1
|
Subrahmanian S, Varshney R, Subramani K, Murphy B, Woolington S, Ahamed J. N-Acetylcysteine Inhibits Aortic Stenosis Progression in a Murine Model by Blocking Shear-Induced Activation of Platelet Latent Transforming Growth Factor Beta 1. Antioxid Redox Signal 2024; 41:e1187-e1196. [PMID: 34619980 PMCID: PMC11693965 DOI: 10.1089/ars.2021.0037] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 09/24/2021] [Accepted: 09/24/2021] [Indexed: 01/08/2023]
Abstract
Objective: Aortic stenosis (AS) is characterized by narrowing of the aortic valve opening, resulting in peak blood flow velocity that induces high wall shear stress (WSS) across the valve. Severe AS leads to heart failure and death. There is no treatment available for AS other than valve replacement. Platelet-derived transforming growth factor beta 1 (TGF-β1) partially contributes to AS progression in mice, and WSS is a potent activator of latent TGF-β1. N-acetylcysteine (NAC) inhibits WSS-induced TGF-β1 activation in vitro. We hypothesize that NAC will inhibit AS progression by inhibiting WSS-induced TGF-β1 activation. Approach: We treated a cohort of Ldlr(-/-)Apob(100/100) low density lipoprotein receptor (LDLR) mice fed a high-fat diet with NAC (2% in drinking water) at different stages of disease progression and measured its effect on AS progression and TGF-β1 activation. Results: Short-term NAC treatment inhibited AS progression in mice with moderate and severe AS relative to controls, but not in LDLR mice lacking platelet-derived TGF-β1 (TGF-β1platlet-KO-LDLR). NAC treatment reduced TGF-β signaling, p-Smad2 and collagen levels, and mesenchymal transition from isolectin B4 and CD45-positive cells in LDLR mice. Mechanistically, NAC treatment resulted in plasma NAC concentrations ranging from 75.5 to 449.2 ng/mL, which were sufficient to block free thiol labeling of plasma proteins and reduce active TGF-β1 levels without substantially affecting reactive oxygen species-modified products in valvular cells. Conclusions: Short-term treatment with NAC inhibits AS progression by inhibiting WSS-induced TGF-β1 activation in the LDLR mouse model of AS, motivating a clinical trial of NAC and/or other thiol-reactive agent(s) as a potential therapy for AS.
Collapse
Affiliation(s)
- Sandeep Subrahmanian
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | - Rohan Varshney
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | - Kumar Subramani
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | - Brennah Murphy
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | - Sean Woolington
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | - Jasimuddin Ahamed
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
2
|
Hamczyk MR, Nevado RM, Gonzalo P, Andrés-Manzano MJ, Nogales P, Quesada V, Rosado A, Torroja C, Sánchez-Cabo F, Dopazo A, Bentzon JF, López-Otín C, Andrés V. Endothelial-to-Mesenchymal Transition Contributes to Accelerated Atherosclerosis in Hutchinson-Gilford Progeria Syndrome. Circulation 2024; 150:1612-1630. [PMID: 39206565 DOI: 10.1161/circulationaha.123.065768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 07/26/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Atherosclerosis is the main medical problem in Hutchinson-Gilford progeria syndrome, a rare premature aging disorder caused by the mutant lamin-A protein progerin. Recently, we found that limiting progerin expression to vascular smooth muscle cells (VSMCs) is sufficient to hasten atherosclerosis and death in Apoe-deficient mice. However, the impact of progerin-driven VSMC defects on endothelial cells (ECs) remained unclear. METHODS Apoe- or Ldlr-deficient C57BL/6J mice with ubiquitous, VSMC-, EC- or myeloid-specific progerin expression fed a normal or high-fat diet were used to study endothelial phenotype during Hutchinson-Gilford progeria syndrome-associated atherosclerosis. Endothelial permeability to low-density lipoproteins was assessed by intravenous injection of fluorescently labeled human low-density lipoprotein and confocal microscopy analysis of the aorta. Leukocyte recruitment to the aortic wall was evaluated by en face immunofluorescence. Endothelial-to-mesenchymal transition (EndMT) was assessed by quantitative polymerase chain reaction and RNA sequencing in the aortic intima and by immunofluorescence in aortic root sections. TGFβ (transforming growth factor β) signaling was analyzed by multiplex immunoassay in serum, by Western blot in the aorta, and by immunofluorescence in aortic root sections. The therapeutic benefit of TGFβ1/SMAD3 pathway inhibition was evaluated in mice by intraperitoneal injection of SIS3 (specific inhibitor of SMAD3), and vascular phenotype was assessed by Oil Red O staining, histology, and immunofluorescence in the aorta and the aortic root. RESULTS Both ubiquitous and VSMC-specific progerin expression in Apoe-null mice provoked alterations in aortic ECs, including increased permeability to low-density lipoprotein and leukocyte recruitment. Atherosclerotic lesions in these progeroid mouse models, but not in EC- and myeloid-specific progeria models, contained abundant cells combining endothelial and mesenchymal features, indicating extensive EndMT triggered by dysfunctional VSMCs. Accordingly, the intima of ubiquitous and VSMC-specific progeroid models at the onset of atherosclerosis presented increased expression of EndMT-linked genes, especially those specific to fibroblasts and extracellular matrix. Aorta in both models showed activation of the TGFβ1/SMAD3 pathway, a major trigger of EndMT, and treatment of VSMC-specific progeroid mice with SIS3 alleviated the aortic phenotype. CONCLUSIONS Progerin-induced VSMC alterations promote EC dysfunction and EndMT through TGFβ1/SMAD3, identifying this process as a candidate target for Hutchinson-Gilford progeria syndrome treatment. These findings also provide insight into the complex role of EndMT during atherogenesis.
Collapse
Affiliation(s)
- Magda R Hamczyk
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología, Universidad de Oviedo, Spain (M.R.H., V.Q., C.L.-O.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Spain (M.R.H., R.M.N., P.G., M.J.A.-M., A.D., V.A.)
| | - Rosa M Nevado
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (R.M.N., P.G., M.J.A.-M., P.N., A.R., C.T., F.S.-C., A.D., J.F.B., V.A.)
| | - Pilar Gonzalo
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Spain (M.R.H., R.M.N., P.G., M.J.A.-M., A.D., V.A.)
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (R.M.N., P.G., M.J.A.-M., P.N., A.R., C.T., F.S.-C., A.D., J.F.B., V.A.)
| | - María J Andrés-Manzano
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Spain (M.R.H., R.M.N., P.G., M.J.A.-M., A.D., V.A.)
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (R.M.N., P.G., M.J.A.-M., P.N., A.R., C.T., F.S.-C., A.D., J.F.B., V.A.)
| | - Paula Nogales
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (R.M.N., P.G., M.J.A.-M., P.N., A.R., C.T., F.S.-C., A.D., J.F.B., V.A.)
| | - Víctor Quesada
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología, Universidad de Oviedo, Spain (M.R.H., V.Q., C.L.-O.)
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (R.M.N., P.G., M.J.A.-M., P.N., A.R., C.T., F.S.-C., A.D., J.F.B., V.A.)
- Centro de Investigación Biomédica en Red de Cáncer, Spain (V.Q.)
| | - Aránzazu Rosado
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (R.M.N., P.G., M.J.A.-M., P.N., A.R., C.T., F.S.-C., A.D., J.F.B., V.A.)
| | - Carlos Torroja
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (R.M.N., P.G., M.J.A.-M., P.N., A.R., C.T., F.S.-C., A.D., J.F.B., V.A.)
| | - Fátima Sánchez-Cabo
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (R.M.N., P.G., M.J.A.-M., P.N., A.R., C.T., F.S.-C., A.D., J.F.B., V.A.)
| | - Ana Dopazo
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Spain (M.R.H., R.M.N., P.G., M.J.A.-M., A.D., V.A.)
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (R.M.N., P.G., M.J.A.-M., P.N., A.R., C.T., F.S.-C., A.D., J.F.B., V.A.)
| | - Jacob F Bentzon
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (R.M.N., P.G., M.J.A.-M., P.N., A.R., C.T., F.S.-C., A.D., J.F.B., V.A.)
- Department of Clinical Medicine, Aarhus University, Denmark (J.F.B.)
| | - Carlos López-Otín
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología, Universidad de Oviedo, Spain (M.R.H., V.Q., C.L.-O.)
- Facultad de Ciencias de la Vida y la Naturaleza, Universidad Nebrija, Madrid, Spain (C.L.-O.)
- Centre de Recherche des Cordeliers, Université de Paris Cité, Sorbonne Université, INSERM U1138, France (C.L.-O.)
| | - Vicente Andrés
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Spain (M.R.H., R.M.N., P.G., M.J.A.-M., A.D., V.A.)
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (R.M.N., P.G., M.J.A.-M., P.N., A.R., C.T., F.S.-C., A.D., J.F.B., V.A.)
| |
Collapse
|
3
|
Niro F, Fernandes S, Cassani M, Apostolico M, Oliver-De La Cruz J, Pereira-Sousa D, Pagliari S, Vinarsky V, Zdráhal Z, Potesil D, Pustka V, Pompilio G, Sommariva E, Rovina D, Maione AS, Bersanini L, Becker M, Rasponi M, Forte G. Fibrotic extracellular matrix impacts cardiomyocyte phenotype and function in an iPSC-derived isogenic model of cardiac fibrosis. Transl Res 2024; 273:58-77. [PMID: 39025226 PMCID: PMC11832458 DOI: 10.1016/j.trsl.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 06/14/2024] [Accepted: 07/14/2024] [Indexed: 07/20/2024]
Abstract
Cardiac fibrosis occurs following insults to the myocardium and is characterized by the abnormal accumulation of non-compliant extracellular matrix (ECM), which compromises cardiomyocyte contractile activity and eventually leads to heart failure. This phenomenon is driven by the activation of cardiac fibroblasts (cFbs) to myofibroblasts and results in changes in ECM biochemical, structural and mechanical properties. The lack of predictive in vitro models of heart fibrosis has so far hampered the search for innovative treatments, as most of the cellular-based in vitro reductionist models do not take into account the leading role of ECM cues in driving the progression of the pathology. Here, we devised a single-step decellularization protocol to obtain and thoroughly characterize the biochemical and micro-mechanical properties of the ECM secreted by activated cFbs differentiated from human induced pluripotent stem cells (iPSCs). We activated iPSC-derived cFbs to the myofibroblast phenotype by tuning basic fibroblast growth factor (bFGF) and transforming growth factor beta 1 (TGF-β1) signalling and confirmed that activated cells acquired key features of myofibroblast phenotype, like SMAD2/3 nuclear shuttling, the formation of aligned alpha-smooth muscle actin (α-SMA)-rich stress fibres and increased focal adhesions (FAs) assembly. Next, we used Mass Spectrometry, nanoindentation, scanning electron and confocal microscopy to unveil the characteristic composition and the visco-elastic properties of the abundant, collagen-rich ECM deposited by cardiac myofibroblasts in vitro. Finally, we demonstrated that the fibrotic ECM activates mechanosensitive pathways in iPSC-derived cardiomyocytes, impacting on their shape, sarcomere assembly, phenotype, and calcium handling properties. We thus propose human bio-inspired decellularized matrices as animal-free, isogenic cardiomyocyte culture substrates recapitulating key pathophysiological changes occurring at the cellular level during cardiac fibrosis.
Collapse
Affiliation(s)
- Francesco Niro
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno; Masaryk University, Faculty of Medicine, Department of Biomedical Sciences, Brno 62500, Czech Republic; School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, UK
| | - Soraia Fernandes
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno
| | - Marco Cassani
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno
| | - Monica Apostolico
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno
| | - Jorge Oliver-De La Cruz
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno; Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Daniel Pereira-Sousa
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno; Masaryk University, Faculty of Medicine, Department of Biomedical Sciences, Brno 62500, Czech Republic
| | - Stefania Pagliari
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno; School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, UK
| | - Vladimir Vinarsky
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno
| | - Zbyněk Zdráhal
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
| | - David Potesil
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
| | - Vaclav Pustka
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
| | - Giulio Pompilio
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Milan, Italy; Dipartimento di Scienze Biomediche, Chirurgiche ed Odontoiatriche, Università degli Studi di Milano, Milan, Italy
| | - Elena Sommariva
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Davide Rovina
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Angela Serena Maione
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | | | | | - Marco Rasponi
- Department of Electronics, Informatics and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Giancarlo Forte
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno; School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, UK.
| |
Collapse
|
4
|
Liu H, Fan P, Jin F, Ren H, Xu F, Li J. Targeting biophysical microenvironment for improved treatment of chronic obstructive pulmonary disease. Trends Mol Med 2023; 29:926-938. [PMID: 37704492 DOI: 10.1016/j.molmed.2023.08.007] [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: 06/01/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 09/15/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is responsible for high disability rates, high death rates, and significant cost to health systems. Growing evidence in recent decades shows significant biophysical microenvironment changes in COPD, impacting lung tissues, cells, and treatment response. Furthermore, such biophysical changes have shown great potential as novel targets for improved therapeutic strategy of COPD, where both pharmacological and non-pharmacological therapies focusing on repairing the biophysical microenvironment of the lung have emerged. We present the first comprehensive review of four distinct biophysical hallmarks [i.e., extracellular matrix (ECM) microarchitecture, stiffness, fluid shear stress, and mechanical stretch] in COPD, the possible involvement of pathological changes, possible effects, and correlated in vitro models and sum up the emerging COPD treatments targeting these biophysical hallmarks.
Collapse
Affiliation(s)
- Han Liu
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan Province and Education Ministry of China, Zhengzhou, Henan 450046, China
| | - Pengbei Fan
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan Province and Education Ministry of China, Zhengzhou, Henan 450046, China
| | - Fanli Jin
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan Province and Education Ministry of China, Zhengzhou, Henan 450046, China
| | - Hui Ren
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, China; Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, China; MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| | - Jiansheng Li
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan Province and Education Ministry of China, Zhengzhou, Henan 450046, China.
| |
Collapse
|
5
|
Tamargo IA, Baek KI, Kim Y, Park C, Jo H. Flow-induced reprogramming of endothelial cells in atherosclerosis. Nat Rev Cardiol 2023; 20:738-753. [PMID: 37225873 PMCID: PMC10206587 DOI: 10.1038/s41569-023-00883-1] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/25/2023] [Indexed: 05/26/2023]
Abstract
Atherosclerotic diseases such as myocardial infarction, ischaemic stroke and peripheral artery disease continue to be leading causes of death worldwide despite the success of treatments with cholesterol-lowering drugs and drug-eluting stents, raising the need to identify additional therapeutic targets. Interestingly, atherosclerosis preferentially develops in curved and branching arterial regions, where endothelial cells are exposed to disturbed blood flow with characteristic low-magnitude oscillatory shear stress. By contrast, straight arterial regions exposed to stable flow, which is associated with high-magnitude, unidirectional shear stress, are relatively well protected from the disease through shear-dependent, atheroprotective endothelial cell responses. Flow potently regulates structural, functional, transcriptomic, epigenomic and metabolic changes in endothelial cells through mechanosensors and mechanosignal transduction pathways. A study using single-cell RNA sequencing and chromatin accessibility analysis in a mouse model of flow-induced atherosclerosis demonstrated that disturbed flow reprogrammes arterial endothelial cells in situ from healthy phenotypes to diseased ones characterized by endothelial inflammation, endothelial-to-mesenchymal transition, endothelial-to-immune cell-like transition and metabolic changes. In this Review, we discuss this emerging concept of disturbed-flow-induced reprogramming of endothelial cells (FIRE) as a potential pro-atherogenic mechanism. Defining the flow-induced mechanisms through which endothelial cells are reprogrammed to promote atherosclerosis is a crucial area of research that could lead to the identification of novel therapeutic targets to combat the high prevalence of atherosclerotic disease.
Collapse
Affiliation(s)
- Ian A Tamargo
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
- Molecular and Systems Pharmacology Program, Emory University, Atlanta, GA, USA
| | - Kyung In Baek
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Yerin Kim
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Christian Park
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA.
- Molecular and Systems Pharmacology Program, Emory University, Atlanta, GA, USA.
- Department of Medicine, Emory University School, Atlanta, GA, USA.
| |
Collapse
|
6
|
Oronsky B, Larson C, Hammond TC, Oronsky A, Kesari S, Lybeck M, Reid TR. A Review of Persistent Post-COVID Syndrome (PPCS). Clin Rev Allergy Immunol 2023; 64:66-74. [PMID: 33609255 PMCID: PMC7896544 DOI: 10.1007/s12016-021-08848-3] [Citation(s) in RCA: 190] [Impact Index Per Article: 95.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2021] [Indexed: 02/07/2023]
Abstract
Persistent post-COVID syndrome, also referred to as long COVID, is a pathologic entity, which involves persistent physical, medical, and cognitive sequelae following COVID-19, including persistent immunosuppression as well as pulmonary, cardiac, and vascular fibrosis. Pathologic fibrosis of organs and vasculature leads to increased mortality and severely worsened quality of life. Inhibiting transforming growth factor beta (TGF-β), an immuno- and a fibrosis modulator, may attenuate these post-COVID sequelae. Current preclinical and clinical efforts are centered on the mechanisms and manifestations of COVID-19 and its presymptomatic and prodromal periods; by comparison, the postdrome, which occurs in the aftermath of COVID-19, which we refer to as persistent post-COVID-syndrome, has received little attention. Potential long-term effects from post-COVID syndrome will assume increasing importance as a surge of treated patients are discharged from the hospital, placing a burden on healthcare systems, patients' families, and society in general to care for these medically devastated COVID-19 survivors. This review explores underlying mechanisms and possible manifestations of persistent post-COVID syndrome, and presents a framework of strategies for the diagnosis and management of patients with suspected or confirmed persistent post-COVID syndrome.
Collapse
Affiliation(s)
- Bryan Oronsky
- EpicentRx Inc, La Jolla, 11099 North Torrey Pines Road, Suite 160, La Jolla, CA 92037 USA
| | - Christopher Larson
- EpicentRx Inc, La Jolla, 11099 North Torrey Pines Road, Suite 160, La Jolla, CA 92037 USA
| | | | | | - Santosh Kesari
- Providence St. John’s Health Center, Santa Monica, CA USA
| | - Michelle Lybeck
- EpicentRx Inc, La Jolla, 11099 North Torrey Pines Road, Suite 160, La Jolla, CA 92037 USA
| | - Tony R. Reid
- EpicentRx Inc, La Jolla, 11099 North Torrey Pines Road, Suite 160, La Jolla, CA 92037 USA
| |
Collapse
|
7
|
Liu H, Fan P, Jin F, Huang G, Guo X, Xu F. Dynamic and static biomechanical traits of cardiac fibrosis. Front Bioeng Biotechnol 2022; 10:1042030. [PMID: 36394025 PMCID: PMC9659743 DOI: 10.3389/fbioe.2022.1042030] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/20/2022] [Indexed: 11/29/2022] Open
Abstract
Cardiac fibrosis is a common pathology in cardiovascular diseases which are reported as the leading cause of death globally. In recent decades, accumulating evidence has shown that the biomechanical traits of fibrosis play important roles in cardiac fibrosis initiation, progression and treatment. In this review, we summarize the four main distinct biomechanical traits (i.e., stretch, fluid shear stress, ECM microarchitecture, and ECM stiffness) and categorize them into two different types (i.e., static and dynamic), mainly consulting the unique characteristic of the heart. Moreover, we also provide a comprehensive overview of the effect of different biomechanical traits on cardiac fibrosis, their transduction mechanisms, and in-vitro engineered models targeting biomechanical traits that will aid the identification and prediction of mechano-based therapeutic targets to ameliorate cardiac fibrosis.
Collapse
Affiliation(s)
- Han Liu
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of China, Zhengzhou, China
| | - Pengbei Fan
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of China, Zhengzhou, China
| | - Fanli Jin
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of China, Zhengzhou, China
| | - Guoyou Huang
- Department of Engineering Mechanics, School of Civil Engineering, Wuhan University, Wuhan, China
| | - Xiaogang Guo
- The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
8
|
Suwittayarak R, Klincumhom N, Ngaokrajang U, Namangkalakul W, Ferreira JN, Pavasant P, Osathanon T. Shear Stress Enhances the Paracrine-Mediated Immunoregulatory Function of Human Periodontal Ligament Stem Cells via the ERK Signalling Pathway. Int J Mol Sci 2022; 23:ijms23137119. [PMID: 35806124 PMCID: PMC9266779 DOI: 10.3390/ijms23137119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/16/2022] [Accepted: 06/23/2022] [Indexed: 11/25/2022] Open
Abstract
Relevant immunomodulatory effects have been proposed following allogeneic cell-based therapy with human periodontal ligament stem cells (hPDLSCs). This study aimed to examine the influence of shear stress on the immunosuppressive capacity of hPDLSCs. Cells were subjected to shear stress at different magnitudes (0.5, 5 and 10 dyn/cm2). The expression of immunosuppressive markers was evaluated in shear stress-induced hPDLSCs using qRT-PCR, western blot, enzyme activity and enzyme-linked immunosorbent assays. The effects of a shear stress-derived condition medium (SS-CM) on T cell proliferation were examined using a resazurin assay. Treg differentiation was investigated using qRT-PCR and flow cytometry analysis. Our results revealed that shear stress increased mRNA expression of IDO and COX2 but not TGF-β1 and IFN-γ. IDO activity, kynurenine and active TGF-β1 increased in SS-CM when compared to the non-shear stress-derived conditioned medium (CTL-CM). The amount of kynurenine in SS-CM was reduced in the presence of cycloheximide and ERK inhibitor. Subsequently, T cell proliferation decreased in SS-CM compared to CTL-CM. Treg differentiation was promoted in SS-CM, indicated by FOXP3, IL-10 expression and CD4+CD25hiCD127lo/− subpopulation. In conclusion, shear stress promotes kynurenine production through ERK signalling in hPDLSC, leading to the inhibition of T cell proliferation and the promotion of Treg cell differentiation.
Collapse
Affiliation(s)
- Ravipha Suwittayarak
- Center of Excellence for Regenerative Dentistry, Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand; (R.S.); (U.N.); (P.P.)
| | - Nuttha Klincumhom
- Center of Excellence for Regenerative Dentistry, Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand; (R.S.); (U.N.); (P.P.)
- Correspondence:
| | - Utapin Ngaokrajang
- Center of Excellence for Regenerative Dentistry, Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand; (R.S.); (U.N.); (P.P.)
| | - Worachat Namangkalakul
- Dental Stem Cell Biology Research Unit, Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand; (W.N.); (T.O.)
| | - João N. Ferreira
- Avatar Biotechnologies for Oral Health and Healthy Longevity Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Prasit Pavasant
- Center of Excellence for Regenerative Dentistry, Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand; (R.S.); (U.N.); (P.P.)
| | - Thanaphum Osathanon
- Dental Stem Cell Biology Research Unit, Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand; (W.N.); (T.O.)
| |
Collapse
|
9
|
Meng F, Cheng H, Qian J, Dai X, Huang Y, Fan Y. In vitro fluidic systems: Applying shear stress on endothelial cells. MEDICINE IN NOVEL TECHNOLOGY AND DEVICES 2022. [DOI: 10.1016/j.medntd.2022.100143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
10
|
Fallon ME, Mathews R, Hinds MT. In Vitro Flow Chamber Design for the Study of Endothelial Cell (Patho)Physiology. J Biomech Eng 2022; 144:020801. [PMID: 34254640 PMCID: PMC8628846 DOI: 10.1115/1.4051765] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 07/06/2021] [Indexed: 02/03/2023]
Abstract
In the native vasculature, flowing blood produces a frictional force on vessel walls that affects endothelial cell function and phenotype. In the arterial system, the vasculature's local geometry directly influences variations in flow profiles and shear stress magnitudes. Straight arterial sections with pulsatile shear stress have been shown to promote an athero-protective endothelial phenotype. Conversely, areas with more complex geometry, such as arterial bifurcations and branch points with disturbed flow patterns and lower, oscillatory shear stress, typically lead to endothelial dysfunction and the pathogenesis of cardiovascular diseases. Many studies have investigated the regulation of endothelial responses to various shear stress environments. Importantly, the accurate in vitro simulation of in vivo hemodynamics is critical to the deeper understanding of mechanotransduction through the proper design and use of flow chamber devices. In this review, we describe several flow chamber apparatuses and their fluid mechanics design parameters, including parallel-plate flow chambers, cone-and-plate devices, and microfluidic devices. In addition, chamber-specific design criteria and relevant equations are defined in detail for the accurate simulation of shear stress environments to study endothelial cell responses.
Collapse
Affiliation(s)
- Meghan E. Fallon
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S Bond Ave CH13B, Portland, OR 97239
| | - Rick Mathews
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S Bond Ave CH13B, Portland, OR 97239
| | - Monica T. Hinds
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S Bond Ave CH13B, Portland, OR 97239
| |
Collapse
|
11
|
Nabeh OA, Matter LM, Khattab MA, Esraa Menshawey. "The possible implication of endothelin in the pathology of COVID-19-induced pulmonary hypertension". Pulm Pharmacol Ther 2021; 71:102082. [PMID: 34601121 PMCID: PMC8483983 DOI: 10.1016/j.pupt.2021.102082] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/23/2021] [Accepted: 09/23/2021] [Indexed: 12/21/2022]
Abstract
COVID-19 pandemic has changed the world dramatically since was first reported in Wuhan city, China [1]. Not only as a respiratory illness that could lead to fatal respiratory failure, but also some evidences suggest that it can propagate as a chronic disease associated with a variety of persistent post COVID-19 pathologies that affect patients' life [2,3]. Pulmonary hypertension (PH) is one of the challenging diseases that may develop as a consequence of SARS-COV-2 infection in some COVID-19 survivors [4,5]. The vasopressor, proliferative, proinflammatory, and prothrombotic actions of endothelin [6] may be encountered in the COVID-19-induced PH pathology. And so, endothelin blockers may have an important role to restrict the development of serious PH outcomes with special precautions considering patients with significant hypoxemia.
Collapse
Affiliation(s)
- Omnia Azmy Nabeh
- M.Sc/ Assistant Lecturer, Department of Medical Pharmacology, Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt; M.Sc, Cardiovascular Medicine, Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt.
| | - Lamiaa Mohammed Matter
- MD/Lecturer, Department of Medical Pharmacology, Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt; Professional Diploma of Family Medicine, Arab Institute for Continuing Professional Development, Arab Medical Union, Egypt.
| | - Mahmoud Ahmed Khattab
- M.Sc/ Assistant Lecturer, Department of Medical Pharmacology, Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt; M.Sc Internal Medicine, Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt.
| | - Esraa Menshawey
- Medical Student, Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt.
| |
Collapse
|
12
|
Herault S, Naser J, Carassiti D, Chooi KY, Nikolopoulou R, Font ML, Patel M, Pedrigi R, Krams R. Mechanosensitive pathways are regulated by mechanosensitive miRNA clusters in endothelial cells. Biophys Rev 2021; 13:787-796. [PMID: 34777618 PMCID: PMC8555030 DOI: 10.1007/s12551-021-00839-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 08/27/2021] [Indexed: 12/15/2022] Open
Abstract
Shear stress is known to affect many processes in (patho-) physiology through a complex, multi-molecular mechanism, termed mechanotransduction. The sheer complexity of the process has raised questions how mechanotransduction is regulated. Here, we comprehensively evaluate the literature about the role of small non-coding miRNA in the regulation of mechanotransduction. Regulation of mRNA by miRNA is rather complex, depending not only on the concentration of mRNA to miRNA, but also on the amount of mRNA competing for a single mRNA. The only mechanism to counteract the latter factor is through overarching structures of miRNA. Indeed, two overarching structures are present miRNA families and miRNA clusters, and both will be discussed in details, regarding the latest literature and a previous conducted study focussed on mechanotransduction. Both the literature and our own data support a new hypothesis that miRNA-clusters predominantly regulate mechanotransduction, affecting 65% of signalling pathways. In conclusion, a new and important mode of regulation of mechanotransduction is proposed, based on miRNA clusters. This finding implicates new avenues for treatment of mechanotransduction and atherosclerosis.
Collapse
Affiliation(s)
- Sean Herault
- School of Engineering and Materials Science, Queen Mary University of London, Room 2.14, London, UK
| | | | - Daniele Carassiti
- School of Engineering and Materials Science, Queen Mary University of London, Room 2.14, London, UK
| | - K. Yean Chooi
- School of Engineering and Materials Science, Queen Mary University of London, Room 2.14, London, UK
| | | | - Marti Llopart Font
- School of Engineering and Materials Science, Queen Mary University of London, Room 2.14, London, UK
| | | | - Ryan Pedrigi
- College of Engineering, Mechanical & Materials Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Rob Krams
- School of Engineering and Materials Science, Queen Mary University of London, Room 2.14, London, UK
| |
Collapse
|
13
|
Dole NS, Yoon J, Monteiro DA, Yang J, Mazur CM, Kaya S, Belair CD, Alliston T. Mechanosensitive miR-100 coordinates TGFβ and Wnt signaling in osteocytes during fluid shear stress. FASEB J 2021; 35:e21883. [PMID: 34569659 PMCID: PMC9153140 DOI: 10.1096/fj.202100930] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 08/12/2021] [Indexed: 11/12/2022]
Abstract
Organism scale mechanical forces elicit cellular scale changes through coordinated regulation of multiple signaling pathways. The mechanisms by which cells integrate signaling to generate a unified biological response remains a major question in mechanobiology. For example, the mechanosensitive response of bone and other tissues requires coordinated signaling by the transforming growth factor beta (TGFβ) and Wnt pathways through mechanisms that are not well‐defined. Here we report a new microRNA‐dependent mechanism that mediates mechanosensitive crosstalk between TGFβ and Wnt signaling in osteocytes exposed to fluid shear stress (FSS). From 60 mechanosensitive microRNA (miRs) identified by small‐RNAseq, miR100 expression is suppressed by in vivo hindlimb loading in the murine tibia and by cellular scale FSS in OCY454 cells. Though FSS activates both TGFβ and Wnt signaling in osteocytes, only TGFβ represses miR‐100 expression. miR‐100, in turn, antagonizes Wnt signaling by targeting and inhibiting expression of Frizzled receptors (FZD5/FZD8). Accordingly, miR‐100 inhibition blunts FSS‐ and TGFβ‐inducible Wnt signaling. Therefore, our results identify FSS‐responsive miRNAs in osteocytes, including one that integrates the mechanosensitive function of two essential signaling pathways in the osteoanabolic response of bone to mechanical load.
Collapse
Affiliation(s)
- Neha S Dole
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Jihee Yoon
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California, USA
| | - David A Monteiro
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Jason Yang
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, California, USA
| | - Courtney M Mazur
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Serra Kaya
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Cassandra D Belair
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, USA.,Department of Urology, University of California, San Francisco, San Francisco, California, USA
| | - Tamara Alliston
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California, USA.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
14
|
Zhou K, Tian KJ, Yan BJ, Gui DD, Luo W, Ren Z, Wei DH, Liu LS, Jiang ZS. A promising field: regulating imbalance of EndMT in cardiovascular diseases. Cell Cycle 2021; 20:1477-1486. [PMID: 34266366 PMCID: PMC8354671 DOI: 10.1080/15384101.2021.1951939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 03/30/2021] [Accepted: 06/30/2021] [Indexed: 10/20/2022] Open
Abstract
Endothelial-mesenchymal transition (EndMT) is widely involved in the occurrence and development of cardiovascular diseases. Although there is no direct evidence, it is very promising as an effective target for the treatment of these diseases. Endothelial cells need to respond to the complex cardiovascular environment through EndMT, but sustained stimuli will cause the imbalance of EndMT. Blocking the signal transduction promoting EndMT is an effective method to control the imbalance of EndMT. In particular, we also discussed the potential role of endothelial cell apoptosis and autophagy in regulating the imbalance of EndMT. In addition, promoting mesenchymal-endothelial transformation (MEndT) is also a method to control the imbalance of EndMT. However, targeting EndMT to treat cardiovascular disease still faces many challenges. By reviewing the research progress of EndMT, we have put forward some insights and translated them into challenges and opportunities for new treatment strategies for cardiovascular diseases.
Collapse
Affiliation(s)
- Kun Zhou
- Key Laboratory for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, Hunan Province, China
| | - Kai-Jiang Tian
- Key Laboratory for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, Hunan Province, China
| | - Bin-Jie Yan
- Key Laboratory for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, Hunan Province, China
| | - Dan-Dan Gui
- Key Laboratory for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, Hunan Province, China
| | - Wen Luo
- Key Laboratory for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, Hunan Province, China
| | - Zhong Ren
- Key Laboratory for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, Hunan Province, China
| | - Dang-Heng Wei
- Key Laboratory for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, Hunan Province, China
| | - Lu-Shan Liu
- Key Laboratory for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, Hunan Province, China
| | - Zhi-Sheng Jiang
- Key Laboratory for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, Hunan Province, China
| |
Collapse
|
15
|
Zhao P, Yao Q, Zhang PJ, The E, Zhai Y, Ao L, Jarrett MJ, Dinarello CA, Fullerton DA, Meng X. Single-cell RNA-seq reveals a critical role of novel pro-inflammatory EndMT in mediating adverse remodeling in coronary artery-on-a-chip. SCIENCE ADVANCES 2021; 7:eabg1694. [PMID: 34417174 PMCID: PMC8378826 DOI: 10.1126/sciadv.abg1694] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 06/30/2021] [Indexed: 05/09/2023]
Abstract
A three-dimensional microengineered human coronary artery-on-a-chip was developed for investigation of the mechanism by which low and oscillatory shear stress (OSS) induces pro-atherogenic changes. Single-cell RNA sequencing revealed that OSS induced distinct changes in endothelial cells (ECs) including pro-inflammatory endothelial-to-mesenchymal transition (EndMT). OSS promoted pro-inflammatory EndMT through the Notch1/p38 MAPK-NF-κB signaling axis. Moreover, OSS-induced EC phenotypic changes resulted in proliferation and extracellular matrix (ECM) protein up-regulation in smooth muscle cells (SMCs) through the RANTES-mediated paracrine mechanism. IL-37 suppressed OSS-induced pro-inflammatory EndMT and thereby abrogated SMC proliferation and ECM protein remodeling. Overall, this study provides insights into endothelial heterogeneity under atheroprone shear stress and identifies the mechanistic role of a novel EC subtype in promoting adverse vascular remodeling. Further, this study demonstrates that anti-inflammatory approach is capable of mitigating vascular pathobiology evoked by atheroprone shear stress.
Collapse
Affiliation(s)
- Peng Zhao
- Department of Surgery, University of Colorado Denver, Aurora, CO, USA
| | - Qingzhou Yao
- Department of Surgery, University of Colorado Denver, Aurora, CO, USA
| | - Pei-Jian Zhang
- Department of Surgery, University of Colorado Denver, Aurora, CO, USA
| | - Erlinda The
- Department of Surgery, University of Colorado Denver, Aurora, CO, USA
| | - Yufeng Zhai
- Department of Surgery, University of Colorado Denver, Aurora, CO, USA
| | - Lihua Ao
- Department of Surgery, University of Colorado Denver, Aurora, CO, USA
| | - Michael J Jarrett
- Department of Surgery, University of Colorado Denver, Aurora, CO, USA
| | | | - David A Fullerton
- Department of Surgery, University of Colorado Denver, Aurora, CO, USA
| | - Xianzhong Meng
- Department of Surgery, University of Colorado Denver, Aurora, CO, USA.
| |
Collapse
|
16
|
Monteiro DA, Dole NS, Campos JL, Kaya S, Schurman CA, Belair CD, Alliston T. Fluid shear stress generates a unique signaling response by activating multiple TGFβ family type I receptors in osteocytes. FASEB J 2021; 35:e21263. [PMID: 33570811 PMCID: PMC7888383 DOI: 10.1096/fj.202001998r] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 11/11/2020] [Accepted: 11/25/2020] [Indexed: 12/18/2022]
Abstract
Bone is a dynamic tissue that constantly adapts to changing mechanical demands. The transforming growth factor beta (TGFβ) signaling pathway plays several important roles in maintaining skeletal homeostasis by both coupling the bone‐forming and bone‐resorbing activities of osteoblasts and osteoclasts and by playing a causal role in the anabolic response of bone to applied loads. However, the extent to which the TGFβ signaling pathway in osteocytes is directly regulated by fluid shear stress (FSS) is unknown, despite work suggesting that fluid flow along canaliculi is a dominant physical cue sensed by osteocytes following bone compression. To investigate the effects of FSS on TGFβ signaling in osteocytes, we stimulated osteocytic OCY454 cells cultured within a microfluidic platform with FSS. We find that FSS rapidly upregulates Smad2/3 phosphorylation and TGFβ target gene expression, even in the absence of added TGFβ. Indeed, relative to treatment with TGFβ, FSS induced a larger increase in levels of pSmad2/3 and Serpine1 that persisted even in the presence of a TGFβ receptor type I inhibitor. Our results show that FSS stimulation rapidly induces phosphorylation of multiple TGFβ family R‐Smads by stimulating multimerization and concurrently activating several TGFβ and BMP type I receptors, in a manner that requires the activity of the corresponding ligand. While the individual roles of the TGFβ and BMP signaling pathways in bone mechanotransduction remain unclear, these results implicate that FSS activates both pathways to generate a downstream response that differs from that achieved by either ligand alone.
Collapse
Affiliation(s)
- David A Monteiro
- Department of Orthopaedic Surgery, University of California, San Francisco, CA, USA.,UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA, USA
| | - Neha S Dole
- Department of Orthopaedic Surgery, University of California, San Francisco, CA, USA
| | - J Luke Campos
- Department of Orthopaedic Surgery, University of California, San Francisco, CA, USA
| | - Serra Kaya
- Department of Orthopaedic Surgery, University of California, San Francisco, CA, USA
| | - Charles A Schurman
- Department of Orthopaedic Surgery, University of California, San Francisco, CA, USA.,UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA, USA
| | - Cassandra D Belair
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA.,Department of Urology, University of California, San Francisco, CA, USA
| | - Tamara Alliston
- Department of Orthopaedic Surgery, University of California, San Francisco, CA, USA.,UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA, USA.,The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA
| |
Collapse
|
17
|
Higgins CE, Tang J, Higgins SP, Gifford CC, Mian BM, Jones DM, Zhang W, Costello A, Conti DJ, Samarakoon R, Higgins PJ. The Genomic Response to TGF-β1 Dictates Failed Repair and Progression of Fibrotic Disease in the Obstructed Kidney. Front Cell Dev Biol 2021; 9:678524. [PMID: 34277620 PMCID: PMC8284093 DOI: 10.3389/fcell.2021.678524] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/07/2021] [Indexed: 12/14/2022] Open
Abstract
Tubulointerstitial fibrosis is a common and diagnostic hallmark of a spectrum of chronic renal disorders. While the etiology varies as to the causative nature of the underlying pathology, persistent TGF-β1 signaling drives the relentless progression of renal fibrotic disease. TGF-β1 orchestrates the multifaceted program of kidney fibrogenesis involving proximal tubular dysfunction, failed epithelial recovery or re-differentiation, capillary collapse and subsequent interstitial fibrosis eventually leading to chronic and ultimately end-stage disease. An increasing complement of non-canonical elements function as co-factors in TGF-β1 signaling. p53 is a particularly prominent transcriptional co-regulator of several TGF-β1 fibrotic-response genes by complexing with TGF-β1 receptor-activated SMADs. This cooperative p53/TGF-β1 genomic cluster includes genes involved in cellular proliferative control, survival, apoptosis, senescence, and ECM remodeling. While the molecular basis for this co-dependency remains to be determined, a subset of TGF-β1-regulated genes possess both p53- and SMAD-binding motifs. Increases in p53 expression and phosphorylation, moreover, are evident in various forms of renal injury as well as kidney allograft rejection. Targeted reduction of p53 levels by pharmacologic and genetic approaches attenuates expression of the involved genes and mitigates the fibrotic response confirming a key role for p53 in renal disorders. This review focuses on mechanisms underlying TGF-β1-induced renal fibrosis largely in the context of ureteral obstruction, which mimics the pathophysiology of pediatric unilateral ureteropelvic junction obstruction, and the role of p53 as a transcriptional regulator within the TGF-β1 repertoire of fibrosis-promoting genes.
Collapse
Affiliation(s)
- Craig E. Higgins
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Jiaqi Tang
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Stephen P. Higgins
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Cody C. Gifford
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Badar M. Mian
- The Urological Institute of Northeastern New York, Albany, NY, United States
- Division of Urology, Department of Surgery, Albany Medical College, Albany, NY, United States
| | - David M. Jones
- Department of Pathology and Laboratory Medicine, Albany Medical College, Albany, NY, United States
| | - Wenzheng Zhang
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Angelica Costello
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - David J. Conti
- Division of Transplantation Surgery, Department of Surgery, Albany Medical College, Albany, NY, United States
| | - Rohan Samarakoon
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Paul J. Higgins
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
- The Urological Institute of Northeastern New York, Albany, NY, United States
- Division of Urology, Department of Surgery, Albany Medical College, Albany, NY, United States
| |
Collapse
|
18
|
Urschel K, Tauchi M, Achenbach S, Dietel B. Investigation of Wall Shear Stress in Cardiovascular Research and in Clinical Practice-From Bench to Bedside. Int J Mol Sci 2021; 22:5635. [PMID: 34073212 PMCID: PMC8198948 DOI: 10.3390/ijms22115635] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 05/20/2021] [Accepted: 05/22/2021] [Indexed: 12/16/2022] Open
Abstract
In the 1900s, researchers established animal models experimentally to induce atherosclerosis by feeding them with a cholesterol-rich diet. It is now accepted that high circulating cholesterol is one of the main causes of atherosclerosis; however, plaque localization cannot be explained solely by hyperlipidemia. A tremendous amount of studies has demonstrated that hemodynamic forces modify endothelial athero-susceptibility phenotypes. Endothelial cells possess mechanosensors on the apical surface to detect a blood stream-induced force on the vessel wall, known as "wall shear stress (WSS)", and induce cellular and molecular responses. Investigations to elucidate the mechanisms of this process are on-going: on the one hand, hemodynamics in complex vessel systems have been described in detail, owing to the recent progress in imaging and computational techniques. On the other hand, investigations using unique in vitro chamber systems with various flow applications have enhanced the understanding of WSS-induced changes in endothelial cell function and the involvement of the glycocalyx, the apical surface layer of endothelial cells, in this process. In the clinical setting, attempts have been made to measure WSS and/or glycocalyx degradation non-invasively, for the purpose of their diagnostic utilization. An increasing body of evidence shows that WSS, as well as serum glycocalyx components, can serve as a predicting factor for atherosclerosis development and, most importantly, for the rupture of plaques in patients with high risk of coronary heart disease.
Collapse
Affiliation(s)
| | | | | | - Barbara Dietel
- Department of Medicine 2—Cardiology and Angiology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Universitätsklinikum, 91054 Erlangen, Germany; (K.U.); (M.T.); (S.A.)
| |
Collapse
|
19
|
Yamada S, Ishikawa M, Nozaki K. Exploring mechanisms of ventricular enlargement in idiopathic normal pressure hydrocephalus: a role of cerebrospinal fluid dynamics and motile cilia. Fluids Barriers CNS 2021; 18:20. [PMID: 33874972 PMCID: PMC8056523 DOI: 10.1186/s12987-021-00243-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 02/13/2021] [Indexed: 11/15/2022] Open
Abstract
Idiopathic normal pressure hydrocephalus (iNPH) is considered an age-dependent chronic communicating hydrocephalus associated with cerebrospinal fluid (CSF) malabsorption; however, the aetiology of ventricular enlargement in iNPH has not yet been elucidated. There is accumulating evidence that support the hypothesis that various alterations in CSF dynamics contribute to ventricle dilatation in iNPH. This review focuses on CSF dynamics associated with ventriculomegaly and summarises the current literature based on three potential aetiology factors: genetic, environmental and hydrodynamic. The majority of gene mutations that cause communicating hydrocephalus were associated with an abnormal structure or dysfunction of motile cilia on the ventricular ependymal cells. Aging, alcohol consumption, sleep apnoea, diabetes and hypertension are candidates for the risk of developing iNPH, although there is no prospective cohort study to investigate the risk factors for iNPH. Alcohol intake may be associated with the dysfunction of ependymal cilia and sustained high CSF sugar concentration due to uncontrolled diabetes increases the fluid viscosity which in turn increases the shear stress on the ventricular wall surface. Sleep apnoea, diabetes and hypertension are known to be associated with the impairment of CSF and interstitial fluid exchange. Oscillatory shear stress to the ventricle wall surfaces is considerably increased by reciprocating bidirectional CSF movements in iNPH. Increased oscillatory shear stress impedes normal cilia beating, leading to motile cilia shedding from the ependymal cells. At the lack of ciliary protection, the ventricular wall is directly exposed to increased oscillatory shear stress. Additionally, increased oscillatory shear stress may be involved in activating the flow-mediated dilation signalling of the ventricular wall. In conclusion, as the CSF stroke volume at the cerebral aqueduct increases, the oscillatory shear stress increases, promoting motor cilia shedding and loss of ependymal cell coverage. These are considered to be the leading causes of ventricular enlargement in iNPH.
Collapse
Affiliation(s)
- Shigeki Yamada
- Department of Neurosurgery, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Shiga, 520-2192, Japan. .,Department of Neurosurgery and Normal Pressure Hydrocephalus Center, Rakuwakai Otowa Hospital, Kyoto, Japan. .,Interfaculty Initiative in Information Studies, Institute of Industrial Science, The University of Tokyo, Tokyo, Japan.
| | - Masatsune Ishikawa
- Department of Neurosurgery and Normal Pressure Hydrocephalus Center, Rakuwakai Otowa Hospital, Kyoto, Japan.,Rakuwa Villa Ilios, Kyoto, Japan
| | - Kazuhiko Nozaki
- Department of Neurosurgery, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Shiga, 520-2192, Japan
| |
Collapse
|
20
|
Brunner G, Roux M, Böhm V, Meiners T. Cellular and molecular changes that predispose skin in chronic spinal cord injury to pressure ulcer formation. Int Wound J 2021; 18:728-737. [PMID: 33723924 PMCID: PMC8450792 DOI: 10.1111/iwj.13575] [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: 11/21/2020] [Revised: 02/09/2021] [Accepted: 02/15/2021] [Indexed: 12/04/2022] Open
Abstract
Patients with spinal cord injury have a predisposition to develop pressure ulcers. Specific characteristics of the patients' skin potentially involved have not yet been identified. The purpose of this investigation was to determine whether loss of neuronal control affects cellular and molecular homeostasis in the skin. Intact afflicted skin, wound edge of pressure ulcers, and control skin were analysed. Platelets, transforming growth factor‐β1, and activin A were identified by immunohistochemistry. Transforming growth factor‐β‐like activity was determined by bioassay, and gene expression by DNA microarray analysis or RT‐PCR. In afflicted skin, enhanced platelet extravasation was detected. Transforming growth factor‐β1 and activin A accumulated in the dermal‐epidermal junction zone. Transforming growth factor‐β‐like activity and activin A expression were increased in intact afflicted skin (compared to control skin) and were further enhanced in pressure ulcers. In vitro, activity was generated by fibroblast‐epithelial cell interactions, which also induced activin A. Thus, loss of neuronal control in spinal cord injury appears to trigger inappropriate wound healing processes in the patients' skin. Plasma leakage and increased transforming growth factor‐β‐like activity combined with shear forces potentially enhance the risk for pressure ulcer formation.
Collapse
Affiliation(s)
- Georg Brunner
- Center for Spinal Cord Injuries, Werner Wicker Hospital, Bad Wildungen, Germany.,Department of Cancer Research, Skin Cancer Center Hornheide, Münster, Germany
| | - Meike Roux
- Department of Cancer Research, Skin Cancer Center Hornheide, Münster, Germany
| | - Volker Böhm
- Center for Spinal Cord Injuries, Werner Wicker Hospital, Bad Wildungen, Germany
| | - Thomas Meiners
- Center for Spinal Cord Injuries, Werner Wicker Hospital, Bad Wildungen, Germany
| |
Collapse
|
21
|
Yamada S, Ito H, Ishikawa M, Yamamoto K, Yamaguchi M, Oshima M, Nozaki K. Quantification of Oscillatory Shear Stress from Reciprocating CSF Motion on 4D Flow Imaging. AJNR Am J Neuroradiol 2021; 42:479-486. [PMID: 33478942 DOI: 10.3174/ajnr.a6941] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 10/05/2020] [Indexed: 01/18/2023]
Abstract
BACKGROUND AND PURPOSE Oscillatory shear stress could not be directly measured in consideration of direction, although cerebrospinal fluid has repetitive movements synchronized with heartbeat. Our aim was to evaluate the important of oscillatory shear stress in the cerebral aqueduct and foramen magnum in idiopathic normal pressure hydrocephalus by comparing it with wall shear stress and the oscillatory shear index in patients with idiopathic normal pressure hydrocephalus. MATERIALS AND METHODS By means of the 4D flow application, oscillatory shear stress, wall shear stress, and the oscillatory shear index were measured in 41 patients with idiopathic normal pressure hydrocephalus, 23 with co-occurrence of idiopathic normal pressure hydrocephalus and Alzheimer-type dementia, and 9 age-matched controls. These shear stress parameters at the cerebral aqueduct were compared with apertures and stroke volumes at the foramen of Magendie and cerebral aqueduct. RESULTS Two wall shear stress magnitude peaks during a heartbeat were changed to periodic oscillation by converting oscillatory shear stress. The mean oscillatory shear stress amplitude and time-averaged wall shear stress values at the dorsal and ventral regions of the cerebral aqueduct in the idiopathic normal pressure hydrocephalus groups were significantly higher than those in controls. Furthermore, those at the ventral region of the cerebral aqueduct in the idiopathic normal pressure hydrocephalus group were also significantly higher than those in the co-occurrence of idiopathic normal pressure hydrocephalus with Alzheimer-type dementia group. The oscillatory shear stress amplitude at the dorsal region of the cerebral aqueduct was significantly associated with foramen of Magendie diameters, whereas it was strongly associated with the stroke volume at the upper end of the cerebral aqueduct rather than that at the foramen of Magendie. CONCLUSIONS Oscillatory shear stress, which reflects wall shear stress vector changes better than the conventional wall shear stress magnitude and the oscillatory shear index, can be directly measured on 4D flow MR imaging. Oscillatory shear stress at the cerebral aqueduct was considerably higher in patients with idiopathic normal pressure hydrocephalus.
Collapse
Affiliation(s)
- S Yamada
- From the Department of Neurosurgery (S.Y., K.N.), Shiga University of Medical Science, Shiga, Japan .,Department of Neurosurgery and Normal Pressure Hydrocephalus Center (S.Y., K.Y., M.Y.), Rakuwakai Otowa Hospital, Kyoto, Japan.,Interfaculty Initiative in Information Studies/Institute of Industrial Science (S.Y., M.O.), The University of Tokyo, Tokyo, Japan
| | - H Ito
- Medical System Research and Development Center (H.I.), Fujifilm Corporation, Tokyo, Japan
| | | | - K Yamamoto
- Department of Neurosurgery and Normal Pressure Hydrocephalus Center (S.Y., K.Y., M.Y.), Rakuwakai Otowa Hospital, Kyoto, Japan
| | - M Yamaguchi
- Department of Neurosurgery and Normal Pressure Hydrocephalus Center (S.Y., K.Y., M.Y.), Rakuwakai Otowa Hospital, Kyoto, Japan
| | - M Oshima
- Interfaculty Initiative in Information Studies/Institute of Industrial Science (S.Y., M.O.), The University of Tokyo, Tokyo, Japan
| | - K Nozaki
- From the Department of Neurosurgery (S.Y., K.N.), Shiga University of Medical Science, Shiga, Japan
| |
Collapse
|
22
|
Sangha GS, Goergen CJ, Prior SJ, Ranadive SM, Clyne AM. Preclinical techniques to investigate exercise training in vascular pathophysiology. Am J Physiol Heart Circ Physiol 2021; 320:H1566-H1600. [PMID: 33385323 PMCID: PMC8260379 DOI: 10.1152/ajpheart.00719.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Atherosclerosis is a dynamic process starting with endothelial dysfunction and inflammation and eventually leading to life-threatening arterial plaques. Exercise generally improves endothelial function in a dose-dependent manner by altering hemodynamics, specifically by increased arterial pressure, pulsatility, and shear stress. However, athletes who regularly participate in high-intensity training can develop arterial plaques, suggesting alternative mechanisms through which excessive exercise promotes vascular disease. Understanding the mechanisms that drive atherosclerosis in sedentary versus exercise states may lead to novel rehabilitative methods aimed at improving exercise compliance and physical activity. Preclinical tools, including in vitro cell assays, in vivo animal models, and in silico computational methods, broaden our capabilities to study the mechanisms through which exercise impacts atherogenesis, from molecular maladaptation to vascular remodeling. Here, we describe how preclinical research tools have and can be used to study exercise effects on atherosclerosis. We then propose how advanced bioengineering techniques can be used to address gaps in our current understanding of vascular pathophysiology, including integrating in vitro, in vivo, and in silico studies across multiple tissue systems and size scales. Improving our understanding of the antiatherogenic exercise effects will enable engaging, targeted, and individualized exercise recommendations to promote cardiovascular health rather than treating cardiovascular disease that results from a sedentary lifestyle.
Collapse
Affiliation(s)
- Gurneet S Sangha
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| | - Craig J Goergen
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana.,Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana
| | - Steven J Prior
- Department of Kinesiology, University of Maryland School of Public Health, College Park, Maryland.,Baltimore Veterans Affairs Geriatric Research, Education, and Clinical Center, Baltimore, Maryland
| | - Sushant M Ranadive
- Department of Kinesiology, University of Maryland School of Public Health, College Park, Maryland
| | - Alisa M Clyne
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| |
Collapse
|
23
|
Hiepen C, Mendez PL, Knaus P. It Takes Two to Tango: Endothelial TGFβ/BMP Signaling Crosstalk with Mechanobiology. Cells 2020; 9:E1965. [PMID: 32858894 PMCID: PMC7564048 DOI: 10.3390/cells9091965] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/19/2020] [Accepted: 08/22/2020] [Indexed: 02/06/2023] Open
Abstract
Bone morphogenetic proteins (BMPs) are members of the transforming growth factor-beta (TGFβ) superfamily of cytokines. While some ligand members are potent inducers of angiogenesis, others promote vascular homeostasis. However, the precise understanding of the molecular mechanisms underlying these functions is still a growing research field. In bone, the tissue in which BMPs were first discovered, crosstalk of TGFβ/BMP signaling with mechanobiology is well understood. Likewise, the endothelium represents a tissue that is constantly exposed to multiple mechanical triggers, such as wall shear stress, elicited by blood flow or strain, and tension from the surrounding cells and to the extracellular matrix. To integrate mechanical stimuli, the cytoskeleton plays a pivotal role in the transduction of these forces in endothelial cells. Importantly, mechanical forces integrate on several levels of the TGFβ/BMP pathway, such as receptors and SMADs, but also global cell-architecture and nuclear chromatin re-organization. Here, we summarize the current literature on crosstalk mechanisms between biochemical cues elicited by TGFβ/BMP growth factors and mechanical cues, as shear stress or matrix stiffness that collectively orchestrate endothelial function. We focus on the different subcellular compartments in which the forces are sensed and integrated into the TGFβ/BMP growth factor signaling.
Collapse
Affiliation(s)
| | | | - Petra Knaus
- Knaus-Lab/Signal Transduction, Institute for Chemistry and Biochemistry, Freie Universitaet Berlin, 14195 Berlin, Germany; (C.H.); (P.-L.M.)
| |
Collapse
|
24
|
da Silva Madaleno C, Jatzlau J, Knaus P. BMP signalling in a mechanical context - Implications for bone biology. Bone 2020; 137:115416. [PMID: 32422297 DOI: 10.1016/j.bone.2020.115416] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 01/12/2023]
Abstract
Bone Morphogenetic Proteins (BMPs) are extracellular multifunctional signalling cytokines and members of the TGFβ super family. These pleiotropic growth factors crucially promote bone formation, remodeling and healing after injury. Additionally, bone homeostasis is systematically regulated by mechanical inputs from the environment, which are incorporated into the bone cells' biochemical response. These inputs range from compression and tension induced by the movement of neighboring muscle, to fluid shear stress induced by interstitial fluid flow in the canaliculi and in the vascular system. Although BMPs are widely applied in a clinic context to promote fracture healing, it is still elusive how mechanical inputs modulate this signalling pathway, hindering an efficient and side-effect free application of these ligands in bone healing. This review aims to summarize the current understanding in how mechanical cues (tension, compression, shear force and hydrostatic pressure) and substrate stiffness modulate BMP signalling. We highlight the time-dependent effects in modulating immediate early up to long-term effects of mechano-BMP crosstalk during bone formation and remodeling, considering the interplay with other already established mechanosensitive pathways, such as MRTF/SRF and Hippo signalling.
Collapse
Affiliation(s)
- Carolina da Silva Madaleno
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany; Berlin Brandenburg School of Regenerative Therapies (BSRT), Charité Universitätsmedizin, Berlin, Germany
| | - Jerome Jatzlau
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Petra Knaus
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany; Berlin Brandenburg School of Regenerative Therapies (BSRT), Charité Universitätsmedizin, Berlin, Germany.
| |
Collapse
|
25
|
Yamada S, Ishikawa M, Ito H, Yamamoto K, Yamaguchi M, Oshima M, Nozaki K. Cerebrospinal fluid dynamics in idiopathic normal pressure hydrocephalus on four-dimensional flow imaging. Eur Radiol 2020; 30:4454-4465. [PMID: 32246220 DOI: 10.1007/s00330-020-06825-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/27/2020] [Accepted: 03/19/2020] [Indexed: 01/18/2023]
Abstract
OBJECTIVES To evaluate complex CSF movements and shear stress in patients with idiopathic normal pressure hydrocephalus (iNPH) on four-dimensional (4D) flow MRI. METHODS Three-dimensional velocities and volumes of the reciprocating CSF movements through 12 ROIs from the foramen of Monro to the upper cervical spine were measured in 41 patients with iNPH, 23 patients with co-occurrence of iNPH and Alzheimer's disease (AD), and 9 age-matched controls, using 4D flow imaging and application. Stroke volume, reversed-flow rate, and shear stress were automatically calculated. Relationships between flow-related parameters and morphological measurements were also assessed. RESULTS Stroke volumes, reversed-flow rates, and shear stress at the cerebral aqueduct were significantly higher in patients with iNPH than in controls. Patients with pure iNPH had significantly higher shear stress at the ventral aspect of the cerebral aqueduct than those with co-occurrence of iNPH and AD. The stroke volume at the upper end of the cerebral aqueduct had the strongest association with the anteroposterior diameter of the lower end of the cerebral aqueduct (r = 0.52). The stroke volume at the foramen of Monro had significant associations with the indices specific to iNPH. The shear stress at the dorsal aspect of the cerebral aqueduct had the strongest association with the diameter of the foramen of Magendie (r = 0.52). CONCLUSIONS Stroke volumes, reversed-flow rates, and shear stress through the cerebral aqueduct on 4D flow MRI are useful parameters for iNPH diagnosis. These findings can aid in elucidating the mechanism of ventricular enlargement in iNPH. KEY POINTS • The CSF stroke volume and bimodal shear stress at the cerebral aqueduct were considerably higher in patients with iNPH. • The patients with pure iNPH had significantly higher shear stress at the ventral aspect of the cerebral aqueduct than those with co-occurrence of iNPH and AD. • The shear stress at the cerebral aqueduct was significantly associated with the diameter of the foramen of Magendie.
Collapse
Affiliation(s)
- Shigeki Yamada
- Department of Neurosurgery, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Shiga, 520-2192, Japan. .,Department of Neurosurgery and Normal Pressure Hydrocephalus Center, Rakuwakai Otowa Hospital, Kyoto, Japan. .,Interfaculty Initiative in Information Studies/Institute of Industrial Science, The University of Tokyo, Tokyo, Japan.
| | - Masatsune Ishikawa
- Department of Neurosurgery and Normal Pressure Hydrocephalus Center, Rakuwakai Otowa Hospital, Kyoto, Japan.,Rakuwa Villa Ilios, Kyoto, Japan
| | - Hirotaka Ito
- Medical System Research & Development Center, FUJIFILM Corporation, Tokyo, Japan
| | - Kazuo Yamamoto
- Department of Neurosurgery and Normal Pressure Hydrocephalus Center, Rakuwakai Otowa Hospital, Kyoto, Japan
| | - Makoto Yamaguchi
- Department of Neurosurgery and Normal Pressure Hydrocephalus Center, Rakuwakai Otowa Hospital, Kyoto, Japan
| | - Marie Oshima
- Interfaculty Initiative in Information Studies/Institute of Industrial Science, The University of Tokyo, Tokyo, Japan
| | - Kazuhiko Nozaki
- Department of Neurosurgery, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Shiga, 520-2192, Japan
| |
Collapse
|