adhesion and cytoskeletal organization, suggesting differing levels of cytoskeletal prestress. However, the functional outcomes of cytoskeletal prestress, particularly in the absence of shear stress, are not extensively studied in ECs. Some evidence suggests that elongated ECs exhibit decreased immunogenicity and enhanced NO production. This study aims to elucidate the signaling pathways governing VEGF-stimulated eNOS regulation in the aligned EC phenotype characterized by elongated and aligned cells within a monolayer. Using anisotropic topographic cues, bovine aortic endothelial cells (BAECs) were elongated and aligned, followed by VEGF treatment in the presence or absence of cytoskeletal tension inhibitors. Phosphorylation of eNOS ser1179, AKT ser437 and FAK Tyr397 in response to VEGF challenge were significantly heightened in aligned ECs compared to unaligned ECs. Moreover this response proved to be robustly tied to cytoskeletal tension as evinced by the abrogation of responses in the presence of the myosin II ATPase inhibitor, blebbistatin. Notably, this work demonstrates for the first time the reliance on FAK phosphorylation in VEGF-mediated eNOS activation and the comparatively greater contribution of the cytoskeletal machinery in propagating VEGF-eNOS signaling in aligned and elongated ECs. This research underscores the importance of utilizing appropriate vascular models in drug development and sheds light on potential mechanisms underlying vascular function and pathology that can help inform vascular graft design.
Collapse
Affiliation(s)
- Aparna Bhattacharyya
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, 3141 Market St., Philadelphia, PA 19104, USA
| | - Kenneth A. Barbee
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, 3141 Market St., Philadelphia, PA 19104, USA
| |
Collapse
|
5
|
Martins FS, Dos Santos MAP, Simões MDG, da Silva TB, de Araújo IDA, Silva AS. Active intervals between sets and exercise of resistance exercises potentiate the magnitude of post-exercise hypotension in middle-aged hypertensive women. Eur J Appl Physiol 2024; 124:2629-2637. [PMID: 38613678 DOI: 10.1007/s00421-024-05474-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 03/14/2024] [Indexed: 04/15/2024]
Abstract
BACKGROUND Walking/run promotes greater post-exercise hypotension (PEH) than resistance exercise (RE) which can be associated nitric oxide production. Then, we investigated if adding walking between RE increases PEH. METHODS Thirteen hypertensive women (48 ± 1.9 years; 26.8 ± 0.3 kg/m2) performed five experimental sessions: 1-traditional RE with passive interval (PIBS); 2-RE with active interval between sets (AIBS); 3-RE with active interval between exercises (AIEX); 4-aerobic exercise (AE); 5-no exercise. Blood pressure was measured before and immediately after exercise and every 10 min of a 60-min post-exercise recovery period. Plasma nitrite (NO) and malondialdehyde (MDA) were also measured. RESULTS AIBS, AIEX, and AE sessions promoted systolic PEH (12 ± 1.53; 16 ± 2.13; 18 ± 1.34 mmHg, respectively) and diastolic PEH (8 ± 1.38; 8 ± 0.79; 8 ± 0.58 mmHg). In AIBS, AIEX and AE sessions, significant PEH was noted at 10 or 20 min; in PIBS, PEH was significant only at 40 min. In the PIBS session, NO did not change, but presented higher values after AIBS, AIEX and AE sessions. MDA showed no changes between sessions. CONCLUSIONS Passive or active intervals in resistance exercise promote PEH in hypertensive women. Active intervals shorten the PEH onset time as additional benefit.
Collapse
Affiliation(s)
| | | | - Márcia Devânia Guedes Simões
- Laboratory of Applied Studies in Physical Training to Performance and Health -LETFADS, Federal University of Paraíba, João Pessoa, Brazil
| | | | - Izabhelle Dantas Agra de Araújo
- Laboratory of Applied Studies in Physical Training to Performance and Health -LETFADS, Federal University of Paraíba, João Pessoa, Brazil
| | | |
Collapse
|
6
|
Yu F, Zong B, Ji L, Sun P, Jia D, Wang R. Free Fatty Acids and Free Fatty Acid Receptors: Role in Regulating Arterial Function. Int J Mol Sci 2024; 25:7853. [PMID: 39063095 PMCID: PMC11277118 DOI: 10.3390/ijms25147853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 07/13/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
The metabolic network's primary sources of free fatty acids (FFAs) are long- and medium-chain fatty acids of triglyceride origin and short-chain fatty acids produced by intestinal microorganisms through dietary fibre fermentation. Recent studies have demonstrated that FFAs not only serve as an energy source for the body's metabolism but also participate in regulating arterial function. Excess FFAs have been shown to lead to endothelial dysfunction, vascular hypertrophy, and vessel wall stiffness, which are important triggers of arterial hypertension and atherosclerosis. Nevertheless, free fatty acid receptors (FFARs) are involved in the regulation of arterial functions, including the proliferation, differentiation, migration, apoptosis, inflammation, and angiogenesis of vascular endothelial cells (VECs) and vascular smooth muscle cells (VSMCs). They actively regulate hypertension, endothelial dysfunction, and atherosclerosis. The objective of this review is to examine the roles and heterogeneity of FFAs and FFARs in the regulation of arterial function, with a view to identifying the points of intersection between their actions and providing new insights into the prevention and treatment of diseases associated with arterial dysfunction, as well as the development of targeted drugs.
Collapse
Affiliation(s)
- Fengzhi Yu
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (F.Y.); (L.J.)
| | - Boyi Zong
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China; (B.Z.); (P.S.)
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China
| | - Lili Ji
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (F.Y.); (L.J.)
| | - Peng Sun
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China; (B.Z.); (P.S.)
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China
| | - Dandan Jia
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (F.Y.); (L.J.)
| | - Ru Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (F.Y.); (L.J.)
| |
Collapse
|
7
|
Liu W, Ding Y, Shen Z, Xu C, Yi W, Wang D, Zhou Y, Zon LI, Liu JX. BF170 hydrochloride enhances the emergence of hematopoietic stem and progenitor cells. Development 2024; 151:dev202476. [PMID: 38940293 DOI: 10.1242/dev.202476] [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: 10/24/2023] [Accepted: 05/14/2024] [Indexed: 06/29/2024]
Abstract
Generation of hematopoietic stem and progenitor cells (HSPCs) ex vivo and in vivo, especially the generation of safe therapeutic HSPCs, still remains inefficient. In this study, we have identified compound BF170 hydrochloride as a previously unreported pro-hematopoiesis molecule, using the differentiation assays of primary zebrafish blastomere cell culture and mouse embryoid bodies (EBs), and we demonstrate that BF170 hydrochloride promoted definitive hematopoiesis in vivo. During zebrafish definitive hematopoiesis, BF170 hydrochloride increases blood flow, expands hemogenic endothelium (HE) cells and promotes HSPC emergence. Mechanistically, the primary cilia-Ca2+-Notch/NO signaling pathway, which is downstream of the blood flow, mediated the effects of BF170 hydrochloride on HSPC induction in vivo. Our findings, for the first time, reveal that BF170 hydrochloride is a compound that enhances HSPC induction and may be applied to the ex vivo expansion of HSPCs.
Collapse
Affiliation(s)
- WenYe Liu
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - YuYan Ding
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Zheng Shen
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Cong Xu
- Stem Cell Program and Hematology/Oncology, Children's Hospital and Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA
| | - William Yi
- Stem Cell Program and Hematology/Oncology, Children's Hospital and Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Ding Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Yi Zhou
- Stem Cell Program and Hematology/Oncology, Children's Hospital and Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Leonard I Zon
- Stem Cell Program and Hematology/Oncology, Children's Hospital and Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA
- Howard Hughes Medical Institute/Children's Hospital, 300 Longwood Avenue, Karp 8, Boston, MA 02115, USA
| | - Jing-Xia Liu
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| |
Collapse
|
8
|
Baek KI, Ryu K. Role of Flow-Sensitive Endothelial Genes in Atherosclerosis and Antiatherogenic Therapeutics Development. J Cardiovasc Transl Res 2024; 17:609-623. [PMID: 38010480 DOI: 10.1007/s12265-023-10463-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/14/2023] [Indexed: 11/29/2023]
Abstract
Atherosclerosis is a chronic inflammatory disease that is the underlying cause of cardiovascular disease which initiates from endothelial dysfunction from genetic and environmental risk factors, including biomechanical forces: blood flow. Endothelial cells (ECs) lining the inner arterial wall regions exposed to disturbed flow are prone to atherosclerosis development, whereas the straight regions exposed to stable flow are spared from the disease. These flow patterns induce genome- and epigenome-wide changes in gene expression in ECs. Through the sweeping changes in gene expression, disturbed flow reprograms ECs from athero-protected cell types under the stable flow condition to pro-atherogenic cell conditions. The pro-atherogenic changes induced by disturbed flow, in combination with additional risk factors such as hypercholesterolemia, lead to the progression of atherosclerosis. The flow-sensitive genes and proteins are critical in understanding the mechanisms and serve as novel targets for antiatherogenic therapeutics.
Collapse
Affiliation(s)
- Kyung In Baek
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Kitae Ryu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
- Department of Biotechnology, The University of Suwon, 17, Wauan-Gil, Bongdam-Eup, Hwaseong-Si, Gyeonggi-Do, 18323, Republic of Korea.
| |
Collapse
|
9
|
Katoh K. Effects of Mechanical Stress on Endothelial Cells In Situ and In Vitro. Int J Mol Sci 2023; 24:16518. [PMID: 38003708 PMCID: PMC10671803 DOI: 10.3390/ijms242216518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 11/26/2023] Open
Abstract
Endothelial cells lining blood vessels are essential for maintaining vascular homeostasis and mediate several pathological and physiological processes. Mechanical stresses generated by blood flow and other biomechanical factors significantly affect endothelial cell activity. Here, we review how mechanical stresses, both in situ and in vitro, affect endothelial cells. We review the basic principles underlying the cellular response to mechanical stresses. We also consider the implications of these findings for understanding the mechanisms of mechanotransducer and mechano-signal transduction systems by cytoskeletal components.
Collapse
Affiliation(s)
- Kazuo Katoh
- Laboratory of Human Anatomy and Cell Biology, Faculty of Health Sciences, Tsukuba University of Technology, Tsukuba 305-8521, Japan
| |
Collapse
|
10
|
Plouffe AA, Fischer KP, Vranish JR. Acute upper and lower limb hemodynamic responses during single sessions of low- versus high-intensity inspiratory muscle strength training. J Appl Physiol (1985) 2023; 135:995-1000. [PMID: 37732375 DOI: 10.1152/japplphysiol.00558.2023] [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: 08/09/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 09/22/2023] Open
Abstract
Inspiratory muscle strength training (IMST) has shown potential to improve both respiratory and cardiovascular function in health and disease. Less is known about acute hemodynamic responses to a single IMST session, therefore we assessed upper and lower limb blood flow via Doppler ultrasound in the brachial and popliteal arteries, respectively. Mean, anterograde, and retrograde blood flow (BF) and shear rate (SR) were assessed relative to baseline during low-intensity (15% maximal inspiratory pressure - PImax) and high-intensity (75% PImax) IMST. During low-intensity IMST, popliteal BF and SR were reduced by ∼10%, and brachial BF and SR were reduced by ∼40%. During high-intensity IMST, popliteal BF and SR were reduced by ∼20%, and brachial BF and SR were reduced by ∼35%. BF and SR responses were not statistically different between low-intensity and high-intensity training for either blood vessel (P > 0.05). In addition, anterograde BF and SR were significantly decreased in the brachial artery for both low-intensity and high-intensity training (P < 0.05), but not the popliteal artery (P > 0.05). Finally, during IMST retrograde BF and SR were significantly increased in both the upper and lower limbs during low-intensity and high-intensity training (P < 0.05). These data provide novel insight into the acute BF and SR responses to a single bout of IMST and may enhance our understanding of the mechanism(s) by which IMST imparts its beneficial chronic effects on cardiovascular function.NEW & NOTEWORTHY Herein, we demonstrate for the first time that upper and lower limb blood flow and shear rate patterns are altered during a single bout of IMST, at low- and high-intensity training. Specifically, anterograde blood flow and shear rate are significantly reduced in the brachial artery, whereas retrograde blood flow is significantly elevated in both the brachial and popliteal arteries. These findings provide insight into the vascular impact of IMST, which may inform future mechanistic studies.
Collapse
Affiliation(s)
- Audrey A Plouffe
- Department of Integrative Physiology and Health Science, Alma College, Alma, Michigan, United States
| | - Kylah P Fischer
- Department of Integrative Physiology and Health Science, Alma College, Alma, Michigan, United States
| | - Jennifer R Vranish
- Department of Integrative Physiology and Health Science, Alma College, Alma, Michigan, United States
| |
Collapse
|
11
|
Allana SS, Kostantinis S, Simsek B, Karacsonyi J, Rempakos A, Alaswad K, Krestyaninov O, Khelimskiid D, Karmpaliotis D, Jaffer FA, Khatri JJ, Poommipanit P, Patel MP, Mahmud E, Koutouzis M, Tsiafoutis I, Gorgulu S, Elbarouni B, Nicholson W, Jaber W, Rinfret S, Rafeh NA, Goktekin O, ElGuindy AM, Sandoval Y, Burke MN, Rangan BV, Brilakis ES. Distal Target Vessel Quality and Outcomes of Chronic Total Occlusion Percutaneous Coronary Intervention. JACC Cardiovasc Interv 2023; 16:1490-1500. [PMID: 37380231 DOI: 10.1016/j.jcin.2023.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 02/01/2023] [Accepted: 03/07/2023] [Indexed: 06/30/2023]
Abstract
BACKGROUND Distal vessel quality is a key parameter in the global chronic total occlusion (CTO) crossing algorithm. OBJECTIVES The study sought to evaluate the association of distal vessel quality with the outcomes of CTO percutaneous coronary intervention. METHODS We examined the clinical and angiographic characteristics and procedural outcomes of 10,028 CTO percutaneous coronary interventions performed at 39 U.S. and non-U.S. centers between 2012 and 2022. A poor-quality distal vessel was defined as <2 mm diameter or with significant diffuse atherosclerotic disease. In-hospital major adverse cardiac events (MACE) included death, myocardial infarction, urgent repeat target vessel revascularization, tamponade requiring pericardiocentesis or surgery, and stroke. RESULTS A total of 33% of all CTO lesions had poor-quality distal vessel. When compared with good-quality distal vessels, CTO lesions with a poor-quality distal vessel had higher J-CTO (Japanese chronic total occlusion) scores (2.7 ± 1.1 vs 2.2 ± 1.3; P < 0.01), lower technical (79.9% vs 86.9%; P < 0.01) and procedural (78.0% vs 86.8%; P < 0.01) success, and higher incidence of MACE (2.5% vs 1.7%; P < 0.01) and perforation (6.4% vs 3.7%; P < 0.01). A poor-quality distal vessel was independently associated with technical failure and MACE. Poor-quality distal vessels were associated with higher use of the retrograde approach (25.2% vs 14.9%; P < 0.01) and higher air kerma radiation dose (2.4 [IQR: 1.3-4.0] Gy vs 2.0 [IQR: 1.1-3.5] Gy; P < 0.01). CONCLUSIONS A poor-quality distal vessel in CTO lesions is associated with higher lesion complexity, higher need for retrograde crossing, lower technical and procedural success, higher incidence of MACE and coronary perforation, and higher radiation dose.
Collapse
Affiliation(s)
- Salman S Allana
- Minneapolis Heart Institute and Minneapolis Heart Institute Foundation, Abbott Northwestern Hospital, Minneapolis, Minnesota, USA
| | - Spyridon Kostantinis
- Minneapolis Heart Institute and Minneapolis Heart Institute Foundation, Abbott Northwestern Hospital, Minneapolis, Minnesota, USA
| | - Bahadir Simsek
- Minneapolis Heart Institute and Minneapolis Heart Institute Foundation, Abbott Northwestern Hospital, Minneapolis, Minnesota, USA
| | - Judit Karacsonyi
- Minneapolis Heart Institute and Minneapolis Heart Institute Foundation, Abbott Northwestern Hospital, Minneapolis, Minnesota, USA
| | - Athanasois Rempakos
- Minneapolis Heart Institute and Minneapolis Heart Institute Foundation, Abbott Northwestern Hospital, Minneapolis, Minnesota, USA
| | | | | | - Dmitrii Khelimskiid
- Meshalkin National Medical Research Center, Ministry of Health of Russian Federation, Novosibirsk, Russian Federation
| | | | | | | | - Paul Poommipanit
- University Hospitals, Case Western Reserve University, Cleveland, Ohio, USA
| | | | | | | | | | | | | | | | - Wissam Jaber
- Emory University Hospital Midtown, Atlanta, Georgia, USA
| | | | | | | | | | - Yader Sandoval
- Minneapolis Heart Institute and Minneapolis Heart Institute Foundation, Abbott Northwestern Hospital, Minneapolis, Minnesota, USA
| | - M Nicholas Burke
- Minneapolis Heart Institute and Minneapolis Heart Institute Foundation, Abbott Northwestern Hospital, Minneapolis, Minnesota, USA
| | - Bavana V Rangan
- Minneapolis Heart Institute and Minneapolis Heart Institute Foundation, Abbott Northwestern Hospital, Minneapolis, Minnesota, USA
| | - Emmanouil S Brilakis
- Minneapolis Heart Institute and Minneapolis Heart Institute Foundation, Abbott Northwestern Hospital, Minneapolis, Minnesota, USA.
| |
Collapse
|
12
|
The role of arginine and endothelial nitric oxide synthase in the pathogenesis of Covid-19 complicated by metabolic syndrome. ACTA BIOMEDICA SCIENTIFICA 2022. [DOI: 10.29413/abs.2022-7.6.6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
This literature review presents the role of endothelial nitric oxide synthase (eNOS) and nitric oxide (NO), as well as arginine, the enzyme substrate, in the disease of metabolic syndrome and COVID-19 (SARS-CoV-2 virus). Metabolic syndrome is a combination of obesity, insulin resistance, hyperglycemia, dyslipidemia and hypertension. It has been shown that in elderly people, patients with obesity, metabolic syndrome, type 2 diabetes mellitus (DM2), and patients with COVID-19, endothelial dysfunction (ED) and vascular endothelial activation are detected. ED is the main cause of a number of pathological conditions during the development of COVID-19 and earlier in patients with metabolic syndrome, while a sharp drop in the level of nitric oxide (NO) is detected due to a decrease in the expression and activity of eNO synthase and enzyme depletion, which leads to a violation of the integrity of bloodvessels, that is, to vasoconstrictive, inflammatory and thrombotic conditions, followed by ischemia of organs and edema of tissues. It should be noted that metabolic syndrome, DM2, hypertension and obesity, in particular, are age-related diseases, and it is known that blood glucose levels increase with age, which reduces the bioavailability of NO in endothelial cells. Defects in the metabolism of NO cause dysfunction in the pulmonary blood vessels, the level of NO decreases, which leads to impaired lung function and coagulopathy. The review presents possible mechanisms of these disorders associated with ED, the release of eNO synthase, changes in phosphorylation and regulation of enzyme activity, as well as insulin resistance. A modern view of the role of the polymorphism of the eNO synthase gene in the development of these pathologies is presented. To increase the level of endothelial NO, drugs are offered that regulate the bioavailability of NO. These include arginine, agonist NO – minoxidil, steroid hormones, statins, metformin. However, further research and clinical trials are needed to develop treatment strategies that increase NO levels in the endothelium.
Collapse
|
13
|
Majumdar U, Choudhury TZ, Manivannan S, Ueyama Y, Basu M, Garg V. Single-cell RNA-sequencing analysis of aortic valve interstitial cells demonstrates the regulation of integrin signaling by nitric oxide. Front Cardiovasc Med 2022; 9:742850. [PMID: 36386365 PMCID: PMC9640371 DOI: 10.3389/fcvm.2022.742850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/30/2022] [Indexed: 11/22/2022] Open
Abstract
Calcific aortic valve disease (CAVD) is an increasingly prevalent condition among the elderly population that is associated with significant morbidity and mortality. Insufficient understanding of the underlying disease mechanisms has hindered the development of pharmacologic therapies for CAVD. Recently, we described nitric oxide (NO) mediated S-nitrosylation as a novel mechanism for preventing the calcific process. We demonstrated that NO donor or an S-nitrosylating agent, S-nitrosoglutathione (GSNO), inhibits spontaneous calcification in porcine aortic valve interstitial cells (pAVICs) and this was supported by single-cell RNA sequencing (scRNAseq) that demonstrated NO donor and GSNO inhibited myofibroblast activation of pAVICs. Here, we investigated novel signaling pathways that are critical for the calcification of pAVICs that are altered by NO and GSNO by performing an in-depth analysis of the scRNA-seq dataset. Transcriptomic analysis revealed 1,247 differentially expressed genes in pAVICs after NO donor or GSNO treatment compared to untreated cells. Pathway-based analysis of the differentially expressed genes revealed an overrepresentation of the integrin signaling pathway, along with the Rho GTPase, Wnt, TGF-β, and p53 signaling pathways. We demonstrate that ITGA8 and VCL, two of the identified genes from the integrin signaling pathway, which are known to regulate cell-extracellular matrix (ECM) communication and focal adhesion, were upregulated in both in vitro and in vivo calcific conditions. Reduced expression of these genes after treatment with NO donor suggests that NO inhibits calcification by targeting myofibroblast adhesion and ECM remodeling. In addition, withdrawal of NO donor after 3 days of exposure revealed that NO-mediated transcriptional and translational regulation is a transient event and requires continuous NO exposure to inhibit calcification. Overall, our data suggest that NO and S-nitrosylation regulate the integrin signaling pathway to maintain healthy cell-ECM interaction and prevent CAVD.
Collapse
Affiliation(s)
- Uddalak Majumdar
- Center for Cardiovascular Research, Nationwide Children’s Hospital, Columbus, OH, United States
- The Heart Center, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Talita Z. Choudhury
- Center for Cardiovascular Research, Nationwide Children’s Hospital, Columbus, OH, United States
- The Heart Center, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Sathiyanarayanan Manivannan
- Center for Cardiovascular Research, Nationwide Children’s Hospital, Columbus, OH, United States
- The Heart Center, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Yukie Ueyama
- Center for Cardiovascular Research, Nationwide Children’s Hospital, Columbus, OH, United States
- The Heart Center, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Madhumita Basu
- Center for Cardiovascular Research, Nationwide Children’s Hospital, Columbus, OH, United States
- The Heart Center, Nationwide Children’s Hospital, Columbus, OH, United States
- Department of Pediatrics, The Ohio State University, Columbus, OH, United States
| | - Vidu Garg
- Center for Cardiovascular Research, Nationwide Children’s Hospital, Columbus, OH, United States
- The Heart Center, Nationwide Children’s Hospital, Columbus, OH, United States
- Department of Pediatrics, The Ohio State University, Columbus, OH, United States
- Department of Molecular Genetics, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
14
|
Hounguè U, Villette C, Tokoudagba JM, Chaker AB, Remila L, Auger C, Heintz D, Gbaguidi FA, Schini-Kerth VB. Carissa edulis Vahl (Apocynaceae) extract, a medicinal plant of Benin pharmacopoeia, induces potent endothelium-dependent relaxation of coronary artery rings involving nitric oxide. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 105:154370. [PMID: 35977457 DOI: 10.1016/j.phymed.2022.154370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 07/14/2022] [Accepted: 07/31/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Hypertension is a major cardiovascular risk factor that affects most countries including those of Africa. Although Carissa edulis Vahl, Diodia scandens Sw. and Cleome gynandra L. are traditionally used in Benin as antihypertensive treatments with some efficacy mentioned by the local population, their biological activity on the cardiovascular system remains poorly studied. AIM The study investigated the vasoreactivity of the plants and assessed the underlying mechanisms using isolated arteries. STUDY DESIGN Aqueous-ethanolic extracts of aerial parts of C. edulis, D. scandens and C. gynandra were prepared by maceration before being subjected to multi-step liquid-liquid fractionation with solvents of increasing polarity. The vasoreactivity of the extracts and fractions were assessed on isolated porcine coronary artery and rat aorta using organ chambers, the role of nitric oxide (NO) using NG-nitro-L-arginine (NO synthase inhibitor), prostanoids using indomethacin (cyclooxygenases inhibitor) and endothelium-dependent hyperpolarization using TRAM-34 plus UCL 1684 (inhibitors of calcium-dependent K+ channels), and the vascular uptake of polyphenols using Neu reagent. RESULTS The aqueous-ethanolic crude extract of C. edulis (CECE) induced potent relaxations that were exclusively endothelium-dependent and more pronounced than those to D. scandens and C. gynandra. The n-butanolic fraction of C. edulis (CEBF) was more active than the cyclohexane, dichloromethane, and ethyl acetate fractions. The relaxation induced by CECE and CEBF were inhibited by NG-nitro-L-arginine and affected neither by TRAM-34 plus UCL 1684 nor by indomethacin. CEBF induced sustained endothelium-dependent relaxations for at least 60 min, and inhibited, in a concentration-dependent manner, contractions to KCl, CaCl2, U46619 and serotonin in rings with endothelium. Analysis of CEBF by LCHRMS indicated the presence of polyphenols, terpenes, and alkaloids. Exposure of coronary artery and aorta rings to CEBF caused the accumulation of polyphenols predominantly in the endothelium. CONCLUSION C. edulis leaf extract induced pronounced endothelium-dependent relaxations and inhibited contractile responses by stimulating the endothelial formation of NO. LCHRMS analysis of the most active fraction, the butanolic fraction, revealed the presence of numerous compounds including polyphenols, terpenes, and alkaloids. The polyphenols of CEBF accumulated preferentially in the endothelium of the arterial wall. Thus, these observations support the folkloric use of C. edulis in hypertension.
Collapse
Affiliation(s)
- Ursula Hounguè
- INSERM UMR 1260, Regenerative Nanomedicine, University of Strasbourg, Strasbourg, France; Medicinal Organic Chemistry Laboratory, Faculty of Health Sciences, University of Abomey-Calavi, Cotonou, Benin.
| | - Claire Villette
- CNRS, Plant Imaging and Mass Spectrometry (PIMS), IBMP, University of Strasbourg, Strasbourg, France.
| | - Jean-Marie Tokoudagba
- Medicinal Organic Chemistry Laboratory, Faculty of Health Sciences, University of Abomey-Calavi, Cotonou, Benin
| | - Ahmed B Chaker
- INSERM UMR 1260, Regenerative Nanomedicine, University of Strasbourg, Strasbourg, France
| | - Lamia Remila
- INSERM UMR 1260, Regenerative Nanomedicine, University of Strasbourg, Strasbourg, France
| | - Cyril Auger
- INSERM UMR 1260, Regenerative Nanomedicine, University of Strasbourg, Strasbourg, France.
| | - Dimitri Heintz
- CNRS, Plant Imaging and Mass Spectrometry (PIMS), IBMP, University of Strasbourg, Strasbourg, France.
| | - Fernand A Gbaguidi
- Medicinal Organic Chemistry Laboratory, Faculty of Health Sciences, University of Abomey-Calavi, Cotonou, Benin
| | - Valérie B Schini-Kerth
- INSERM UMR 1260, Regenerative Nanomedicine, University of Strasbourg, Strasbourg, France.
| |
Collapse
|
15
|
Demos C, Johnson J, Andueza A, Park C, Kim Y, Villa-Roel N, Kang DW, Kumar S, Jo H. Sox13 is a novel flow-sensitive transcription factor that prevents inflammation by repressing chemokine expression in endothelial cells. Front Cardiovasc Med 2022; 9:979745. [PMID: 36247423 PMCID: PMC9561411 DOI: 10.3389/fcvm.2022.979745] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease and occurs preferentially in arterial regions exposed to disturbed blood flow (d-flow) while the stable flow (s-flow) regions are spared. D-flow induces endothelial inflammation and atherosclerosis by regulating endothelial gene expression partly through the flow-sensitive transcription factors (FSTFs). Most FSTFs, including the well-known Kruppel-like factors KLF2 and KLF4, have been identified from in vitro studies using cultured endothelial cells (ECs). Since many flow-sensitive genes and pathways are lost or dysregulated in ECs during culture, we hypothesized that many important FSTFs in ECs in vivo have not been identified. We tested the hypothesis by analyzing our recent gene array and single-cell RNA sequencing (scRNAseq) and chromatin accessibility sequencing (scATACseq) datasets generated using the mouse partial carotid ligation model. From the analyses, we identified 30 FSTFs, including the expected KLF2/4 and novel FSTFs. They were further validated in mouse arteries in vivo and cultured human aortic ECs (HAECs). These results revealed 8 FSTFs, SOX4, SOX13, SIX2, ZBTB46, CEBPβ, NFIL3, KLF2, and KLF4, that are conserved in mice and humans in vivo and in vitro. We selected SOX13 for further studies because of its robust flow-sensitive regulation, preferential expression in ECs, and unknown flow-dependent function. We found that siRNA-mediated knockdown of SOX13 increased endothelial inflammatory responses even under the unidirectional laminar shear stress (ULS, mimicking s-flow) condition. To understand the underlying mechanisms, we conducted an RNAseq study in HAECs treated with SOX13 siRNA under shear conditions (ULS vs. oscillatory shear mimicking d-flow). We found 94 downregulated and 40 upregulated genes that changed in a shear- and SOX13-dependent manner. Several cytokines, including CXCL10 and CCL5, were the most strongly upregulated genes in HAECs treated with SOX13 siRNA. The robust induction of CXCL10 and CCL5 was further validated by qPCR and ELISA in HAECs. Moreover, the treatment of HAECs with Met-CCL5, a specific CCL5 receptor antagonist, prevented the endothelial inflammation responses induced by siSOX13. In addition, SOX13 overexpression prevented the endothelial inflammation responses. In summary, SOX13 is a novel conserved FSTF, which represses the expression of pro-inflammatory chemokines in ECs under s-flow. Reduction of endothelial SOX13 triggers chemokine expression and inflammatory responses, a major proatherogenic pathway.
Collapse
Affiliation(s)
- Catherine Demos
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA, United States
| | - Janie Johnson
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA, United States
| | - Aitor Andueza
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA, United States
| | - Christian Park
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA, United States
| | - Yerin Kim
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA, United States
| | - Nicolas Villa-Roel
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA, United States
| | - Dong-Won Kang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA, United States
| | - Sandeep Kumar
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA, United States
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA, United States
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA, United States
| |
Collapse
|
16
|
Sheldon H, Zhang W, Bridges E, Ang KH, Lin S, Masiero M, Li D, Handford PA, Whiteman P, Fischer R, Buffa F, Vatish M, Banham AH, Harris AL. ELTD1 is present in extracellular vesicles derived from endothelial cells as a cleaved extracellular domain which induces in vivo angiogenesis. JOURNAL OF EXTRACELLULAR BIOLOGY 2022; 1:e52. [PMID: 38939053 PMCID: PMC11080856 DOI: 10.1002/jex2.52] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 06/29/2024]
Abstract
ELTD1/ADGRL4 is an adhesion GPCR with an important role in angiogenesis. We recently identified a role for ELTD1 in wound repair and inflammation. Activation of ELTD1 in endothelial cells results in a type II EMT to myofibroblast-like cells that have enhanced angiogenic ability. Furthermore, expression of Eltd1 in murine breast cancer cells increases tumour growth by increasing blood vessel size and perfusion and by creating an immunosuppressive microenvironment. As extracellular vesicles (EVs) are known to be involved in vascular development, growth and maturation we investigated the composition and functional effects of the EVs isolated from ELTD1 expressing cells to elucidate their role in these processes. A highly glycosylated form of the extracellular domain (ECD) of ELTD1 is readily incorporated into EVs. Using mass spectrometry-based proteomics we identified proteins that are enriched in ELTD1-EVs and are involved in haemostasis and immune responses. ELTD1 enriched EVs were pro-angiogenic in vivo and in vitro and the presence of the ECD alone induced endothelial sprouting. In endothelial cells experiencing laminar flow, ELTD1 levels were reduced in the EVs when they are quiescent, showing a relationship between ELTD1 and the activation state of the endothelium. Using FACS, we detected a significant increase in vesicular ELTD1 in the plasma of patients with preeclampsia, a condition characterized by endothelial dysfunction. These data confirm a role for ELTD1 in wound repair and inflammation and reveal its potential as a biomarker of vessel dysfunction.
Collapse
Affiliation(s)
- Helen Sheldon
- Cancer Research UK Molecular Oncology LaboratoriesWeatherall Institute of Molecular MedicineUniversity of OxfordJohn Radcliffe HospitalOxfordUK
| | - Wei Zhang
- Nuffield Department of Women's & Reproductive Health, Women's CentreUniversity of OxfordJohn Radcliffe HospitalOxfordUK
| | - Esther Bridges
- Cancer Research UK Molecular Oncology LaboratoriesWeatherall Institute of Molecular MedicineUniversity of OxfordJohn Radcliffe HospitalOxfordUK
| | - Koon Hwee Ang
- Cancer Research UK Molecular Oncology LaboratoriesWeatherall Institute of Molecular MedicineUniversity of OxfordJohn Radcliffe HospitalOxfordUK
| | - Salwa Lin
- Cancer Research UK Molecular Oncology LaboratoriesWeatherall Institute of Molecular MedicineUniversity of OxfordJohn Radcliffe HospitalOxfordUK
| | - Massimo Masiero
- Nuffield Division of Clinical Laboratory SciencesRadcliffe Department of MedicineJohn Radcliffe HospitalOxfordUK
| | - Demin Li
- Nuffield Division of Clinical Laboratory SciencesRadcliffe Department of MedicineJohn Radcliffe HospitalOxfordUK
| | | | - Pat Whiteman
- Department of BiochemistryUniversity of OxfordOxfordUK
| | - Roman Fischer
- Nuffield Department of MedicineTarget Discovery InstituteOxford University, NDM Research BuildingOxfordUK
| | - Francesca Buffa
- Department of OncologyUniversity of OxfordChurchill HospitalOxfordUK
| | - Manu Vatish
- Nuffield Department of Women's & Reproductive Health, Women's CentreUniversity of OxfordJohn Radcliffe HospitalOxfordUK
| | - Alison H. Banham
- Nuffield Division of Clinical Laboratory SciencesRadcliffe Department of MedicineJohn Radcliffe HospitalOxfordUK
| | - Adrian L. Harris
- Cancer Research UK Molecular Oncology LaboratoriesWeatherall Institute of Molecular MedicineUniversity of OxfordJohn Radcliffe HospitalOxfordUK
| |
Collapse
|
17
|
Ghim M, Yang SW, David KRZ, Eustaquio J, Warboys CM, Weinberg PD. NO Synthesis but Not Apoptosis, Mitosis or Inflammation Can Explain Correlations between Flow Directionality and Paracellular Permeability of Cultured Endothelium. Int J Mol Sci 2022; 23:8076. [PMID: 35897652 PMCID: PMC9332325 DOI: 10.3390/ijms23158076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/09/2022] [Accepted: 07/13/2022] [Indexed: 12/10/2022] Open
Abstract
Haemodynamic wall shear stress varies from site to site within the arterial system and is thought to cause local variation in endothelial permeability to macromolecules. Our aim was to investigate mechanisms underlying the changes in paracellular permeability caused by different patterns of shear stress in long-term culture. We used the swirling well system and a substrate-binding tracer that permits visualisation of transport at the cellular level. Permeability increased in the centre of swirled wells, where flow is highly multidirectional, and decreased towards the edge, where flow is more uniaxial, compared to static controls. Overall, there was a reduction in permeability. There were also decreases in early- and late-stage apoptosis, proliferation and mitosis, and there were significant correlations between the first three and permeability when considering variation from the centre to the edge under flow. However, data from static controls did not fit the same relation, and a cell-by-cell analysis showed that <5% of uptake under shear was associated with each of these events. Nuclear translocation of NF-κB p65 increased and then decreased with the duration of applied shear, as did permeability, but the spatial correlation between them was not significant. Application of an NO synthase inhibitor abolished the overall decrease in permeability caused by chronic shear and the difference in permeability between the centre and the edge of the well. Hence, shear and paracellular permeability appear to be linked by NO synthesis and not by apoptosis, mitosis or inflammation. The effect was mediated by an increase in transport through tricellular junctions.
Collapse
Affiliation(s)
| | | | | | | | | | - Peter D. Weinberg
- Department of Bioengineering, Imperial College London, London SW7 2AZ, UK; (M.G.); (S.-W.Y.); (K.R.Z.D.); (J.E.); (C.M.W.)
| |
Collapse
|
18
|
Ando J, Yamamoto K. Hemodynamic Forces, Endothelial Mechanotransduction, and Vascular Diseases. Magn Reson Med Sci 2022; 21:258-266. [PMID: 34024868 PMCID: PMC9680547 DOI: 10.2463/mrms.rev.2021-0018] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/23/2021] [Indexed: 11/09/2022] Open
Abstract
Cells in the tissues and organs of a living body are subjected to mechanical forces, such as pressure, friction, and tension from their surrounding environment. Cells are equipped with a mechanotransduction mechanism by which they perceive mechanical forces and transmit information into the cell interior, thereby causing physiological or pathogenetic mechano-responses. Endothelial cells (ECs) lining the inner surface of blood vessels are constantly exposed to shear stress caused by blood flow and a cyclic strain caused by intravascular pressure. A number of studies have shown that ECs are sensitive to changes in these hemodynamic forces and alter their morphology and function, sometimes by modifying gene expression. The mechanism of endothelial mechanotransduction has been elucidated, and the plasma membrane has recently been shown to act as a mechanosensor. The lipid order and cholesterol content of plasma membranes change immediately upon the exposure of ECs to hemodynamic forces, resulting in a change in membrane fluidity. These changes in a plasma membrane's physical properties affect the conformation and function of various ion channels, receptors, and microdomains (such as caveolae and primary cilia), thereby activating a wide variety of downstream signaling pathways. Such endothelial mechanotransduction works to maintain circulatory homeostasis; however, errors in endothelial mechanotransduction can cause abnormalities in vascular physiological function, leading to the initiation and progression of various vascular diseases, such as hypertension, thrombosis, aneurysms, and atherosclerosis. Recent advances in detailed imaging technology and computational fluid dynamics analysis have enabled us to evaluate the hemodynamic forces acting on vascular tissue accurately, contributing greatly to our understanding of vascular mechanotransduction and the pathogenesis of vascular diseases, as well as the development of new therapies for vascular diseases.
Collapse
Affiliation(s)
- Joji Ando
- Laboratory of Biomedical Engineering, School of Medicine, Dokkyo Medical University, Mibu, Tochigi, Japan
| | - Kimiko Yamamoto
- Laboratory of System Physiology, Department of Biomedical Engineering, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
19
|
Zhang Y, He P, Wang G, Liang M, Xie D, Nie J, Liu C, Song Y, Liu L, Wang B, Li J, Zhang Y, Wang X, Huo Y, Hou FF, Xu X, Qin X. Interaction of Serum Alkaline Phosphatase and Folic Acid Treatment on Chronic Kidney Disease Progression in Treated Hypertensive Adults. Front Pharmacol 2022; 12:753803. [PMID: 35095485 PMCID: PMC8793861 DOI: 10.3389/fphar.2021.753803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 12/22/2021] [Indexed: 12/27/2022] Open
Abstract
The relation of alkaline phosphatase (ALP) with chronic kidney disease (CKD) is still uncertain. We aimed to examine the prospective association between serum ALP and CKD progression, and the modifying effect of serum ALP on folic acid in preventing CKD progression in treated hypertensive patients. This is a post-hoc analysis of 12,734 hypertensive adults with relevant measurements and without liver disease at baseline from the renal sub-study of the China Stroke Primary Prevention Trial, where participants were randomly assigned to daily treatments of 10 mg enalapril and 0.8 mg folic acid, or 10 mg enalapril alone. The primary outcome was CKD progression, defined as a decrease in estimated glomerular filtration rate (eGFR) of ≥30% and to a level of <60 ml/min/1.73 m2 if baseline eGFR was ≥60 ml/min/1.73 m2; or a decrease in eGFR of ≥50% if baseline eGFR was <60 ml/min/1.73 m2; or end-stage renal disease. Over a median of 4.4 years, in the enalapril only group, participants with baseline serum ALP≥110IU/L (quartile 4) had a significantly higher risk of CKD progression (3.4% vs 2.3%; adjusted OR,1.61; 95%CI:1.11, 2.32), compared with those with ALP<110IU/L. For those with enalapril and folic acid treatment, compared with the enalapril only treatment, the risk of CKD progression was reduced from 3.4 to 2.1% (adjusted OR, 0.53; 95%CI:0.34, 0.83) among participants with baseline ALP≥110IU/L, whereas there was no significant effect among those with ALP<110IU/L. In hypertensive patients, higher serum ALP was associated with increased risk of CKD progression, and this risk was reduced by 47% with folic acid treatment.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- State Key Laboratory of Organ Failure Research, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Division of Nephrology, Nanfang Hospital, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Renal Failure Research, Southern Medical University, Guangzhou, China
| | - Panpan He
- State Key Laboratory of Organ Failure Research, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Division of Nephrology, Nanfang Hospital, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Renal Failure Research, Southern Medical University, Guangzhou, China
| | - Guobao Wang
- State Key Laboratory of Organ Failure Research, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Division of Nephrology, Nanfang Hospital, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Renal Failure Research, Southern Medical University, Guangzhou, China
| | - Min Liang
- State Key Laboratory of Organ Failure Research, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Division of Nephrology, Nanfang Hospital, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Renal Failure Research, Southern Medical University, Guangzhou, China
| | - Di Xie
- State Key Laboratory of Organ Failure Research, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Division of Nephrology, Nanfang Hospital, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Renal Failure Research, Southern Medical University, Guangzhou, China
| | - Jing Nie
- State Key Laboratory of Organ Failure Research, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Division of Nephrology, Nanfang Hospital, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Renal Failure Research, Southern Medical University, Guangzhou, China
| | - Chengzhang Liu
- Institute of Biomedicine, Anhui Medical University, Hefei, China
| | - Yun Song
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Lishun Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Binyan Wang
- Institute of Biomedicine, Anhui Medical University, Hefei, China
| | - Jianping Li
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Yan Zhang
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Xiaobin Wang
- Department of Population, Family and Reproductive Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Yong Huo
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Fan Fan Hou
- State Key Laboratory of Organ Failure Research, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Division of Nephrology, Nanfang Hospital, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Renal Failure Research, Southern Medical University, Guangzhou, China
| | - Xiping Xu
- State Key Laboratory of Organ Failure Research, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Division of Nephrology, Nanfang Hospital, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Renal Failure Research, Southern Medical University, Guangzhou, China
| | - Xianhui Qin
- State Key Laboratory of Organ Failure Research, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Division of Nephrology, Nanfang Hospital, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Renal Failure Research, Southern Medical University, Guangzhou, China
| |
Collapse
|
20
|
Samaha D, Diaconu V, Bouchard JF, Desalliers C, Dupont A. Effect of Latanoprostene Bunod on Optic Nerve Head Blood Flow. Optom Vis Sci 2022; 99:172-176. [PMID: 34889858 DOI: 10.1097/opx.0000000000001842] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
SIGNIFICANCE Topical latanoprostene bunod increases capillary oxygen saturation and blood volume at the optic nerve head in healthy individuals. PURPOSE This study aimed to evaluate the effect of topical latanoprostene bunod on optic nerve blood volume and oxygen saturation in a population of healthy participants. METHODS In this prospective double-blind crossover study, 23 healthy participants aged from 21 to 62 years were recruited. Optic nerve head capillary blood volume (ONHvol) and oxygen saturation (ONHSaO2) baselines were measured over a period of 2 hours using multichannel spectroscopic reflectometry and were remeasured after a 7-day once-daily instillation regimen of either latanoprost 0.005% or latanoprostene bunod 0.024%. After a 30-day washout period, participants were crossed over to the alternate product, and measurements were repeated. Participants were used as their own baselines to calculate variation in ONHvol and ONHSaO2 across time and pharmacological agents. The Friedman test was used to establish significant differences in optic nerve head parameters from baseline values, and Conover post hoc analysis was carried for multiple between-group comparisons. RESULTS Latanoprostene bunod 0.024% induced a significant increase of 4% in ONHSaO2 compared with latanoprost 0.005% (P < .001). Furthermore, latanoprostene bunod increased ONHvol levels by more than twofold at all time points (P < .001 at T60, T90, and T120). The increase in ONHvol was 66.2% higher than levels achieved with latanoprost at T60 (P = .001), 47% higher at T90 (P < .001), and 45% higher at T120 (P < .01). CONCLUSIONS Latanoprostene bunod 0.024% induces a significant increase in optic nerve head blood volume and oxygen saturation in healthy subjects, when compared with latanoprost 0.005%. Future studies are needed to evaluate whether similar responses are elicited in patients suffering from glaucomatous optic neuropathy.
Collapse
Affiliation(s)
| | - Vasile Diaconu
- Montreal University School of Optometry, Montreal, Quebec, Canada
| | | | | | - Ariane Dupont
- Montreal University School of Optometry, Montreal, Quebec, Canada
| |
Collapse
|
21
|
Loeb E, El Asmar K, Trabado S, Gressier F, Colle R, Rigal A, Martin S, Verstuyft C, Fève B, Chanson P, Becquemont L, Corruble E. Nitric Oxide Synthase activity in major depressive episodes before and after antidepressant treatment: Results of a large case-control treatment study. Psychol Med 2022; 52:80-89. [PMID: 32524920 DOI: 10.1017/s0033291720001749] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Nitric oxide synthase (NOS) activity, an enzyme potentially involved in the major depressive episodes (MDE), could be indirectly measured by the L-Citrulline/L-Arginine ratio (L-Cit/L-Arg). The aim of this study was: (1) to compare the NOS activity of patients with a MDE to that of healthy controls (HC); (2) to assess its change after antidepressant treatment. METHODS A total of 460 patients with a current MDE in a context of major depressive disorder (MDD) were compared to 895 HC for NOS activity (L-Cit/L-Arg plasma ratio). L-Arg and L-Cit plasma levels were measured using a MS-based liquid chromatography method. Depressed patients were assessed at baseline, and after 3 and 6 months of antidepressant treatment for depression severity and clinical response. RESULTS Depressed patients had a lower NOS activity than HC at baseline [0.31 ± 0.09 v. 0.38 ± 0.12; 95% confidence interval (CI) -0.084 to -0.062, p < 0.0001]. Lower NOS activity at baseline predicted a higher response rate [odds ratio (OR) = 29.20; 95% CI 1.58-536.37; p = 0.023]. NOS activity in depressed patients increased significantly up to 0.34 ± 0.08 after antidepressant treatment (Est = 0.0034; 95% CI 0.0002-0.0067; p = 0.03). CONCLUSIONS Depressed patients have a decreased NOS activity that improves after antidepressant treatment and predicts drug response. NOS activity may be a promising biomarker for MDE in a context of MDD.
Collapse
Affiliation(s)
- E Loeb
- INSERM CESP - Equipe 'Moods'- Univ Paris-Saclay, 94275 Le Kremlin Bicêtre, France
- Service de Psychiatrie- Hôpital Bicêtre- GH Paris Saclay- APHP, 94275 Le Kremlin Bicêtre, France
- Faculté de Médecine Paris-Saclay, 94275 Le Kremlin Bicêtre, France
| | - K El Asmar
- INSERM CESP - Equipe 'Moods'- Univ Paris-Saclay, 94275 Le Kremlin Bicêtre, France
| | - S Trabado
- Faculté de Médecine Paris-Saclay, 94275 Le Kremlin Bicêtre, France
- Inserm U1185 - Univ Paris-Sud, 94275 Le Kremlin Bicêtre, France
- Service de Génétique moléculaire, Pharmacogénétique et Hormonologie- CHU de Bicêtre- APHP, 94275 Le Kremlin Bicêtre, France
| | - F Gressier
- INSERM CESP - Equipe 'Moods'- Univ Paris-Saclay, 94275 Le Kremlin Bicêtre, France
- Service de Psychiatrie- Hôpital Bicêtre- GH Paris Saclay- APHP, 94275 Le Kremlin Bicêtre, France
- Faculté de Médecine Paris-Saclay, 94275 Le Kremlin Bicêtre, France
| | - R Colle
- INSERM CESP - Equipe 'Moods'- Univ Paris-Saclay, 94275 Le Kremlin Bicêtre, France
- Service de Psychiatrie- Hôpital Bicêtre- GH Paris Saclay- APHP, 94275 Le Kremlin Bicêtre, France
- Faculté de Médecine Paris-Saclay, 94275 Le Kremlin Bicêtre, France
| | - A Rigal
- INSERM CESP - Equipe 'Moods'- Univ Paris-Saclay, 94275 Le Kremlin Bicêtre, France
- Service de Psychiatrie- Hôpital Bicêtre- GH Paris Saclay- APHP, 94275 Le Kremlin Bicêtre, France
- Faculté de Médecine Paris-Saclay, 94275 Le Kremlin Bicêtre, France
| | - S Martin
- INSERM CESP - Equipe 'Moods'- Univ Paris-Saclay, 94275 Le Kremlin Bicêtre, France
- Service de Psychiatrie- Hôpital Bicêtre- GH Paris Saclay- APHP, 94275 Le Kremlin Bicêtre, France
- Faculté de Médecine Paris-Saclay, 94275 Le Kremlin Bicêtre, France
| | - C Verstuyft
- INSERM CESP - Equipe 'Moods'- Univ Paris-Saclay, 94275 Le Kremlin Bicêtre, France
- Faculté de Médecine Paris-Saclay, 94275 Le Kremlin Bicêtre, France
- Service de Génétique moléculaire, Pharmacogénétique et Hormonologie- CHU de Bicêtre- APHP, 94275 Le Kremlin Bicêtre, France
| | - B Fève
- Sorbonne Université-INSERM UMR S_938, Centre de Recherche Saint-Antoine, 75012Paris, France
- Service d'Endocrinologie- Hôpital Saint-Antoine- APHP, 75012Paris, France
- Institut Hospitalo-Universitaire ICAN, 75012Paris, France
| | - P Chanson
- Faculté de Médecine Paris-Saclay, 94275 Le Kremlin Bicêtre, France
- Inserm U1185 - Univ Paris-Sud, 94275 Le Kremlin Bicêtre, France
- Service d'Endocrinologie et des Maladies de la Reproduction- CHU de Bicêtre- APHP, 94275 Le Kremlin Bicêtre, France
| | - L Becquemont
- INSERM CESP - Equipe 'Moods'- Univ Paris-Saclay, 94275 Le Kremlin Bicêtre, France
- Faculté de Médecine Paris-Saclay, 94275 Le Kremlin Bicêtre, France
- Service de Génétique moléculaire, Pharmacogénétique et Hormonologie- CHU de Bicêtre- APHP, 94275 Le Kremlin Bicêtre, France
| | - E Corruble
- INSERM CESP - Equipe 'Moods'- Univ Paris-Saclay, 94275 Le Kremlin Bicêtre, France
- Service de Psychiatrie- Hôpital Bicêtre- GH Paris Saclay- APHP, 94275 Le Kremlin Bicêtre, France
- Faculté de Médecine Paris-Saclay, 94275 Le Kremlin Bicêtre, France
| |
Collapse
|
22
|
Kant S, Tran KV, Kvandova M, Caliz AD, Yoo HJ, Learnard H, Dolan AC, Craige SM, Hall JD, Jiménez JM, St. Hilaire C, Schulz E, Kröller-Schön S, Keaney JF. PGC1α Regulates the Endothelial Response to Fluid Shear Stress via Telomerase Reverse Transcriptase Control of Heme Oxygenase-1. Arterioscler Thromb Vasc Biol 2022; 42:19-34. [PMID: 34789002 PMCID: PMC8702461 DOI: 10.1161/atvbaha.121.317066] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
OBJECTIVE Fluid shear stress (FSS) is known to mediate multiple phenotypic changes in the endothelium. Laminar FSS (undisturbed flow) is known to promote endothelial alignment to flow, which is key to stabilizing the endothelium and rendering it resistant to atherosclerosis and thrombosis. The molecular pathways responsible for endothelial responses to FSS are only partially understood. In this study, we determine the role of PGC1α (peroxisome proliferator gamma coactivator-1α)-TERT (telomerase reverse transcriptase)-HMOX1 (heme oxygenase-1) during shear stress in vitro and in vivo. Approach and Results: Here, we have identified PGC1α as a flow-responsive gene required for endothelial flow alignment in vitro and in vivo. Compared with oscillatory FSS (disturbed flow) or static conditions, laminar FSS (undisturbed flow) showed increased PGC1α expression and its transcriptional coactivation. PGC1α was required for laminar FSS-induced expression of TERT in vitro and in vivo via its association with ERRα(estrogen-related receptor alpha) and KLF (Kruppel-like factor)-4 on the TERT promoter. We found that TERT inhibition attenuated endothelial flow alignment, elongation, and nuclear polarization in response to laminar FSS in vitro and in vivo. Among the flow-responsive genes sensitive to TERT status, HMOX1 was required for endothelial alignment to laminar FSS. CONCLUSIONS These data suggest an important role for a PGC1α-TERT-HMOX1 axis in the endothelial stabilization response to laminar FSS.
Collapse
Affiliation(s)
- Shashi Kant
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
- Equal contribution
| | - Khanh-Van Tran
- Division of Cardiovascular Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655
- Equal contribution
| | - Miroslava Kvandova
- Department of Cardiology, University Medical Center, Mainz, Germany
- Equal contribution
| | - Amada D. Caliz
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Hyung-Jin Yoo
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Heather Learnard
- Division of Cardiovascular Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655
| | - Ana C. Dolan
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Siobhan M. Craige
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blackburg, VA 24061
| | - Joshua D. Hall
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, MA 01003
| | - Juan M. Jiménez
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, MA 01003
| | - Cynthia St. Hilaire
- Division of Cardiology, Departments of Medicine and Bioengineering, and the Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261
| | - Eberhard Schulz
- Department of Cardiology, Allgemeines Krankenhaus, Celle, Germany
| | | | - John F. Keaney
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
23
|
Zhang Y, Li H, Xie D, Li J, Zhang Y, Wang B, Liu C, Song Y, Wang X, Huo Y, Hou FF, Xu X, Qin X. Positive Association Between Serum Alkaline Phosphatase and First Stroke in Hypertensive Adults. Front Cardiovasc Med 2021; 8:749196. [PMID: 34957239 PMCID: PMC8702620 DOI: 10.3389/fcvm.2021.749196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 11/22/2021] [Indexed: 11/30/2022] Open
Abstract
The relation of alkaline phosphatase (ALP) with stroke risk remains uncertain. We aimed to examine the association between serum ALP and the risk of first stroke, and explore the possible effect modifiers in the association, among adults with hypertension. A total of 19,747 participants with baseline ALP measurements and without liver disease at baseline from the China Stroke Primary Prevention Trial (CSPPT) were included. The primary outcome was a first stroke. Over a median follow-up of 4.5 years, there was a positive association between serum ALP levels and the risk of first stroke (per SD increment, adjusted HR, 1.10; 95%CI: 1.01, 1.20). When serum ALP was evaluated as quartiles, a significantly higher risk of first stroke was observed in those in quartile 2–4 (ALP ≥79 IU/L; adjusted HR, 1.38; 95% CI: 1.11, 1.71), compared with participants in quartile 1 (ALP <79 IU/L). Similar results were found for first ischemic or hemorrhagic stroke. Similar findings were also found in those with a normal range of baseline ALP levels (20–140 IU/L) (per SD increment, adjusted HR, 1.15; 95%CI: 1.05, 1.27). None of the variables, including sex, age, body mass index, smoking, alcohol drinking, blood pressure, total cholesterol, fasting glucose levels at baseline, and blood pressure levels during the treatment period, significantly modified the association. In summary, our study suggests that higher serum ALP levels, even in normal range, were significantly related to higher risk of first stroke among Chinese hypertensive adults.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,National Clinical Research Center for Kidney Disease, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Guangzhou, China.,Guangdong Provincial Institute of Nephrology, Guangzhou, China.,Guangdong Provincial Clinical Research Center for Kidney Disease, Guangzhou, China.,Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Huan Li
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,National Clinical Research Center for Kidney Disease, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Guangzhou, China.,Guangdong Provincial Institute of Nephrology, Guangzhou, China.,Guangdong Provincial Clinical Research Center for Kidney Disease, Guangzhou, China.,Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Di Xie
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,National Clinical Research Center for Kidney Disease, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Guangzhou, China.,Guangdong Provincial Institute of Nephrology, Guangzhou, China.,Guangdong Provincial Clinical Research Center for Kidney Disease, Guangzhou, China.,Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Jianping Li
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Yan Zhang
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Binyan Wang
- Institute of Biomedicine, Anhui Medical University, Hefei, China
| | - Chengzhang Liu
- Institute of Biomedicine, Anhui Medical University, Hefei, China
| | - Yun Song
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Xiaobin Wang
- Department of Population, Family and Reproductive Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Yong Huo
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Fan Fan Hou
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,National Clinical Research Center for Kidney Disease, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Guangzhou, China.,Guangdong Provincial Institute of Nephrology, Guangzhou, China.,Guangdong Provincial Clinical Research Center for Kidney Disease, Guangzhou, China.,Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Xiping Xu
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,National Clinical Research Center for Kidney Disease, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Guangzhou, China.,Guangdong Provincial Institute of Nephrology, Guangzhou, China.,Guangdong Provincial Clinical Research Center for Kidney Disease, Guangzhou, China.,Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Xianhui Qin
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,National Clinical Research Center for Kidney Disease, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Guangzhou, China.,Guangdong Provincial Institute of Nephrology, Guangzhou, China.,Guangdong Provincial Clinical Research Center for Kidney Disease, Guangzhou, China.,Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| |
Collapse
|
24
|
Serreli G, Le Sayec M, Diotallevi C, Teissier A, Deiana M, Corona G. Conjugated Metabolites of Hydroxytyrosol and Tyrosol Contribute to the Maintenance of Nitric Oxide Balance in Human Aortic Endothelial Cells at Physiologically Relevant Concentrations. Molecules 2021; 26:molecules26247480. [PMID: 34946563 PMCID: PMC8707355 DOI: 10.3390/molecules26247480] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 12/17/2022] Open
Abstract
Nitric oxide (NO) is an important signaling molecule involved in many pathophysiological processes. NO mediates vasodilation and blood flow in the arteries, and its action contributes to maintaining vascular homeostasis by inhibiting vascular smooth muscle contraction and growth, platelet aggregation, and leukocyte adhesion to the endothelium. Dietary antioxidants and their metabolites have been found to be directly and/or indirectly involved in the modulation of the intracellular signals that lead to the production of NO. The purpose of this study was to investigate the contribution of conjugated metabolites of hydroxytyrosol (HT) and tyrosol (TYR) to the release of NO at the vascular level, and the related mechanism of action, in comparison to their parental forms. Experiments were performed in human aortic endothelial cells (HAEC) to evaluate the superoxide production, the release of NO and production of cyclic guanosine monophosphate (cGMP), the activation of serine/threonine-protein kinase 1 (Akt1), and the activation state of endothelial nitric oxide synthase (eNOS). It was observed that the tested phenolic compounds enhanced NO and cGMP concentration, inhibiting its depletion caused by superoxide overproduction. Moreover, some of them enhanced the activation of Akt (TYR, HT metabolites) and eNOS (HT, HVA, TYR-S, HT-3S). Overall, the obtained data showed that these compounds promote NO production and availability, suggesting that HT and TYR conjugated metabolites may contribute to the effects of parental extra virgin olive oil (EVOO) phenolics in the prevention of cardiovascular diseases.
Collapse
Affiliation(s)
- Gabriele Serreli
- School of Life and Health Sciences, University of Roehampton, London SW15 4JD, UK;
- Department of Biomedical Sciences, University of Cagliari, Monserrato, 09042 Cagliari, Italy;
| | - Melanie Le Sayec
- School of Life and Health Sciences, University of Roehampton, London SW15 4JD, UK;
| | - Camilla Diotallevi
- School of Life and Health Sciences, University of Roehampton, London SW15 4JD, UK;
| | - Alice Teissier
- School of Life and Health Sciences, University of Roehampton, London SW15 4JD, UK;
| | - Monica Deiana
- Department of Biomedical Sciences, University of Cagliari, Monserrato, 09042 Cagliari, Italy;
| | - Giulia Corona
- School of Life and Health Sciences, University of Roehampton, London SW15 4JD, UK;
- Correspondence: ; Tel.: +44-(0)20-8392-3622
| |
Collapse
|
25
|
Wei SY, Shih YT, Wu HY, Wang WL, Lee PL, Lee CI, Lin CY, Chen YJ, Chien S, Chiu JJ. Endothelial Yin Yang 1 Phosphorylation at S118 Induces Atherosclerosis Under Flow. Circ Res 2021; 129:1158-1174. [PMID: 34747636 DOI: 10.1161/circresaha.121.319296] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Rationale: Disturbed flow occurring in arterial branches and curvatures induces vascular endothelial cell (EC) dysfunction and atherosclerosis. We postulated that disturbed flow plays important roles in modulating phosphoprotein expression profiles to regulate endothelial functions and atherogenesis. Objective: The goal of this study is to discover novel site-specific phosphorylation alterations induced by disturbed flow in ECs to contribute to atherosclerosis. Methods and Results: Quantitative phosphoproteomics analysis of ECs exposed to disturbed flow with low and oscillatory shear stress (OS, 0.5plusminus4 dynes/cm2) vs. pulsatile flow with high shear stress (PS, 124plusminus dynes/cm2) revealed that OS induces serine (S)118 phosphorylation of Yin Yang 1 (phospho-YY1S118) in ECs. Elevated phospho-YY1S118 level in ECs was further confirmed to be present in the disturbed flow regions in experimental animals and human atherosclerotic arteries. This disturbed flow-induced EC phospho-YY1S118 is mediated by casein kinase 2α (CK2α) through its direct interaction with YY1. Yeast two-hybrid library screening and in situ proximity ligation assays demonstrate that phospho-YY1S118 directly binds zinc finger with KRAB and SCAN domains 4 (ZKSCAN4) to induce promoter activity and gene expression of human double minute 2 (HDM2), which consequently induces EC proliferation through down-regulations of p53 and p21CIP1. Administration of apolipoprotein E-deficient (ApoE-/-) mice with CK2-specific inhibitor tetrabromocinnamic acid or atorvastatin inhibits atherosclerosis formation through down-regulations of EC phospho-YY1S118 and HDM2. Generation of novel transgenic mice bearing EC-specific overexpression of S118-non-phosphorylatable mutant of YY1 in ApoE-/- mice confirms the critical role of phospho-YY1S118 in promoting atherosclerosis through EC HDM2. Conclusions: Our findings provide new insights into the mechanisms by which disturbed flow induces endothelial phospho-YY1S118 to promote atherosclerosis, thus indicating phospho-YY1S118 as a potential molecular target for atherosclerosis treatment.
Collapse
Affiliation(s)
- Shu-Yi Wei
- Institute of Cellular and System Medicine, National Health Research Institutes, TAIWAN
| | - Yu-Tsung Shih
- Institute of Cellular and System Medicine, National Health Research Institutes, TAIWAN
| | - Hsin-Yi Wu
- Instrumentation Center, National Taiwan University, TAIWAN
| | - Wei-Li Wang
- Institute of Cellular and System Medicine, TAIWAN
| | - Pei Ling Lee
- Institute of Cellular and System Medicine, National Health Research Institutes, TAIWAN
| | - Chih-I Lee
- Institute of Cellular and System Medicine, National Health Research Institutes, TAIWAN
| | - Chia-Yu Lin
- National Health Research Institutes, Taiwan, TAIWAN
| | | | - Shu Chien
- Bioengineering, University of California, San Diego, UNITED STATES
| | - Jeng-Jiann Chiu
- Institute of Cellular and System Medicine, National Health Research Institutes, TAIWAN
| |
Collapse
|
26
|
A validated reduced-order dynamic model of nitric oxide regulation in coronary arteries. Comput Biol Med 2021; 139:104958. [PMID: 34717232 DOI: 10.1016/j.compbiomed.2021.104958] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 09/30/2021] [Accepted: 10/16/2021] [Indexed: 01/15/2023]
Abstract
Nitric Oxide (NO) provides myocardial oxygen demands of the heart during exercise and cardiac pacing and also prevents cardiovascular diseases such as atherosclerosis and platelet adhesion and aggregation. However, the direct in vivo measurement of NO in coronary arteries is still challenging. To address this matter, a mathematical model of dynamic changes of calcium and NO concentration in the coronary artery was developed for the first time. The model is able to simulate the effect of NO release in coronary arteries and its impact on the hemodynamics of the coronary arterial tree and also to investigate the vasodilation effects of arteries during cardiac pacing. For these purposes, flow rate, time-averaged wall shear stress, dilation percent, NO concentration, and Calcium (Ca2+) concentration within coronary arteries were obtained. In addition, the impact of hematocrit on the flow rate of the coronary artery was studied. It was seen that the behavior of flow rate, wall shear stress, and Ca2+ is biphasic, but the behavior of NO concentration and the dilation percent is triphasic. Also, by increasing the Hematocrit, the blood flow reduces slightly. The results were compared with several experimental measurements to validate the model qualitatively and quantitatively. It was observed that the presented model is well capable of predicting the behavior of arteries after releasing NO during cardiac pacing. Such a study would be a valuable tool to understand the mechanisms underlying vessel damage, and thereby to offer insights for the prevention or treatment of cardiovascular diseases.
Collapse
|
27
|
Engler AJ, Wang Y. Editorial: Understanding molecular interactions that underpin vascular mechanobiology. APL Bioeng 2021; 5:030401. [PMID: 34258496 PMCID: PMC8253597 DOI: 10.1063/5.0058611] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 06/15/2021] [Indexed: 02/04/2023] Open
Abstract
Cells are exposed to a variety of mechanical forces in their daily lives, especially endothelial cells that are stretched from vessel distention and are exposed to hemodynamic shear stress from a blood flow. Exposure to excessive forces can induce a disease, but the molecular details on how these cells perceive forces, transduce them into biochemical signals and genetic events, i.e., mechanotransduction, and integrate them into physiological or pathological changes remain unclear. However, seminal studies in endothelial cells over the past several decades have begun to elucidate some of these signals. These studies have been highlighted in APL Bioengineering and elsewhere, describing a complex temporal pattern where forces are sensed immediately by ion channels and force-dependent conformational changes in surface proteins, followed by biochemical cascades, cytoskeletal contraction, and nuclear remodeling that can affect long-term changes in endothelial morphology and fate. Key examples from the endothelial literature that have established these pathways include showing that integrins and Flk-1 or VE-cadherin act as shear stress transducers, activating downstream proteins such as Cbl and Nckβ or Src, respectively. In this Editorial, we summarize a recent literature highlighting these accomplishments, noting the engineering tools and analysis methods used in these discoveries while also highlighting unanswered questions.
Collapse
|
28
|
Ferulic Acid Derivatives and Avenanthramides Modulate Endothelial Function through Maintenance of Nitric Oxide Balance in HUVEC Cells. Nutrients 2021; 13:nu13062026. [PMID: 34204635 PMCID: PMC8231282 DOI: 10.3390/nu13062026] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/07/2021] [Accepted: 06/10/2021] [Indexed: 02/06/2023] Open
Abstract
Wholegrain oats contain a variety of phenolic compounds thought to help maintain healthy vascular function, through the maintenance of local levels of the vasodilator nitric oxide (NO). Thus, the full molecular mechanisms involved are not yet clear. With this work we aim to understand the possible cellular mechanisms by which avenanthramides and ferulic acid derivatives, present in oats, may help maintain a healthy vascular function through the modulation of the NO pathway. Primary Human Umbilical Vein Endothelial Cells (HUVEC) were exposed to ferulic acid, isoferulic acid, hydroferulic acid, ferulic acid 4-O-glucuronide, isoferulic acid 3-O-sulfate, dihydroferulic acid 4-O-glucuronide, avenanthramide A, avenanthramide B and avenanthramide C (1 μM) or vehicle (methanol) for 24 h. Apocynin and Nω-Nitro-L-arginine (L-NNA) were additionally included as controls. NO and cyclic GMP (cGMP) levels, superoxide production and the activation of the Akt1/eNOS pathway were assessed. The statistical analysis was performed using one-way ANOVA followed by a Tukey post-hoc t-test. Apocynin and all phenolic compounds increased NO levels in HUVEC cells (increased DAF2-DA fluorescence and cGMP), and significantly reduced superoxide levels. Protein expression results highlighted an increase in the Akt1 activation state, and increased eNOS expression. Overall, our results indicated that the glucuronide metabolites do not enhance NO production through the Akt1/eNOS pathway, thus all compounds tested are able to reduce NO degradation through reduced superoxide formation.
Collapse
|
29
|
Ku KH, Dubinsky MK, Sukumar AN, Subramaniam N, Feasson MYM, Nair R, Tran E, Steer BM, Knight BJ, Marsden PA. In Vivo Function of Flow-Responsive Cis-DNA Elements of eNOS Gene: A Role for Chromatin-Based Mechanisms. Circulation 2021; 144:365-381. [PMID: 33910388 DOI: 10.1161/circulationaha.120.051078] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND eNOS (endothelial nitric oxide synthase) is an endothelial cell (EC)-specific gene predominantly expressed in medium- to large-sized arteries where ECs experience atheroprotective laminar flow with high shear stress. Disturbed flow with lower average shear stress decreases eNOS transcription, which leads to the development of atherosclerosis, especially at bifurcations and curvatures of arteries. This prototypic arterial EC gene contains 2 distinct flow-responsive cis-DNA elements in the promoter, the shear stress response element (SSRE) and the KLF (Krüppel-like factor) element. Previous in vitro studies suggested their positive regulatory functions on flow-induced transcription of EC genes including eNOS. However, the in vivo function of these cis-DNA elements remains unknown. METHODS Insertional transgenic mice with a mutation at each flow-responsive cis-DNA element were generated using a murine eNOS promoter-β-galactosidase reporter by linker-scanning mutagenesis and compared with episomal-based mutations in vitro. DNA methylation at the eNOS proximal promoter in mouse ECs was assessed by bisulfite sequencing or pyrosequencing. RESULTS Wild type mice with a functional eNOS promoter-reporter transgene exhibited reduced endothelial reporter expression in the atheroprone regions of disturbed flow (n=5). It is surprising that the SSRE mutation abrogated reporter expression in ECs and was associated with aberrant hypermethylation at the eNOS proximal promoter (n=7). Reporter gene silencing was independent of transgene copy number and integration position, indicating that the SSRE is a critical cis-element necessary for eNOS transcription in vivo. The KLF mutation demonstrated an integration site-specific decrease in eNOS transcription, again with marked promoter methylation (n=8), suggesting that the SSRE alone is not sufficient for eNOS transcription in vivo. In wild type mice, the native eNOS promoter was significantly hypermethylated in ECs from the atheroprone regions where eNOS expression was markedly repressed by chronic disturbed flow, demonstrating that eNOS expression is regulated by flow-dependent DNA methylation that is region-specific in the arterial endothelium in vivo. CONCLUSIONS We report, for the first time, that the SSRE and KLF elements are critical flow sensors necessary for a transcriptionally permissive, hypomethylated eNOS promoter in ECs under chronic shear stress in vivo. Moreover, eNOS expression is regulated by flow-dependent epigenetic mechanisms, which offers novel mechanistic insight on eNOS gene regulation in atherogenesis.
Collapse
Affiliation(s)
- Kyung Ha Ku
- Department of Laboratory Medicine and Pathobiology (K.H.K., M.Y.M.F., R.N., E.T., B.J.K., P.A.M.).,Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute (K.H.K., M.K.D., A.N.S., N.S., M.Y.M.F., R.N., B.M.B., P.A.M.)
| | - Michelle K Dubinsky
- Institute of Medical Science (M.K.D., A.N.S., N.S., P.A.M.) University of Toronto, Ontario, Canada.,Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute (K.H.K., M.K.D., A.N.S., N.S., M.Y.M.F., R.N., B.M.B., P.A.M.)
| | - Aravin N Sukumar
- Institute of Medical Science (M.K.D., A.N.S., N.S., P.A.M.) University of Toronto, Ontario, Canada.,Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute (K.H.K., M.K.D., A.N.S., N.S., M.Y.M.F., R.N., B.M.B., P.A.M.)
| | - Noeline Subramaniam
- Institute of Medical Science (M.K.D., A.N.S., N.S., P.A.M.) University of Toronto, Ontario, Canada.,Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute (K.H.K., M.K.D., A.N.S., N.S., M.Y.M.F., R.N., B.M.B., P.A.M.)
| | - Manon Y M Feasson
- Department of Laboratory Medicine and Pathobiology (K.H.K., M.Y.M.F., R.N., E.T., B.J.K., P.A.M.).,Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute (K.H.K., M.K.D., A.N.S., N.S., M.Y.M.F., R.N., B.M.B., P.A.M.)
| | - Ranju Nair
- Department of Laboratory Medicine and Pathobiology (K.H.K., M.Y.M.F., R.N., E.T., B.J.K., P.A.M.).,Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute (K.H.K., M.K.D., A.N.S., N.S., M.Y.M.F., R.N., B.M.B., P.A.M.)
| | - Eileen Tran
- Department of Laboratory Medicine and Pathobiology (K.H.K., M.Y.M.F., R.N., E.T., B.J.K., P.A.M.)
| | - Brent M Steer
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute (K.H.K., M.K.D., A.N.S., N.S., M.Y.M.F., R.N., B.M.B., P.A.M.)
| | - Britta J Knight
- Department of Laboratory Medicine and Pathobiology (K.H.K., M.Y.M.F., R.N., E.T., B.J.K., P.A.M.)
| | - Philip A Marsden
- Department of Laboratory Medicine and Pathobiology (K.H.K., M.Y.M.F., R.N., E.T., B.J.K., P.A.M.).,Institute of Medical Science (M.K.D., A.N.S., N.S., P.A.M.) University of Toronto, Ontario, Canada.,Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute (K.H.K., M.K.D., A.N.S., N.S., M.Y.M.F., R.N., B.M.B., P.A.M.).,Department of Medicine (P.A.M.), St Michael's Hospital, Toronto, Ontario, Canada
| |
Collapse
|
30
|
Lillo MA, Gaete PS, Puebla M, Burboa PC, Poblete I, Figueroa XF. Novel Pannexin-1-Coupled Signaling Cascade Involved in the Control of Endothelial Cell Function and NO-Dependent Relaxation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:2678134. [PMID: 33688389 PMCID: PMC7914086 DOI: 10.1155/2021/2678134] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 01/03/2021] [Accepted: 01/13/2021] [Indexed: 12/11/2022]
Abstract
Deletion of pannexin-1 (Panx-1) leads not only to a reduction in endothelium-derived hyperpolarization but also to an increase in NO-mediated vasodilation. Therefore, we evaluated the participation of Panx-1-formed channels in the control of membrane potential and [Ca2+]i of endothelial cells. Changes in NO-mediated vasodilation, membrane potential, superoxide anion (O2 ·-) formation, and endothelial cell [Ca2+]i were analyzed in rat isolated mesenteric arterial beds and primary cultures of mesenteric endothelial cells. Inhibition of Panx-1 channels with probenecid (1 mM) or the Panx-1 blocking peptide 10Panx (60 μM) evoked an increase in the ACh (100 nM)-induced vasodilation of KCl-contracted mesenteries and in the phosphorylation level of endothelial NO synthase (eNOS) at serine 1177 (P-eNOSS1177) and Akt at serine 473 (P-AktS473). In addition, probenecid or 10Panx application activated a rapid, tetrodotoxin (TTX, 300 nM)-sensitive, membrane potential depolarization and [Ca2+]i increase in endothelial cells. Interestingly, the endothelial cell depolarization was converted into a transient spike after removing Ca2+ ions from the buffer solution and in the presence of 100 μM mibefradil or 10 μM Ni2+. As expected, Ni2+ also abolished the increment in [Ca2+]i. Expression of Nav1.2, Nav1.6, and Cav3.2 isoforms of voltage-dependent Na+ and Ca2+ channels was confirmed by immunocytochemistry. Furthermore, the Panx-1 channel blockade was associated with an increase in O2 ·- production. Treatment with 10 μM TEMPOL or 100 μM apocynin prevented the increase in O2 ·- formation, ACh-induced vasodilation, P-eNOSS1177, and P-AktS473 observed in response to Panx-1 inhibition. These findings indicate that the Panx-1 channel blockade triggers a novel complex signaling pathway initiated by the sequential activation of TTX-sensitive Nav channels and Cav3.2 channels, leading to an increase in NO-mediated vasodilation through a NADPH oxidase-dependent P-eNOSS1177, which suggests that Panx-1 may be involved in the endothelium-dependent control of arterial blood pressure.
Collapse
Affiliation(s)
- Mauricio A. Lillo
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8330025, Chile
| | - Pablo S. Gaete
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8330025, Chile
| | - Mariela Puebla
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina-Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Pía C. Burboa
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8330025, Chile
| | - Inés Poblete
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8330025, Chile
| | - Xavier F. Figueroa
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8330025, Chile
| |
Collapse
|
31
|
Mao YJ, Wu JB, Yang ZQ, Zhang YH, Huang ZJ. Nitric oxide donating anti-glaucoma drugs: advances and prospects. Chin J Nat Med 2021; 18:275-283. [PMID: 32402405 DOI: 10.1016/s1875-5364(20)30035-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Indexed: 12/11/2022]
Abstract
Glaucoma is a disease that causes irreversible blindness. Reducing intraocular pressure (IOP) is the main treatment at present. Nitric oxide (NO), an endogenous gas signaling molecule, can increase aqueous humor outflow facility, inhibit aqueous humor production thereby reducing IOP, as well as regulate eye blood flow and protect the optic nerve. Therefore, NO donating anti-glaucoma drugs have broad research prospects. In this review, we summarize NO-mediated therapy for glaucoma, and the state of the art of some NO donating molecules, including latanoprostene bunod in market and some other candidate compounds, for the intervention of glaucoma, as well as prospects and challenges ahead in this field.
Collapse
Affiliation(s)
- Yu-Jie Mao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, China
| | - Jian-Bing Wu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, China
| | - Ze-Qiu Yang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, China
| | - Yi-Hua Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, China
| | - Zhang-Jian Huang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
32
|
Babendreyer A, Rojas-González DM, Giese AA, Fellendorf S, Düsterhöft S, Mela P, Ludwig A. Differential Induction of the ADAM17 Regulators iRhom1 and 2 in Endothelial Cells. Front Cardiovasc Med 2020; 7:610344. [PMID: 33335915 PMCID: PMC7736406 DOI: 10.3389/fcvm.2020.610344] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 11/10/2020] [Indexed: 12/23/2022] Open
Abstract
Background: Endothelial function significantly depends on the proteolytic release of surface expressed signal molecules, their receptors and adhesion molecules via the metalloproteinase ADAM17. The pseudoproteases iRhom1 and 2 independently function as adapter proteins for ADAM17 and are essential for the maturation, trafficking, and activity regulation of ADAM17. Bioinformatic data confirmed that immune cells predominantly express iRhom2 while endothelial cells preferentially express iRhom1. Objective: Here, we investigate possible reasons for higher iRhom1 expression and potential inflammatory regulation of iRhom2 in endothelial cells and analyze the consequences for ADAM17 maturation and function. Methods: Primary endothelial cells were cultured in absence and presence of flow with and without inflammatory cytokines (TNFα and INFγ). Regulation of iRhoms was studied by qPCR, involved signaling pathways were studied with transcriptional inhibitors and consequences were analyzed by assessment of ADAM17 maturation, surface expression and cleavage of the ADAM17 substrate junctional adhesion molecule JAM-A. Results: Endothelial iRhom1 is profoundly upregulated by physiological shear stress. This is accompanied by a homeostatic phenotype driven by the transcription factor KLF2 which is, however, only partially responsible for regulation of iRhom1. By contrast, iRhom2 is most prominently upregulated by inflammatory cytokines. This correlates with an inflammatory phenotype driven by the transcription factors NFκB and AP-1 of which AP-1 is most relevant for iRhom2 regulation. Finally, shear stress exposure and inflammatory stimulation have independent and no synergistic effects on ADAM17 maturation, surface expression and JAM-A shedding. Conclusion: Conditions of shear stress and inflammation differentially upregulate iRhom1 and 2 in primary endothelial cells which then results in independent regulation of ADAM17.
Collapse
Affiliation(s)
- Aaron Babendreyer
- Institute of Molecular Pharmacology, University Hospital Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Diana M Rojas-González
- Department of Mechanical Engineering, Munich School of BioEngineering, Technical University of Munich, Garching, Germany
| | - Anja Adelina Giese
- Institute of Molecular Pharmacology, University Hospital Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Sandra Fellendorf
- Institute of Molecular Pharmacology, University Hospital Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Stefan Düsterhöft
- Institute of Molecular Pharmacology, University Hospital Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Petra Mela
- Department of Mechanical Engineering, Munich School of BioEngineering, Technical University of Munich, Garching, Germany
| | - Andreas Ludwig
- Institute of Molecular Pharmacology, University Hospital Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| |
Collapse
|
33
|
Activity of Gas Transmitters in Vessels of the Anterior Abdominal Wall after Implantation of a Polypropylene Mesh. Bull Exp Biol Med 2020; 169:811-814. [PMID: 33123916 DOI: 10.1007/s10517-020-04984-6] [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: 12/24/2019] [Indexed: 10/23/2022]
Abstract
The distribution of NO and H2S in the arterial vessels of the anterior abdominal wall after implantation of a polypropylene mesh was studied by immunohistochemical methods at different stages of healing of the surgical wound in mature male Wistar rats. The presence of enzymes of NO and H2S synthesis in the wall of arterial vessels of the soft tissues of the anterior abdominal wall has been established. It has been shown that endothelial NO synthase is localized exclusively in the endothelium of both large and small vessels. Cystathionine γ lyase in small vessels is located only in the endothelial lining, whereas in large arteries and vessels of medium caliber, it is located in the endothelium and in myocytes. Inducible NO synthase appears in the artery wall only in animals with implanted polypropylene mesh by day 5 of the postoperative period, reaching the maximum by day 10. The content and localization of cystathionine γ lyase in the vascular wall of sham-operated and experimental rats did not much differ from the control values.
Collapse
|
34
|
Zhang Y, Zhou C, Li J, Zhang Y, Xie D, Liang M, Wang B, Song Y, Wang X, Huo Y, Hou FF, Xu X, Qin X. Serum alkaline phosphatase levels and the risk of new-onset diabetes in hypertensive adults. Cardiovasc Diabetol 2020; 19:186. [PMID: 33099298 PMCID: PMC7585682 DOI: 10.1186/s12933-020-01161-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 10/17/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND The association between alkaline phosphatase (ALP) and incident diabetes remains uncertain. Our study aimed to investigate the prospective relation of serum ALP with the risk of new-onset diabetes, and explore possible effect modifiers, in hypertensive adults. METHODS A total 14,393 hypertensive patients with available ALP measurements and without diabetes and liver disease at baseline were included from the China Stroke Primary Prevention Trial (CSPPT). The primary outcome was new-onset diabetes, defined as physician-diagnosed diabetes or use of glucose-lowering drugs during follow-up, or fasting glucose ≥ 7.0 mmol/L at the exit visit. The secondary study outcome was new-onset impaired fasting glucose (IFG), defined as FG < 6.1 mmol/L at baseline and ≥ 6.1 but < 7.0 mmol/L at the exit visit. RESULTS Over a median of 4.5 years follow-up, 1549 (10.8%) participants developed diabetes. Overall, there was a positive relation of serum ALP and the risk of new-onset diabetes (per SD increment, adjusted OR, 1.07; 95% CI: 1.01, 1.14) and new-onset IFG (per SD increment, adjusted OR, 1.07; 95% CI: 1.02, 1.14). Moreover, a stronger positive association between baseline ALP (per SD increment) with new-onset diabetes was found in participants with total homocysteine (tHcy) < 10 μmol/L (adjusted OR, 1.24; 95% CI: 1.10, 1.40 vs. ≥ 10 μmol/L: adjusted OR, 1.03; 95% CI: 0.96, 1.10; P-interaction = 0.007) or FG ≥ 5.9 mmol/L (adjusted OR, 1.16; 95% CI: 1.07, 1.27 vs. < 5.9 mmol/L: adjusted OR, 1.00; 95% CI: 0.93, 1.08; P-interaction = 0.009) CONCLUSIONS: In this non-diabetic, hypertensive population, higher serum ALP was significantly associated with the increased risk of new-onset diabetes, especially in those with lower tHcy or higher FG levels. Clinical Trial Registration-URL Trial registration: NCT00794885 (clinicaltrials.gov). Retrospectively registered November 20, 2008.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Division of Nephrology, Nanfang Hospital, Southern Medical UniversityNational Clinical Research Center for Kidney DiseaseState Key Laboratory of Organ Failure ResearchGuangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510515, China
| | - Chun Zhou
- Division of Nephrology, Nanfang Hospital, Southern Medical UniversityNational Clinical Research Center for Kidney DiseaseState Key Laboratory of Organ Failure ResearchGuangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510515, China
| | - Jianping Li
- Department of Cardiology, Peking University First Hospital, Beijing, 100034, China
| | - Yan Zhang
- Department of Cardiology, Peking University First Hospital, Beijing, 100034, China
| | - Di Xie
- Division of Nephrology, Nanfang Hospital, Southern Medical UniversityNational Clinical Research Center for Kidney DiseaseState Key Laboratory of Organ Failure ResearchGuangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510515, China
| | - Min Liang
- Division of Nephrology, Nanfang Hospital, Southern Medical UniversityNational Clinical Research Center for Kidney DiseaseState Key Laboratory of Organ Failure ResearchGuangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510515, China
| | - Binyan Wang
- Institute of Biomedicine, Anhui Medical University, Hefei, 230032, China
| | - Yun Song
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Xiaobin Wang
- Department of Population, Family and Reproductive Health, Johns Hopkins University Bloomberg School of Public Health, 615 N. Wolfe Street, E4132, Baltimore, MD, 21205-2179, USA
| | - Yong Huo
- Department of Cardiology, Peking University First Hospital, Beijing, 100034, China
| | - Fan Fan Hou
- Division of Nephrology, Nanfang Hospital, Southern Medical UniversityNational Clinical Research Center for Kidney DiseaseState Key Laboratory of Organ Failure ResearchGuangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510515, China
| | - Xiping Xu
- Division of Nephrology, Nanfang Hospital, Southern Medical UniversityNational Clinical Research Center for Kidney DiseaseState Key Laboratory of Organ Failure ResearchGuangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510515, China.
| | - Xianhui Qin
- Division of Nephrology, Nanfang Hospital, Southern Medical UniversityNational Clinical Research Center for Kidney DiseaseState Key Laboratory of Organ Failure ResearchGuangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510515, China.
| |
Collapse
|
35
|
Saternos H, Ley S, AbouAlaiwi W. Primary Cilia and Calcium Signaling Interactions. Int J Mol Sci 2020; 21:E7109. [PMID: 32993148 PMCID: PMC7583801 DOI: 10.3390/ijms21197109] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/23/2020] [Accepted: 09/24/2020] [Indexed: 02/06/2023] Open
Abstract
The calcium ion (Ca2+) is a diverse secondary messenger with a near-ubiquitous role in a vast array of cellular processes. Cilia are present on nearly every cell type in either a motile or non-motile form; motile cilia generate fluid flow needed for a variety of biological processes, such as left-right body patterning during development, while non-motile cilia serve as the signaling powerhouses of the cell, with vital singling receptors localized to their ciliary membranes. Much of the research currently available on Ca2+-dependent cellular actions and primary cilia are tissue-specific processes. However, basic stimuli-sensing pathways, such as mechanosensation, chemosensation, and electrical sensation (electrosensation), are complex processes entangled in many intersecting pathways; an overview of proposed functions involving cilia and Ca2+ interplay will be briefly summarized here. Next, we will focus on summarizing the evidence for their interactions in basic cellular activities, including the cell cycle, cell polarity and migration, neuronal pattering, glucose-mediated insulin secretion, biliary regulation, and bone formation. Literature investigating the role of cilia and Ca2+-dependent processes at a single-cellular level appears to be scarce, though overlapping signaling pathways imply that cilia and Ca2+ interact with each other on this level in widespread and varied ways on a perpetual basis. Vastly different cellular functions across many different cell types depend on context-specific Ca2+ and cilia interactions to trigger the correct physiological responses, and abnormalities in these interactions, whether at the tissue or the single-cell level, can result in diseases known as ciliopathies; due to their clinical relevance, pathological alterations of cilia function and Ca2+ signaling will also be briefly touched upon throughout this review.
Collapse
Affiliation(s)
| | | | - Wissam AbouAlaiwi
- Department of Pharmacology and Experimental Therapeutics, University of Toledo Health Science Campus, Toledo, OH 43614, USA; (H.S.); (S.L.)
| |
Collapse
|
36
|
Sarvasti D, Lalenoh I, Oepangat E, Purwowiyoto BS, Santoso A, Romdoni R. Cardiovascular Protection Variables Based on Exercise Intensity in Stable Coronary Heart Disease Patients After Coronary Stenting: A Comparative Study. Vasc Health Risk Manag 2020; 16:257-270. [PMID: 32753874 PMCID: PMC7352377 DOI: 10.2147/vhrm.s259190] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/15/2020] [Indexed: 11/23/2022] Open
Abstract
Purpose Our study aimed at determining and comparing the mechanism of cardiovascular protection variables in moderate-intensity continuous training (MICT) and high-intensity interval training (HIIT) in patients with stable coronary heart disease (CHD) after coronary stenting. Participants and Methods This experimental study used the same subject and cross-over design, involving eleven stable CHD patients after coronary stenting. These were randomly divided into two groups; MICT for 29 minutes at 50–60% heart rate reserve and HIIT with 4x4 minute intervals at 60–80% heart rate reserve, each followed by three minutes of active recovery at 40–50% heart rate reserve. These were conducted three times a week for two weeks. The participants’ levels of adrenaline, noradrenaline, endothelial nitric oxide synthase (eNOS), extracellular superoxide dismutase (EC-SOD) activity assayed, and flow-mediated dilatation (FMD) were examined before and after treatments were completed. Results The HIIT significantly increased the levels of noradrenaline and eNOS compared with MICT (p<0.05). Also, HIIT was better in maintaining EC-SOD activity and FMD compared with MICT (p<0.05). Through the noradrenalin pathway, HIIT had a direct and significant effect on eNOS and FMD (p<0.05) but MICT, through the noradrenaline pathways, had a direct and significant effect on eNOS (p<0.05), and through the EC-SOD activity pathways had a direct and significant effect on FMD (p<0.05). MICT reduced EC-SOD activity and also decreased the FMD value. Conclusion HIIT is superior to MICT in increasing cardiovascular protection by increasing the concentrations of noradrenalin and eNOS, maintaining EC-SOD activity, and FMD in stable CHD patients after coronary stenting.
Collapse
Affiliation(s)
- Dyana Sarvasti
- Department of Internal Medicine, Faculty of Medicine Widya Mandala Catholic University, Surabaya, Indonesia
| | - Isabella Lalenoh
- Department of Cardiology, Siloam Hospital TB Simatupang, Jakarta, Indonesia
| | - Emanoel Oepangat
- Department of Cardiology, Siloam Hospital TB Simatupang, Jakarta, Indonesia
| | - Budhi Setianto Purwowiyoto
- Department of Cardiology and Vascular Medicine, Faculty of Medicine University of Indonesia National Cardiovascular Center Harapan Kita, Jakarta, Indonesia
| | - Anwar Santoso
- Department of Cardiology and Vascular Medicine, Faculty of Medicine University of Indonesia National Cardiovascular Center Harapan Kita, Jakarta, Indonesia
| | - Rochmad Romdoni
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Airlangga University - Dr. Soetomo District General Hospital, Surabaya, Indonesia
| |
Collapse
|
37
|
Sciacqua A, Tripepi G, Perticone M, Cassano V, Fiorentino TV, Pititto GN, Maio R, Miceli S, Andreozzi F, Sesti G, Perticone F. Alkaline phosphatase affects renal function in never-treated hypertensive patients: effect modification by age. Sci Rep 2020; 10:9847. [PMID: 32555235 PMCID: PMC7299930 DOI: 10.1038/s41598-020-66911-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 04/06/2020] [Indexed: 11/16/2022] Open
Abstract
Several studies in patients with chronic kidney disease or normal renal function have shown that high levels of tissue non-specific alkaline phosphatase (ALP) are associated with an increased risk of all cause and cardiovascular (CV) mortality. Considering the independent prognostic role of renal function, we investigated the possible association between ALP levels and estimated glomerular filtration rate (e-GFR) in a large cohort of hypertensive subjects. We enrolled 2157 never-treated uncomplicated hypertensive patients with ALP levels within normal range. In the whole population, e-GFR was strongly related to ALP (r = −0.43, P < 0.0001) with similar magnitude in females and in males, resulting ALP the second independent predictor of renal function. In a multiple linear regression model, both on crude (P < 0.001) and adjusted (P = 0.01) analyses age significantly modified the effect of a fixed increase in ALP (20 UI/L) on renal function so that the reduction in e-GFR associated to a 20 UI/L increase in ALP was of lower magnitude in younger patients and progressively of higher extent from 20 years of age onwards. In conclusion, present data indicate a significant relationship between ALP levels and e-GFR in uncomplicated hypertensive patients that is modulated by age and that persisted after adjusting for several confounders.
Collapse
Affiliation(s)
- Angela Sciacqua
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy.
| | - Giovanni Tripepi
- CNR-IFC, Istituto di Fisiologia Clinica, Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, Reggio Calabria, Italy
| | - Maria Perticone
- Department of Experimental and Clinical Medicine, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Velia Cassano
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Teresa V Fiorentino
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Gerardo N Pititto
- ASST Sette Laghi, Internal Medicine Unit, University of Insubria, Varese, Italy
| | - Raffaele Maio
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Sofia Miceli
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Francesco Andreozzi
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Giorgio Sesti
- Department of Clinical and Molecular Medicine, University of Rome-Sapienza, Rome, Italy
| | - Francesco Perticone
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| |
Collapse
|
38
|
Liddle L, Burleigh MC, Monaghan C, Muggeridge DJ, Easton C. Venous occlusion during blood collection decreases plasma nitrite but not nitrate concentration in humans. Nitric Oxide 2020; 102:21-27. [PMID: 32535185 DOI: 10.1016/j.niox.2020.06.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 05/04/2020] [Accepted: 06/08/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND To maintain vascular tone and blood flow when tissue oxygenation is reduced, nitrite anions are reduced to nitric oxide (NO). From a practical perspective, it is unclear how the application of a tourniquet during blood collection might influence measurement of NO metabolites. Accordingly, this study evaluated the effect of venous occlusion on plasma nitrite and nitrate during venous blood collection. METHODS Fifteen healthy participants completed two trials that were preceded by the ingestion of nitrate-rich beetroot juice (BRJ; total of ~8.4 mmol nitrate) or no supplementation (control). In both trials, blood was collected using a venepuncture needle while a tourniquet was applied to the upper arm and using an indwelling intravenous cannula, from the opposing arm. The venepuncture samples were collected at 35 s post occlusion. Changes in the oxygenation of forearm flexor muscles were assessed using near-infrared spectroscopy. Plasma nitrite and nitrate were analysed using gas-phase chemiluminescence. RESULTS In the control trial, plasma nitrite was significantly elevated when collected via the cannula (179 ± 67 nM) compared to venepuncture (112 ± 51 nM, P = 0.03). The ingestion of BRJ increased plasma nitrite and values remained higher when sampled from the cannula (473 ± 164 nM) compared to venepuncture (387 ± 136 nM, P < 0.001). Plasma nitrate did not differ between collection methods in either trial (all P > 0.05). The delta changes in total-, deoxy-, and oxy-haemoglobin were all significantly greater during venepuncture sample compared to the cannula sample at the point of blood collection (all P < 0.05). CONCLUSIONS Venous occlusion during venepuncture blood collection lowers plasma nitrite concentration, potentially due to localised changes in haemoglobin concentration and/or a suppression of endogenous NO synthesis. Accordingly, the method of blood collection to enable measurements of NO metabolites should be carefully considered and consistently reported by researchers.
Collapse
Affiliation(s)
- Luke Liddle
- Institute for Clinical Exercise and Health Science, University of the West of Scotland, Hamilton, UK
| | - Mia C Burleigh
- Institute for Clinical Exercise and Health Science, University of the West of Scotland, Hamilton, UK
| | - Chris Monaghan
- Institute for Clinical Exercise and Health Science, University of the West of Scotland, Hamilton, UK
| | - David J Muggeridge
- Institute of Health Research & Innovation, Division of Biomedical Science, University of the Highlands and Islands, Inverness, UK
| | - Chris Easton
- Institute for Clinical Exercise and Health Science, University of the West of Scotland, Hamilton, UK.
| |
Collapse
|
39
|
Odelin G, Faure E, Maurel-Zaffran C, Zaffran S. Krox20 Regulates Endothelial Nitric Oxide Signaling in Aortic Valve Development and Disease. J Cardiovasc Dev Dis 2019; 6:jcdd6040039. [PMID: 31684048 PMCID: PMC6955692 DOI: 10.3390/jcdd6040039] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/25/2019] [Accepted: 10/31/2019] [Indexed: 12/18/2022] Open
Abstract
Among the aortic valve diseases, the bicuspid aortic valve (BAV) occurs when the aortic valve has two leaflets (cusps), rather than three, and represents the most common form of congenital cardiac malformation, affecting 1–2% of the population. Despite recent advances, the etiology of BAV is poorly understood. We have recently shown that Krox20 is expressed in endothelial and cardiac neural crest derivatives that normally contribute to aortic valve development and that lack of Krox20 in these cells leads to aortic valve defects including partially penetrant BAV formation. Dysregulated expression of endothelial nitric oxide synthase (Nos3) is associated with BAV. To investigate the relationship between Krox20 and Nos3 during aortic valve development, we performed inter-genetic cross. While single heterozygous mice had normal valve formation, the compound Krox20+/−;Nos3+/− mice had BAV malformations displaying an in vivo genetic interaction between these genes for normal valve morphogenesis. Moreover, in vivo and in vitro experiments demonstrate that Krox20 directly binds to Nos3 proximal promoter to activate its expression. Our data suggests that Krox20 is a regulator of nitric oxide in endothelial-derived cells in the development of the aortic valve and concludes on the interaction of Krox20 and Nos3 in BAV formation.
Collapse
Affiliation(s)
- Gaëlle Odelin
- Aix Marseille University, INSERM, Marseille Medical Genetics, U1251, 13005 Marseille, France.
| | - Emilie Faure
- Aix Marseille University, INSERM, Marseille Medical Genetics, U1251, 13005 Marseille, France.
| | | | - Stéphane Zaffran
- Aix Marseille University, INSERM, Marseille Medical Genetics, U1251, 13005 Marseille, France.
| |
Collapse
|
40
|
Dalla K, Bech‐Hanssen O, Ricksten S. Impact of norepinephrine on right ventricular afterload and function in septic shock-a strain echocardiography study. Acta Anaesthesiol Scand 2019; 63:1337-1345. [PMID: 31361336 PMCID: PMC7159388 DOI: 10.1111/aas.13454] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 07/06/2019] [Accepted: 07/23/2019] [Indexed: 12/25/2022]
Abstract
Background In this observational study, the effects of norepinephrine‐induced changes in mean arterial pressure (MAP) on right ventricular (RV) systolic function, afterload and pulmonary haemodynamics were studied in septic shock patients. We hypothesised that RV systolic function improves at higher doses of norepinephrine/MAP levels. Methods Eleven patients with septic shock requiring norepinephrine after fluid resuscitation were included <24 hours after ICU arrival. Study enrolment and insertion of a pulmonary artery catheter was performed after written informed consent from the next of kin. Norepinephrine infusion was titrated to target mean arterial pressures (MAP) of 60, 75 and 90 mmHg in a random sequential order. At each target MAP, strain—and conventional echocardiographic—and pulmonary haemodynamic variables were measured. RV afterload was assessed as effective pulmonary arterial elastance, (Epa) and pulmonary vascular resistance index, (PVRI). RV free wall peak strain was the primary end‐point. Results At highest compared to lowest norepinephrine dose/MAP level, RV free wall peak strain increased from −19% to −25% (32%, P = .003), accompanied by increased tricuspid annular plane systolic excursion (22%, P = .01). At the highest norepinephrine dose/MAP, RV end‐diastolic area index (16%, P < .001), central venous pressure (38%, P < .001), stroke volume index (7%, P = .001), mean pulmonary artery pressure (19%, P < .001) and RV stroke work index (15%, P = .045) increased, with no effects on PVRI or Epa. Cardiac index did not change, assessed by thermodilution (P = .079) and echocardiography (P = .054). Conclusions Higher doses of norepinephrine to a target MAP of 90 mm Hg improved RV systolic function while RV afterload was not affected.
Collapse
Affiliation(s)
- Keti Dalla
- Department of Anaesthesiology and Intensive Care Medicine, Sahlgrenska Academy University of Gothenburg, Sahlgrenska University Hospital Gothenburg Sweden
| | - Odd Bech‐Hanssen
- Department of Clinical Physiology, Sahlgrenska Academy University of Gothenburg, Sahlgrenska University Hospital Gothenburg Sweden
| | - Sven‐Erik Ricksten
- Department of Anaesthesiology and Intensive Care Medicine, Sahlgrenska Academy University of Gothenburg, Sahlgrenska University Hospital Gothenburg Sweden
| |
Collapse
|
41
|
Zemskov EA, Lu Q, Ornatowski W, Klinger CN, Desai AA, Maltepe E, Yuan JXJ, Wang T, Fineman JR, Black SM. Biomechanical Forces and Oxidative Stress: Implications for Pulmonary Vascular Disease. Antioxid Redox Signal 2019; 31:819-842. [PMID: 30623676 PMCID: PMC6751394 DOI: 10.1089/ars.2018.7720] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Oxidative stress in the cell is characterized by excessive generation of reactive oxygen species (ROS). Superoxide (O2-) and hydrogen peroxide (H2O2) are the main ROS involved in the regulation of cellular metabolism. As our fundamental understanding of the underlying causes of lung disease has increased it has become evident that oxidative stress plays a critical role. Recent Advances: A number of cells in the lung both produce, and respond to, ROS. These include vascular endothelial and smooth muscle cells, fibroblasts, and epithelial cells as well as the cells involved in the inflammatory response, including macrophages, neutrophils, eosinophils. The redox system is involved in multiple aspects of cell metabolism and cell homeostasis. Critical Issues: Dysregulation of the cellular redox system has consequential effects on cell signaling pathways that are intimately involved in disease progression. The lung is exposed to biomechanical forces (fluid shear stress, cyclic stretch, and pressure) due to the passage of blood through the pulmonary vessels and the distension of the lungs during the breathing cycle. Cells within the lung respond to these forces by activating signal transduction pathways that alter their redox state with both physiologic and pathologic consequences. Future Directions: Here, we will discuss the intimate relationship between biomechanical forces and redox signaling and its role in the development of pulmonary disease. An understanding of the molecular mechanisms induced by biomechanical forces in the pulmonary vasculature is necessary for the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Evgeny A Zemskov
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Qing Lu
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Wojciech Ornatowski
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Christina N Klinger
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Ankit A Desai
- Department of Medicine, Indiana University, Indianapolis, Indiana
| | - Emin Maltepe
- Department of Pediatrics, University of California, San Francisco, San Francisco, California
| | - Jason X-J Yuan
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Ting Wang
- Department of Internal Medicine, The University of Arizona Health Sciences, Phoenix, Arizona
| | - Jeffrey R Fineman
- Department of Pediatrics, University of California, San Francisco, San Francisco, California
| | - Stephen M Black
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| |
Collapse
|
42
|
Cui J, Liu Y, Fu BM. Numerical study on the dynamics of primary cilium in pulsatile flows by the immersed boundary-lattice Boltzmann method. Biomech Model Mechanobiol 2019; 19:21-35. [PMID: 31256275 DOI: 10.1007/s10237-019-01192-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 06/21/2019] [Indexed: 10/26/2022]
Abstract
An explicit immersed boundary-lattice Boltzmann method is applied to numerically investigate the dynamics of primary cilium in pulsatile blood flows with two-way fluid-structure interaction considered. To well characterize the effect of cilium basal body on cilium dynamics, the cilium base is modeled as a nonlinear rotational spring attached to the cilium's basal end as proposed by Resnick (Biophys J 109:18-25, 2015. https://doi.org/10.1016/j.bpj.2015.05.031). After several careful validations, the fluid-cilium interaction system is investigated in detail at various pulsatile flow conditions that are characterized by peak Reynolds numbers ([Formula: see text]) and Womersley numbers ([Formula: see text]). The periodic flapping of primary cilium observed in our simulations is very similar to the in vivo ciliary oscillation captured by O'Connor et al. (Cilia 2:8, 2013. https://doi.org/10.1186/2046-2530-2-8). The cilium's dynamics is found to be closely related to the [Formula: see text] and [Formula: see text]. Increase the [Formula: see text] or decrease the [Formula: see text] bring to an increase in the cilium's flapping amplitude, tip angular speed, basal rotation, and maximum tensile stress. It is also demonstrated that by reducing the [Formula: see text] or enhancing the [Formula: see text] to a certain level, one can shift the flapping pattern of cilium from its original two-side one to a one-side one, making the stretch only happen on one particular side. During the flapping process, the location of the maximum tensile stress is not always found at the basal region; instead, it is able to propagate from time to time within a certain distance to the base. Due to the obstruction of the primary cilium, the distribution of wall shear stress no longer remains uniform as in the absence of cilia. It oscillates in space with the minimum magnitude which is always found near where the cilium is located. The presence of cilium also reduces the overall level of wall shear stress, especially at the region near the cilium's anchor point.
Collapse
Affiliation(s)
- Jingyu Cui
- Research Centre for Fluid-Structure Interactions, Department of Mechanical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Yang Liu
- Research Centre for Fluid-Structure Interactions, Department of Mechanical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong.
| | - Bingmei M Fu
- Department of Biomedical Engineering, City College of New York, New York City, USA
| |
Collapse
|
43
|
Iring A, Jin YJ, Albarrán-Juárez J, Siragusa M, Wang S, Dancs PT, Nakayama A, Tonack S, Chen M, Künne C, Sokol AM, Günther S, Martínez A, Fleming I, Wettschureck N, Graumann J, Weinstein LS, Offermanns S. Shear stress-induced endothelial adrenomedullin signaling regulates vascular tone and blood pressure. J Clin Invest 2019; 129:2775-2791. [PMID: 31205027 DOI: 10.1172/jci123825] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 05/01/2019] [Indexed: 12/22/2022] Open
Abstract
Hypertension is a primary risk factor for cardiovascular diseases including myocardial infarction and stroke. Major determinants of blood pressure are vasodilatory factors such as nitric oxide (NO) released from the endothelium under the influence of fluid shear stress exerted by the flowing blood. Several endothelial signaling processes mediating fluid shear stress-induced formation and release of vasodilatory factors have been described. It is, however, still poorly understood how fluid shear stress induces these endothelial responses. Here we show that the endothelial mechanosensitive cation channel PIEZO1 mediated fluid shear stress-induced release of adrenomedullin, which in turn activated its Gs-coupled receptor. The subsequent increase in cAMP levels promoted the phosphorylation of endothelial NO synthase (eNOS) at serine 633 through protein kinase A (PKA), leading to the activation of the enzyme. This Gs/PKA-mediated pathway synergized with the AKT-mediated pathways leading to eNOS phosphorylation at serine 1177. Mice with endothelium-specific deficiency of adrenomedullin, the adrenomedullin receptor, or Gαs showed reduced flow-induced eNOS activation and vasodilation and developed hypertension. Our data identify fluid shear stress-induced PIEZO1 activation as a central regulator of endothelial adrenomedullin release and establish the adrenomedullin receptor and subsequent Gs-mediated formation of cAMP as a critical endothelial mechanosignaling pathway regulating basal endothelial NO formation, vascular tone, and blood pressure.
Collapse
Affiliation(s)
- Andras Iring
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Young-June Jin
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Julián Albarrán-Juárez
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Mauro Siragusa
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany.,German Centre for Cardiovascular Research (DZHK), Rhine-Main site, Frankfurt and Bad Nauheim, Germany
| | - ShengPeng Wang
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Yanta District, Xi'an, China
| | - Péter T Dancs
- Institute of Clinical Experimental Research, Semmelweis University, Budapest, Hungary
| | - Akiko Nakayama
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Sarah Tonack
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Min Chen
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | | | - Anna M Sokol
- Scientific Service Group Biomolecular Mass Spectrometry, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | | | - Alfredo Martínez
- Oncology Area, Center for Biomedical Research of La Rioja (CIBIR), Logroño, Spain
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany.,German Centre for Cardiovascular Research (DZHK), Rhine-Main site, Frankfurt and Bad Nauheim, Germany
| | - Nina Wettschureck
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,German Centre for Cardiovascular Research (DZHK), Rhine-Main site, Frankfurt and Bad Nauheim, Germany.,Centre for Molecular Medicine, Medical Faculty, Goethe University, Frankfurt am Main, Germany
| | - Johannes Graumann
- German Centre for Cardiovascular Research (DZHK), Rhine-Main site, Frankfurt and Bad Nauheim, Germany.,Scientific Service Group Biomolecular Mass Spectrometry, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Lee S Weinstein
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,German Centre for Cardiovascular Research (DZHK), Rhine-Main site, Frankfurt and Bad Nauheim, Germany.,Centre for Molecular Medicine, Medical Faculty, Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
44
|
Kang JJ, Treadwell TA, Bodary PF, Shayman JA. Voluntary wheel running activates Akt/AMPK/eNOS signaling cascades without improving profound endothelial dysfunction in mice deficient in α-galactosidase A. PLoS One 2019; 14:e0217214. [PMID: 31120949 PMCID: PMC6533039 DOI: 10.1371/journal.pone.0217214] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 05/07/2019] [Indexed: 11/19/2022] Open
Abstract
Fabry disease is caused by loss of activity of the lysosomal hydrolase α-galactosidase A (GLA). Premature life-threatening complications in Fabry patients arise from cardiovascular disease, including stroke and myocardial infarction. Exercise training has been shown to improve endothelial dysfunction in various settings including coronary artery disease. However, the effects of exercise training on endothelial dysfunction in Fabry disease have not been investigated. Gla knockout mice were single-housed in a cage equipped with a voluntary wheel (EX) or no wheel (SED) for 12 weeks. Exercised mice ran 10 km/day on average during the voluntary running intervention (VR) period. Despite significantly higher food intake in EX than SED, body weights of EX and SED remained stable during the VR period. After the completion of VR, citrate synthase activity in gastrocnemius muscle was significantly higher in EX than SED. VR resulted in greater phosphorylation of Akt (S473) and AMPK (T172) in the aorta of EX compared to SED measured by western blot. Furthermore, VR significantly enhanced eNOS protein expression and phosphorylation at S1177 by 20% and 50% in the aorta of EX when compared with SED. Similarly, plasma nitrate and nitrite levels were 77% higher in EX than SED. In contrast, measures of anti- and pro-oxidative enzymes (superoxide dismutase and p67phox subunit of NADPH oxidase) and overall oxidative stress (plasma oxidized glutathione) were not different between groups. Although the aortic endothelial relaxation to acetylcholine was slightly increased in EX, it did not reach statistical significance. This study provides the first evidence that VR improves Akt/AMPK/eNOS signaling cascades, but not endothelial function in the aorta of aged Gla deficient mice.
Collapse
Affiliation(s)
- Justin J. Kang
- School of Kinesiology, University of Michigan, Ann Arbor, MI, United States of America
| | - Taylour A. Treadwell
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, United States of America
| | - Peter F. Bodary
- School of Kinesiology, University of Michigan, Ann Arbor, MI, United States of America
| | - James A. Shayman
- Division of Nephrology, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, United States of America
- * E-mail:
| |
Collapse
|
45
|
Kumar S, Williams D, Sur S, Wang JY, Jo H. Role of flow-sensitive microRNAs and long noncoding RNAs in vascular dysfunction and atherosclerosis. Vascul Pharmacol 2019; 114:76-92. [PMID: 30300747 PMCID: PMC6905428 DOI: 10.1016/j.vph.2018.10.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 09/19/2018] [Accepted: 10/05/2018] [Indexed: 02/07/2023]
Abstract
Atherosclerosis is the primary underlying cause of myocardial infarction, ischemic stroke, and peripheral artery disease. The disease preferentially occurs in arterial regions exposed to disturbed blood flow, in part, by altering expression of flow-sensitive coding- and non-coding genes. In this review, we summarize the role of noncoding RNAs, [microRNAs (miRNAs) and long noncoding RNAs(lncRNAs)], as regulators of gene expression and outline their relationship to the pathogenesis of atherosclerosis. While miRNAs are small noncoding genes that post-transcriptionally regulate gene expression by targeting mRNA transcripts, the lncRNAs regulate gene expression by diverse mechanisms, which are still emerging and incompletely understood. We focused on multiple flow-sensitive miRNAs such as, miR-10a, -19a, -23b, -17~92, -21, -663, -92a, -143/145, -101, -126, -712, -205, and -155 that play a critical role in endothelial function and atherosclerosis by targeting inflammation, cell cycle, proliferation, migration, apoptosis, and nitric oxide signaling. Flow-dependent regulation of lncRNAs is just emerging, and their role in vascular dysfunction and atherosclerosis is unknown. Here, we discuss the flow-sensitive lncRNA STEEL along with other lncRNAs studied in the context of vascular pathophysiology and atherosclerosis such as MALAT1, MIAT1, ANRIL, MYOSLID, MEG3, SENCR, SMILR, LISPR1, and H19. Also discussed is the use of these noncoding RNAs as potential biomarkers and therapeutics to reduce and regress atherosclerosis.
Collapse
Affiliation(s)
- Sandeep Kumar
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, USA
| | - Darian Williams
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, USA
| | - Sanjoli Sur
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, USA
| | - Jun-Yao Wang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, USA
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, USA; Division of Cardiology, Emory University, Atlanta, USA.
| |
Collapse
|
46
|
Looft‐Wilson RC, Todd SE, Berberich KM, Wolfert MR. Flow does not alter eNOS phosphoryation at Ser1179 or Thr495 in preconstricted mouse mesenteric arteries. Physiol Rep 2018; 6:e13864. [PMID: 30247813 PMCID: PMC6129772 DOI: 10.14814/phy2.13864] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 08/17/2018] [Accepted: 08/20/2018] [Indexed: 12/23/2022] Open
Abstract
In arteries, endothelium-dependent vasodilatory agonists and flow-induced shear stress cause vasodilation largely by activation of the endothelial enzyme eNOS, which generates nitric oxide that relaxes vascular smooth muscle. Agonists activate eNOS in part through increased phosphorylation at Ser1179 and decreased phosphorylation at Thr495. We previously found that preconstriction of intact, isolated mouse mesenteric arteries with phenylephrine also caused increased Ser1179 and decreased Thr495 eNOS phosphorylation, and sequential treatment with the vasodilatory agonist acetylcholine did not cause any further change in phosphorylation at these sites, despite producing vasodilation. The present study tests the hypothesis that luminal flow in these arteries preconstricted with phenylephrine also produces vasodilation without phosphorylation changes at these sites. First-order mesenteric arteries, isolated from male C57/BL6 mice (7-20 weeks of age) anesthetized with pentobarbital (50 mg/kg, i.p.), were cannulated, pressurized, and treated with stepped increases in luminal flow (15-120 μL/min). Flow resulted in dilation that plateaued at ~60 μL/min (31.3 ± 3.0% dilation) and was significantly (P < 0.001) NOS-dependent at all flow rates (determined by 10-4 mol/L L-NAME treatment). In separate arteries, preconstriction with phenylephrine (10-5 mol/L) resulted in increased eNOS phosphorylation at Ser1179 (P < 0.05) and decreased phosphorylation at Thr495, but subsequent flow at 60 μL/min for 5 or 15 min did not cause further changes in phosphorylation, despite causing dilation. Thus, flow-induced dilation does not require changes in these eNOS phosphorylation sites beyond those induced by alpha1-adrenergic stimulation with phenylephrine, indicating that eNOS is activated by other mechanisms during acute flow-induced dilation of preconstricted arteries.
Collapse
Affiliation(s)
- Robin C. Looft‐Wilson
- Department of Kinesiology and Health SciencesThe College of William & MaryWilliamsburgVirginia
| | - Sarah E. Todd
- Department of Kinesiology and Health SciencesThe College of William & MaryWilliamsburgVirginia
| | - Kristen M. Berberich
- Department of Kinesiology and Health SciencesThe College of William & MaryWilliamsburgVirginia
| | - Madeline R. Wolfert
- Department of Kinesiology and Health SciencesThe College of William & MaryWilliamsburgVirginia
| |
Collapse
|
47
|
Saternos HC, AbouAlaiwi WA. Signaling interplay between primary cilia and nitric oxide: A mini review. Nitric Oxide 2018; 80:108-112. [PMID: 30099097 DOI: 10.1016/j.niox.2018.08.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 06/01/2018] [Accepted: 08/06/2018] [Indexed: 01/12/2023]
Abstract
New discoveries into the functional role of primary cilia are on the rise. In little more than 20 years, research has shown the once vestigial organelle is a signaling powerhouse involved in a vast number of essential cellular processes. In the same decade that interest in primary cilia was burgeoning, nitric oxide won molecule of the year and a Nobel prize for its role as a near ubiquitous signaling molecule. Although primary cilia and nitric oxide are both involved in signaling, a direct relationship has not been investigated; however, after a quick review of the literature, parallels between their functions can be drawn. This review aims to suggest a possible interplay between primary cilia and nitric oxide signaling especially in the areas of vascular tissue homeostasis and cellular proliferation.
Collapse
Affiliation(s)
- Hannah C Saternos
- University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Department of Pharmacology and Experimental Therapeutics, USA
| | - Wissam A AbouAlaiwi
- University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Department of Pharmacology and Experimental Therapeutics, USA.
| |
Collapse
|
48
|
Wareham LK, Buys ES, Sappington RM. The nitric oxide-guanylate cyclase pathway and glaucoma. Nitric Oxide 2018; 77:75-87. [PMID: 29723581 DOI: 10.1016/j.niox.2018.04.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 04/18/2018] [Accepted: 04/23/2018] [Indexed: 01/12/2023]
Abstract
Glaucoma is a prevalent optic neuropathy characterized by the progressive dysfunction and loss of retinal ganglion cells (RGCs) and their optic nerve axons, which leads to irreversible visual field loss. Multiple risk factors for the disease have been identified, but elevated intraocular pressure (IOP) remains the primary risk factor amenable to treatment. Reducing IOP however does not always prevent glaucomatous neurodegeneration, and many patients progress with the disease despite having IOP in the normal range. There is increasing evidence that nitric oxide (NO) is a direct regulator of IOP and that dysfunction of the NO-Guanylate Cyclase (GC) pathway is associated with glaucoma incidence. NO has shown promise as a novel therapeutic with targeted effects that: 1) lower IOP; 2) increase ocular blood flow; and 3) confer neuroprotection. The various effects of NO in the eye appear to be mediated through the activation of the GC- guanosine 3:5'-cyclic monophosphate (cGMP) pathway and its effect on downstream targets, such as protein kinases and Ca2+ channels. Although NO-donor compounds are promising as therapeutics for IOP regulation, they may not be ideal to harness the neuroprotective potential of NO signaling. Here we review evidence that supports direct targeting of GC as a novel pleiotrophic treatment for the disease, without the need for direct NO application. The identification and targeting of other factors that contribute to glaucoma would be beneficial to patients, particularly those that do not respond well to IOP-dependent interventions.
Collapse
Affiliation(s)
- Lauren K Wareham
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | - Rebecca M Sappington
- Department of Ophthalmology and Visual Sciences, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
49
|
Pattern of vascular remodeling of distal reference segment after recanalization of chronic total occlusion, long-term angiographic follow-up. Egypt Heart J 2018; 69:161-163. [PMID: 29622971 PMCID: PMC5839350 DOI: 10.1016/j.ehj.2016.10.002] [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: 06/03/2016] [Accepted: 10/08/2016] [Indexed: 11/24/2022] Open
Abstract
Chronic total occlusion (CTO) is probably caused by thrombus and lipid-rich cholesterol esters that are replaced over time by collagen and calcium deposition. Experimental models showed endothelial cell necrosis in response to vessel ligation, whereas more recent models suggest that the endothelium might retain viability guiding the subsequent development of CTO, including CTO neo-revascularization, which occurs within the lumen and in various layers of the vessel wall, by the release of paracrine substances. It is uncertain whether after CTO recanalization the recovery of anterograde reverses endothelial dysfunction, thus promoting vasodilation and positive remodeling.
Collapse
|
50
|
Atkinson L, Yusuf MZ, Aburima A, Ahmed Y, Thomas SG, Naseem KM, Calaminus SDJ. Reversal of stress fibre formation by Nitric Oxide mediated RhoA inhibition leads to reduction in the height of preformed thrombi. Sci Rep 2018; 8:3032. [PMID: 29445102 PMCID: PMC5813033 DOI: 10.1038/s41598-018-21167-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 01/24/2018] [Indexed: 01/10/2023] Open
Abstract
Evidence has emerged to suggest that thrombi are dynamic structures with distinct areas of differing platelet activation and inhibition. We hypothesised that Nitric oxide (NO), a platelet inhibitor, can modulate the actin cytoskeleton reversing platelet spreading, and therefore reduce the capability of thrombi to withstand a high shear environment. Our data demonstrates that GSNO, DEANONOate, and a PKG-activating cGMP analogue reversed stress fibre formation and increased actin nodule formation in adherent platelets. This effect is sGC dependent and independent of ADP and thromboxanes. Stress fibre formation is a RhoA dependent process and NO induced RhoA inhibition, however, it did not phosphorylate RhoA at ser188 in spread platelets. Interestingly NO and PGI2 synergise to reverse stress fibre formation at physiologically relevant concentrations. Analysis of high shear conditions indicated that platelets activated on fibrinogen, induced stress fibre formation, which was reversed by GSNO treatment. Furthermore, preformed thrombi on collagen post perfused with GSNO had a 30% reduction in thrombus height in comparison to the control. This study demonstrates that NO can reverse key platelet functions after their initial activation and identifies a novel mechanism for controlling excessive thrombosis.
Collapse
Affiliation(s)
- L Atkinson
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull, HU6 7RX, UK
| | - M Z Yusuf
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull, HU6 7RX, UK
| | - A Aburima
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull, HU6 7RX, UK
| | - Y Ahmed
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull, HU6 7RX, UK
| | - S G Thomas
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK.,Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham, UK
| | - K M Naseem
- Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, LS2 9JT, UK
| | - S D J Calaminus
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull, HU6 7RX, UK.
| |
Collapse
|