51
|
Proteomic Insights into Cardiac Fibrosis: From Pathophysiological Mechanisms to Therapeutic Opportunities. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27248784. [PMID: 36557919 PMCID: PMC9781843 DOI: 10.3390/molecules27248784] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022]
Abstract
Cardiac fibrosis is a common pathophysiologic process in nearly all forms of heart disease which refers to excessive deposition of extracellular matrix proteins by cardiac fibroblasts. Activated fibroblasts are the central cellular effectors in cardiac fibrosis, and fibrotic remodelling can cause several cardiac dysfunctions either by reducing the ejection fraction due to a stiffened myocardial matrix, or by impairing electric conductance. Recently, there is a rising focus on the proteomic studies of cardiac fibrosis for pathogenesis elucidation and potential biomarker mining. This paper summarizes the current knowledge of molecular mechanisms underlying cardiac fibrosis, discusses the potential of imaging and circulating biomarkers available to recognize different phenotypes of this lesion, reviews the currently available and potential future therapies that allow individualized management in reversing progressive fibrosis, as well as the recent progress on proteomic studies of cardiac fibrosis. Proteomic approaches using clinical specimens and animal models can provide the ability to track pathological changes and new insights into the mechanisms underlining cardiac fibrosis. Furthermore, spatial and cell-type resolved quantitative proteomic analysis may also serve as a minimally invasive method for diagnosing cardiac fibrosis and allowing for the initiation of prophylactic treatment.
Collapse
|
52
|
Ho DLL, Lee S, Du J, Weiss JD, Tam T, Sinha S, Klinger D, Devine S, Hamfeldt A, Leng HT, Herrmann JE, He M, Fradkin LG, Tan TK, Standish D, Tomasello P, Traul D, Dianat N, Ladi R, Vicard Q, Katikireddy K, Skylar‐Scott MA. Large-Scale Production of Wholly Cellular Bioinks via the Optimization of Human Induced Pluripotent Stem Cell Aggregate Culture in Automated Bioreactors. Adv Healthc Mater 2022; 11:e2201138. [PMID: 36314397 PMCID: PMC10234214 DOI: 10.1002/adhm.202201138] [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] [Received: 05/13/2022] [Revised: 10/10/2022] [Indexed: 01/28/2023]
Abstract
Combining the sustainable culture of billions of human cells and the bioprinting of wholly cellular bioinks offers a pathway toward organ-scale tissue engineering. Traditional 2D culture methods are not inherently scalable due to cost, space, and handling constraints. Here, the suspension culture of human induced pluripotent stem cell-derived aggregates (hAs) is optimized using an automated 250 mL stirred tank bioreactor system. Cell yield, aggregate morphology, and pluripotency marker expression are maintained over three serial passages in two distinct cell lines. Furthermore, it is demonstrated that the same optimized parameters can be scaled to an automated 1 L stirred tank bioreactor system. This 4-day culture results in a 16.6- to 20.4-fold expansion of cells, generating approximately 4 billion cells per vessel, while maintaining >94% expression of pluripotency markers. The pluripotent aggregates can be subsequently differentiated into derivatives of the three germ layers, including cardiac aggregates, and vascular, cortical and intestinal organoids. Finally, the aggregates are compacted into a wholly cellular bioink for rheological characterization and 3D bioprinting. The printed hAs are subsequently differentiated into neuronal and vascular tissue. This work demonstrates an optimized suspension culture-to-3D bioprinting pipeline that enables a sustainable approach to billion cell-scale organ engineering.
Collapse
Affiliation(s)
- Debbie L. L. Ho
- Department of BioengineeringStanford UniversityStanfordCA94305USA
| | - Stacey Lee
- Department of BioengineeringStanford UniversityStanfordCA94305USA
| | - Jianyi Du
- Department of BioengineeringStanford UniversityStanfordCA94305USA
| | | | - Tony Tam
- Department of BioengineeringStanford UniversityStanfordCA94305USA
| | - Soham Sinha
- Department of BioengineeringStanford UniversityStanfordCA94305USA
| | - Danielle Klinger
- Department of BioengineeringStanford UniversityStanfordCA94305USA
| | - Sean Devine
- Sartorius Stedim North America Inc565 Johnson AvenueBohemiaNY11716USA
| | - Art Hamfeldt
- Sartorius Stedim North America Inc565 Johnson AvenueBohemiaNY11716USA
| | - Hope T. Leng
- Department of BioengineeringStanford UniversityStanfordCA94305USA
| | - Jessica E. Herrmann
- Department of BioengineeringStanford UniversityStanfordCA94305USA
- School of MedicineStanford UniversityStanfordCA94305USA
| | - Mengdi He
- Materials Science and EngineeringStanford UniversityStanfordCA94305USA
| | - Lee G. Fradkin
- Department of BioengineeringStanford UniversityStanfordCA94305USA
| | - Tze Kai Tan
- Institute of Stem Cell Biology and Regenerative MedicineStanford University School of MedicineStanfordCA94305USA
- Department of GeneticsStanford University School of MedicineStanfordCA94305USA
- Department of PathologyStanford University School of MedicineStanfordCA94305USA
| | - David Standish
- Sartorius Stedim North America Inc565 Johnson AvenueBohemiaNY11716USA
| | - Peter Tomasello
- Sartorius Stedim North America Inc565 Johnson AvenueBohemiaNY11716USA
| | - Donald Traul
- Sartorius Stedim North America Inc565 Johnson AvenueBohemiaNY11716USA
| | - Noushin Dianat
- Sartorius Stedim France S.A.SZone Industrielle les PaludsAvenue de Jouques CS 71058Aubagne Cedex13781France
| | - Rukmini Ladi
- Sartorius Stedim North America Inc565 Johnson AvenueBohemiaNY11716USA
| | - Quentin Vicard
- Sartorius Stedim France S.A.SZone Industrielle les PaludsAvenue de Jouques CS 71058Aubagne Cedex13781France
| | | | - Mark A. Skylar‐Scott
- Department of BioengineeringStanford UniversityStanfordCA94305USA
- Basic Science and Engineering InitiativeChildren's Heart CenterStanford UniversityStanfordCA94305USA
- Chan Zuckerberg BiohubSan FranciscoCA94158USA
| |
Collapse
|
53
|
Chin IL, Amos SE, Jeong JH, Hool L, Hwang Y, Choi YS. Mechanosensation mediates volume adaptation of cardiac cells and spheroids in 3D. Mater Today Bio 2022; 16:100391. [PMID: 36042852 PMCID: PMC9420370 DOI: 10.1016/j.mtbio.2022.100391] [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: 05/09/2022] [Revised: 08/02/2022] [Accepted: 08/02/2022] [Indexed: 12/27/2022]
Abstract
With the adoption of 3-dimensional (3D) cell culture for in vitro modelling of cardiac function and regenerative medicine applications, there is an increased need to understand cardiomyocyte mechanosensation in 3D. With existing studies of cardiomyocyte mechanosensation primarily focussed on the behaviour of individual cells in a 2-Dimensional context, it is unclear whether mechanosensation is the same in a 3D, multicellular context. In this study, H9C2 cardiac-derived myoblasts were encapsulated as individual cells and as cell spheroids within stiffness gradient gelatin methacryloyl (GelMA) hydrogels to investigate individual and collective cardiac cell mechanosensation in 3D. Over a 3.68–17.52 kPa stiffness range, it was found that H9C2 cells have a limited capacity to adapt their volume to increasing substrate stiffness, demonstrated by the lack of changes in cell volume and shape across the stiffness gradient. Morphological trends were reflected by the expression of the mechanomarkers YAP, MRTF-A and Lamin-A, which were better correlated with cell and nuclear volume than with substrate stiffness. The localisation of YAP and MRTF-A were dependent on the relative volumes of the cytoplasm and nucleus while Lamin-A expression was elevated with increasing cytoplasmic and nuclear volumes. When cultured as spheroids rather than as individual cells, H9C2 cells adopted a distinct morphology with comparably smaller nuclei than individually cultured cells, while retaining the same overall cell volume. As spheroids, H9C2 cells were sensitive to stiffness cues, shown by decreasing YAP and MRTF-A nuclear localisation, increasing Lamin-A expression, and increasing vinculin expression with increasing substrate stiffness. Like the individually cultured H9C2 cells, mechanomarker expression was correlated to volume adaptation. With increasing cytoplasmic volume, YAP and MRTF-A became less nuclear localised, vinculin expression was increased, and with increasing nuclear volume, the Lamin-A expression fincreased. Together, these data suggest that cardiac cell volume adaptation may be enhanced by cell-cell interactions.
Collapse
Affiliation(s)
- Ian L Chin
- School of Human Sciences, The University of Western Australia, Perth, WA, Australia
| | - Sebastian E Amos
- School of Human Sciences, The University of Western Australia, Perth, WA, Australia
| | - Ji Hoon Jeong
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan-si, Chungnam-do, 31151, Republic of Korea.,Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si, Chungnam-do, 31538, Republic of Korea
| | - Livia Hool
- School of Human Sciences, The University of Western Australia, Perth, WA, Australia.,Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
| | - Yongsung Hwang
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan-si, Chungnam-do, 31151, Republic of Korea.,Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si, Chungnam-do, 31538, Republic of Korea
| | - Yu Suk Choi
- School of Human Sciences, The University of Western Australia, Perth, WA, Australia
| |
Collapse
|
54
|
Watson MC, Williams C, Wang RM, Perreault LR, Sullivan KE, Stoppel WL, Black LD. Extracellular matrix and cyclic stretch alter fetal cardiomyocyte proliferation and maturation in a rodent model of heart hypoplasia. Front Cardiovasc Med 2022; 9:993310. [PMID: 36518682 PMCID: PMC9744115 DOI: 10.3389/fcvm.2022.993310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 10/10/2022] [Indexed: 01/22/2024] Open
Abstract
Introduction Birth defects, particularly those that affect development of the heart, are a leading cause of morbidity and mortality in infants and young children. Babies born with heart hypoplasia (heart hypoplasia) disorders often have a poor prognosis. It remains unclear whether cardiomyocytes from hypoplastic hearts retain the potential to recover growth, although this knowledge would be beneficial for developing therapies for heart hypoplasia disorders. The objective of this study was to determine the proliferation and maturation potential of cardiomyocytes from hypoplastic hearts and whether these behaviors are influenced by biochemical signaling from the extracellular matrix (ECM) and cyclic mechanical stretch. Method Congenital diaphragmatic hernia (CDH)-associated heart hypoplasia was induced in rat fetuses by maternal exposure to nitrofen. Hearts were isolated from embryonic day 21 nitrofen-treated fetuses positive for CDH (CDH+) and from fetuses without nitrofen administration during gestation. Results and discussion CDH+ hearts were smaller and had decreased myocardial proliferation, along with evidence of decreased maturity compared to healthy hearts. In culture, CDH+ cardiomyocytes remained immature and demonstrated increased proliferative capacity compared to their healthy counterparts. Culture on ECM derived from CDH+ hearts led to a significant reduction in proliferation for both CDH+ and healthy cardiomyocytes. Healthy cardiomyocytes were dosed with exogenous nitrofen to examine whether nitrofen may have an aberrant effect on the proliferative ability of cardiomyocyte, yet no significant change in proliferation was observed. When subjected to stretch, CDH+ cardiomyocytes underwent lengthening of sarcomeres while healthy cardiomyocyte sarcomeres were unaffected. Taken together, our results suggest that alterations to environmental cues such as ECM and stretch may be important factors in the pathological progression of heart hypoplasia.
Collapse
Affiliation(s)
- Matthew C. Watson
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
- Department of Mechanical Engineering, Tufts University, Medford, MA, United States
| | - Corin Williams
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Raymond M. Wang
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Luke R. Perreault
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Kelly E. Sullivan
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Whitney L. Stoppel
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Lauren D. Black
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
- Cellular, Molecular, and Developmental Biology Program, Sackler School for Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
| |
Collapse
|
55
|
Xie S, Yang Y, Luo Z, Li X, Liu J, Zhang B, Li W. Role of non-cardiomyocytes in anticancer drug-induced cardiotoxicity: A systematic review. iScience 2022; 25:105283. [PMID: 36300001 PMCID: PMC9589207 DOI: 10.1016/j.isci.2022.105283] [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] [Indexed: 11/19/2022] Open
Abstract
Cardiotoxicity induced by anticancer drugs interferes with the continuation of optimal treatment, inducing life-threatening risks or leading to long-term morbidity. The heart is a complex pluricellular organ comprised of cardiomyocytes and non-cardiomyocytes. Although the study of these cell populations has been often focusing on cardiomyocytes, the contributions of non-cardiomyocytes to development and disease are increasingly being appreciated as both dynamic and essential. This review summarized the role of non-cardiomyocytes in anticancer drug-induced cardiotoxicity, including the mechanism of direct damage to resident non-cardiomyocytes, cardiomyocytes injury caused by paracrine modality, myocardial inflammation induced by transient cell populations and the protective agents that focused on non-cardiomyocytes.
Collapse
Affiliation(s)
- Suifen Xie
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Institute of Clinical Pharmacy, Central South University, Changsha, Hunan 410011, China
| | - Yuanying Yang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Institute of Clinical Pharmacy, Central South University, Changsha, Hunan 410011, China
| | - Ziheng Luo
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
| | - Xiangyun Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Institute of Clinical Pharmacy, Central South University, Changsha, Hunan 410011, China
| | - Jian Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Institute of Clinical Pharmacy, Central South University, Changsha, Hunan 410011, China
| | - Bikui Zhang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Institute of Clinical Pharmacy, Central South University, Changsha, Hunan 410011, China
| | - Wenqun Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Institute of Clinical Pharmacy, Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
56
|
Azimzadeh O, Moertl S, Ramadan R, Baselet B, Laiakis EC, Sebastian S, Beaton D, Hartikainen JM, Kaiser JC, Beheshti A, Salomaa S, Chauhan V, Hamada N. Application of radiation omics in the development of adverse outcome pathway networks: an example of radiation-induced cardiovascular disease. Int J Radiat Biol 2022; 98:1722-1751. [PMID: 35976069 DOI: 10.1080/09553002.2022.2110325] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Epidemiological studies have indicated that exposure of the heart to doses of ionizing radiation as low as 0.5 Gy increases the risk of cardiac morbidity and mortality with a latency period of decades. The damaging effects of radiation to myocardial and endothelial structures and functions have been confirmed radiobiologically at high dose, but much less is known at low dose. Integration of radiation biology and epidemiology data is a recommended approach to improve the radiation risk assessment process. The adverse outcome pathway (AOP) framework offers a comprehensive tool to compile and translate mechanistic information into pathological endpoints which may be relevant for risk assessment at the different levels of a biological system. Omics technologies enable the generation of large volumes of biological data at various levels of complexity, from molecular pathways to functional organisms. Given the quality and quantity of available data across levels of biology, omics data can be attractive sources of information for use within the AOP framework. It is anticipated that radiation omics studies could improve our understanding of the molecular mechanisms behind the adverse effects of radiation on the cardiovascular system. In this review, we explored the available omics studies on radiation-induced cardiovascular disease (CVD) and their applicability to the proposed AOP for CVD. RESULTS The results of 80 omics studies published on radiation-induced CVD over the past 20 years have been discussed in the context of the AOP of CVD proposed by Chauhan et al. Most of the available omics data on radiation-induced CVD are from proteomics, transcriptomics, and metabolomics, whereas few datasets were available from epigenomics and multi-omics. The omics data presented here show great promise in providing information for several key events of the proposed AOP of CVD, particularly oxidative stress, alterations of energy metabolism, extracellular matrix and vascular remodeling. CONCLUSIONS The omics data presented here shows promise to inform the various levels of the proposed AOP of CVD. However, the data highlight the urgent need of designing omics studies to address the knowledge gap concerning different radiation scenarios, time after exposure and experimental models. This review presents the evidence to build a qualitative omics-informed AOP and provides views on the potential benefits and challenges in using omics data to assess risk-related outcomes.
Collapse
Affiliation(s)
- Omid Azimzadeh
- Federal Office for Radiation Protection (BfS), Section Radiation Biology, 85764 Neuherberg, Germany
| | - Simone Moertl
- Federal Office for Radiation Protection (BfS), Section Radiation Biology, 85764 Neuherberg, Germany
| | - Raghda Ramadan
- Institute for Environment, Health and Safety, Radiobiology Unit, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Bjorn Baselet
- Institute for Environment, Health and Safety, Radiobiology Unit, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Evagelia C Laiakis
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA.,Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, DC 20057, USA
| | | | | | - Jaana M Hartikainen
- School of Medicine, Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland, Kuopio, Finland
| | - Jan Christian Kaiser
- Helmholtz Zentrum München, Institute of Radiation Medicine (HMGU-IRM), 85764 Neuherberg, Germany
| | - Afshin Beheshti
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Sisko Salomaa
- Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland
| | - Vinita Chauhan
- Environmental Health Science Research Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Nobuyuki Hamada
- Biology and Environmental Chemistry Division, Sustainable System Research Laboratory, Central Research Institute of Electric Power Industry (CRIEPI), Komae, Tokyo 201-8511, Japan
| |
Collapse
|
57
|
Fatoyinbo HO, Brown RG, Simpson DJW, van Brunt B. Pattern Formation in a Spatially Extended Model of Pacemaker Dynamics in Smooth Muscle Cells. Bull Math Biol 2022; 84:86. [PMID: 35804271 PMCID: PMC9270316 DOI: 10.1007/s11538-022-01043-1] [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: 10/26/2021] [Accepted: 06/16/2022] [Indexed: 11/30/2022]
Abstract
Spatiotemporal patterns are common in biological systems. For electrically coupled cells, previous studies of pattern formation have mainly used applied current as the primary bifurcation parameter. The purpose of this paper is to show that applied current is not needed to generate spatiotemporal patterns for smooth muscle cells. The patterns can be generated solely by external mechanical stimulation (transmural pressure). To do this we study a reaction-diffusion system involving the Morris-Lecar equations and observe a wide range of spatiotemporal patterns for different values of the model parameters. Some aspects of these patterns are explained via a bifurcation analysis of the system without coupling - in particular Type I and Type II excitability both occur. We show the patterns are not due to a Turing instability and that the spatially extended model exhibits spatiotemporal chaos. We also use travelling wave coordinates to analyse travelling waves.
Collapse
Affiliation(s)
- H. O. Fatoyinbo
- School of Mathematical and Computational Sciences, Massey University, Palmerston North, New Zealand
| | - R. G. Brown
- School of Mathematical and Computational Sciences, Massey University, Palmerston North, New Zealand
| | - D. J. W. Simpson
- School of Mathematical and Computational Sciences, Massey University, Palmerston North, New Zealand
| | - B. van Brunt
- School of Mathematical and Computational Sciences, Massey University, Palmerston North, New Zealand
| |
Collapse
|
58
|
Abelanet A, Camoin M, Rubin S, Bougaran P, Delobel V, Pernot M, Forfar I, Guilbeau-Frugier C, Galès C, Bats ML, Renault MA, Dufourcq P, Couffinhal T, Duplàa C. Increased Capillary Permeability in Heart Induces Diastolic Dysfunction Independently of Inflammation, Fibrosis, or Cardiomyocyte Dysfunction. Arterioscler Thromb Vasc Biol 2022; 42:745-763. [PMID: 35510550 DOI: 10.1161/atvbaha.121.317319] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND While endothelial dysfunction is suggested to contribute to heart failure with preserved ejection fraction pathophysiology, understanding the importance of the endothelium alone, in the pathogenesis of diastolic abnormalities has not yet been fully elucidated. Here, we investigated the consequences of specific endothelial dysfunction on cardiac function, independently of any comorbidity or risk factor (diabetes or obesity) and their potential effect on cardiomyocyte. METHODS The ubiquitine ligase Pdzrn3, expressed in endothelial cells (ECs), was shown to destabilize tight junction. A genetic mouse model in which Pdzrn3 is overexpressed in EC (iEC-Pdzrn3) in adults was developed. RESULTS EC-specific Pdzrn3 expression increased cardiac leakage of IgG and fibrinogen blood-born molecules. The induced edema demonstrated features of diastolic dysfunction, with increased end-diastolic pressure, alteration of dP/dt min, increased natriuretic peptides, in addition to limited exercise capacity, without major signs of cardiac fibrosis and inflammation. Electron microscopic images showed edema with disrupted EC-cardiomyocyte interactions. RNA sequencing analysis of gene expression in cardiac EC demonstrated a decrease in genes coding for endothelial extracellular matrix proteins, which could be related to the fragile blood vessel phenotype. Irregularly shaped capillaries with hemorrhages were found in heart sections of iEC-Pdzrn3 mice. We also found that a high-fat diet was not sufficient to provoke diastolic dysfunction; high-fat diet aggravated cardiac inflammation, associated with an altered cardiac metabolic signature in EC-Pdzrn3 mice, reminiscent of heart failure with preserved ejection fraction features. CONCLUSIONS An increase of endothelial permeability is responsible for mediating diastolic dysfunction pathophysiology and for aggravating detrimental effects of a high-fat diet on cardiac inflammation and metabolism.
Collapse
Affiliation(s)
- Alice Abelanet
- University of Bordeaux, INSERM, Biologie des maladies cardiovasculaires, U1034, Pessac, France (A.A., M.C., S.R., P.B., V.D., M.P., I.F., M.L.B., M.-A.R., P.D., T.C., C.D.)
| | - Marion Camoin
- University of Bordeaux, INSERM, Biologie des maladies cardiovasculaires, U1034, Pessac, France (A.A., M.C., S.R., P.B., V.D., M.P., I.F., M.L.B., M.-A.R., P.D., T.C., C.D.).,CHU de Bordeaux, Pessac, France (M.C., S.R., M.P., M.L.B., P.D., T.C.)
| | - Sebastien Rubin
- University of Bordeaux, INSERM, Biologie des maladies cardiovasculaires, U1034, Pessac, France (A.A., M.C., S.R., P.B., V.D., M.P., I.F., M.L.B., M.-A.R., P.D., T.C., C.D.).,CHU de Bordeaux, Pessac, France (M.C., S.R., M.P., M.L.B., P.D., T.C.)
| | - Pauline Bougaran
- University of Bordeaux, INSERM, Biologie des maladies cardiovasculaires, U1034, Pessac, France (A.A., M.C., S.R., P.B., V.D., M.P., I.F., M.L.B., M.-A.R., P.D., T.C., C.D.)
| | - Valentin Delobel
- University of Bordeaux, INSERM, Biologie des maladies cardiovasculaires, U1034, Pessac, France (A.A., M.C., S.R., P.B., V.D., M.P., I.F., M.L.B., M.-A.R., P.D., T.C., C.D.)
| | - Mathieu Pernot
- University of Bordeaux, INSERM, Biologie des maladies cardiovasculaires, U1034, Pessac, France (A.A., M.C., S.R., P.B., V.D., M.P., I.F., M.L.B., M.-A.R., P.D., T.C., C.D.).,CHU de Bordeaux, Pessac, France (M.C., S.R., M.P., M.L.B., P.D., T.C.)
| | - Isabelle Forfar
- University of Bordeaux, INSERM, Biologie des maladies cardiovasculaires, U1034, Pessac, France (A.A., M.C., S.R., P.B., V.D., M.P., I.F., M.L.B., M.-A.R., P.D., T.C., C.D.)
| | - Céline Guilbeau-Frugier
- Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, INSERM U1048, I2MC, France (C.G.-F., C.G.)
| | - Céline Galès
- Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, INSERM U1048, I2MC, France (C.G.-F., C.G.)
| | - Marie Lise Bats
- University of Bordeaux, INSERM, Biologie des maladies cardiovasculaires, U1034, Pessac, France (A.A., M.C., S.R., P.B., V.D., M.P., I.F., M.L.B., M.-A.R., P.D., T.C., C.D.).,CHU de Bordeaux, Pessac, France (M.C., S.R., M.P., M.L.B., P.D., T.C.)
| | - Marie-Ange Renault
- University of Bordeaux, INSERM, Biologie des maladies cardiovasculaires, U1034, Pessac, France (A.A., M.C., S.R., P.B., V.D., M.P., I.F., M.L.B., M.-A.R., P.D., T.C., C.D.)
| | - Pascale Dufourcq
- University of Bordeaux, INSERM, Biologie des maladies cardiovasculaires, U1034, Pessac, France (A.A., M.C., S.R., P.B., V.D., M.P., I.F., M.L.B., M.-A.R., P.D., T.C., C.D.).,CHU de Bordeaux, Pessac, France (M.C., S.R., M.P., M.L.B., P.D., T.C.)
| | - Thierry Couffinhal
- University of Bordeaux, INSERM, Biologie des maladies cardiovasculaires, U1034, Pessac, France (A.A., M.C., S.R., P.B., V.D., M.P., I.F., M.L.B., M.-A.R., P.D., T.C., C.D.).,CHU de Bordeaux, Pessac, France (M.C., S.R., M.P., M.L.B., P.D., T.C.)
| | - Cécile Duplàa
- University of Bordeaux, INSERM, Biologie des maladies cardiovasculaires, U1034, Pessac, France (A.A., M.C., S.R., P.B., V.D., M.P., I.F., M.L.B., M.-A.R., P.D., T.C., C.D.)
| |
Collapse
|
59
|
Galli A, Montree RJH, Que S, Peri E, Vullings R. An Overview of the Sensors for Heart Rate Monitoring Used in Extramural Applications. SENSORS (BASEL, SWITZERLAND) 2022; 22:4035. [PMID: 35684656 PMCID: PMC9185322 DOI: 10.3390/s22114035] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 06/02/2023]
Abstract
This work presents an overview of the main strategies that have been proposed for non-invasive monitoring of heart rate (HR) in extramural and home settings. We discuss three categories of sensing according to what physiological effect is used to measure the pulsatile activity of the heart, and we focus on an illustrative sensing modality for each of them. Therefore, electrocardiography, photoplethysmography, and mechanocardiography are presented as illustrative modalities to sense electrical activity, mechanical activity, and the peripheral effect of heart activity. In this paper, we describe the physical principles underlying the three categories and the characteristics of the different types of sensors that belong to each class, and we touch upon the most used software strategies that are currently adopted to effectively and reliably extract HR. In addition, we investigate the strengths and weaknesses of each category linked to the different applications in order to provide the reader with guidelines for selecting the most suitable solution according to the requirements and constraints of the application.
Collapse
Affiliation(s)
- Alessandra Galli
- Department of Information Engineering, University of Padova, I-35131 Padova, Italy;
| | - Roel J. H. Montree
- Department of Electrical Engineering, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands; (R.J.H.M.); (S.Q.); (E.P.)
| | - Shuhao Que
- Department of Electrical Engineering, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands; (R.J.H.M.); (S.Q.); (E.P.)
| | - Elisabetta Peri
- Department of Electrical Engineering, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands; (R.J.H.M.); (S.Q.); (E.P.)
| | - Rik Vullings
- Department of Electrical Engineering, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands; (R.J.H.M.); (S.Q.); (E.P.)
| |
Collapse
|
60
|
Hussain MWA, Garg P, Yazji JH, Alomari M, Alamouti-fard E, Wadiwala I, Jacob S. Is a Bioengineered Heart From Recipient Tissues the Answer to the Shortage of Donors in Heart Transplantation? Cureus 2022; 14:e25329. [PMID: 35637923 PMCID: PMC9132496 DOI: 10.7759/cureus.25329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/25/2022] [Indexed: 11/08/2022] Open
Abstract
With the increase in life expectancy worldwide, end-organ failure is becoming more prevalent. In addition, improving post-transplant outcomes has contributed to soaring demand for organs. Unfortunately, thousands have died waiting on the transplant list due to the critical shortage of organs. The success of bioengineered hearts may eventually lead to the production of limitless organs using the patient’s own cells that can be transplanted into them without the need for immunosuppressive medications. Despite being in its infancy, scientists are making tremendous strides in “growing” an artificial heart in the lab. We discuss these processes involved in bioengineering a human-compatible heart in this review. The components of a functional heart must be replicated in a bioengineered heart to make it viable. This review aims to discuss the advances that have already been made and the future challenges of bioengineering a human heart suitable for transplantation.
Collapse
|
61
|
Caturano A, Vetrano E, Galiero R, Salvatore T, Docimo G, Epifani R, Alfano M, Sardu C, Marfella R, Rinaldi L, Sasso FC. Cardiac Hypertrophy: From Pathophysiological Mechanisms to Heart Failure Development. Rev Cardiovasc Med 2022; 23:165. [PMID: 39077592 PMCID: PMC11273913 DOI: 10.31083/j.rcm2305165] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/23/2022] [Accepted: 02/28/2022] [Indexed: 07/31/2024] Open
Abstract
Cardiac hypertrophy develops in response to increased workload to reduce ventricular wall stress and maintain function and efficiency. Pathological hypertrophy can be adaptive at the beginning. However, if the stimulus persists, it may progress to ventricular chamber dilatation, contractile dysfunction, and heart failure, resulting in poorer outcome and increased social burden. The main pathophysiological mechanisms of pathological hypertrophy are cell death, fibrosis, mitochondrial dysfunction, dysregulation of Ca 2 + -handling proteins, metabolic changes, fetal gene expression reactivation, impaired protein and mitochondrial quality control, altered sarcomere structure, and inadequate angiogenesis. Diabetic cardiomyopathy is a condition in which cardiac pathological hypertrophy mainly develop due to insulin resistance and subsequent hyperglycaemia, associated with altered fatty acid metabolism, altered calcium homeostasis and inflammation. In this review, we summarize the underlying molecular mechanisms of pathological hypertrophy development and progression, which can be applied in the development of future novel therapeutic strategies in both reversal and prevention.
Collapse
Affiliation(s)
- Alfredo Caturano
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| | - Erica Vetrano
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| | - Raffaele Galiero
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| | - Teresa Salvatore
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| | - Giovanni Docimo
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| | - Raffaella Epifani
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| | - Maria Alfano
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| | - Celestino Sardu
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| | - Raffaele Marfella
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| | - Luca Rinaldi
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| | - Ferdinando Carlo Sasso
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| |
Collapse
|
62
|
Williams MAC, Mair DB, Lee W, Lee E, Kim DH. Engineering Three-Dimensional Vascularized Cardiac Tissues. TISSUE ENGINEERING. PART B, REVIEWS 2022; 28:336-350. [PMID: 33559514 PMCID: PMC9063162 DOI: 10.1089/ten.teb.2020.0343] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 02/08/2021] [Indexed: 12/23/2022]
Abstract
Heart disease is one of the largest burdens to human health worldwide and has very limited therapeutic options. Engineered three-dimensional (3D) vascularized cardiac tissues have shown promise in rescuing cardiac function in diseased hearts and may serve as a whole organ replacement in the future. One of the major obstacles in reconstructing these thick myocardial tissues to a clinically applicable scale is the integration of functional vascular networks capable of providing oxygen and nutrients throughout whole engineered constructs. Without perfusion of oxygen and nutrient flow throughout the entire engineered tissue not only is tissue viability compromised, but also overall tissue functionality is lost. There are many supporting technologies and approaches that have been developed to create vascular networks such as 3D bioprinting, co-culturing hydrogels, and incorporation of soluble angiogenic factors. In this state-of-the-art review, we discuss some of the most current engineered vascular cardiac tissues reported in the literature and future directions in the field. Impact statement The field of cardiac tissue engineering is rapidly evolving and is now closer than ever to having engineered tissue models capable of predicting preclinical responses to therapeutics, modeling diseases, and being used as a means of rescuing cardiac function following injuries to the native myocardium. However, a major obstacle of engineering thick cardiac tissue remains to be the integration of functional vasculature. In this review, we highlight seminal and recently published works that have influenced and pushed the field of cardiac tissue engineering toward achieving vascularized functional tissues.
Collapse
Affiliation(s)
| | - Devin B. Mair
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Wonjae Lee
- Department of Neurosurgery, Stanford School of Medicine, Stanford, California, USA
| | - Esak Lee
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
63
|
Germena G, Zelarayán LC, Hinkel R. Cellular Chitchatting: Exploring the Role of Exosomes as Cardiovascular Risk Factors. Front Cell Dev Biol 2022; 10:860005. [PMID: 35433670 PMCID: PMC9008366 DOI: 10.3389/fcell.2022.860005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 03/16/2022] [Indexed: 11/24/2022] Open
Abstract
Exosomes are small bi-lipid membranous vesicles (30–150 nm) containing different biological material such as proteins, lipids and nucleic acid. These small vesicles, inducing a cell to cell signaling pathway, are able to mediate multidirectional crosstalk to maintain homeostasis or modulate disease processes. With their various contents, exosomes sort and transfer specific information from their origin to a recipient cell, from a tissue or organ in the close proximity or at distance, generating an intra-inter tissue or organ communication. In the last decade exosomes have been identified in multiple organs and fluids under different pathological conditions. In particular, while the content and the abundance of exosome is now a diagnostic marker for cardiovascular diseases, their role in context-specific physiological and pathophysiological conditions in the cardiovascular system remains largely unknown. We summarize here the current knowledge on the role of exosomes as mediators of cardiovascular diseases in several pathophysiological conditions such as atherosclerosis and diabetes. In addition, we describe evidence of intercellular connection among multiple cell type (cardiac, vasculature, immune cells) as well as the challenge of their in vivo analysis.
Collapse
Affiliation(s)
- Giulia Germena
- Laboratory Animal Science Unit, Leibniz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
- *Correspondence: Giulia Germena, ; Rabea Hinkel,
| | - Laura Cecilia Zelarayán
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany
| | - Rabea Hinkel
- Laboratory Animal Science Unit, Leibniz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
- Institute for Animal Hygiene, Animal Welfare and Farm Animal Behaviour (ITTN), Stiftung Tierärztliche Hochschule Hannover, University of Veterinary Medicine, Hannover, Germany
- *Correspondence: Giulia Germena, ; Rabea Hinkel,
| |
Collapse
|
64
|
Progress in Bioengineering Strategies for Heart Regenerative Medicine. Int J Mol Sci 2022; 23:ijms23073482. [PMID: 35408844 PMCID: PMC8998628 DOI: 10.3390/ijms23073482] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/20/2022] [Accepted: 03/21/2022] [Indexed: 02/05/2023] Open
Abstract
The human heart has the least regenerative capabilities among tissues and organs, and heart disease continues to be a leading cause of mortality in the industrialized world with insufficient therapeutic options and poor prognosis. Therefore, developing new therapeutic strategies for heart regeneration is a major goal in modern cardiac biology and medicine. Recent advances in stem cell biology and biotechnologies such as human pluripotent stem cells (hPSCs) and cardiac tissue engineering hold great promise for opening novel paths to heart regeneration and repair for heart disease, although these areas are still in their infancy. In this review, we summarize and discuss the recent progress in cardiac tissue engineering strategies, highlighting stem cell engineering and cardiomyocyte maturation, development of novel functional biomaterials and biofabrication tools, and their therapeutic applications involving drug discovery, disease modeling, and regenerative medicine for heart disease.
Collapse
|
65
|
Mathematical modelling of autoimmune myocarditis and the effects of immune checkpoint inhibitors. J Theor Biol 2022; 537:111002. [PMID: 35007511 DOI: 10.1016/j.jtbi.2021.111002] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 12/20/2021] [Accepted: 12/27/2021] [Indexed: 12/26/2022]
Abstract
Autoimmune myocarditis is a rare, but frequently fatal, side effect of immune checkpoint inhibitors (ICIs), a class of cancer therapies. Despite extensive experimental work on the causes, development and progression of this disease, much still remains unknown about the importance of the different immunological pathways involved. We present a mathematical model of autoimmune myocarditis and the effects of ICIs on its development and progression to either resolution or chronic inflammation. From this, we gain a better understanding of the role of immune cells, cytokines and other components of the immune system in driving the cardiotoxicity of ICIs. We parameterise the model using existing data from the literature, and show that qualitative model behaviour is consistent with disease characteristics seen in patients in an ICI-free context. The bifurcation structures of the model show how the presence of ICIs increases the risk of developing autoimmune myocarditis. This predictive modelling approach is a first step towards determining treatment regimens that balance the benefits of treating cancer with the risk of developing autoimmune myocarditis.
Collapse
|
66
|
Graham C, Sethu P. Myocardial Fibrosis: Cell Signaling and In Vitro Modeling. CARDIOVASCULAR SIGNALING IN HEALTH AND DISEASE 2022:287-321. [DOI: 10.1007/978-3-031-08309-9_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
67
|
Corker A, Neff LS, Broughton P, Bradshaw AD, DeLeon-Pennell KY. Organized Chaos: Deciphering Immune Cell Heterogeneity's Role in Inflammation in the Heart. Biomolecules 2021; 12:11. [PMID: 35053159 PMCID: PMC8773626 DOI: 10.3390/biom12010011] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/10/2021] [Accepted: 12/18/2021] [Indexed: 12/24/2022] Open
Abstract
During homeostasis, immune cells perform daily housekeeping functions to maintain heart health by acting as sentinels for tissue damage and foreign particles. Resident immune cells compose 5% of the cellular population in healthy human ventricular tissue. In response to injury, there is an increase in inflammation within the heart due to the influx of immune cells. Some of the most common immune cells recruited to the heart are macrophages, dendritic cells, neutrophils, and T-cells. In this review, we will discuss what is known about cardiac immune cell heterogeneity during homeostasis, how these cell populations change in response to a pathology such as myocardial infarction or pressure overload, and what stimuli are regulating these processes. In addition, we will summarize technologies used to evaluate cell heterogeneity in models of cardiovascular disease.
Collapse
Affiliation(s)
- Alexa Corker
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, Charleston, SC 29425, USA; (A.C.); (L.S.N.); (P.B.); (A.D.B.)
| | - Lily S. Neff
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, Charleston, SC 29425, USA; (A.C.); (L.S.N.); (P.B.); (A.D.B.)
| | - Philip Broughton
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, Charleston, SC 29425, USA; (A.C.); (L.S.N.); (P.B.); (A.D.B.)
| | - Amy D. Bradshaw
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, Charleston, SC 29425, USA; (A.C.); (L.S.N.); (P.B.); (A.D.B.)
- Ralph H. Johnson Veterans Affairs Medical Center, Medical University of South Carolina, Charleston, SC 29401, USA
| | - Kristine Y. DeLeon-Pennell
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, Charleston, SC 29425, USA; (A.C.); (L.S.N.); (P.B.); (A.D.B.)
- Ralph H. Johnson Veterans Affairs Medical Center, Medical University of South Carolina, Charleston, SC 29401, USA
| |
Collapse
|
68
|
Taghdiri N, Calcagno DM, Fu Z, Huang K, Kohler RH, Weissleder R, Coleman TP, King KR. Macrophage calcium reporter mice reveal immune cell communication in vitro and in vivo. CELL REPORTS METHODS 2021; 1:100132. [PMID: 35079727 PMCID: PMC8786215 DOI: 10.1016/j.crmeth.2021.100132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 08/26/2021] [Accepted: 11/19/2021] [Indexed: 01/01/2023]
Abstract
Cell communication underlies emergent functions in diverse cell types and tissues. Recent evidence suggests that macrophages are organized in communicating networks, but new tools are needed to quantitatively characterize the resulting cellular conversations. Here, we infer cell communication from spatiotemporal correlations of intracellular calcium dynamics that are non-destructively imaged across cell populations expressing genetically encoded calcium indicators. We describe a hematopoietic calcium reporter mouse (Csf1rCreGCaMP5fl) and a computational analysis pipeline for inferring communication between reporter cells based on "excess synchrony." We observed signals suggestive of cell communication in macrophages treated with immune-stimulatory DNA in vitro and tumor-associated immune cells imaged in a dorsal window chamber model in vivo. Together, the methods described here expand the toolkit for discovery of cell communication events in macrophages and other immune cells.
Collapse
Affiliation(s)
- Nika Taghdiri
- Department of Bioengineering, Jacobs School of Engineering, University of California, San Diego, 9500 Gilman Dr. MC 0412, La Jolla, CA 92093, USA
| | - David M. Calcagno
- Department of Bioengineering, Jacobs School of Engineering, University of California, San Diego, 9500 Gilman Dr. MC 0412, La Jolla, CA 92093, USA
| | - Zhenxing Fu
- Division of Cardiology and Cardiovascular Institute, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Kenneth Huang
- Division of Cardiology and Cardiovascular Institute, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Rainer H. Kohler
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA, USA
| | - Todd P. Coleman
- Department of Bioengineering, Jacobs School of Engineering, University of California, San Diego, 9500 Gilman Dr. MC 0412, La Jolla, CA 92093, USA
| | - Kevin R. King
- Department of Bioengineering, Jacobs School of Engineering, University of California, San Diego, 9500 Gilman Dr. MC 0412, La Jolla, CA 92093, USA
- Division of Cardiology and Cardiovascular Institute, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
69
|
Gunawan F, Priya R, Stainier DYR. Sculpting the heart: Cellular mechanisms shaping valves and trabeculae. Curr Opin Cell Biol 2021; 73:26-34. [PMID: 34147705 DOI: 10.1016/j.ceb.2021.04.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 04/30/2021] [Indexed: 12/13/2022]
Abstract
The transformation of the heart from a simple tube to a complex organ requires the orchestration of several morphogenetic processes. Two structures critical for cardiac function, the cardiac valves and the trabecular network, are formed through extensive tissue morphogenesis-endocardial cell migration, deadhesion and differentiation into fibroblast-like cells during valve formation, and cardiomyocyte delamination and apico-basal depolarization during trabeculation. Here, we review current knowledge of how these specialized structures acquire their shape by focusing on the underlying cellular behaviors and molecular mechanisms, highlighting findings from in vivo models and briefly discussing the recent advances in cardiac cell culture and organoids.
Collapse
Affiliation(s)
- Felix Gunawan
- Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, Bad Nauheim 61231, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany; Excellence Cluster Cardio-Pulmonary Institute (CPI), Bad Nauheim, Frankfurt, Giessen, Germany.
| | - Rashmi Priya
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| | - Didier Y R Stainier
- Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, Bad Nauheim 61231, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany; Excellence Cluster Cardio-Pulmonary Institute (CPI), Bad Nauheim, Frankfurt, Giessen, Germany.
| |
Collapse
|
70
|
Jæger KH, Edwards AG, Giles WR, Tveito A. From Millimeters to Micrometers; Re-introducing Myocytes in Models of Cardiac Electrophysiology. Front Physiol 2021; 12:763584. [PMID: 34777021 PMCID: PMC8578869 DOI: 10.3389/fphys.2021.763584] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 09/30/2021] [Indexed: 11/13/2022] Open
Abstract
Computational modeling has contributed significantly to present understanding of cardiac electrophysiology including cardiac conduction, excitation-contraction coupling, and the effects and side-effects of drugs. However, the accuracy of in silico analysis of electrochemical wave dynamics in cardiac tissue is limited by the homogenization procedure (spatial averaging) intrinsic to standard continuum models of conduction. Averaged models cannot resolve the intricate dynamics in the vicinity of individual cardiomyocytes simply because the myocytes are not present in these models. Here we demonstrate how recently developed mathematical models based on representing every myocyte can significantly increase the accuracy, and thus the utility of modeling electrophysiological function and dysfunction in collections of coupled cardiomyocytes. The present gold standard of numerical simulation for cardiac electrophysiology is based on the bidomain model. In the bidomain model, the extracellular (E) space, the cell membrane (M) and the intracellular (I) space are all assumed to be present everywhere in the tissue. Consequently, it is impossible to study biophysical processes taking place close to individual myocytes. The bidomain model represents the tissue by averaging over several hundred myocytes and this inherently limits the accuracy of the model. In our alternative approach both E, M, and I are represented in the model which is therefore referred to as the EMI model. The EMI model approach allows for detailed analysis of the biophysical processes going on in functionally important spaces very close to individual myocytes, although at the cost of significantly increased CPU-requirements.
Collapse
Affiliation(s)
| | | | - Wayne R Giles
- Simula Research Laboratory, Lysaker, Norway
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
| | | |
Collapse
|
71
|
Thakur V, Alcoreza N, Cazares J, Chattopadhyay M. Changes in Stress-Mediated Markers in a Human Cardiomyocyte Cell Line under Hyperglycemia. Int J Mol Sci 2021; 22:10802. [PMID: 34639171 PMCID: PMC8509354 DOI: 10.3390/ijms221910802] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 09/30/2021] [Accepted: 10/01/2021] [Indexed: 01/04/2023] Open
Abstract
Diabetes is a major risk factor for cardiovascular diseases, especially cardiomyopathy, a condition in which the smooth muscles of the heart become thick and rigid, affecting the functioning of cardiomyocytes, the contractile cells of the heart. Uncontrolled elevated glucose levels over time can result in oxidative stress, which could lead to inflammation and altered epigenetic mechanisms. In the current study, we investigated whether hyperglycemia can modify cardiac function by directly affecting these changes in cardiomyocytes. To evaluate the adverse effect of high glucose, we measured the levels of gap junction protein, connexin 43, which is responsible for modulating cardiac electric activities and Troponin I, a part of the troponin complex in the heart muscles, commonly used as cardiac markers of ischemic heart disease. AC16 human cardiomyocyte cells were used in this study. Under hyperglycemic conditions, these cells demonstrated altered levels of connexin 43 and Troponin-I after 24 h of exposure. We also examined hyperglycemia induced changes in epigenetic markers: H3K9me1, Sirtuin-1 (SIRT1), and histone deacetylase (HDAC)-2 as well as in inflammatory and stress-related mediators, such as heat shock protein (HSP)-60, receptor for advanced glycation end products (RAGE), toll-like receptor (TLR)-4, high mobility group box (HMGB)-1 and CXC chemokine receptor (CXCR)-4. Cardiomyocytes exposed to 25mM glucose resulted in the downregulation of HSP60 and SIRT1 after 48 h. We further examined that hyperglycemia mediated the decrease in the gap junction protein CX43, as well as CXC chemokine receptor CXCR4 which may affect the physiological functions of the cardiomyocytes when exposed to high glucose for 24 and 48 h. Upregulated expression of DNA-binding nuclear protein HMGB1, along with changes in histone methylation marker H3K9me1 have demonstrated hyperglycemia-induced damage to cardiomyocyte at 24 h of exposure. Our study established that 24 to 48 h of hyperglycemic exposure could stimulate stress-mediated inflammatory mediators in cardiomyocytes in vitro. These stress-related changes in hyperglycemia-induced cardiomyocytes may further initiate an increase in injury markers which eventually could alter the epigenetic processes. Therefore, epigenetic and inflammatory mechanisms in conjunction with alterations in a downstream signaling pathway could have a direct effect on the functionality of the cardiomyocytes exposed to high glucose during short and long-term exposures.
Collapse
Affiliation(s)
- Vikram Thakur
- Center of Emphasis in Diabetes and Metabolism, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA;
| | - Narah Alcoreza
- Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (N.A.); (J.C.)
| | - Jasmine Cazares
- Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (N.A.); (J.C.)
| | - Munmun Chattopadhyay
- Center of Emphasis in Diabetes and Metabolism, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA;
- Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (N.A.); (J.C.)
| |
Collapse
|
72
|
Guo H, Liu L, Nishiga M, Cong L, Wu JC. Deciphering pathogenicity of variants of uncertain significance with CRISPR-edited iPSCs. Trends Genet 2021; 37:1109-1123. [PMID: 34509299 DOI: 10.1016/j.tig.2021.08.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 10/20/2022]
Abstract
Genetic variants play an important role in conferring risk for cardiovascular diseases (CVDs). With the rapid development of next-generation sequencing (NGS), thousands of genetic variants associated with CVDs have been identified by genome-wide association studies (GWAS), but the function of more than 40% of genetic variants is still unknown. This gap of knowledge is a barrier to the clinical application of the genetic information. However, determining the pathogenicity of a variant of uncertain significance (VUS) is challenging due to the lack of suitable model systems and accessible technologies. By combining clustered regularly interspaced short palindromic repeats (CRISPR) and human induced pluripotent stem cells (iPSCs), unprecedented advances are now possible in determining the pathogenicity of VUS in CVDs. Here, we summarize recent progress and new strategies in deciphering pathogenic variants for CVDs using CRISPR-edited human iPSCs.
Collapse
Affiliation(s)
- Hongchao Guo
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lichao Liu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Masataka Nishiga
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Le Cong
- Department of Pathology and Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
73
|
Lityagina O, Dobreva G. The LINC Between Mechanical Forces and Chromatin. Front Physiol 2021; 12:710809. [PMID: 34408666 PMCID: PMC8365421 DOI: 10.3389/fphys.2021.710809] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 07/14/2021] [Indexed: 12/26/2022] Open
Abstract
The heart continually senses and responds to mechanical stimuli that balance cardiac structure and activity. Tensile forces, compressive forces, and shear stress are sensed by the different cardiac cell types and converted into signals instructing proper heart morphogenesis, postnatal growth, and function. Defects in mechanotransduction, the ability of cells to convert mechanical stimuli into biochemical signals, are implicated in cardiovascular disease development and progression. In this review, we summarize the current knowledge on how mechanical forces are transduced to chromatin through the tensed actomyosin cytoskeleton, the linker of nucleoskeleton and cytoskeleton (LINC) complex and the nuclear lamina. We also discuss the functional significance of the LINC complex in cardiovascular disease.
Collapse
Affiliation(s)
- Olga Lityagina
- Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Gergana Dobreva
- Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, Mannheim, Germany
| |
Collapse
|
74
|
GDF15 and Cardiac Cells: Current Concepts and New Insights. Int J Mol Sci 2021; 22:ijms22168889. [PMID: 34445593 PMCID: PMC8396208 DOI: 10.3390/ijms22168889] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 08/12/2021] [Accepted: 08/13/2021] [Indexed: 02/06/2023] Open
Abstract
Growth and differentiation factor 15 (GDF15) belongs to the transforming growth factor-β (TGF-β) superfamily of proteins. Glial-derived neurotrophic factor (GDNF) family receptor α-like (GFRAL) is an endogenous receptor for GDF15 detected selectively in the brain. GDF15 is not normally expressed in the tissue but is prominently induced by “injury”. Serum levels of GDF15 are also increased by aging and in response to cellular stress and mitochondrial dysfunction. It acts as an inflammatory marker and plays a role in the pathogenesis of cardiovascular diseases, metabolic disorders, and neurodegenerative processes. Identified as a new heart-derived endocrine hormone that regulates body growth, GDF15 has a local cardioprotective role, presumably due to its autocrine/paracrine properties: antioxidative, anti-inflammatory, antiapoptotic. GDF15 expression is highly induced in cardiomyocytes after ischemia/reperfusion and in the heart within hours after myocardial infarction (MI). Recent studies show associations between GDF15, inflammation, and cardiac fibrosis during heart failure and MI. However, the reason for this increase in GDF15 production has not been clearly identified. Experimental and clinical studies support the potential use of GDF15 as a novel therapeutic target (1) by modulating metabolic activity and (2) promoting an adaptive angiogenesis and cardiac regenerative process during cardiovascular diseases. In this review, we comment on new aspects of the biology of GDF15 as a cardiac hormone and show that GDF15 may be a predictive biomarker of adverse cardiac events.
Collapse
|
75
|
Guo F, Hall AR, Tape CJ, Ling S, Pointon A. Intra- and intercellular signaling pathways associated with drug-induced cardiac pathophysiology. Trends Pharmacol Sci 2021; 42:675-687. [PMID: 34092416 DOI: 10.1016/j.tips.2021.05.004] [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: 12/10/2020] [Revised: 04/20/2021] [Accepted: 05/06/2021] [Indexed: 11/30/2022]
Abstract
Cardiac physiology and homeostasis are maintained by the interaction of multiple cell types, via both intra- and intercellular signaling pathways. Perturbations in these signaling pathways induced by oncology therapies can reduce cardiac function, ultimately leading to heart failure. As cancer survival increases, related cardiovascular complications are becoming increasingly prevalent, thus identifying the perturbations and cell signaling drivers of cardiotoxicity is increasingly important. Here, we discuss the homotypic and heterotypic cellular interactions that form the basis of intra- and intercellular cardiac signaling pathways, and how oncological agents disrupt these pathways, leading to heart failure. We also highlight the emerging systems biology techniques that can be applied, enabling a deeper understanding of the intra- and intercellular signaling pathways across multiple cell types associated with cardiovascular toxicity.
Collapse
Affiliation(s)
- Fei Guo
- Functional and Mechanistic Safety, Clinical Pharmacology and Safety Sciences, Research and Development, AstraZeneca, Cambridge, UK; Cell Communication Laboratory, Department of Oncology, University College London Cancer Institute, London, WC1E 6DD, UK
| | - Andrew R Hall
- Functional and Mechanistic Safety, Clinical Pharmacology and Safety Sciences, Research and Development, AstraZeneca, Cambridge, UK
| | - Christopher J Tape
- Cell Communication Laboratory, Department of Oncology, University College London Cancer Institute, London, WC1E 6DD, UK
| | - Stephanie Ling
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, Research and Development, AstraZeneca, Cambridge, UK
| | - Amy Pointon
- Functional and Mechanistic Safety, Clinical Pharmacology and Safety Sciences, Research and Development, AstraZeneca, Cambridge, UK.
| |
Collapse
|
76
|
Lafci Büyükkahraman M, Sabine GK, Kojouharov HV, Chen-Charpentier BM, McMahan SR, Liao J. Using models to advance medicine: mathematical modeling of post-myocardial infarction left ventricular remodeling. Comput Methods Biomech Biomed Engin 2021; 25:298-307. [PMID: 34266318 DOI: 10.1080/10255842.2021.1953487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The heart is an organ with limited capacity for regeneration and repair. The irreversible cell death and corresponding diminished ability of the heart to repair after myocardial infarction (MI), is a leading cause of morbidity and mortality worldwide. In this paper, a new mathematical model is presented to study the left ventricular (LV) remodeling and associated events after MI. The model accurately describes and predicts the interactions among heart cells and the immune system post-MI in the absence of medical interventions. The resulting system of nonlinear ordinary differential equations is studied both analytically and numerically in order to demonstrate the functionality and performance of the new model. To the best of our knowledge, this model is the only one of its kind to consider and correctly apply all of the known factors in diseased heart LV modeling. This model has the potential to provide researchers with a predictive computational tool to better understand the MI pathology and develop various cell-based treatment options, with benefits of lowering the cost and reducing the development time.
Collapse
Affiliation(s)
- Mehtap Lafci Büyükkahraman
- Department of Mathematics, The University of Texas at Arlington, Arlington, TX, USA.,Department of Mathematics, Uşak University, Uşak, Turkey
| | - Gavin K Sabine
- Department of Mathematics, The University of Texas at Arlington, Arlington, TX, USA
| | - Hristo V Kojouharov
- Department of Mathematics, The University of Texas at Arlington, Arlington, TX, USA
| | | | - Sara R McMahan
- Department of Bioengineering, The University of Texas at Arlington, Arlington, TX, USA
| | - Jun Liao
- Department of Bioengineering, The University of Texas at Arlington, Arlington, TX, USA
| |
Collapse
|
77
|
Brown JL, Lawrence MM, Borowik A, Oliver L, Peelor FF, Van Remmen H, Miller BF. Tumor burden negatively impacts protein turnover as a proteostatic process in noncancerous liver, heart, and muscle, but not brain. J Appl Physiol (1985) 2021; 131:72-82. [PMID: 34013745 PMCID: PMC8325617 DOI: 10.1152/japplphysiol.01026.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 04/28/2021] [Accepted: 05/17/2021] [Indexed: 12/24/2022] Open
Abstract
Cancer survivors are more susceptible to pathologies such as hypertension, liver disease, depression, and coronary artery disease when compared with individuals who have never been diagnosed with cancer. Therefore, it is important to understand how tumor burden negatively impacts nontumor-bearing tissues that may impact future disease susceptibility. We hypothesized that the energetic costs of a tumor would compromise proteostatic maintenance in other tissues. Therefore, the purpose of this study was to determine if tumor burden changes protein synthesis and proliferation rates in heart, brain, and liver. One million Lewis lung carcinoma (LLC) cells or phosphate-buffered saline (PBS, sham) were injected into the hind flank of female mice at ∼4.5 mo of age, and the tumor developed for 3 wk. Rates of proliferation and protein synthesis were measured in heart, brain, liver, and tumor tissue. Compared with sham, rates of protein synthesis (structural/nuclear, cytosolic, mitochondrial, and collagen) relative to proliferation were lower in the heart and liver of LLC mice, but higher in the brain of LLC mice. In the tumor tissue, the ratio of protein synthesis to DNA synthesis was approximately 1.0 showing that protein synthesis in the tumor was used for proliferation with little proteostatic maintenance. We further provide evidence that the differences in tissue responses may be due to energetic stress. We concluded that the decrease in proteostatic maintenance in liver, heart, and muscle might contribute to the increased risk of disease in cancer survivors.NEW & NOTEWORTHY We present data showing that simultaneously measuring protein synthesis and cell proliferation can help in the understanding of protein turnover as a proteostatic process in response to tumor burden. In some tissues, like hepatic, cardiac, and skeletal muscle, there was a decrease in the protein to DNA synthesis ratio indicating less proteostatic maintenance. In contrast, the brain maintained or even increased this protein to DNA synthesis ratio indicating more proteostatic maintenance.
Collapse
Affiliation(s)
- Jacob L Brown
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Marcus M Lawrence
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
- Department of Kinesiology and Outdoor Recreation, Southern Utah University, Cedar City, Utah
| | - Agnieszka Borowik
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Lauren Oliver
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
- Oklahoma University Health Science Center, Oklahoma City, Oklahoma
| | - Fredrick F Peelor
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Holly Van Remmen
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
- Oklahoma City VA Medical Center, Oklahoma City, Oklahoma
| | - Benjamin F Miller
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| |
Collapse
|
78
|
Kim H, Wang M, Paik DT. Endothelial-Myocardial Angiocrine Signaling in Heart Development. Front Cell Dev Biol 2021; 9:697130. [PMID: 34277641 PMCID: PMC8281241 DOI: 10.3389/fcell.2021.697130] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 06/10/2021] [Indexed: 12/23/2022] Open
Abstract
Vascular endothelial cells are a multifunctional cell type with organotypic specificity in their function and structure. In this review, we discuss various subpopulations of endothelial cells in the mammalian heart, which spatiotemporally regulate critical cellular and molecular processes of heart development via unique sets of angiocrine signaling pathways. In particular, elucidation of intercellular communication among the functional cell types in the developing heart has recently been accelerated by the use of single-cell sequencing. Specifically, we overview the heterogeneic nature of cardiac endothelial cells and their contribution to heart tube and chamber formation, myocardial trabeculation and compaction, and endocardial cushion and valve formation via angiocrine pathways.
Collapse
Affiliation(s)
- Hyeonyu Kim
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States.,Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Mingqiang Wang
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States.,Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - David T Paik
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States.,Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
79
|
King O, Sunyovszki I, Terracciano CM. Vascularisation of pluripotent stem cell-derived myocardium: biomechanical insights for physiological relevance in cardiac tissue engineering. Pflugers Arch 2021; 473:1117-1136. [PMID: 33855631 PMCID: PMC8245389 DOI: 10.1007/s00424-021-02557-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/15/2021] [Accepted: 03/18/2021] [Indexed: 12/22/2022]
Abstract
The myocardium is a diverse environment, requiring coordination between a variety of specialised cell types. Biochemical crosstalk between cardiomyocytes (CM) and microvascular endothelial cells (MVEC) is essential to maintain contractility and healthy tissue homeostasis. Yet, as myocytes beat, heterocellular communication occurs also through constantly fluctuating biomechanical stimuli, namely (1) compressive and tensile forces generated directly by the beating myocardium, and (2) pulsatile shear stress caused by intra-microvascular flow. Despite endothelial cells (EC) being highly mechanosensitive, the role of biomechanical stimuli from beating CM as a regulatory mode of myocardial-microvascular crosstalk is relatively unexplored. Given that cardiac biomechanics are dramatically altered during disease, and disruption of myocardial-microvascular communication is a known driver of pathological remodelling, understanding the biomechanical context necessary for healthy myocardial-microvascular interaction is of high importance. The current gap in understanding can largely be attributed to technical limitations associated with reproducing dynamic physiological biomechanics in multicellular in vitro platforms, coupled with limited in vitro viability of primary cardiac tissue. However, differentiation of CM from human pluripotent stem cells (hPSC) has provided an unlimited source of human myocytes suitable for designing in vitro models. This technology is now converging with the diverse field of tissue engineering, which utilises in vitro techniques designed to enhance physiological relevance, such as biomimetic extracellular matrix (ECM) as 3D scaffolds, microfluidic perfusion of vascularised networks, and complex multicellular architectures generated via 3D bioprinting. These strategies are now allowing researchers to design in vitro platforms which emulate the cell composition, architectures, and biomechanics specific to the myocardial-microvascular microenvironment. Inclusion of physiological multicellularity and biomechanics may also induce a more mature phenotype in stem cell-derived CM, further enhancing their value. This review aims to highlight the importance of biomechanical stimuli as determinants of CM-EC crosstalk in cardiac health and disease, and to explore emerging tissue engineering and hPSC technologies which can recapitulate physiological dynamics to enhance the value of in vitro cardiac experimentation.
Collapse
Affiliation(s)
- Oisín King
- National Heart & Lung Institute, Imperial College London, Hammersmith Campus, ICTEM 4th floor, Du Cane Road, London, W12 0NN, UK.
| | - Ilona Sunyovszki
- National Heart & Lung Institute, Imperial College London, Hammersmith Campus, ICTEM 4th floor, Du Cane Road, London, W12 0NN, UK
| | - Cesare M Terracciano
- National Heart & Lung Institute, Imperial College London, Hammersmith Campus, ICTEM 4th floor, Du Cane Road, London, W12 0NN, UK
| |
Collapse
|
80
|
Ren Y, Yang X, Ma Z, Sun X, Zhang Y, Li W, Yang H, Qiang L, Yang Z, Liu Y, Deng C, Zhou L, Wang T, Lin J, Li T, Wu T, Wang J. Developments and Opportunities for 3D Bioprinted Organoids. Int J Bioprint 2021; 7:364. [PMID: 34286150 PMCID: PMC8287496 DOI: 10.18063/ijb.v7i3.364] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 05/08/2021] [Indexed: 12/11/2022] Open
Abstract
Organoids developed from pluripotent stem cells or adult stem cells are three-dimensional cell cultures possessing certain key characteristics of their organ counterparts, and they can mimic certain biological developmental processes of organs in vitro. Therefore, they have promising applications in drug screening, disease modeling, and regenerative repair of tissues and organs. However, the construction of organoids currently faces numerous challenges, such as breakthroughs in scale size, vascularization, better reproducibility, and precise architecture in time and space. Recently, the application of bioprinting has accelerated the process of organoid construction. In this review, we present current bioprinting techniques and the application of bioinks and summarize examples of successful organoid bioprinting. In the future, a multidisciplinary combination of developmental biology, disease pathology, cell biology, and materials science will aid in overcoming the obstacles pertaining to the bioprinting of organoids. The combination of bioprinting and organoids with a focus on structure and function can facilitate further development of real organs.
Collapse
Affiliation(s)
- Ya Ren
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital Affiliated Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Rd, Shanghai 200011, China
- Southwest JiaoTong University College of Medicine, No. 111 North 1 Section of Second Ring Road, Chengdu 610036, China
| | - Xue Yang
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital Affiliated Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Rd, Shanghai 200011, China
- Southwest JiaoTong University College of Medicine, No. 111 North 1 Section of Second Ring Road, Chengdu 610036, China
| | - Zhengjiang Ma
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital Affiliated Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Rd, Shanghai 200011, China
| | - Xin Sun
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital Affiliated Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Rd, Shanghai 200011, China
| | - Yuxin Zhang
- Department of Rehabilitation Medicine, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Huangpu District, Shanghai 200011, China
| | - Wentao Li
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital Affiliated Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Rd, Shanghai 200011, China
| | - Han Yang
- Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Rd, Shanghai 200030, China
| | - Lei Qiang
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610036, China
| | - Zezheng Yang
- Department of Orthopedics, The Fifth People’s Hospital of Shanghai, Fudan University, Minhang District, Shanghai 200240, P. R. China
| | - Yihao Liu
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital Affiliated Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Rd, Shanghai 200011, China
| | - Changxu Deng
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital Affiliated Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Rd, Shanghai 200011, China
| | - Liang Zhou
- Center for Medicine Intelligent and Development, China Hospital Development Institute, Shanghai Jiao Tong University, Shanghai 200020, China
| | - Tianchang Wang
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital Affiliated Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Rd, Shanghai 200011, China
| | - Jingsheng Lin
- Department of Information Technology, Ruijin Hospital Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Tao Li
- Department of Orthopaedics, Xinhua Hospital affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, P. R. China
| | - Tao Wu
- Shanghai University of Medicine and Health Sciences, Shanghai 200120, China
| | - Jinwu Wang
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital Affiliated Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Rd, Shanghai 200011, China
- Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Rd, Shanghai 200030, China
| |
Collapse
|
81
|
Liu L, Zhao Q, Lin L, Yang G, Yu L, Zhuo L, Yang Y, Xu Y. Myeloid MKL1 Disseminates Cues to Promote Cardiac Hypertrophy in Mice. Front Cell Dev Biol 2021; 9:583492. [PMID: 33898415 PMCID: PMC8063155 DOI: 10.3389/fcell.2021.583492] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 01/04/2021] [Indexed: 12/11/2022] Open
Abstract
Cardiac hypertrophy is a key pathophysiological process in the heart in response to stress cues. Although taking place in cardiomyocytes, the hypertrophic response is influenced by other cell types, both within the heart and derived from circulation. In the present study we investigated the myeloid-specific role of megakaryocytic leukemia 1 (MKL1) in cardiac hypertrophy. Following transverse aortic constriction (TAC), myeloid MKL1 conditional knockout (MFCKO) mice exhibit an attenuated phenotype of cardiac hypertrophy compared to the WT mice. In accordance, the MFCKO mice were protected from excessive cardiac inflammation and fibrosis as opposed to the WT mice. Conditioned media collected from macrophages enhanced the pro-hypertrophic response in cardiomyocytes exposed to endothelin in an MKL1-dependent manner. Of interest, expression levels of macrophage derived miR-155, known to promote cardiac hypertrophy, were down-regulated in the MFCKO mice compared to the WT mice. MKL1 depletion or inhibition repressed miR-155 expression in macrophages. Mechanistically, MKL1 interacted with NF-κB to activate miR-155 transcription in macrophages. In conclusion, our data suggest that MKL1 may contribute to pathological hypertrophy via regulating macrophage-derived miR-155 transcription.
Collapse
Affiliation(s)
- Li Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China.,Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Qianwen Zhao
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Lin Lin
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Guang Yang
- Department of Pathology, Suzhou Municipal Hospital Affiliated with Nanjing Medical University, Suzhou, China
| | - Liming Yu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Lili Zhuo
- Department of Geriatrics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuyu Yang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China.,Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| | - Yong Xu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China.,Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| |
Collapse
|
82
|
Lee J, Mehrotra S, Zare-Eelanjegh E, Rodrigues RO, Akbarinejad A, Ge D, Amato L, Kiaee K, Fang Y, Rosenkranz A, Keung W, Mandal BB, Li RA, Zhang T, Lee H, Dokmeci MR, Zhang YS, Khademhosseini A, Shin SR. A Heart-Breast Cancer-on-a-Chip Platform for Disease Modeling and Monitoring of Cardiotoxicity Induced by Cancer Chemotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2004258. [PMID: 33094918 PMCID: PMC8049959 DOI: 10.1002/smll.202004258] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/12/2020] [Indexed: 05/02/2023]
Abstract
Cardiotoxicity is one of the most serious side effects of cancer chemotherapy. Current approaches to monitoring of chemotherapy-induced cardiotoxicity (CIC) as well as model systems that develop in vivo or in vitro CIC platforms fail to notice early signs of CIC. Moreover, breast cancer (BC) patients with preexisting cardiac dysfunctions may lead to different incident levels of CIC. Here, a model is presented for investigating CIC where not only induced pluripotent stem cell (iPSC)-derived cardiac tissues are interacted with BC tissues on a dual-organ platform, but electrochemical immuno-aptasensors can also monitor cell-secreted multiple biomarkers. Fibrotic stages of iPSC-derived cardiac tissues are promoted with a supplement of transforming growth factor-β 1 to assess the differential functionality in healthy and fibrotic cardiac tissues after treatment with doxorubicin (DOX). The production trend of biomarkers evaluated by using the immuno-aptasensors well-matches the outcomes from conventional enzyme-linked immunosorbent assay, demonstrating the accuracy of the authors' sensing platform with much higher sensitivity and lower detection limits for early monitoring of CIC and BC progression. Furthermore, the versatility of this platform is demonstrated by applying a nanoparticle-based DOX-delivery system. The proposed platform would potentially help allow early detection and prediction of CIC in individual patients in the future.
Collapse
Affiliation(s)
- Junmin Lee
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| | - Shreya Mehrotra
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Elaheh Zare-Eelanjegh
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Raquel O Rodrigues
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Center for MicroElectromechanical Systems (CMEMS-UMinho), University of Minho, Campus de Azurém, Guimarães, 4800-058, Portugal
| | - Alireza Akbarinejad
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Chemistry, Faculty of Basic Sciences, Tarbiat Modares University, Tehran, 14115-175, Iran
| | - David Ge
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Luca Amato
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Kiavash Kiaee
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Mechanical Engineering, Stevens Institute of Technology, Hoboken, NJ, 07030, USA
| | - YongCong Fang
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Aliza Rosenkranz
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Wendy Keung
- Dr. Li Dak Sum Research Centre, The University of Hong Kong, Pokfulam, Hong Kong
| | - Biman B Mandal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Ronald A Li
- Dr. Li Dak Sum Research Centre, The University of Hong Kong, Pokfulam, Hong Kong
- Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
- Ming Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Shatin, Hong Kong
| | - Ting Zhang
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - HeaYeon Lee
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- MARA Nanotech New York, inc., New York, NY, 10031-9101, USA
| | - Mehmet Remzi Dokmeci
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
- Department of Radiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Ali Khademhosseini
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
- Department of Radiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Chemical and Biomolecular Engineering, Henry Samueli School of Engineering and Applied Sciences, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Su Ryon Shin
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| |
Collapse
|
83
|
Campostrini G, Meraviglia V, Giacomelli E, van Helden RW, Yiangou L, Davis RP, Bellin M, Orlova VV, Mummery CL. Generation, functional analysis and applications of isogenic three-dimensional self-aggregating cardiac microtissues from human pluripotent stem cells. Nat Protoc 2021; 16:2213-2256. [PMID: 33772245 PMCID: PMC7611409 DOI: 10.1038/s41596-021-00497-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 01/11/2021] [Indexed: 02/01/2023]
Abstract
Tissue-like structures from human pluripotent stem cells containing multiple cell types are transforming our ability to model and understand human development and disease. Here we describe a protocol to generate cardiomyocytes (CMs), cardiac fibroblasts (CFs) and cardiac endothelial cells (ECs), the three principal cell types in the heart, from human induced pluripotent stem cells (hiPSCs) and combine them in three-dimensional (3D) cardiac microtissues (MTs). We include details of how to differentiate, isolate, cryopreserve and thaw the component cells and how to construct and analyze the MTs. The protocol supports hiPSC-CM maturation and allows replacement of one or more of the three heart cell types in the MTs with isogenic variants bearing disease mutations. Differentiation of each cell type takes ~30 d, while MT formation and maturation requires another 20 d. No specialist equipment is needed and the method is inexpensive, requiring just 5,000 cells per MT.
Collapse
Affiliation(s)
- Giulia Campostrini
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Viviana Meraviglia
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Elisa Giacomelli
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Ruben W.J. van Helden
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Loukia Yiangou
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Richard P. Davis
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Milena Bellin
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands,Department of Biology, University of Padua, 35121 Padua, Italy,Veneto Institute of Molecular Medicine, 35129 Padua, Italy,Correspondence to , or
| | - Valeria V. Orlova
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands,Correspondence to , or
| | - Christine L. Mummery
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands,Department of Applied Stem Cell Technologies, University of Twente, The Netherlands,Correspondence to , or
| |
Collapse
|
84
|
Guimbal S, Cornuault L, Rouault P, Hollier PL, Chapouly C, Bats ML, Imbault J, Gadeau AP, Couffinhal T, Renault MA. Mast Cells Are the Trigger of Small Vessel Disease and Diastolic Dysfunction in Diabetic Obese Mice. Arterioscler Thromb Vasc Biol 2021; 41:e193-e207. [PMID: 33567863 DOI: 10.1161/atvbaha.121.315900] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
[Figure: see text].
Collapse
MESH Headings
- Animals
- Cell Degranulation
- Cells, Cultured
- Coronary Vessels/immunology
- Coronary Vessels/metabolism
- Coronary Vessels/pathology
- Diabetes Mellitus/genetics
- Diabetes Mellitus/immunology
- Diabetes Mellitus/metabolism
- Diastole
- Disease Models, Animal
- Female
- Heart Failure/etiology
- Heart Failure/immunology
- Heart Failure/metabolism
- Heart Failure/physiopathology
- Histamine Release
- Humans
- Mast Cells/immunology
- Mast Cells/metabolism
- Mast Cells/pathology
- Mice, Knockout
- Myocardium/immunology
- Myocardium/metabolism
- Myocardium/pathology
- Obesity/complications
- Obesity/immunology
- Obesity/metabolism
- Receptors, Leptin/deficiency
- Receptors, Leptin/genetics
- Ventricular Dysfunction, Left/etiology
- Ventricular Dysfunction, Left/immunology
- Ventricular Dysfunction, Left/metabolism
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Function, Left
- Mice
Collapse
Affiliation(s)
- Sarah Guimbal
- University of Bordeaux, Inserm, Biology of Cardiovascular Diseases, U1034, CHU de Bordeaux, Pessac, France
| | - Lauriane Cornuault
- University of Bordeaux, Inserm, Biology of Cardiovascular Diseases, U1034, CHU de Bordeaux, Pessac, France
| | - Paul Rouault
- University of Bordeaux, Inserm, Biology of Cardiovascular Diseases, U1034, CHU de Bordeaux, Pessac, France
| | - Pierre-Louis Hollier
- University of Bordeaux, Inserm, Biology of Cardiovascular Diseases, U1034, CHU de Bordeaux, Pessac, France
| | - Candice Chapouly
- University of Bordeaux, Inserm, Biology of Cardiovascular Diseases, U1034, CHU de Bordeaux, Pessac, France
| | - Marie-Lise Bats
- University of Bordeaux, Inserm, Biology of Cardiovascular Diseases, U1034, CHU de Bordeaux, Pessac, France
| | - Julien Imbault
- University of Bordeaux, Inserm, Biology of Cardiovascular Diseases, U1034, CHU de Bordeaux, Pessac, France
| | - Alain-Pierre Gadeau
- University of Bordeaux, Inserm, Biology of Cardiovascular Diseases, U1034, CHU de Bordeaux, Pessac, France
| | - Thierry Couffinhal
- University of Bordeaux, Inserm, Biology of Cardiovascular Diseases, U1034, CHU de Bordeaux, Pessac, France
| | - Marie-Ange Renault
- University of Bordeaux, Inserm, Biology of Cardiovascular Diseases, U1034, CHU de Bordeaux, Pessac, France
| |
Collapse
|
85
|
Shi M, Zhao F, Sun L, Tang F, Gao W, Xie W, Cao X, Zhuang J, Chen X. Bioactive glass activates VEGF paracrine signaling of cardiomyocytes to promote cardiac angiogenesis. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 124:112077. [PMID: 33947569 DOI: 10.1016/j.msec.2021.112077] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 03/13/2021] [Accepted: 03/20/2021] [Indexed: 12/28/2022]
Abstract
The heart contains a wide range of cell types, which are not isolated but interact with one another via multifarious paracrine, autocrine and endocrine factors. In terms of cardiac angiogenesis, previous studies have proved that regulating the communication between cardiomyocytes and endothelial cells is efficacious to promote capillary formation. Firstly, this study investigated the effect and underlying mechanism of bioactive glass (BG) acted on vascular endothelial growth factor (VEGF) paracrine signaling in cardiomyocytes. We found that bioactive ions released from BG significantly promoted the VEGF production and secretion of cardiomyocytes. Subsequently, we proved that cardiomyocyte-derived VEGF played an important role in mediating the behavior of endothelial cells. Further research showed that the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/hypoxia-inducible factor 1α (HIF-1α) signaling pathway was upregulated by BG, which was involved in VEGF expression of cardiomyocytes. This study revealed that by means of modulating cellular crosstalk via paracrine signaling of host cells in heart is a new direction for the application of BGs in cardiac angiogenesis.
Collapse
Affiliation(s)
- Miao Shi
- School of Medicine, South China University of Technology, Guangzhou 510006, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, PR China; Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China
| | - Fujian Zhao
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China; School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, PR China; Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China
| | - Luyao Sun
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China; School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, PR China; Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China
| | - Fengling Tang
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China; School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, PR China; Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China
| | - Wendong Gao
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China; School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, PR China; Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China
| | - Weihan Xie
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China; School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, PR China; Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China
| | - Xiaodong Cao
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China; School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, PR China; Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China
| | - Jian Zhuang
- School of Medicine, South China University of Technology, Guangzhou 510006, PR China; Guangdong General Hospital, Guangzhou 510080, PR China.
| | - Xiaofeng Chen
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China; School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, PR China; Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China.
| |
Collapse
|
86
|
Chang YC, Mirhaidari G, Kelly J, Breuer C. Current Challenges and Solutions to Tissue Engineering of Large-scale Cardiac Constructs. Curr Cardiol Rep 2021; 23:47. [PMID: 33733317 DOI: 10.1007/s11886-021-01474-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/11/2021] [Indexed: 01/06/2023]
Abstract
PURPOSE OF REVIEW Large-scale tissue engineering of cardiac constructs is a rapidly advancing field; however, there are several barriers still associated with the creation and clinical application of large-scale engineered cardiac tissues. We provide an overview of the current challenges and recently (within the last 5 years) described promising solutions to overcoming said challenges. RECENT FINDINGS The five major criteria yet to be met for clinical application of engineered cardiac tissues are successful electrochemical/mechanical cell coupling, efficient maturation of cardiomyocytes, functional vascularization of large tissues, balancing appropriate immune response, and large-scale generation of constructs. Promising solutions include the use of carbon/graphene in conjunction with existing scaffold designs, utilization of biological hormones, 3D bioprinting, and gene editing. While some of the described barriers to generation of large-scale cardiac tissue have seen encouraging advancements, there is no solution that yet achieves all 5 described criteria. It is vital then to consider a combination of techniques to achieve the optimal construct. Critically, following the demonstration of a viable construct, there remain important considerations to address associated with good manufacturing practices and establishing a standard for clinical trials.
Collapse
Affiliation(s)
- Yu-Chun Chang
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Research Building III, Columbus, OH, 43215, USA.,Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Gabriel Mirhaidari
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Research Building III, Columbus, OH, 43215, USA.,Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, OH, USA
| | - John Kelly
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Research Building III, Columbus, OH, 43215, USA.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA.,The Heart Center, Nationwide Children's Hospital, Columbus, OH, USA
| | - Christopher Breuer
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Research Building III, Columbus, OH, 43215, USA. .,Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA. .,Department of Surgery, Nationwide Children's Hospital, Columbus, OH, USA.
| |
Collapse
|
87
|
James EC, Tomaskovic-Crook E, Crook JM. Bioengineering Clinically Relevant Cardiomyocytes and Cardiac Tissues from Pluripotent Stem Cells. Int J Mol Sci 2021; 22:ijms22063005. [PMID: 33809429 PMCID: PMC8001925 DOI: 10.3390/ijms22063005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 12/23/2022] Open
Abstract
The regenerative capacity of cardiomyocytes is insufficient to functionally recover damaged tissue, and as such, ischaemic heart disease forms the largest proportion of cardiovascular associated deaths. Human-induced pluripotent stem cells (hiPSCs) have enormous potential for developing patient specific cardiomyocytes for modelling heart disease, patient-based cardiac toxicity testing and potentially replacement therapy. However, traditional protocols for hiPSC-derived cardiomyocytes yield mixed populations of atrial, ventricular and nodal-like cells with immature cardiac properties. New insights gleaned from embryonic heart development have progressed the precise production of subtype-specific hiPSC-derived cardiomyocytes; however, their physiological immaturity severely limits their utility as model systems and their use for drug screening and cell therapy. The long-entrenched challenges in this field are being addressed by innovative bioengingeering technologies that incorporate biophysical, biochemical and more recently biomimetic electrical cues, with the latter having the potential to be used to both direct hiPSC differentiation and augment maturation and the function of derived cardiomyocytes and cardiac tissues by mimicking endogenous electric fields.
Collapse
Affiliation(s)
- Emma Claire James
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, Wollongong 2500, Australia;
| | - Eva Tomaskovic-Crook
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, Wollongong 2500, Australia;
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong 2500, Australia
- Correspondence: (E.T.-C.); (J.M.C.)
| | - Jeremy Micah Crook
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, Wollongong 2500, Australia;
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong 2500, Australia
- Department of Surgery, St Vincent’s Hospital, The University of Melbourne, Fitzroy 3065, Australia
- Correspondence: (E.T.-C.); (J.M.C.)
| |
Collapse
|
88
|
Hendrickson T, Mancino C, Whitney L, Tsao C, Rahimi M, Taraballi F. Mimicking cardiac tissue complexity through physical cues: A review on cardiac tissue engineering approaches. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 33:102367. [PMID: 33549819 DOI: 10.1016/j.nano.2021.102367] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 01/06/2021] [Accepted: 01/15/2021] [Indexed: 02/08/2023]
Abstract
Cardiovascular diseases are the number one killer in the world.1,2 Currently, there are no clinical treatments to regenerate damaged cardiac tissue, leaving patients to develop further life-threatening cardiac complications. Cardiac tissue has multiple functional demands including vascularization, contraction, and conduction that require many synergic components to properly work. Most of these functions are a direct result of the cardiac tissue structure and composition, and, for this reason, tissue engineering strongly proposed to develop substitute engineered heart tissues (EHTs). EHTs usually have combined pluripotent stem cells and supporting scaffolds with the final aim to repair or replace the damaged native tissue. However, as simple as this idea is, indeed, it resulted, after many attempts in the field, to be very challenging. Without design complexity, EHTs remain unable to mature fully and integrate into surrounding heart tissue resulting in minimal in vivo effects.3 Lately, there has been a growing body of evidence that a complex, multifunctional approach through implementing scaffold designs, cellularization, and molecular release appears to be essential in the development of a functional cardiac EHTs.4-6 This review covers the advancements in EHTs developments focusing on how to integrate contraction, conduction, and vascularization mimics and how combinations have resulted in improved designs thus warranting further investigation to develop a clinically applicable treatment.
Collapse
Affiliation(s)
- Troy Hendrickson
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston Methodist, Houston, TX, USA; Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA; Texas A&M MD/PhD Program, Texas A&M Health Science Center, College Station, TX, USA
| | - Chiara Mancino
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston Methodist, Houston, TX, USA; Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, (MI), Italy
| | - Lauren Whitney
- Texas A&M Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Chris Tsao
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston Methodist, Houston, TX, USA
| | - Maham Rahimi
- Department of Cardiovascular Surgery, Houston Methodist, Houston, TX, USA
| | - Francesca Taraballi
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston Methodist, Houston, TX, USA; Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA.
| |
Collapse
|
89
|
Dorn LE, Lawrence W, Petrosino JM, Xu X, Hund TJ, Whitson BA, Stratton MS, Janssen PML, Mohler PJ, Schlosser A, Sorensen GL, Accornero F. Microfibrillar-Associated Protein 4 Regulates Stress-Induced Cardiac Remodeling. Circ Res 2021; 128:723-737. [PMID: 33530700 DOI: 10.1161/circresaha.120.317146] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Lisa E Dorn
- Physiology and Cell Biology (L.E.D., W.L., J.M.P., M.S.S., P.M.L.J., P.J.M., F.A.), The Ohio State University Wexner Medical Center, Columbus
| | - William Lawrence
- Physiology and Cell Biology (L.E.D., W.L., J.M.P., M.S.S., P.M.L.J., P.J.M., F.A.), The Ohio State University Wexner Medical Center, Columbus
| | - Jennifer M Petrosino
- Physiology and Cell Biology (L.E.D., W.L., J.M.P., M.S.S., P.M.L.J., P.J.M., F.A.), The Ohio State University Wexner Medical Center, Columbus
| | - Xianyao Xu
- Biomedical Engineering, The Ohio State University, Columbus (X.X., T.J.H.)
| | - Thomas J Hund
- Biomedical Engineering, The Ohio State University, Columbus (X.X., T.J.H.)
| | - Bryan A Whitson
- Bob and Corrine Frick Center for Heart Failure and Arrhythmia (B.A.W., P.J.M.), The Ohio State University Wexner Medical Center, Columbus.,Dorothy M. Davis Heart and Lung Research Institute and Surgery (B.A.W.), The Ohio State University Wexner Medical Center, Columbus
| | - Matthew S Stratton
- Physiology and Cell Biology (L.E.D., W.L., J.M.P., M.S.S., P.M.L.J., P.J.M., F.A.), The Ohio State University Wexner Medical Center, Columbus
| | - Paul M L Janssen
- Physiology and Cell Biology (L.E.D., W.L., J.M.P., M.S.S., P.M.L.J., P.J.M., F.A.), The Ohio State University Wexner Medical Center, Columbus
| | - Peter J Mohler
- Physiology and Cell Biology (L.E.D., W.L., J.M.P., M.S.S., P.M.L.J., P.J.M., F.A.), The Ohio State University Wexner Medical Center, Columbus.,Bob and Corrine Frick Center for Heart Failure and Arrhythmia (B.A.W., P.J.M.), The Ohio State University Wexner Medical Center, Columbus
| | - Anders Schlosser
- Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense (A.S., G.L.S.)
| | - Grith L Sorensen
- Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense (A.S., G.L.S.)
| | - Federica Accornero
- Physiology and Cell Biology (L.E.D., W.L., J.M.P., M.S.S., P.M.L.J., P.J.M., F.A.), The Ohio State University Wexner Medical Center, Columbus
| |
Collapse
|
90
|
Endothelial ERG alleviates cardiac fibrosis via blocking endothelin-1-dependent paracrine mechanism. Cell Biol Toxicol 2021; 37:873-890. [PMID: 33469864 DOI: 10.1007/s10565-021-09581-5] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 01/07/2021] [Indexed: 12/12/2022]
Abstract
Cardiac endothelium communicates closely with adjacent cardiac cells by multiple cytokines and plays critical roles in regulating fibroblasts proliferation, activation, and collagen synthesis during cardiac fibrosis. E26 transformation-specific (ETS)-related gene (ERG) belongs to the ETS transcriptional factor family and is required for endothelial cells (ECs) homeostasis and cardiac development. This study aims at investigating the potential role and molecular basis of ERG in fibrotic remodeling within the adult heart. We observed that ERG was abundant in murine hearts, especially in cardiac ECs, but decreased during cardiac fibrosis. ERG knockdown within murine hearts caused spontaneously cardiac fibrosis and dysfunction, accompanied by the activation of multiple Smad-dependent and independent pathways. However, the direct silence of ERG in cardiac fibroblasts did not affect the expression of fibrotic markers. Intriguingly, ERG knockdown in human umbilical vein endothelial cells (HUVECs) promoted the secretion of endothelin-1 (ET-1), which subsequently accelerated the proliferation, phenotypic transition, and collagen synthesis of cardiac fibroblasts in a paracrine manner. Suppressing ET-1 with either a neutralizing antibody or a receptor blocker abolished ERG knockdown-mediated deleterious effect in vivo and in vitro. This pro-fibrotic effect was also negated by RGD (Arg-Gly-Asp)-peptide magnetic nanoparticles target delivery of ET-1 small interfering RNA to ECs in mice. More importantly, we proved that endothelial ERG overexpression notably prevented pressure overload-induced cardiac fibrosis. Collectively, endothelial ERG alleviates cardiac fibrosis via blocking ET-1-dependent paracrine mechanism and it functions as a candidate for treating cardiac fibrosis. • ERG is abundant in murine hearts, especially in cardiac ECs, but decreased during fibrotic remodeling. • ERG knockdown causes spontaneously cardiac fibrosis and dysfunction. • ERG silence in HUVECs promotes the secretion of endothelin-1, which in turn activates cardiac fibroblasts in a paracrine manner. • Endothelial ERG overexpression prevents pressure overload-induced cardiac fibrosis.
Collapse
|
91
|
Turner A, Aggarwal P, Matter A, Olson B, Gu CC, Hunt SC, Lewis CE, Arnett DK, Lorier R, Broeckel U. Donor-specific phenotypic variation in hiPSC cardiomyocyte-derived exosomes impacts endothelial cell function. Am J Physiol Heart Circ Physiol 2021; 320:H954-H968. [PMID: 33416449 DOI: 10.1152/ajpheart.00463.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Exosomes are an important mechanism of cell-cell interaction in the cardiovascular system, both in maintaining homeostasis and in stress response. Interindividual differences that alter content in exosomes may play a role in cardiovascular disease pathology. To study the effect of interindividual cardiomyocyte (CM) variation, we characterized exosomal content in phenotypically diverse human induced pluripotent stem cell-derived CMs (hiPSC-CMs). Cell lines were generated from six participants in the HyperGEN cohort: three with left ventricular hypertrophy (LVH) and three with normal left ventricular mass (LVM). Sequence analysis of the intracellular and exosomal RNA populations showed distinct expression pattern differences between hiPSC-CM lines derived from individuals with LVH and those with normal LVM. Functional analysis of hiPSC-endothelial cells (hiPSC-ECs) treated with exosomes from both hiPSC-CM groups showed significant variation in response, including differences in tube formation, migration, and proliferation. Overall, treatment of hiPSC-ECs with exosomes resulted in significant expression changes associated with angiogenesis and endothelial cell vasculogenesis. However, the hiPSC-ECs treated with exosomes from the LVH-affected donors exhibited significantly increased proliferation but decreased tube formation and migration, suggesting angiogenic dysregulation.NEW & NOTEWORTHY The intracellular RNA and the miRNA content in exosomes are significantly different in hiPSC-CMs derived from LVH-affected individuals compared with those from unaffected individuals. Treatment of endothelial cells with these exosomes functionally affects cellular phenotypes in a donor-specific manner. These findings provide novel insight into underlying mechanisms of hypertrophic cell signaling between different cell types. With a growing interest in stem cells and exosomes for cardiovascular therapeutic use, this also provides information important for regenerative medicine.
Collapse
Affiliation(s)
- Amy Turner
- Section of Genomic Pediatrics, Department of Pediatrics, Medicine and Physiology, Children's Research Institute and Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Praful Aggarwal
- Section of Genomic Pediatrics, Department of Pediatrics, Medicine and Physiology, Children's Research Institute and Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Andrea Matter
- Section of Genomic Pediatrics, Department of Pediatrics, Medicine and Physiology, Children's Research Institute and Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Benjamin Olson
- Section of Genomic Pediatrics, Department of Pediatrics, Medicine and Physiology, Children's Research Institute and Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Molecular Genetics and Genomics, Washington University, St. Louis, Missouri
| | - C Charles Gu
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri
| | - Steven C Hunt
- Department of Genetic Medicine, Weill Cornell Medicine, Doha, Qatar.,Division of Epidemiology, University of Utah School of Medicine, Salt Lake City, Utah
| | - Cora E Lewis
- Division of Preventive Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Donna K Arnett
- Department of Epidemiology, College of Public Health, University of Kentucky, Lexington, Kentucky
| | - Rachel Lorier
- Section of Genomic Pediatrics, Department of Pediatrics, Medicine and Physiology, Children's Research Institute and Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Ulrich Broeckel
- Section of Genomic Pediatrics, Department of Pediatrics, Medicine and Physiology, Children's Research Institute and Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
92
|
Gladka MM, Kohela A, Molenaar B, Versteeg D, Kooijman L, Monshouwer-Kloots J, Kremer V, Vos HR, Huibers MMH, Haigh JJ, Huylebroeck D, Boon RA, Giacca M, van Rooij E. Cardiomyocytes stimulate angiogenesis after ischemic injury in a ZEB2-dependent manner. Nat Commun 2021; 12:84. [PMID: 33398012 PMCID: PMC7782784 DOI: 10.1038/s41467-020-20361-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 11/20/2020] [Indexed: 12/25/2022] Open
Abstract
The disruption in blood supply due to myocardial infarction is a critical determinant for infarct size and subsequent deterioration in function. The identification of factors that enhance cardiac repair by the restoration of the vascular network is, therefore, of great significance. Here, we show that the transcription factor Zinc finger E-box-binding homeobox 2 (ZEB2) is increased in stressed cardiomyocytes and induces a cardioprotective cross-talk between cardiomyocytes and endothelial cells to enhance angiogenesis after ischemia. Single-cell sequencing indicates ZEB2 to be enriched in injured cardiomyocytes. Cardiomyocyte-specific deletion of ZEB2 results in impaired cardiac contractility and infarct healing post-myocardial infarction (post-MI), while cardiomyocyte-specific ZEB2 overexpression improves cardiomyocyte survival and cardiac function. We identified Thymosin β4 (TMSB4) and Prothymosin α (PTMA) as main paracrine factors released from cardiomyocytes to stimulate angiogenesis by enhancing endothelial cell migration, and whose regulation is validated in our in vivo models. Therapeutic delivery of ZEB2 to cardiomyocytes in the infarcted heart induces the expression of TMSB4 and PTMA, which enhances angiogenesis and prevents cardiac dysfunction. These findings reveal ZEB2 as a beneficial factor during ischemic injury, which may hold promise for the identification of new therapies.
Collapse
Affiliation(s)
- Monika M Gladka
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre, Utrecht, The Netherlands
| | - Arwa Kohela
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre, Utrecht, The Netherlands
| | - Bas Molenaar
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre, Utrecht, The Netherlands
| | - Danielle Versteeg
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre, Utrecht, The Netherlands
- Department of Cardiology, University Medical Center, Utrecht, The Netherlands
| | - Lieneke Kooijman
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre, Utrecht, The Netherlands
| | - Jantine Monshouwer-Kloots
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre, Utrecht, The Netherlands
| | - Veerle Kremer
- Department of Physiology, Amsterdam University Medical Center VU, Amsterdam, The Netherlands
- Department of Medical Biochemistry, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Harmjan R Vos
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center, Utrecht, The Netherlands
| | - Manon M H Huibers
- Department of Pathology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Jody J Haigh
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, Canada
| | - Danny Huylebroeck
- Department of Cell Biology, Erasmus University Medical Centre, Rotterdam, The Netherlands
- Department of Development and Regeneration, University of Leuven, Leuven, Belgium
| | - Reinier A Boon
- Department of Physiology, Amsterdam University Medical Center VU, Amsterdam, The Netherlands
- Institute for Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany
- German Center for Cardiovascular Research (DZHK), Frankfurt am Main, Germany
| | - Mauro Giacca
- School of Cardiovascular Medicine and Sciences, King's College London, London, UK
| | - Eva van Rooij
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre, Utrecht, The Netherlands.
- Department of Cardiology, University Medical Center, Utrecht, The Netherlands.
| |
Collapse
|
93
|
Wei X, Zhuang L, Li H, He C, Wan H, Hu N, Wang P. Advances in Multidimensional Cardiac Biosensing Technologies: From Electrophysiology to Mechanical Motion and Contractile Force. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2005828. [PMID: 33230867 DOI: 10.1002/smll.202005828] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Indexed: 06/11/2023]
Abstract
Cardiovascular disease is currently a leading killer to human, while drug-induced cardiotoxicity remains the main cause of the withdrawal and attrition of drugs. Taking clinical correlation and throughput into account, cardiomyocyte is perfect as in vitro cardiac model for heart disease modeling, drug discovery, and cardiotoxicity assessment by accurately measuring the physiological multiparameters of cardiomyocytes. Remarkably, cardiomyocytes present both electrophysiological and biomechanical characteristics due to the unique excitation-contraction coupling, which plays a significant role in studying the cardiomyocytes. This review mainly focuses on the recent advances of biosensing technologies for the 2D and 3D cardiac models with three special properties: electrophysiology, mechanical motion, and contractile force. These high-performance multidimensional cardiac models are popular and effective to rebuild and mimic the heart in vitro. To help understand the high-quality and accurate physiologies, related detection techniques are highly demanded, from microtechnology to nanotechnology, from extracellular to intracellular recording, from multiple cells to single cell, and from planar to 3D models. Furthermore, the characteristics, advantages, limitations, and applications of these cardiac biosensing technologies, as well as the future development prospects should contribute to the systematization and expansion of knowledge.
Collapse
Affiliation(s)
- Xinwei Wei
- Department of Biomedical Engineering, Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University, Hangzhou, 310027, China
- State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Liujing Zhuang
- Department of Biomedical Engineering, Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University, Hangzhou, 310027, China
- State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Hongbo Li
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou, 510006, China
| | - Chuanjiang He
- Department of Biomedical Engineering, Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University, Hangzhou, 310027, China
| | - Hao Wan
- Department of Biomedical Engineering, Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University, Hangzhou, 310027, China
- State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Ning Hu
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou, 510006, China
| | - Ping Wang
- Department of Biomedical Engineering, Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University, Hangzhou, 310027, China
- State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| |
Collapse
|
94
|
AlBadri A, Wei J, Quesada O, Mehta PK, Xiao Y, Ko YA, Anderson RD, Petersen J, Azarbal B, Samuels B, Henry TD, Cook-Wiens G, Handberg EM, Van Eyk J, Pepine CJ, Merz CNB. Coronary Vascular Function and Cardiomyocyte Injury: A Report From the WISE-CVD. Arterioscler Thromb Vasc Biol 2020; 40:3015-3021. [PMID: 33028098 PMCID: PMC8079158 DOI: 10.1161/atvbaha.120.314260] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 09/14/2020] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Women with symptoms or signs of myocardial ischemia but no obstructive coronary artery disease (INOCA) often have coronary vascular dysfunction and elevated risk for adverse cardiovascular events. We hypothesized that u-hscTnI (ultra-high-sensitivity cardiac troponin I), a sensitive indicator of ischemic cardiomyocyte injury, is associated with coronary vascular dysfunction in women with INOCA. Approach and Results: Women (N=263) with INOCA enrolled in the WISE-CVD study (Women's Ischemic Syndrome Evaluation-Coronary Vascular Dysfunction) underwent invasive coronary vascular function testing and u-hscTnI measurements (Simoa HD-1 Analyzer; Quanterix Corporation, Lexington, MA). Logistic regression models, adjusted for traditional cardiovascular risk factors were used to evaluate associations between u-hscTnI and coronary vascular function. Women with coronary vascular dysfunction (microvascular constriction and limited coronary epicardial dilation) had higher plasma u-hscTnI levels (both P=0.001). u-hscTnI levels were associated with microvascular constriction (odds ratio, 1.38 per doubling of u-hscTnI [95% CI, 1.03-1.84]; P=0.033) and limited coronary epicardial dilation (odds ratio, 1.37 per doubling of u-hscTnI [95% CI, 1.04-1.81]; P=0.026). u-hscTnI levels were not associated with microvascular dilation or coronary epicardial constriction. CONCLUSIONS Our findings indicate that higher u-hscTnI is associated with coronary vascular dysfunction in women with INOCA. This suggests that ischemic cardiomyocyte injury in the setting of coronary vascular dysfunction has the potential to contribute to adverse cardiovascular outcomes observed in these women. Additional studies are needed to confirm and investigate mechanisms underlying these findings in INOCA. Registration: URL: https://www.clinicaltrials.gov; Unique identifier: NCT00832702.
Collapse
Affiliation(s)
- Ahmed AlBadri
- Emory Clinical Cardiovascular Research Institute, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Janet Wei
- Barbra Streisand Women’s Heart Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA
| | - Odayme Quesada
- Barbra Streisand Women’s Heart Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA
| | - Puja K. Mehta
- Emory Clinical Cardiovascular Research Institute, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Yi Xiao
- Emory Clinical Cardiovascular Research Institute, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Yi-An Ko
- Emory Clinical Cardiovascular Research Institute, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - R. David Anderson
- Division of Cardiovascular Medicine, University of Florida College of Medicine, Gainesville, FL
| | - John Petersen
- Division of Cardiovascular Medicine, University of Florida College of Medicine, Gainesville, FL
| | - Babak Azarbal
- Barbra Streisand Women’s Heart Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA
| | - Bruce Samuels
- Barbra Streisand Women’s Heart Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA
| | - Timothy D. Henry
- Barbra Streisand Women’s Heart Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA
| | - Galen Cook-Wiens
- Biostatistics and Bioinformatics Research Center, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Eileen M. Handberg
- Division of Cardiovascular Medicine, University of Florida College of Medicine, Gainesville, FL
| | - Jennifer Van Eyk
- Barbra Streisand Women’s Heart Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA
| | - Carl J. Pepine
- Division of Cardiovascular Medicine, University of Florida College of Medicine, Gainesville, FL
| | - C. Noel Bairey Merz
- Barbra Streisand Women’s Heart Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA
| |
Collapse
|
95
|
Chandika P, Heo SY, Kim TH, Oh GW, Kim GH, Kim MS, Jung WK. Recent advances in biological macromolecule based tissue-engineered composite scaffolds for cardiac tissue regeneration applications. Int J Biol Macromol 2020; 164:2329-2357. [DOI: 10.1016/j.ijbiomac.2020.08.054] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/01/2020] [Accepted: 08/06/2020] [Indexed: 12/11/2022]
|
96
|
Camici PG, Tschöpe C, Di Carli MF, Rimoldi O, Van Linthout S. Coronary microvascular dysfunction in hypertrophy and heart failure. Cardiovasc Res 2020; 116:806-816. [PMID: 31999329 DOI: 10.1093/cvr/cvaa023] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 12/05/2019] [Accepted: 01/23/2020] [Indexed: 12/12/2022] Open
Abstract
Left ventricular (LV) hypertrophy (LVH) is a growth in left myocardial mass mainly caused by increased cardiomyocyte size. LVH can be a physiological adaptation to physical exercise or a pathological condition either primary, i.e. genetic, or secondary to LV overload. Patients with both primary and secondary LVH have evidence of coronary microvascular dysfunction (CMD). The latter is mainly due to capillary rarefaction and adverse remodelling of intramural coronary arterioles due to medial wall thickening with an increased wall/lumen ratio. An important feature of this phenomenon is the diffuse nature of this remodelling, which generally affects the coronary microvessels in the whole of the left ventricle. Patients with LVH secondary to arterial hypertension can develop both heart failure with preserved ejection fraction (HFpEF) and heart failure with reduced ejection fraction (HFrEF). These patients can develop HFrEF via a 'direct pathway' with an interval myocardial infarction and also in its absence. On the other hand, patients can develop HFpEF that can then progress to HFrEF with or without interval myocardial infarction. A similar evolution towards LV dysfunction and both HFpEF and HFrEF can occur in patients with hypertrophic cardiomyopathy, the most common genetic cardiomyopathy with a phenotype characterized by massive LVH. In this review article, we will discuss both the experimental and clinical studies explaining the mechanisms responsible for CMD in LVH as well as the evidence linking CMD with HFpEF and HFrEF.
Collapse
Affiliation(s)
- Paolo G Camici
- Vita Salute University and San Raffaele Hospital, Milano, Italy
| | - Carsten Tschöpe
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany.,Department of Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
| | - Marcelo F Di Carli
- Cardiovascular Imaging Program, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ornella Rimoldi
- Vita Salute University and San Raffaele Hospital, Milano, Italy.,CNR IBFM, Segrate, Italy
| | - Sophie Van Linthout
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| |
Collapse
|
97
|
Mota C, Camarero-Espinosa S, Baker MB, Wieringa P, Moroni L. Bioprinting: From Tissue and Organ Development to in Vitro Models. Chem Rev 2020; 120:10547-10607. [PMID: 32407108 PMCID: PMC7564098 DOI: 10.1021/acs.chemrev.9b00789] [Citation(s) in RCA: 186] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Indexed: 02/08/2023]
Abstract
Bioprinting techniques have been flourishing in the field of biofabrication with pronounced and exponential developments in the past years. Novel biomaterial inks used for the formation of bioinks have been developed, allowing the manufacturing of in vitro models and implants tested preclinically with a certain degree of success. Furthermore, incredible advances in cell biology, namely, in pluripotent stem cells, have also contributed to the latest milestones where more relevant tissues or organ-like constructs with a certain degree of functionality can already be obtained. These incredible strides have been possible with a multitude of multidisciplinary teams around the world, working to make bioprinted tissues and organs more relevant and functional. Yet, there is still a long way to go until these biofabricated constructs will be able to reach the clinics. In this review, we summarize the main bioprinting activities linking them to tissue and organ development and physiology. Most bioprinting approaches focus on mimicking fully matured tissues. Future bioprinting strategies might pursue earlier developmental stages of tissues and organs. The continuous convergence of the experts in the fields of material sciences, cell biology, engineering, and many other disciplines will gradually allow us to overcome the barriers identified on the demanding path toward manufacturing and adoption of tissue and organ replacements.
Collapse
Affiliation(s)
- Carlos Mota
- Department of Complex Tissue Regeneration,
MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Sandra Camarero-Espinosa
- Department of Complex Tissue Regeneration,
MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Matthew B. Baker
- Department of Complex Tissue Regeneration,
MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Paul Wieringa
- Department of Complex Tissue Regeneration,
MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Lorenzo Moroni
- Department of Complex Tissue Regeneration,
MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6211 LK Maastricht, The Netherlands
| |
Collapse
|
98
|
An Investigation of Fibulin-2 in Hypertrophic Cardiomyopathy. Int J Mol Sci 2020; 21:ijms21197176. [PMID: 33003281 PMCID: PMC7583916 DOI: 10.3390/ijms21197176] [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: 09/01/2020] [Revised: 09/20/2020] [Accepted: 09/22/2020] [Indexed: 11/17/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common inherited heart muscle disease, with a prevalence of at least 1 in 500 in the general population. The disease is pleiotropic and is characterized by an increased stiffness of the myocardium, partly due to changes in the extracellular matrix (ECM), with elevated levels of interstitial fibrosis. Myocardial fibrosis is linked to impaired diastolic function and possibly phenotypic heterogeneity of HCM. The ECM consists of a very large number of proteins, which actively interact with each other as well as with myocardial cells. The role of other multiple components of the ECM in HCM has not been defined. Fibulin-2 is a glycoprotein component of the ECM, which plays an important role during embryogenesis of the heart; however, its role in adult myocardium has not been adequately studied. We here describe, for the first time, abnormal expression of fibulin-2 in the myocardium in patients with HCM as compared to normal controls. This abnormal expression was localized in the cytoplasm of myocardial cells and in the interstitial fibroblasts. In addition, fibulin-2 levels, measured by ELISA, were significantly elevated in the serum of patients with HCM as compared to normal controls.
Collapse
|
99
|
Shan D, Guo S, Wu HK, Lv F, Jin L, Zhang M, Xie P, Wang Y, Song Y, Wu F, Lan F, Hu X, Cao CM, Zhang Y, Xiao RP. Cardiac Ischemic Preconditioning Promotes MG53 Secretion Through H 2O 2-Activated Protein Kinase C-δ Signaling. Circulation 2020; 142:1077-1091. [PMID: 32677469 DOI: 10.1161/circulationaha.119.044998] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 06/16/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Ischemic heart disease is the leading cause of morbidity and mortality worldwide. Ischemic preconditioning (IPC) is the most powerful intrinsic protection against cardiac ischemia/reperfusion injury. Previous studies have shown that a multifunctional TRIM family protein, MG53 (mitsugumin 53; also called TRIM72), not only plays an essential role in IPC-mediated cardioprotection against ischemia/reperfusion injury but also ameliorates mechanical damage. In addition to its intracellular actions, as a myokine/cardiokine, MG53 can be secreted from the heart and skeletal muscle in response to metabolic stress. However, it is unknown whether IPC-mediated cardioprotection is causally related to MG53 secretion and, if so, what the underlying mechanism is. METHODS Using proteomic analysis in conjunction with genetic and pharmacological approaches, we examined MG53 secretion in response to IPC and explored the underlying mechanism using rodents in in vivo, isolated perfused hearts, and cultured neonatal rat ventricular cardiomyocytes. Moreover, using recombinant MG53 proteins, we investigated the potential biological function of secreted MG53 in the context of IPC and ischemia/reperfusion injury. RESULTS We found that IPC triggered robust MG53 secretion in rodents in vivo, perfused hearts, and cultured cardiac myocytes without causing cell membrane leakage. Mechanistically, IPC promoted MG53 secretion through H2O2-evoked activation of protein kinase-C-δ. Specifically, IPC-induced myocardial MG53 secretion was mediated by H2O2-triggered phosphorylation of protein kinase-C-δ at Y311, which is necessary and sufficient to facilitate MG53 secretion. Functionally, systemic delivery of recombinant MG53 proteins to mimic elevated circulating MG53 not only restored IPC function in MG53-deficient mice but also protected rodent hearts from ischemia/reperfusion injury even in the absence of IPC. Moreover, oxidative stress by H2O2 augmented MG53 secretion, and MG53 knockdown exacerbated H2O2-induced cell injury in human embryonic stem cell-derived cardiomyocytes, despite relatively low basal expression of MG53 in human heart. CONCLUSIONS We conclude that IPC and oxidative stress can trigger MG53 secretion from the heart via an H2O2-protein kinase-C-δ-dependent mechanism and that extracellular MG53 can participate in IPC protection against cardiac ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Dan Shan
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine (D.S., S.G., H.-K.W., F. Lv, L.J., M.Z., P.X., Y.W., Y.S., X.H., C.-M.C., Y.Z., R.-P.X.), Peking University, China
| | - Sile Guo
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine (D.S., S.G., H.-K.W., F. Lv, L.J., M.Z., P.X., Y.W., Y.S., X.H., C.-M.C., Y.Z., R.-P.X.), Peking University, China
| | - Hong-Kun Wu
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine (D.S., S.G., H.-K.W., F. Lv, L.J., M.Z., P.X., Y.W., Y.S., X.H., C.-M.C., Y.Z., R.-P.X.), Peking University, China
| | - Fengxiang Lv
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine (D.S., S.G., H.-K.W., F. Lv, L.J., M.Z., P.X., Y.W., Y.S., X.H., C.-M.C., Y.Z., R.-P.X.), Peking University, China
| | - Li Jin
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine (D.S., S.G., H.-K.W., F. Lv, L.J., M.Z., P.X., Y.W., Y.S., X.H., C.-M.C., Y.Z., R.-P.X.), Peking University, China
| | - Mao Zhang
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine (D.S., S.G., H.-K.W., F. Lv, L.J., M.Z., P.X., Y.W., Y.S., X.H., C.-M.C., Y.Z., R.-P.X.), Peking University, China
| | - Peng Xie
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine (D.S., S.G., H.-K.W., F. Lv, L.J., M.Z., P.X., Y.W., Y.S., X.H., C.-M.C., Y.Z., R.-P.X.), Peking University, China
| | - Yimei Wang
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine (D.S., S.G., H.-K.W., F. Lv, L.J., M.Z., P.X., Y.W., Y.S., X.H., C.-M.C., Y.Z., R.-P.X.), Peking University, China
| | - Ying Song
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine (D.S., S.G., H.-K.W., F. Lv, L.J., M.Z., P.X., Y.W., Y.S., X.H., C.-M.C., Y.Z., R.-P.X.), Peking University, China
| | - Fujian Wu
- Beijing Laboratory for Cardiovascular Precision Medicine, The Key Laboratory of Remodeling-Related Cardiovascular Disease, Ministry of Education, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Anzhen Hospital, Capital Medical University, China (F.W., F. Lan)
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, China (F.W., F. Lan)
| | - Feng Lan
- Beijing Laboratory for Cardiovascular Precision Medicine, The Key Laboratory of Remodeling-Related Cardiovascular Disease, Ministry of Education, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Anzhen Hospital, Capital Medical University, China (F.W., F. Lan)
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, China (F.W., F. Lan)
| | - Xinli Hu
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine (D.S., S.G., H.-K.W., F. Lv, L.J., M.Z., P.X., Y.W., Y.S., X.H., C.-M.C., Y.Z., R.-P.X.), Peking University, China
| | - Chun-Mei Cao
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine (D.S., S.G., H.-K.W., F. Lv, L.J., M.Z., P.X., Y.W., Y.S., X.H., C.-M.C., Y.Z., R.-P.X.), Peking University, China
| | - Yan Zhang
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine (D.S., S.G., H.-K.W., F. Lv, L.J., M.Z., P.X., Y.W., Y.S., X.H., C.-M.C., Y.Z., R.-P.X.), Peking University, China
| | - Rui-Ping Xiao
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine (D.S., S.G., H.-K.W., F. Lv, L.J., M.Z., P.X., Y.W., Y.S., X.H., C.-M.C., Y.Z., R.-P.X.), Peking University, China
- Beijing City Key Laboratory of Cardiometabolic Molecular Medicine (R.-P.X.), Peking University, China
- Peking-Tsinghua Center for Life Sciences, Beijing, China (R.-P.X.)
| |
Collapse
|
100
|
Wang T, Maltez MT, Lee HW, Ahmad M, Wang HW, Leenen FHH. Effect of exercise training on the FNDC5/BDNF pathway in spontaneously hypertensive rats. Physiol Rep 2020; 7:e14323. [PMID: 31883222 PMCID: PMC6934876 DOI: 10.14814/phy2.14323] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Increased sympathetic activity contributes to the development of cardiovascular diseases such as hypertension. Exercise training lowers sympathetic activity and is beneficial for the prevention and treatment of hypertension and associated cognitive impairment. Increased BDNF expression in skeletal muscle, heart, and brain may contribute to these actions of exercise, but the mechanisms by which this occurs are unknown. We postulated that hypertension is associated with decreased hippocampal BDNF, which can be restored by exercise‐mediated upregulation of fibronectin type‐II domain‐containing 5 (FNDC5). Spontaneously hypertensive rats (SHR) and normotensive Wistar–Kyoto rats (WKY) were subjected to 5 weeks of motorized treadmill training. BDNF and FNDC5 expressions were measured in the left ventricle (LV), quadriceps, soleus muscle, and brain areas. Exercise training reduced blood pressure (BP) in both strains. BDNF and FNDC5 protein in the LV were increased in SHR, but exercise increased only BDNF protein in both strains. BDNF mRNA, but not protein, was increased in the quadriceps of SHR, and BDNF mRNA and protein were decreased by exercise in both groups. FNDC5 protein was higher in SHR in both the quadriceps and soleus muscle, whereas exercise increased FNDC5 protein only in the quadriceps in both strains. BDNF mRNA was lower in the dentate gyrus (DG) of SHR, which was normalized by exercise. BDNF mRNA expression in the DG negatively correlated with BP. No differences in FNDC5 expression were observed in the brain, suggesting that enhanced BDNF signaling may contribute to the cardiovascular and neurological benefits of exercise training, and these processes involve peripheral, but not central, FNDC5.
Collapse
Affiliation(s)
- Tao Wang
- Brain and Heart Research Group, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Melissa T Maltez
- Brain and Heart Research Group, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Heow Won Lee
- Brain and Heart Research Group, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Monir Ahmad
- Brain and Heart Research Group, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Hong-Wei Wang
- Brain and Heart Research Group, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Frans H H Leenen
- Brain and Heart Research Group, University of Ottawa Heart Institute, Ottawa, ON, Canada
| |
Collapse
|