151
|
Raucci FJ, Singh AP, Soslow J, Markham LW, Zhong L, Aljafar W, Lessiohadi N, Awgulewitsch CP, Umbarkar P, Zhang Q, Cannon PL, Buchowski M, Roland JT, Carrier EJ, Burnette WB, Hatzopoulos AK, Lal H, Galindo CL. The BDNF rs6265 Polymorphism is a Modifier of Cardiomyocyte Contractility and Dilated Cardiomyopathy. Int J Mol Sci 2020; 21:E7466. [PMID: 33050457 PMCID: PMC7593910 DOI: 10.3390/ijms21207466] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 09/25/2020] [Accepted: 10/08/2020] [Indexed: 12/22/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is a neuronal growth and survival factor that harbors cardioprotective qualities that may attenuate dilated cardiomyopathy. In ~30% of the population, BDNF has a common, nonsynonymous single nucleotide polymorphism rs6265 (Val66Met), which might be correlated with increased risk of cardiovascular events. We previously showed that BDNF correlates with better cardiac function in Duchenne muscular dystrophy (DMD) patients. However, the effect of the Val66Met polymorphism on cardiac function has not been determined. The goal of the current study was to determine the effects of rs6265 on BDNF biomarker suitability and DMD cardiac functions more generally. We assessed cardiovascular and skeletal muscle function in human DMD patients segregated by polymorphic allele. We also compared echocardiographic, electrophysiologic, and cardiomyocyte contractility in C57/BL-6 wild-type mice with rs6265 polymorphism and in mdx/mTR (mDMD) mouse model of DMD. In human DMD patients, plasma BDNF levels had a positive correlation with left ventricular function, opposite to that seen in rs6265 carriers. There was also a substantial decrease in skeletal muscle function in carriers compared to the Val homozygotes. Surprisingly, the opposite was true when cardiac function of DMD carriers and non-carriers were compared. On the other hand, Val66Met wild-type mice had only subtle functional differences at baseline but significantly decreased cardiomyocyte contractility. Our results indicate that the Val66Met polymorphism alters myocyte contractility, conferring worse skeletal muscle function but better cardiac function in DMD patients. Moreover, these results suggest a mechanism for the relative preservation of cardiac tissues compared to skeletal muscle in DMD patients and underscores the complexity of BDNF signaling in response to mechanical workload.
Collapse
Affiliation(s)
- Frank J. Raucci
- Thomas P. Graham Division of Pediatric Cardiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (F.J.R.J.); (J.S.)
- Division of Pediatric Cardiology, Department of Pediatrics, Children’s Hospital of Richmond at Virginia Commonwealth University, Richmond, VA 23219, USA
| | - Anand Prakash Singh
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama Birmingham School of Medicine, Birmingham, AL 35233, USA; (A.P.S.); (P.U.); (Q.Z.); (H.L.)
| | - Jonathan Soslow
- Thomas P. Graham Division of Pediatric Cardiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (F.J.R.J.); (J.S.)
| | - Larry W. Markham
- Division of Cardiology, Department of Pediatrics, Riley Hospital for Children at Indiana University Health, Indianapolis, IN 46202, USA;
| | - Lin Zhong
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (L.Z.); (W.A.); (N.L.); (C.P.A.); (P.L.C.); (J.T.R.); (E.J.C.); (A.K.H.)
| | - Wejdan Aljafar
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (L.Z.); (W.A.); (N.L.); (C.P.A.); (P.L.C.); (J.T.R.); (E.J.C.); (A.K.H.)
| | - Natasja Lessiohadi
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (L.Z.); (W.A.); (N.L.); (C.P.A.); (P.L.C.); (J.T.R.); (E.J.C.); (A.K.H.)
| | - Cassandra P. Awgulewitsch
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (L.Z.); (W.A.); (N.L.); (C.P.A.); (P.L.C.); (J.T.R.); (E.J.C.); (A.K.H.)
| | - Prachi Umbarkar
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama Birmingham School of Medicine, Birmingham, AL 35233, USA; (A.P.S.); (P.U.); (Q.Z.); (H.L.)
| | - Qinkun Zhang
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama Birmingham School of Medicine, Birmingham, AL 35233, USA; (A.P.S.); (P.U.); (Q.Z.); (H.L.)
| | - Presley L. Cannon
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (L.Z.); (W.A.); (N.L.); (C.P.A.); (P.L.C.); (J.T.R.); (E.J.C.); (A.K.H.)
| | - Maciej Buchowski
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Joseph T. Roland
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (L.Z.); (W.A.); (N.L.); (C.P.A.); (P.L.C.); (J.T.R.); (E.J.C.); (A.K.H.)
| | - Erica J. Carrier
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (L.Z.); (W.A.); (N.L.); (C.P.A.); (P.L.C.); (J.T.R.); (E.J.C.); (A.K.H.)
| | - William B. Burnette
- Division of Pediatric Neurology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Antonis K. Hatzopoulos
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (L.Z.); (W.A.); (N.L.); (C.P.A.); (P.L.C.); (J.T.R.); (E.J.C.); (A.K.H.)
| | - Hind Lal
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama Birmingham School of Medicine, Birmingham, AL 35233, USA; (A.P.S.); (P.U.); (Q.Z.); (H.L.)
| | - Cristi L. Galindo
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (L.Z.); (W.A.); (N.L.); (C.P.A.); (P.L.C.); (J.T.R.); (E.J.C.); (A.K.H.)
- Department of Biology, Western Kentucky University, Bowling Green, KY 42101, USA
| |
Collapse
|
152
|
Pettinato AM, Ladha FA, Mellert DJ, Legere N, Cohn R, Romano R, Thakar K, Chen YS, Hinson JT. Development of a Cardiac Sarcomere Functional Genomics Platform to Enable Scalable Interrogation of Human TNNT2 Variants. Circulation 2020; 142:2262-2275. [PMID: 33025817 DOI: 10.1161/circulationaha.120.047999] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Pathogenic TNNT2 variants are a cause of hypertrophic and dilated cardiomyopathies, which promote heart failure by incompletely understood mechanisms. The precise functional significance for 87% of TNNT2 variants remains undetermined, in part, because of a lack of functional genomics studies. The knowledge of which and how TNNT2 variants cause hypertrophic and dilated cardiomyopathies could improve heart failure risk determination, treatment efficacy, and therapeutic discovery, and provide new insights into cardiomyopathy pathogenesis, as well. METHODS We created a toolkit of human induced pluripotent stem cell models and functional assays using CRISPR/Cas9 to study TNNT2 variant pathogenicity and pathophysiology. Using human induced pluripotent stem cell-derived cardiomyocytes in cardiac microtissue and single-cell assays, we functionally interrogated 51 TNNT2 variants, including 30 pathogenic/likely pathogenic variants and 21 variants of uncertain significance. We used RNA sequencing to determine the transcriptomic consequences of pathogenic TNNT2 variants and adapted CRISPR/Cas9 to engineer a transcriptional reporter assay to assist prediction of TNNT2 variant pathogenicity. We also studied variant-specific pathophysiology using a thin filament-directed calcium reporter to monitor changes in myofilament calcium affinity. RESULTS Hypertrophic cardiomyopathy-associated TNNT2 variants caused increased cardiac microtissue contraction, whereas dilated cardiomyopathy-associated variants decreased contraction. TNNT2 variant-dependent changes in sarcomere contractile function induced graded regulation of 101 gene transcripts, including MAPK (mitogen-activated protein kinase) signaling targets, HOPX, and NPPB. We distinguished pathogenic TNNT2 variants from wildtype controls using a sarcomere functional reporter engineered by inserting tdTomato into the endogenous NPPB locus. On the basis of a combination of NPPB reporter activity and cardiac microtissue contraction, our study provides experimental support for the reclassification of 2 pathogenic/likely pathogenic variants and 2 variants of uncertain significance. CONCLUSIONS Our study found that hypertrophic cardiomyopathy-associated TNNT2 variants increased cardiac microtissue contraction, whereas dilated cardiomyopathy-associated variants decreased contraction, both of which paralleled changes in myofilament calcium affinity. Transcriptomic changes, including NPPB levels, directly correlated with sarcomere function and can be used to predict TNNT2 variant pathogenicity.
Collapse
Affiliation(s)
| | - Feria A Ladha
- University of Connecticut Health Center (A.M.P., F.A.L., R.R., J.T.H.)
| | - David J Mellert
- The Jackson Laboratory for Genomic Medicine (D.J.M., N.L., R.C., K.T., Y.-S.C., J.T.H.)
| | - Nicholas Legere
- The Jackson Laboratory for Genomic Medicine (D.J.M., N.L., R.C., K.T., Y.-S.C., J.T.H.)
| | - Rachel Cohn
- The Jackson Laboratory for Genomic Medicine (D.J.M., N.L., R.C., K.T., Y.-S.C., J.T.H.)
| | - Robert Romano
- University of Connecticut Health Center (A.M.P., F.A.L., R.R., J.T.H.)
| | - Ketan Thakar
- The Jackson Laboratory for Genomic Medicine (D.J.M., N.L., R.C., K.T., Y.-S.C., J.T.H.)
| | - Yu-Sheng Chen
- The Jackson Laboratory for Genomic Medicine (D.J.M., N.L., R.C., K.T., Y.-S.C., J.T.H.)
| | - J Travis Hinson
- University of Connecticut Health Center (A.M.P., F.A.L., R.R., J.T.H.).,The Jackson Laboratory for Genomic Medicine (D.J.M., N.L., R.C., K.T., Y.-S.C., J.T.H.).,Calhoun Cardiology Center, UConn Health (J.T.H.), Farmington
| |
Collapse
|
153
|
X-Ray Structural Analysis of Single Adult Cardiomyocytes: Tomographic Imaging and Microdiffraction. Biophys J 2020; 119:1309-1323. [PMID: 32937109 PMCID: PMC7567981 DOI: 10.1016/j.bpj.2020.08.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/06/2020] [Accepted: 08/17/2020] [Indexed: 02/08/2023] Open
Abstract
We present a multiscale imaging approach to characterize the structure of isolated adult murine cardiomyocytes based on a combination of full-field three-dimensional coherent x-ray imaging and scanning x-ray diffraction. Using these modalities, we probe the structure from the molecular to the cellular scale. Holographic projection images on freeze-dried cells have been recorded using highly coherent and divergent x-ray waveguide radiation. Phase retrieval and tomographic reconstruction then yield the three-dimensional electron density distribution with a voxel size below 50 nm. In the reconstruction volume, myofibrils, sarcomeric organization, and mitochondria can be visualized and quantified within a single cell without sectioning. Next, we use microfocusing optics by compound refractive lenses to probe the diffraction signal of the actomyosin lattice. Comparison between recordings of chemically fixed and untreated, living cells indicate that the characteristic lattice distances shrink by ∼10% upon fixation.
Collapse
|
154
|
Veldhuizen J, Cutts J, Brafman DA, Migrino RQ, Nikkhah M. Engineering anisotropic human stem cell-derived three-dimensional cardiac tissue on-a-chip. Biomaterials 2020; 256:120195. [DOI: 10.1016/j.biomaterials.2020.120195] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 05/15/2020] [Accepted: 06/09/2020] [Indexed: 01/02/2023]
|
155
|
Obal D, Wu JC. Induced pluripotent stem cells as a platform to understand patient-specific responses to opioids and anaesthetics. Br J Pharmacol 2020; 177:4581-4594. [PMID: 32767563 PMCID: PMC7520445 DOI: 10.1111/bph.15228] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 07/22/2020] [Accepted: 07/27/2020] [Indexed: 12/26/2022] Open
Abstract
Recent advances in human induced pluripotent stem cell (iPSC) technology may provide unprecedented opportunities to study patient-specific responses to anaesthetics and opioids. In this review, we will (1) examine the advantages and limitations of iPSC technology, (2) summarize studies using iPSCs that have contributed to our current understanding of anaesthetics and opioid action on the cardiovascular system and central nervous system (CNS), and (3) describe how iPSC technology can be used to further develop personalized analgesic and sedative pharmacotherapies with reduced or minimal detrimental cardiovascular effects.
Collapse
Affiliation(s)
- Detlef Obal
- Stanford Cardiovascular InstituteStanford UniversityStanfordCaliforniaUSA
- Department of Anesthesiology, Pain, and Perioperative MedicineStanford UniversityStanfordCaliforniaUSA
- Outcomes Research ConsortiumClevelandOhioUSA
| | - Joseph C. Wu
- Stanford Cardiovascular InstituteStanford UniversityStanfordCaliforniaUSA
- Department of Medicine, Division of Cardiovascular MedicineStanford UniversityStanfordCaliforniaUSA
- Department of RadiologyStanford UniversityStanfordCaliforniaUSA
| |
Collapse
|
156
|
Li W, Han JL, Entcheva E. Syncytium cell growth increases Kir2.1 contribution in human iPSC-cardiomyocytes. Am J Physiol Heart Circ Physiol 2020; 319:H1112-H1122. [PMID: 32986966 PMCID: PMC7789971 DOI: 10.1152/ajpheart.00148.2020] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) enable cardiotoxicity testing and personalized medicine. However, their maturity is of concern, including relatively depolarized resting membrane potential and more spontaneous activity compared with adult cardiomyocytes, implicating low or lacking inward rectifier potassium current (Ik1). Here, protein quantification confirms Kir2.1 expression in hiPSC-CM syncytia, albeit several times lower than in adult heart tissue. We find that hiPSC-CM culture density influences Kir2.1 expression at the mRNA level (potassium inwardly rectifying channel subfamily J member 2) and at the protein level and its associated electrophysiology phenotype. Namely, all-optical cardiac electrophysiology and pharmacological treatments reveal reduction of spontaneous and irregular activity and increase in action potential upstroke in denser cultures. Blocking Ik1-like currents with BaCl2 increased spontaneous frequency and blunted action potential upstrokes during pacing in a dose-dependent manner only in the highest-density cultures, in line with Ik1’s role in regulating the resting membrane potential. Our results emphasize the importance of syncytial growth of hiPSC-CMs for more physiologically relevant phenotype and the power of all-optical electrophysiology to study cardiomyocytes in their multicellular setting. NEW & NOTEWORTHY We identify cell culture density and cell-cell contact as an important factor in determining the expression of a key ion channel at the transcriptional and the protein levels, KCNJ2/Kir2.1, and its contribution to the electrophysiology of human induced pluripotent stem cell-derived cardiomyocytes. Our results indicate that studies on isolated cells, out of tissue context, may underestimate the cellular ion channel properties being characterized.
Collapse
Affiliation(s)
- Weizhen Li
- Department of Biomedical Engineering, George Washington University, Washington, District of Columbia
| | - Julie L Han
- Department of Biomedical Engineering, George Washington University, Washington, District of Columbia
| | - Emilia Entcheva
- Department of Biomedical Engineering, George Washington University, Washington, District of Columbia
| |
Collapse
|
157
|
Ito M, Nomura S, Morita H, Komuro I. Trends and Limitations in the Assessment of the Contractile Properties of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes From Patients With Dilated Cardiomyopathy. Front Cardiovasc Med 2020; 7:154. [PMID: 33102534 PMCID: PMC7494730 DOI: 10.3389/fcvm.2020.00154] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/22/2020] [Indexed: 12/14/2022] Open
Abstract
The application of human induced pluripotent stem cell-derived cardiomyocytes (hiPSCMs) from patients is expected in disease modeling and drug screening in vitro. Dilated cardiomyopathy (DCM) is an intractable disease characterized by the impairment of systolic function and leads to severe heart failure. A number of researchers have focused on disease modeling of DCM and reproduced its pathologic phenotypes in hiPSCMs, but a robust method to evaluate the contractile properties of cardiomyocytes in vitro has not been standardized. In addition, it is unknown whether the throughput of measurements and analyses could be increased sufficiently for compound screening. Here, we reviewed the articles in which the contractile abnormalities of DCM hiPSCMs were recapitulated and assessed the trends and problems in sample preparation and evaluation. We found that single-cell level analysis was ineffective in some cases, and a tissue engineering approach has become dominant recently because of its increased efficiency in reproducing impaired contractility. We also examined two commercially available automated measurement devices with moderate throughput for motion analysis using two-dimensional hiPSCM sheets composed of originally established DCM hiPSCMs. As a result, both of the tested devices, an impedance analyzer and a video image-based cell motion analyzer, were not effective in detecting the expected reduction of contractility in the DCM clone. These findings collectively suggest that a tissue engineering approach could expand the potential of disease modeling with hiPSCMs, and so far, appropriate methods for in vitro force measurement with sufficient throughput, but without sacrificing physiological fidelity, are awaited.
Collapse
Affiliation(s)
- Masamichi Ito
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Seitaro Nomura
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Morita
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
158
|
Lavine KJ, Greenberg MJ. Beyond genomics-technological advances improving the molecular characterization and precision treatment of heart failure. Heart Fail Rev 2020; 26:405-415. [PMID: 32885327 DOI: 10.1007/s10741-020-10021-5] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/25/2020] [Indexed: 01/04/2023]
Abstract
Dilated cardiomyopathy (DCM) is a major cause of heart failure and cardiovascular mortality. In the past 20 years, there has been an overwhelming focus on developing therapeutics that target common downstream disease pathways thought to be involved in all forms of heart failure independent of the initial etiology. While this strategy is effective at the population level, individual responses vary tremendously and only approximately one third of patients receive benefit from modern heart failure treatments. In this perspective, we propose that DCM should be considered as a collection of diseases with a common phenotype of left ventricular dilation and systolic dysfunction rather than a single disease entity, and that mechanism-based classification of disease subtypes will revolutionize our understanding and clinical approach towards DCM. We discuss how these efforts are central to realizing the potential of precision medicine and how they are empowered by the development of new tools that allow investigators to strategically employ genomic and transcriptomic information. Finally, we outline an investigational strategy to (1) define DCM at the patient level, (2) develop new tools to model and mechanistically dissect subtypes of human heart failure, and (3) harness these insights for the development of precision therapeutics.
Collapse
Affiliation(s)
- Kory J Lavine
- Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine, Washington University School of Medicine, 660 S. Euclid Ave., Campus Box 8086, St. Louis, MO, 63110, USA.
| | - Michael J Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, 660 S. Euclid Ave., Campus Box 8231, St. Louis, MO, 63110, USA.
| |
Collapse
|
159
|
Pioner JM, Guan X, Klaiman JM, Racca AW, Pabon L, Muskheli V, Macadangdang J, Ferrantini C, Hoopmann MR, Moritz RL, Kim DH, Tesi C, Poggesi C, Murry CE, Childers MK, Mack DL, Regnier M. Absence of full-length dystrophin impairs normal maturation and contraction of cardiomyocytes derived from human-induced pluripotent stem cells. Cardiovasc Res 2020; 116:368-382. [PMID: 31049579 DOI: 10.1093/cvr/cvz109] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 03/20/2019] [Accepted: 04/17/2019] [Indexed: 12/30/2022] Open
Abstract
AIMS Heart failure invariably affects patients with various forms of muscular dystrophy (MD), but the onset and molecular sequelae of altered structure and function resulting from full-length dystrophin (Dp427) deficiency in MD heart tissue are poorly understood. To better understand the role of dystrophin in cardiomyocyte development and the earliest phase of Duchenne muscular dystrophy (DMD) cardiomyopathy, we studied human cardiomyocytes differentiated from induced pluripotent stem cells (hiPSC-CMs) obtained from the urine of a DMD patient. METHODS AND RESULTS The contractile properties of patient-specific hiPSC-CMs, with no detectable dystrophin (DMD-CMs with a deletion of exon 50), were compared to CMs containing a CRISPR-Cas9 mediated deletion of a single G base at position 263 of the dystrophin gene (c.263delG-CMs) isogenic to the parental line of hiPSC-CMs from a healthy individual. We hypothesized that the absence of a dystrophin-actin linkage would adversely affect myofibril and cardiomyocyte structure and function. Cardiomyocyte maturation was driven by culturing long-term (80-100 days) on a nanopatterned surface, which resulted in hiPSC-CMs with adult-like dimensions and aligned myofibrils. CONCLUSIONS Our data demonstrate that lack of Dp427 results in reduced myofibril contractile tension, slower relaxation kinetics, and to Ca2+ handling abnormalities, similar to DMD cells, suggesting either retarded or altered maturation of cardiomyocyte structures associated with these functions. This study offers new insights into the functional consequences of Dp427 deficiency at an early stage of cardiomyocyte development in both patient-derived and CRISPR-generated models of dystrophin deficiency.
Collapse
Affiliation(s)
- J Manuel Pioner
- Experimental and Clinical Medicine, Div. of Physiology, University of Florence, Florence, Italy
| | - Xuan Guan
- Bioengineering, University of Washington, Seattle, WA, USA
| | | | - Alice W Racca
- School of Biosciences, University of Kent, Canterbury, UK
| | - Lil Pabon
- Pathology, University of Washington, Seattle, WA, USA.,Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, USA
| | - Veronica Muskheli
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA
| | | | - Cecilia Ferrantini
- Experimental and Clinical Medicine, Div. of Physiology, University of Florence, Florence, Italy
| | | | | | - Deok-Ho Kim
- Bioengineering, University of Washington, Seattle, WA, USA.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, USA
| | - Chiara Tesi
- Experimental and Clinical Medicine, Div. of Physiology, University of Florence, Florence, Italy
| | - Corrado Poggesi
- Experimental and Clinical Medicine, Div. of Physiology, University of Florence, Florence, Italy
| | - Charles E Murry
- Bioengineering, University of Washington, Seattle, WA, USA.,Pathology, University of Washington, Seattle, WA, USA.,Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, USA
| | - Martin K Childers
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, USA.,Rehabilitation Medicine, University of Washington, Seattle, WA, USA
| | - David L Mack
- Bioengineering, University of Washington, Seattle, WA, USA.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, USA.,Rehabilitation Medicine, University of Washington, Seattle, WA, USA
| | - Michael Regnier
- Bioengineering, University of Washington, Seattle, WA, USA.,Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, USA
| |
Collapse
|
160
|
Reza N, Musunuru K, Owens AT. From Hypertrophy to Heart Failure: What Is New in Genetic Cardiomyopathies. Curr Heart Fail Rep 2020; 16:157-167. [PMID: 31243690 DOI: 10.1007/s11897-019-00435-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE The purpose of this review is to provide an update on the recent advances in the research and clinical care of patients with the major phenotypes of inherited cardiomyopathies-hypertrophic, dilated, and arrhythmogenic. Developments in genetics, risk stratification, therapies, and disease modeling will be discussed. RECENT Diagnostic, prognostic, and therapeutic tools which incorporate genetic and genomic data are being steadily incorporated into the routine clinical care of patients with genetic cardiomyopathies. Human pluripotent stem cells are a breakthrough model system for the study of genetic variation associated with inherited cardiovascular disease. Next-generation sequencing technology and molecular-based diagnostics and therapeutics have emerged as valuable tools to improve the recognition and care of patients with hypertrophic, dilated, and arrhythmogenic cardiomyopathies. Improved adjudication of variant pathogenicity and management of genotype-positive/phenotype-negative individuals are imminent challenges in this realm of precision medicine.
Collapse
Affiliation(s)
- Nosheen Reza
- Division of Cardiovascular Medicine, Department of Medicine, and Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Cardiovascular Institute, Philadelphia, PA, 19104, USA.
| | - Kiran Musunuru
- Division of Cardiovascular Medicine, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, 11 South Pavilion, Room 11-134, 3400 Civic Center Boulevard, Philadelphia, PA, 19104, USA
| | - Anjali Tiku Owens
- Division of Cardiovascular Medicine, Department of Medicine, and Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Cardiovascular Institute, Philadelphia, PA, 19104, USA
| |
Collapse
|
161
|
Ballan N, Shaheen N, Keller GM, Gepstein L. Single-Cell Mechanical Analysis of Human Pluripotent Stem Cell-Derived Cardiomyocytes for Drug Testing and Pathophysiological Studies. Stem Cell Reports 2020; 15:587-596. [PMID: 32763158 PMCID: PMC7486198 DOI: 10.1016/j.stemcr.2020.07.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 07/08/2020] [Accepted: 07/08/2020] [Indexed: 01/20/2023] Open
Abstract
Current platforms for studying the mechanical properties of human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) as single cells do not measure forces directly, require numerous assumptions, and cannot study cell mechanics at different loading conditions. We present a method for directly measuring the active and passive forces generated by single-cell hPSC-CMs at different stretch levels. Utilizing this technique, single hPSC-CMs exhibited positive length-tension relationship and appropriate inotropic, klinotropic, and lusitropic changes in response to pharmacological treatments (isoproterenol and verapamil). The unique potential of the approach for drug testing and disease modeling was exemplified by doxorubicin and omecamtiv mecarbil drug studies revealing their known actions to suppress (doxorubicin) or augment (omecamtiv mecarbil at low dose) cardiomyocyte contractility, respectively. Finally, mechanistic insights were gained regarding the cellular effects of these drugs as doxorubicin treatment led to cellular mechanical alternans and high doses of omecamtiv mecarbil suppressed contractility and worsened the cellular diastolic properties. A unique approach for evaluating the mechanical properties of single-cell hPSC-CMs Both active and passive forces can be directly measured at different stretch levels The new approach can be used to evaluate drug effects and pathological conditions
Collapse
Affiliation(s)
- Nimer Ballan
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, POB 9649, Haifa 3109601, Israel
| | - Naim Shaheen
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, POB 9649, Haifa 3109601, Israel
| | - Gordon M Keller
- McEwen Stem Cell Institute and Princess Margaret Cancer Center, UHN, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Lior Gepstein
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, POB 9649, Haifa 3109601, Israel; Cardiolology Department, Rambam Health Care Campus, Haifa, Israel.
| |
Collapse
|
162
|
Robinson P, Sparrow AJ, Patel S, Malinowska M, Reilly SN, Zhang YH, Casadei B, Watkins H, Redwood C. Dilated cardiomyopathy mutations in thin-filament regulatory proteins reduce contractility, suppress systolic Ca 2+, and activate NFAT and Akt signaling. Am J Physiol Heart Circ Physiol 2020; 319:H306-H319. [PMID: 32618513 PMCID: PMC7473929 DOI: 10.1152/ajpheart.00272.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Dilated cardiomyopathy (DCM) is clinically characterized by dilated ventricular cavities and reduced ejection fraction, leading to heart failure and increased thromboembolic risk. Mutations in thin-filament regulatory proteins can cause DCM and have been shown in vitro to reduce contractility and myofilament Ca2+-affinity. In this work we have studied the functional consequences of mutations in cardiac troponin T (R131W), cardiac troponin I (K36Q) and α-tropomyosin (E40K) using adenovirally transduced isolated guinea pig left ventricular cardiomyocytes. We find significantly reduced fractional shortening with reduced systolic Ca2+. Contraction and Ca2+ reuptake times were slowed, which contrast with some findings in murine models of myofilament Ca2+ desensitization. We also observe increased sarcoplasmic reticulum (SR) Ca2+ load and smaller fractional SR Ca2+ release. This corresponds to a reduction in SR Ca2+-ATPase activity and increase in sodium-calcium exchanger activity. We also observe dephosphorylation and nuclear translocation of the nuclear factor of activated T cells (NFAT), with concordant RAC-α-serine/threonine protein kinase (Akt) phosphorylation but no change to extracellular signal-regulated kinase activation in chronically paced cardiomyocytes expressing DCM mutations. These changes in Ca2+ handling and signaling are common to all three mutations, indicating an analogous pathway of disease pathogenesis in thin-filament sarcomeric DCM. Previous work has shown that changes to myofilament Ca2+ sensitivity caused by DCM mutations are qualitatively opposite from hypertrophic cardiomyopathy (HCM) mutations in the same genes. However, we find several common pathways such as increased relaxation times and NFAT activation that are also hallmarks of HCM. This suggests more complex intracellular signaling underpinning DCM, driven by the primary mutation.NEW & NOTEWORTHY Dilated cardiomyopathy (DCM) is a frequently occurring cardiac disorder with a degree of genetic inheritance. We have found that DCM mutations in proteins that regulate the contractile machinery cause alterations to contraction, calcium-handling, and some new signaling pathways that provide stimuli for disease development. We have used guinea pig cells that recapitulate human calcium-handling and introduced the mutations using adenovirus gene transduction to look at the initial triggers of disease before remodeling.
Collapse
Affiliation(s)
- Paul Robinson
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
- British Heart Foundation, Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Alexander J Sparrow
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
- British Heart Foundation, Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Suketu Patel
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
- British Heart Foundation, Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Marta Malinowska
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
- British Heart Foundation, Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Svetlana N Reilly
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
- British Heart Foundation, Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Yin-Hua Zhang
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
- British Heart Foundation, Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Barbara Casadei
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
- British Heart Foundation, Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Hugh Watkins
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
- British Heart Foundation, Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Charles Redwood
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
- British Heart Foundation, Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
163
|
Li Q, Wang J, Wu Q, Cao N, Yang HT. Perspective on human pluripotent stem cell-derived cardiomyocytes in heart disease modeling and repair. Stem Cells Transl Med 2020; 9:1121-1128. [PMID: 32725800 PMCID: PMC7519762 DOI: 10.1002/sctm.19-0340] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 03/12/2020] [Accepted: 03/29/2020] [Indexed: 12/20/2022] Open
Abstract
Heart diseases (HDs) are the leading cause of morbidity and mortality worldwide. Despite remarkable clinical progress made, current therapies cannot restore the lost myocardium, and the correlation of genotype to phenotype of many HDs is poorly modeled. In the past two decades, with the rapid developments of human pluripotent stem cell (hPSC) biology and technology that allow the efficient preparation of cardiomyocytes from individual patients, tremendous efforts have been made for using hPSC‐derived cardiomyocytes in preclinical and clinical cardiac therapy as well as in dissection of HD mechanisms to develop new methods for disease prediction and treatment. However, their applications have been hampered by several obstacles. Here, we discuss recent advances, remaining challenges, and the potential solutions to advance this field.
Collapse
Affiliation(s)
- Qiang Li
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (CAS), Shanghai, People's Republic of China.,Institute for Stem Cell and Regeneration, CAS, Beijing, People's Republic of China
| | - Jia Wang
- Program of Stem Cells and Regenerative Medicine, The Fifth Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangdong, People's Republic of China.,Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-Sen University), Ministry of Education, Guangzhou, People's Republic of China
| | - Qiang Wu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (CAS), Shanghai, People's Republic of China.,Institute for Stem Cell and Regeneration, CAS, Beijing, People's Republic of China
| | - Nan Cao
- Program of Stem Cells and Regenerative Medicine, The Fifth Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangdong, People's Republic of China.,Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-Sen University), Ministry of Education, Guangzhou, People's Republic of China
| | - Huang-Tian Yang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (CAS), Shanghai, People's Republic of China.,Institute for Stem Cell and Regeneration, CAS, Beijing, People's Republic of China
| |
Collapse
|
164
|
Gong Y, Chen Z, Yang L, Ai X, Yan B, Wang H, Qiu L, Tan Y, Witman N, Wang W, Zhao Y, Fu W. Intrinsic Color Sensing System Allows for Real-Time Observable Functional Changes on Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes. ACS NANO 2020; 14:8232-8246. [PMID: 32609489 DOI: 10.1021/acsnano.0c01745] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Stem-cell based in vitro differentiation for disease modeling offers great value to explore the molecular and functional underpinnings driving many types of cardiomyopathy and congenital heart diseases. Nevertheless, one major caveat in the application of in vitro differentiation of human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (hiPSC-CMs) involves the immature phenotype of the CMs. Most of the existing methods need complex apparatus and require laborious procedures in order to monitor the cardiac differentiation/maturation process and often result in cell death. Here we developed an intrinsic color sensing system utilizing a microgroove structural color methacrylated gelatin film, which allows us to monitor the cardiac differentiation process of hiPSC-derived cardiac progenitor cells in real time. Subsequently this system can be employed as an assay system to live monitor induced functional changes on hiPSC-CMs stemming from drug treatment, the effects of which are simply revealed through color diversity. Our research shows that early intervention of cardiac differentiation through simple physical cues can enhance cardiac differentiation and maturation to some extent. Our system also simplifies the previous complex experimental processes for evaluating the physiological effects of successful differentiation and drug treatment and lays a solid foundation for future transformational applications.
Collapse
Affiliation(s)
- Yiqi Gong
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China
| | - Zhuoyue Chen
- Department of Clinical Laboratory, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Li Yang
- Department of Anesthesiology, Fudan University Shanghai Cancer Center and Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong An Road, Shanghai 200032, China
| | - Xuefeng Ai
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China
| | - Bingqian Yan
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China
- Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China
| | - Huijing Wang
- Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China
| | - Liya Qiu
- Shanghai Institute of Technical Physics of the Chinese Academy of Sciences, Shanghai 200083, China
| | - Yao Tan
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China
- Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China
| | - Nevin Witman
- Department of Medicine and Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Wei Wang
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China
| | - Yuanjin Zhao
- Department of Clinical Laboratory, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Wei Fu
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China
- Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China
- Shanghai Key Laboratory of Tissue Engineering, Shanghai ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China
| |
Collapse
|
165
|
Filippo Buono M, von Boehmer L, Strang J, P. Hoerstrup S, Y. Emmert M, Nugraha B. Human Cardiac Organoids for Modeling Genetic Cardiomyopathy. Cells 2020; 9:cells9071733. [PMID: 32698471 PMCID: PMC7409052 DOI: 10.3390/cells9071733] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/10/2020] [Accepted: 07/13/2020] [Indexed: 12/12/2022] Open
Abstract
Genetic cardiomyopathies are characterized by changes in the function and structure of the myocardium. The development of a novel in vitro model could help to better emulate healthy and diseased human heart conditions and may improve the understanding of disease mechanisms. In this study, for the first time, we demonstrated the generation of cardiac organoids using a triculture approach of human induced pluripotent stem-cell-derived cardiomyocytes (hiPS-CMs)-from healthy subjects and cardiomyopathy patients-human cardiac microvascular endothelial cells (HCMECs) and human cardiac fibroblasts (HCFs). We assessed the organoids' suitability as a 3D cellular model for the representation of phenotypical features of healthy and cardiomyopathic hearts. We observed clear differences in structure and beating behavior between the organoid groups, depending on the type of hiPS-CMs (healthy versus cardiomyopathic) used. Organoids may thus prove a promising tool for the design and testing of patient-specific treatments as well as provide a platform for safer and more efficacious drug development.
Collapse
Affiliation(s)
- Michele Filippo Buono
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland; (M.F.B.); (L.v.B.); (S.P.H.)
| | - Lisa von Boehmer
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland; (M.F.B.); (L.v.B.); (S.P.H.)
| | - Jaan Strang
- Zurich University of Applied Sciences, 8820 Wädenswil, Switzerland;
| | - Simon P. Hoerstrup
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland; (M.F.B.); (L.v.B.); (S.P.H.)
- Wyss Translational Center Zurich, 8006 Zurich, Switzerland
| | - Maximilian Y. Emmert
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland; (M.F.B.); (L.v.B.); (S.P.H.)
- Wyss Translational Center Zurich, 8006 Zurich, Switzerland
- Department of Cardiovascular Surgery, University Hospital Zurich, 8091 Zurich, Switzerland
- Department of Cardiovascular Surgery, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany
- Correspondence: (M.Y.E.); (B.N.); Tel.: +41-44-634-5610 (M.Y.E.); +41-635-8533 (B.N.)
| | - Bramasta Nugraha
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland; (M.F.B.); (L.v.B.); (S.P.H.)
- Correspondence: (M.Y.E.); (B.N.); Tel.: +41-44-634-5610 (M.Y.E.); +41-635-8533 (B.N.)
| |
Collapse
|
166
|
Sharma A, Garcia G, Wang Y, Plummer JT, Morizono K, Arumugaswami V, Svendsen CN. Human iPSC-Derived Cardiomyocytes Are Susceptible to SARS-CoV-2 Infection. CELL REPORTS MEDICINE 2020; 1:100052. [PMID: 32835305 PMCID: PMC7323681 DOI: 10.1016/j.xcrm.2020.100052] [Citation(s) in RCA: 220] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/27/2020] [Accepted: 06/18/2020] [Indexed: 12/27/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is a pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). COVID-19 is defined by respiratory symptoms, but cardiac complications including viral myocarditis are also prevalent. Although ischemic and inflammatory responses caused by COVID-19 can detrimentally affect cardiac function, the direct impact of SARS-CoV-2 infection on human cardiomyocytes is not well understood. Here, we utilize human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) as a model to examine the mechanisms of cardiomyocyte-specific infection by SARS-CoV-2. Microscopy and RNA sequencing demonstrate that SARS-CoV-2 can enter hiPSC-CMs via ACE2. Viral replication and cytopathic effect induce hiPSC-CM apoptosis and cessation of beating after 72 h of infection. SARS-CoV-2 infection activates innate immune response and antiviral clearance gene pathways, while inhibiting metabolic pathways and suppressing ACE2 expression. These studies show that SARS-CoV-2 can infect hiPSC-CMs in vitro, establishing a model for elucidating infection mechanisms and potentially a cardiac-specific antiviral drug screening platform. Human iPSC-derived cardiomyocytes are susceptible to SARS-CoV-2 infection ACE2 antibody blunts SARS-CoV-2 infection in cardiomyocytes Infected human iPSC-derived cardiomyocytes activate viral clearance pathways
Collapse
Affiliation(s)
- Arun Sharma
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.,Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Gustavo Garcia
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yizhou Wang
- Genomics Core, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jasmine T Plummer
- Genomics Core, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Kouki Morizono
- Division of Hematology and Oncology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.,UCLA AIDS Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Vaithilingaraja Arumugaswami
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Clive N Svendsen
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
167
|
Guo J, Huebsch N. Modeling the Response of Heart Muscle to Mechanical Stimulation In Vitro. ACTA ACUST UNITED AC 2020. [DOI: 10.1007/s43152-020-00007-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
168
|
Mazzola M, Di Pasquale E. Toward Cardiac Regeneration: Combination of Pluripotent Stem Cell-Based Therapies and Bioengineering Strategies. Front Bioeng Biotechnol 2020; 8:455. [PMID: 32528940 PMCID: PMC7266938 DOI: 10.3389/fbioe.2020.00455] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/21/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases represent the major cause of morbidity and mortality worldwide. Multiple studies have been conducted so far in order to develop treatments able to prevent the progression of these pathologies. Despite progress made in the last decade, current therapies are still hampered by poor translation into actual clinical applications. The major drawback of such strategies is represented by the limited regenerative capacity of the cardiac tissue. Indeed, after an ischaemic insult, the formation of fibrotic scar takes place, interfering with mechanical and electrical functions of the heart. Hence, the ability of the heart to recover after ischaemic injury depends on several molecular and cellular pathways, and the imbalance between them results into adverse remodeling, culminating in heart failure. In this complex scenario, a new chapter of regenerative medicine has been opened over the past 20 years with the discovery of induced pluripotent stem cells (iPSCs). These cells share the same characteristic of embryonic stem cells (ESCs), but are generated from patient-specific somatic cells, overcoming the ethical limitations related to ESC use and providing an autologous source of human cells. Similarly to ESCs, iPSCs are able to efficiently differentiate into cardiomyocytes (CMs), and thus hold a real regenerative potential for future clinical applications. However, cell-based therapies are subjected to poor grafting and may cause adverse effects in the failing heart. Thus, over the last years, bioengineering technologies focused their attention on the improvement of both survival and functionality of iPSC-derived CMs. The combination of these two fields of study has burst the development of cell-based three-dimensional (3D) structures and organoids which mimic, more realistically, the in vivo cell behavior. Toward the same path, the possibility to directly induce conversion of fibroblasts into CMs has recently emerged as a promising area for in situ cardiac regeneration. In this review we provide an up-to-date overview of the latest advancements in the application of pluripotent stem cells and tissue-engineering for therapeutically relevant cardiac regenerative approaches, aiming to highlight outcomes, limitations and future perspectives for their clinical translation.
Collapse
Affiliation(s)
- Marta Mazzola
- Stem Cell Unit, Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy
| | - Elisa Di Pasquale
- Stem Cell Unit, Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy.,Institute of Genetic and Biomedical Research (IRGB) - UOS of Milan, National Research Council (CNR), Milan, Italy
| |
Collapse
|
169
|
Gigli M, Merlo M, Graw SL, Barbati G, Rowland TJ, Slavov DB, Stolfo D, Haywood ME, Dal Ferro M, Altinier A, Ramani F, Brun F, Cocciolo A, Puggia I, Morea G, McKenna WJ, La Rosa FG, Taylor MRG, Sinagra G, Mestroni L. Genetic Risk of Arrhythmic Phenotypes in Patients With Dilated Cardiomyopathy. J Am Coll Cardiol 2020; 74:1480-1490. [PMID: 31514951 DOI: 10.1016/j.jacc.2019.06.072] [Citation(s) in RCA: 199] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 06/13/2019] [Accepted: 06/29/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Genotype-phenotype correlations in dilated cardiomyopathy (DCM) and, in particular, the effects of gene variants on clinical outcomes remain poorly understood. OBJECTIVES The purpose of this study was to investigate the prognostic role of genetic variant carrier status in a large cohort of DCM patients. METHODS A total of 487 DCM patients were analyzed by next-generation sequencing and categorized the disease genes into functional gene groups. The following composite outcome measures were assessed: 1) all-cause mortality; 2) heart failure-related death, heart transplantation, or destination left ventricular assist device implantation (DHF/HTx/VAD); and 3) sudden cardiac death/sustained ventricular tachycardia/ventricular fibrillation (SCD/VT/VF). RESULTS A total of 183 pathogenic/likely pathogenic variants were found in 178 patients (37%): 54 (11%) Titin; 19 (4%) Lamin A/C (LMNA); 24 (5%) structural cytoskeleton-Z disk genes; 16 (3.5%) desmosomal genes; 46 (9.5%) sarcomeric genes; 8 (1.6%) ion channel genes; and 11 (2.5%) other genes. All-cause mortality was no different between variant carriers and noncarriers (p = 0.99). A trend toward worse SCD/VT/VF (p = 0.062) and DHF/HTx/VAD (p = 0.061) was found in carriers. Carriers of desmosomal and LMNA variants experienced the highest rate of SCD/VT/VF, which was independent of the left ventricular ejection fraction. CONCLUSIONS Desmosomal and LMNA gene variants identify the subset of DCM patients who are at greatest risk for SCD and life-threatening ventricular arrhythmias, regardless of the left ventricular ejection fraction.
Collapse
Affiliation(s)
- Marta Gigli
- Cardiovascular Department, Azienda Sanitaria-Universitaria Integrata Trieste "ASUITS," Trieste, Italy; Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Marco Merlo
- Cardiovascular Department, Azienda Sanitaria-Universitaria Integrata Trieste "ASUITS," Trieste, Italy
| | - Sharon L Graw
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Giulia Barbati
- Biostatistics Unit, Department of Medical Sciences, University of Trieste, Trieste, Italy
| | - Teisha J Rowland
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Dobromir B Slavov
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Davide Stolfo
- Cardiovascular Department, Azienda Sanitaria-Universitaria Integrata Trieste "ASUITS," Trieste, Italy
| | - Mary E Haywood
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Matteo Dal Ferro
- Cardiovascular Department, Azienda Sanitaria-Universitaria Integrata Trieste "ASUITS," Trieste, Italy
| | - Alessandro Altinier
- Cardiovascular Department, Azienda Sanitaria-Universitaria Integrata Trieste "ASUITS," Trieste, Italy
| | - Federica Ramani
- Cardiovascular Department, Azienda Sanitaria-Universitaria Integrata Trieste "ASUITS," Trieste, Italy
| | - Francesca Brun
- Cardiovascular Department, Azienda Sanitaria-Universitaria Integrata Trieste "ASUITS," Trieste, Italy
| | - Andrea Cocciolo
- Cardiovascular Department, Azienda Sanitaria-Universitaria Integrata Trieste "ASUITS," Trieste, Italy; Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Ilaria Puggia
- Cardiovascular Department, Azienda Sanitaria-Universitaria Integrata Trieste "ASUITS," Trieste, Italy; Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Gaetano Morea
- Cardiovascular Department, Azienda Sanitaria-Universitaria Integrata Trieste "ASUITS," Trieste, Italy; Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - William J McKenna
- Institute of Cardiovascular Science, University College London, London, United Kingdom; Heart Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Francisco G La Rosa
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Matthew R G Taylor
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| | - Gianfranco Sinagra
- Cardiovascular Department, Azienda Sanitaria-Universitaria Integrata Trieste "ASUITS," Trieste, Italy
| | - Luisa Mestroni
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| |
Collapse
|
170
|
Pioner JM, Fornaro A, Coppini R, Ceschia N, Sacconi L, Donati MA, Favilli S, Poggesi C, Olivotto I, Ferrantini C. Advances in Stem Cell Modeling of Dystrophin-Associated Disease: Implications for the Wider World of Dilated Cardiomyopathy. Front Physiol 2020; 11:368. [PMID: 32477154 PMCID: PMC7235370 DOI: 10.3389/fphys.2020.00368] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 03/30/2020] [Indexed: 12/26/2022] Open
Abstract
Familial dilated cardiomyopathy (DCM) is mostly caused by mutations in genes encoding cytoskeletal and sarcomeric proteins. In the pediatric population, DCM is the predominant type of primitive myocardial disease. A severe form of DCM is associated with mutations in the DMD gene encoding dystrophin, which are the cause of Duchenne Muscular Dystrophy (DMD). DMD-associated cardiomyopathy is still poorly understood and orphan of a specific therapy. In the last 5 years, a rise of interest in disease models using human induced pluripotent stem cells (hiPSCs) has led to more than 50 original studies on DCM models. In this review paper, we provide a comprehensive overview on the advances in DMD cardiomyopathy disease modeling and highlight the most remarkable findings obtained from cardiomyocytes differentiated from hiPSCs of DMD patients. We will also describe how hiPSCs based studies have contributed to the identification of specific myocardial disease mechanisms that may be relevant in the pathogenesis of DCM, representing novel potential therapeutic targets.
Collapse
Affiliation(s)
- Josè Manuel Pioner
- Division of Physiology, Department of Experimental and Clinical Medicine, Università degli Studi di Firenze, Florence, Italy
| | | | - Raffaele Coppini
- Department of NeuroFarBa, Università degli Studi di Firenze, Florence, Italy
| | - Nicole Ceschia
- Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy
| | - Leonardo Sacconi
- LENS, Università degli Studi di Firenze and National Institute of Optics (INO-CNR), Florence, Italy
| | | | - Silvia Favilli
- Pediatric Cardiology, Meyer Children's Hospital, Florence, Italy
| | - Corrado Poggesi
- Division of Physiology, Department of Experimental and Clinical Medicine, Università degli Studi di Firenze, Florence, Italy
| | - Iacopo Olivotto
- Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy
| | - Cecilia Ferrantini
- Division of Physiology, Department of Experimental and Clinical Medicine, Università degli Studi di Firenze, Florence, Italy
| |
Collapse
|
171
|
Ramachandra CJA, Chua J, Cong S, Kp MMJ, Shim W, Wu JC, Hausenloy DJ. Human-induced pluripotent stem cells for modelling metabolic perturbations and impaired bioenergetics underlying cardiomyopathies. Cardiovasc Res 2020; 117:694-711. [PMID: 32365198 DOI: 10.1093/cvr/cvaa125] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/23/2020] [Accepted: 04/24/2020] [Indexed: 12/17/2022] Open
Abstract
Normal cardiac contractile and relaxation functions are critically dependent on a continuous energy supply. Accordingly, metabolic perturbations and impaired mitochondrial bioenergetics with subsequent disruption of ATP production underpin a wide variety of cardiac diseases, including diabetic cardiomyopathy, dilated cardiomyopathy, hypertrophic cardiomyopathy, anthracycline cardiomyopathy, peripartum cardiomyopathy, and mitochondrial cardiomyopathies. Crucially, there are no specific treatments for preventing the onset or progression of these cardiomyopathies to heart failure, one of the leading causes of death and disability worldwide. Therefore, new treatments are needed to target the metabolic disturbances and impaired mitochondrial bioenergetics underlying these cardiomyopathies in order to improve health outcomes in these patients. However, investigation of the underlying mechanisms and the identification of novel therapeutic targets have been hampered by the lack of appropriate animal disease models. Furthermore, interspecies variation precludes the use of animal models for studying certain disorders, whereas patient-derived primary cell lines have limited lifespan and availability. Fortunately, the discovery of human-induced pluripotent stem cells has provided a promising tool for modelling cardiomyopathies via human heart tissue in a dish. In this review article, we highlight the use of patient-derived iPSCs for studying the pathogenesis underlying cardiomyopathies associated with metabolic perturbations and impaired mitochondrial bioenergetics, as the ability of iPSCs for self-renewal and differentiation makes them an ideal platform for investigating disease pathogenesis in a controlled in vitro environment. Continuing progress will help elucidate novel mechanistic pathways, and discover novel therapies for preventing the onset and progression of heart failure, thereby advancing a new era of personalized therapeutics for improving health outcomes in patients with cardiomyopathy.
Collapse
Affiliation(s)
- Chrishan J A Ramachandra
- National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, Singapore 169609, Singapore.,Cardiovascular and Metabolic Disorders Programme, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Jasper Chua
- National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, Singapore 169609, Singapore.,Faculty of Science, National University of Singapore, 6 Science Drive 2, Singapore 117546, Singapore
| | - Shuo Cong
- National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, Singapore 169609, Singapore.,Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, 111 Yixueyuan Road, Xuhui District, Shanghai 200032, China
| | - Myu Mai Ja Kp
- National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, Singapore 169609, Singapore
| | - Winston Shim
- Health and Social Sciences Cluster, Singapore Institute of Technology, 10 Dover Drive, Singapore 138683, Singapore
| | - Joseph C Wu
- Cardiovascular Institute, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Medicine, Stanford University, Stanford, CA 94305, USA.,Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Derek J Hausenloy
- National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, Singapore 169609, Singapore.,Cardiovascular and Metabolic Disorders Programme, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore.,Yong Loo Lin Medical School, National University of Singapore, 10 Medical Drive, Singapore 11759, Singapore.,The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, Bloomsbury, London WC1E 6HX, UK.,Cardiovascular Research Centre, College of Medical and Health Sciences, Asia University, No. 500, Liufeng Road, Wufeng District, Taichung City 41354,Taiwan
| |
Collapse
|
172
|
Zhao MT, Shao NY, Garg V. Subtype-specific cardiomyocytes for precision medicine: Where are we now? Stem Cells 2020; 38:822-833. [PMID: 32232889 DOI: 10.1002/stem.3178] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 03/06/2020] [Accepted: 03/16/2020] [Indexed: 11/12/2022]
Abstract
Patient-derived pluripotent stem cells (PSCs) have greatly transformed the current understanding of human heart development and cardiovascular disease. Cardiomyocytes derived from personalized PSCs are powerful tools for modeling heart disease and performing patient-based cardiac toxicity testing. However, these PSC-derived cardiomyocytes (PSC-CMs) are a mixed population of atrial-, ventricular-, and pacemaker-like cells in the dish, hindering the future of precision cardiovascular medicine. Recent insights gleaned from the developing heart have paved new avenues to refine subtype-specific cardiomyocytes from patients with known pathogenic genetic variants and clinical phenotypes. Here, we discuss the recent progress on generating subtype-specific (atrial, ventricular, and nodal) cardiomyocytes from the perspective of embryonic heart development and how human pluripotent stem cells will expand our current knowledge on molecular mechanisms of cardiovascular disease and the future of precision medicine.
Collapse
Affiliation(s)
- Ming-Tao Zhao
- Center for Cardiovascular Research, The Abigail Wexner Research Institute and The Heart Center, Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Ning-Yi Shao
- Faculty of Health Sciences, University of Macau, Macau, People's Republic of China
| | - Vidu Garg
- Center for Cardiovascular Research, The Abigail Wexner Research Institute and The Heart Center, Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, USA.,Department of Molecular Genetics, The Ohio State University College of Medicine, Columbus, Ohio, USA
| |
Collapse
|
173
|
Sharma A, Garcia G, Arumugaswami V, Svendsen CN. Human iPSC-Derived Cardiomyocytes are Susceptible to SARS-CoV-2 Infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020. [PMID: 32511402 DOI: 10.1101/2020.04.21.051912] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is a viral pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). COVID-19 is predominantly defined by respiratory symptoms, but cardiac complications including arrhythmias, heart failure, and viral myocarditis are also prevalent. Although the systemic ischemic and inflammatory responses caused by COVID-19 can detrimentally affect cardiac function, the direct impact of SARS-CoV-2 infection on human cardiomyocytes is not well-understood. We used human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) as a model system to examine the mechanisms of cardiomyocyte-specific infection by SARS-CoV-2. Microscopy and immunofluorescence demonstrated that SARS-CoV-2 can enter and replicate within hiPSC-CMs, localizing at perinuclear locations within the cytoplasm. Viral cytopathic effect induced hiPSC-CM apoptosis and cessation of beating after 72 hours of infection. These studies show that SARS-CoV-2 can infect hiPSC-CMs in vitro , establishing a model for elucidating the mechanisms of infection and potentially a cardiac-specific antiviral drug screening platform.
Collapse
|
174
|
Rampoldi A, Singh M, Wu Q, Duan M, Jha R, Maxwell JT, Bradner JM, Zhang X, Saraf A, Miller GW, Gibson G, Brown LA, Xu C. Cardiac Toxicity From Ethanol Exposure in Human-Induced Pluripotent Stem Cell-Derived Cardiomyocytes. Toxicol Sci 2020; 169:280-292. [PMID: 31059573 DOI: 10.1093/toxsci/kfz038] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Alcohol use prior to and during pregnancy remains a significant societal problem and can lead to developmental fetal abnormalities including compromised myocardia function and increased risk for heart disease later in life. Alcohol-induced cardiac toxicity has traditionally been studied in animal-based models. These models have limitations due to physiological differences from human cardiomyocytes (CMs) and are also not suitable for high-throughput screening. We hypothesized that human-induced pluripotent stem cell-derived CMs (hiPSC-CMs) could serve as a useful tool to study alcohol-induced cardiac defects and/or toxicity. In this study, hiPSC-CMs were treated with ethanol at doses corresponding to the clinically relevant levels of alcohol intoxication. hiPSC-CMs exposed to ethanol showed a dose-dependent increase in cellular damage and decrease in cell viability, corresponding to increased production of reactive oxygen species. Furthermore, ethanol exposure also generated dose-dependent increased irregular Ca2+ transients and contractility in hiPSC-CMs. RNA-seq analysis showed significant alteration in genes belonging to the potassium voltage-gated channel family or solute carrier family, partially explaining the irregular Ca2+ transients and contractility in ethanol-treated hiPSC-CMs. RNA-seq also showed significant upregulation in the expression of genes associated with collagen and extracellular matrix modeling, and downregulation of genes involved in cardiovascular system development and actin filament-based process. These results suggest that hiPSC-CMs can be a novel and physiologically relevant system for the study of alcohol-induced cardiac toxicity.
Collapse
Affiliation(s)
- Antonio Rampoldi
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Monalisa Singh
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Qingling Wu
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, Georgia.,Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia
| | - Meixue Duan
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia
| | - Rajneesh Jha
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Joshua T Maxwell
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Joshua M Bradner
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | | | - Anita Saraf
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, Georgia.,Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - Gary W Miller
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Greg Gibson
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia
| | - Lou Ann Brown
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Chunhui Xu
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, Georgia.,Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia
| |
Collapse
|
175
|
Human induced pluripotent stem cell-derived cardiomyocytes reveal abnormal TGFβ signaling in type 2 diabetes mellitus. J Mol Cell Cardiol 2020; 142:53-64. [PMID: 32251671 DOI: 10.1016/j.yjmcc.2020.03.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 03/23/2020] [Accepted: 03/30/2020] [Indexed: 12/17/2022]
Abstract
Diabetes mellitus is a serious metabolic condition associated with a multitude of cardiovascular complications. Moreover, the prevalence of diabetes in heart failure populations is higher than that in control populations. However, the role of cardiomyocyte alterations in type 2 diabetes mellitus (T2DM) has not been well characterized and the underlying mechanisms remain elusive. In this study, two patients who were diagnosed as T2DM were recruited and patient-specific induced pluripotent stem cells (iPSCs) were generated from urine epithelial cells using nonintegrated Sendai virus. The iPSC lines derived from five healthy subjects were used as controls. All iPSCs were differentiated into cardiomyocytes (iPSC-CMs) using the monolayer-based differentiation protocol. T2DM iPSC-CMs exhibited various disease phenotypes, including cellular hypertrophy and lipid accumulation. Moreover, T2DM iPSC-CMs exhibited higher susceptibility to high-glucose/high-lipid challenge than control iPSC-CMs, manifesting an increase in apoptosis. RNA-Sequencing analysis revealed a differential transcriptome profile and abnormal activation of TGFβ signaling pathway in T2DM iPSC-CMs. We went on to show that inhibition of TGFβ significantly rescued the hypertrophic phenotype in T2DM iPSC-CMs. In conclusion, we demonstrate that the iPSC-CM model is able to recapitulate cellular phenotype of T2DM. Our results indicate that iPSC-CMs can therefore serve as a suitable model for investigating molecular mechanisms underlying diabetic cardiomyopathies and for screening therapeutic drugs.
Collapse
|
176
|
Cong S, Ramachandra CJ, Mai Ja KPM, Yap J, Shim W, Wei L, Hausenloy DJ. Mechanisms underlying diabetic cardiomyopathy: From pathophysiology to novel therapeutic targets. CONDITIONING MEDICINE 2020; 3:82-97. [PMID: 34169234 PMCID: PMC8221238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Diabetic cardiomyopathy (DC) is defined as a clinical condition of cardiac dysfunction that occurs in the absence of coronary atherosclerosis, valvular disease, and hypertension in patients with diabetes mellitus (DM). Despite the increasing worldwide prevalence of DC, due to the global epidemic of DM, the underlying pathophysiology of DC has not been fully elucidated. In addition, the clinical criteria for diagnosing DC have not been established, and specific therapeutic options are not currently available. The current paradigm suggests the impaired cardiomyocyte function arises due to a number of DM-related metabolic disturbances including hyperglycemia, hyperinsulinemia, and hyperlipidemia, which lead to diastolic dysfunction and signs and symptoms of heart failure. Other factors, which have been implicated in the progression of DC, include mitochondrial dysfunction, increased oxidative stress, impaired calcium handling, inflammation, and cardiomyocyte apoptosis. Herein, we review the current theories surrounding the occurrence and progression of DC, and discuss the recent advances in diagnostic methodologies and therapeutic strategies. Moreover, apart from conventional animal DC models, we highlight alternative disease models for studying DC such as the use of patient-derived human induced pluripotent stem cells (hiPSCs) for studying the mechanisms underlying DC. The ability to obtain hiPSC-derived cardiomyocytes from DM patients with a DC phenotype could help identify novel therapeutic targets for preventing and delaying the progression of DC, and for improving clinical outcomes in DM patients.
Collapse
Affiliation(s)
- Shuo Cong
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
| | - Chrishan J.A. Ramachandra
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
- Cardiovascular and Metabolic Disorders Programme, Duke-NUS Medical School, Singapore
| | - KP Myu Mai Ja
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
| | - Jonathan Yap
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, USA
| | - Winston Shim
- Health and Social Sciences Cluster, Singapore Institute of Technology, Singapore
| | - Lai Wei
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Engineering Research Centre of Cardiac Valve, Shanghai, China
| | - Derek J. Hausenloy
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
- Cardiovascular and Metabolic Disorders Programme, Duke-NUS Medical School, Singapore
- Yong Loo Lin Medical School, National University of Singapore, Singapore
- The Hatter Cardiovascular Institute, University College London, London, UK
- Cardiovascular Research Centre, College of Medical and Health Sciences, Asia University, Taiwan
| |
Collapse
|
177
|
Can Patient Pluripotent Stem Cell–Derived Cardiomyocytes Provide Useful Modeling on Arrhythmias of DMD Cardiomyopathy? J Am Coll Cardiol 2020; 75:1175-1177. [DOI: 10.1016/j.jacc.2020.01.038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 01/27/2020] [Accepted: 01/28/2020] [Indexed: 01/05/2023]
|
178
|
Jiang B, Yan L, Shamul JG, Hakun M, He X. Stem cell therapy of myocardial infarction: a promising opportunity in bioengineering. ADVANCED THERAPEUTICS 2020; 3:1900182. [PMID: 33665356 PMCID: PMC7928435 DOI: 10.1002/adtp.201900182] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Indexed: 02/06/2023]
Abstract
Myocardial infarction (MI) is a life-threatening disease resulting from irreversible death of cardiomyocytes (CMs) and weakening of the heart blood-pumping function. Stem cell-based therapies have been studied for MI treatment over the last two decades with promising outcome. In this review, we critically summarize the past work in this field to elucidate the advantages and disadvantages of treating MI using pluripotent stem cells (PSCs) including both embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), adult stem cells, and cardiac progenitor cells. The main advantage of the latter is their cytokine production capability to modulate immune responses and control the progression of healing. However, human adult stem cells have very limited (if not 'no') capacity to differentiate into functional CMs in vitro or in vivo. In contrast, PSCs can be differentiated into functional CMs although the protocols for the cardiac differentiation of PSCs are mainly for adherent cells under 2D culture. Derivation of PSC-CMs in 3D, allowing for large-scale production of CMs via modulation of the Wnt/β-catenin signal pathway with defined chemicals and medium, may be desired for clinical translation. Furthermore, the technology of purification and maturation of the PSC-CMs may need further improvements to eliminate teratoma formation after in vivo implantation of the PSC-CMs for treating MI. In addition, in vitro derived PSC-CMs may have mechanical and electrical mismatch with the patient's cardiac tissue, which causes arrhythmia. This supports the use of PSC-derived cells committed to cardiac lineage without beating for implantation to treat MI. In this case, the PSC derived cells may utilize the mechanical, electrical, and chemical cues in the heart to further differentiate into mature/functional CMs in situ. Another major challenge facing stem cell therapy of MI is the low retention/survival of stem cells or their derivatives (e.g., PSC-CMs) in the heart for MI treatment after injection in vivo. This may be resolved by using biomaterials to engineer stem cells for reduced immunogenicity, immobilization of the cells in the heart, and increased integration with the host cardiac tissue. Biomaterials have also been applied in the derivation of CMs in vitro to increase the efficiency and maturation of differentiation. Collectively, a lot has been learned from the past failure of simply injecting intact stem cells or their derivatives in vivo for treating MI, and bioengineering stem cells with biomaterials is expected to be a valuable strategy for advancing stem cell therapy towards its widespread application for treating MI in the clinic.
Collapse
Affiliation(s)
- Bin Jiang
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Li Yan
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - James G Shamul
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Maxwell Hakun
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Xiaoming He
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| |
Collapse
|
179
|
Nakao S, Ihara D, Hasegawa K, Kawamura T. Applications for Induced Pluripotent Stem Cells in Disease Modelling and Drug Development for Heart Diseases. Eur Cardiol 2020; 15:1-10. [PMID: 32180835 PMCID: PMC7066852 DOI: 10.15420/ecr.2019.03] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 08/09/2019] [Indexed: 12/22/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) are derived from reprogrammed somatic cells by the introduction of defined transcription factors. They are characterised by a capacity for self-renewal and pluripotency. Human (h)iPSCs are expected to be used extensively for disease modelling, drug screening and regenerative medicine. Obtaining cardiac tissue from patients with mutations for genetic studies and functional analyses is a highly invasive procedure. In contrast, disease-specific hiPSCs are derived from the somatic cells of patients with specific genetic mutations responsible for disease phenotypes. These disease-specific hiPSCs are a better tool for studies of the pathophysiology and cellular responses to therapeutic agents. This article focuses on the current understanding, limitations and future direction of disease-specific hiPSC-derived cardiomyocytes for further applications.
Collapse
Affiliation(s)
- Shu Nakao
- Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Kusatsu, Japan.,Global Innovation Research Organization, Ritsumeikan University, Kusatsu, Japan.,Division of Translational Research, Kyoto Medical Center, National Hospital Organization, Kyoto, Japan
| | - Dai Ihara
- Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Kusatsu, Japan.,Global Innovation Research Organization, Ritsumeikan University, Kusatsu, Japan
| | - Koji Hasegawa
- Division of Translational Research, Kyoto Medical Center, National Hospital Organization, Kyoto, Japan
| | - Teruhisa Kawamura
- Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Kusatsu, Japan.,Global Innovation Research Organization, Ritsumeikan University, Kusatsu, Japan.,Division of Translational Research, Kyoto Medical Center, National Hospital Organization, Kyoto, Japan
| |
Collapse
|
180
|
Law ML, Cohen H, Martin AA, Angulski ABB, Metzger JM. Dysregulation of Calcium Handling in Duchenne Muscular Dystrophy-Associated Dilated Cardiomyopathy: Mechanisms and Experimental Therapeutic Strategies. J Clin Med 2020; 9:jcm9020520. [PMID: 32075145 PMCID: PMC7074327 DOI: 10.3390/jcm9020520] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 02/06/2020] [Indexed: 02/07/2023] Open
Abstract
: Duchenne muscular dystrophy (DMD) is an X-linked recessive disease resulting in the loss of dystrophin, a key cytoskeletal protein in the dystrophin-glycoprotein complex. Dystrophin connects the extracellular matrix with the cytoskeleton and stabilizes the sarcolemma. Cardiomyopathy is prominent in adolescents and young adults with DMD, manifesting as dilated cardiomyopathy (DCM) in the later stages of disease. Sarcolemmal instability, leading to calcium mishandling and overload in the cardiac myocyte, is a key mechanistic contributor to muscle cell death, fibrosis, and diminished cardiac contractile function in DMD patients. Current therapies for DMD cardiomyopathy can slow disease progression, but they do not directly target aberrant calcium handling and calcium overload. Experimental therapeutic targets that address calcium mishandling and overload include membrane stabilization, inhibition of stretch-activated channels, ryanodine receptor stabilization, and augmentation of calcium cycling via modulation of the Serca2a/phospholamban (PLN) complex or cytosolic calcium buffering. This paper addresses what is known about the mechanistic basis of calcium mishandling in DCM, with a focus on DMD cardiomyopathy. Additionally, we discuss currently utilized therapies for DMD cardiomyopathy, and review experimental therapeutic strategies targeting the calcium handling defects in DCM and DMD cardiomyopathy.
Collapse
Affiliation(s)
- Michelle L. Law
- Department of Family and Consumer Sciences, Robbins College of Health and Human Sciences, Baylor University, Waco, TX 76706, USA;
| | - Houda Cohen
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (H.C.); (A.A.M.); (A.B.B.A.)
| | - Ashley A. Martin
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (H.C.); (A.A.M.); (A.B.B.A.)
| | - Addeli Bez Batti Angulski
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (H.C.); (A.A.M.); (A.B.B.A.)
| | - Joseph M. Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (H.C.); (A.A.M.); (A.B.B.A.)
- Correspondence: ; Tel.: +1-612-625-5902; Fax: +1-612-625-5149
| |
Collapse
|
181
|
Altered microtubule structure, hemichannel localization and beating activity in cardiomyocytes expressing pathologic nuclear lamin A/C. Heliyon 2020; 6:e03175. [PMID: 32021920 PMCID: PMC6992992 DOI: 10.1016/j.heliyon.2020.e03175] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 10/31/2019] [Accepted: 01/02/2020] [Indexed: 12/21/2022] Open
Abstract
Given the clinical effect of laminopathies, understanding lamin mechanical properties will benefit the treatment of heart failure. Here we report a mechano-dynamic study of LMNA mutations in neonatal rat ventricular myocytes (NRVM) using single cell spectroscopy with Atomic Force Microscopy (AFM) and measured changes in beating force, frequency and contractile amplitude of selected mutant-expressing cells within cell clusters. Furthermore, since beat-to-beat variations can provide clues on the origin of arrhythmias, we analyzed the beating rate variability using a time-domain method which provides a Poincaré plot. Data were further correlated to cell phenotypes. Immunofluorescence and calcium imaging analysis showed that mutant lamin changed NRVMs beating force and frequency. Additionally, we noted an altered microtubule network organization with shorter filament length, and defective hemichannel membrane localization (Connexin 43). These data highlight the interconnection between nucleoskeleton, cytoskeleton and sarcolemmal structures, and the transcellular consequences of mutant lamin protein in the pathogenesis of the cardiac laminopathies.
Collapse
|
182
|
Pourrier M, Fedida D. The Emergence of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes (hiPSC-CMs) as a Platform to Model Arrhythmogenic Diseases. Int J Mol Sci 2020; 21:ijms21020657. [PMID: 31963859 PMCID: PMC7013748 DOI: 10.3390/ijms21020657] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/13/2020] [Accepted: 01/15/2020] [Indexed: 12/13/2022] Open
Abstract
There is a need for improved in vitro models of inherited cardiac diseases to better understand basic cellular and molecular mechanisms and advance drug development. Most of these diseases are associated with arrhythmias, as a result of mutations in ion channel or ion channel-modulatory proteins. Thus far, the electrophysiological phenotype of these mutations has been typically studied using transgenic animal models and heterologous expression systems. Although they have played a major role in advancing the understanding of the pathophysiology of arrhythmogenesis, more physiological and predictive preclinical models are necessary to optimize the treatment strategy for individual patients. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have generated much interest as an alternative tool to model arrhythmogenic diseases. They provide a unique opportunity to recapitulate the native-like environment required for mutated proteins to reproduce the human cellular disease phenotype. However, it is also important to recognize the limitations of this technology, specifically their fetal electrophysiological phenotype, which differentiates them from adult human myocytes. In this review, we provide an overview of the major inherited arrhythmogenic cardiac diseases modeled using hiPSC-CMs and for which the cellular disease phenotype has been somewhat characterized.
Collapse
Affiliation(s)
- Marc Pourrier
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada;
- IonsGate Preclinical Services Inc., Vancouver, BC V6T 1Z3, Canada
- Correspondence:
| | - David Fedida
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada;
| |
Collapse
|
183
|
Dai Y, Amenov A, Ignatyeva N, Koschinski A, Xu H, Soong PL, Tiburcy M, Linke WA, Zaccolo M, Hasenfuss G, Zimmermann WH, Ebert A. Troponin destabilization impairs sarcomere-cytoskeleton interactions in iPSC-derived cardiomyocytes from dilated cardiomyopathy patients. Sci Rep 2020; 10:209. [PMID: 31937807 PMCID: PMC6959358 DOI: 10.1038/s41598-019-56597-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 09/13/2019] [Indexed: 12/14/2022] Open
Abstract
The sarcomeric troponin-tropomyosin complex is a critical mediator of excitation-contraction coupling, sarcomeric stability and force generation. We previously reported that induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) from patients with a dilated cardiomyopathy (DCM) mutation, troponin T (TnT)-R173W, display sarcomere protein misalignment and impaired contractility. Yet it is not known how TnT mutation causes dysfunction of sarcomere microdomains and how these events contribute to misalignment of sarcomeric proteins in presence of DCM TnT-R173W. Using a human iPSC-CM model combined with CRISPR/Cas9-engineered isogenic controls, we uncovered that TnT-R173W destabilizes molecular interactions of troponin with tropomyosin, and limits binding of PKA to local sarcomere microdomains. This attenuates troponin phosphorylation and dysregulates local sarcomeric microdomains in DCM iPSC-CMs. Disrupted microdomain signaling impairs MYH7-mediated, AMPK-dependent sarcomere-cytoskeleton filament interactions and plasma membrane attachment. Small molecule-based activation of AMPK can restore TnT microdomain interactions, and partially recovers sarcomere protein misalignment as well as impaired contractility in DCM TnT-R173W iPSC-CMs. Our findings suggest a novel therapeutic direction targeting sarcomere- cytoskeleton interactions to induce sarcomere re-organization and contractile recovery in DCM.
Collapse
Affiliation(s)
- Yuanyuan Dai
- Heart Center, Department of Cardiology and Pneumology, Goettingen, Germany.,DZHK (German Center for Cardiovascular Research), partner site, Goettingen, Germany
| | - Asset Amenov
- Heart Center, Department of Cardiology and Pneumology, Goettingen, Germany.,DZHK (German Center for Cardiovascular Research), partner site, Goettingen, Germany
| | - Nadezda Ignatyeva
- Heart Center, Department of Cardiology and Pneumology, Goettingen, Germany.,DZHK (German Center for Cardiovascular Research), partner site, Goettingen, Germany
| | - Andreas Koschinski
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK
| | - Hang Xu
- Heart Center, Department of Cardiology and Pneumology, Goettingen, Germany.,DZHK (German Center for Cardiovascular Research), partner site, Goettingen, Germany
| | - Poh Loong Soong
- Institute of Pharmacology, University of Goettingen, Robert-Koch-Str. 40, 37075, Goettingen, Germany.,DZHK (German Center for Cardiovascular Research), partner site, Goettingen, Germany
| | - Malte Tiburcy
- Institute of Pharmacology, University of Goettingen, Robert-Koch-Str. 40, 37075, Goettingen, Germany.,DZHK (German Center for Cardiovascular Research), partner site, Goettingen, Germany
| | - Wolfgang A Linke
- Heart Center, Department of Cardiology and Pneumology, Goettingen, Germany.,DZHK (German Center for Cardiovascular Research), partner site, Goettingen, Germany.,Institute of Physiology II, University of Muenster, Muenster, Germany
| | - Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK
| | - Gerd Hasenfuss
- Heart Center, Department of Cardiology and Pneumology, Goettingen, Germany.,DZHK (German Center for Cardiovascular Research), partner site, Goettingen, Germany
| | - Wolfram-Hubertus Zimmermann
- Institute of Pharmacology, University of Goettingen, Robert-Koch-Str. 40, 37075, Goettingen, Germany.,DZHK (German Center for Cardiovascular Research), partner site, Goettingen, Germany
| | - Antje Ebert
- Heart Center, Department of Cardiology and Pneumology, Goettingen, Germany. .,DZHK (German Center for Cardiovascular Research), partner site, Goettingen, Germany.
| |
Collapse
|
184
|
Ramchand J, Wallis M, Macciocca I, Lynch E, Farouque O, Martyn M, Phelan D, Chong B, Lockwood S, Weintraub R, Thompson T, Trainer A, Zentner D, Vohra J, Chetrit M, Hare DL, James P. Prospective Evaluation of the Utility of Whole Exome Sequencing in Dilated Cardiomyopathy. J Am Heart Assoc 2020; 9:e013346. [PMID: 31931689 PMCID: PMC7033851 DOI: 10.1161/jaha.119.013346] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background Dilated cardiomyopathy may be heritable but shows extensive genetic heterogeneity. The utility of whole exome sequencing as a first-line genetic test for patients with dilated cardiomyopathy in a contemporary "real-world" setting has not been specifically established. Using whole exome sequencing with rigorous, evidence-based variant interpretation, we aimed to identify the prevalence of a molecular diagnosis in patients with dilated cardiomyopathy in a clinical setting. Methods and Results Whole exome sequencing was performed in eligible patients (n=83) with idiopathic or familial dilated cardiomyopathy. Variants were prioritized for curation in up to 247 genes and classified using American College of Medical Genetics and Genomics-based criteria. Ten (12%) had a pathogenic or likely pathogenic variant. Eight (10%) participants had truncating TTN variants classified as variants of uncertain significance. Five (6%) participants had variants of unknown significance according to strict American College of Medical Genetics and Genomics criteria but classified as either pathogenic or likely pathogenic by other clinical laboratories. Pathogenic or likely pathogenic variants were found in 8 genes (all within tier 1 genes), 2 (20%) of which are not included in a standard commercially available dilated cardiomyopathy panel. Using our bioinformatics pipeline, there was an average of 0.74 variants of uncertain significance per case with ≈0.75 person-hours needed to interpret each of these variants. Conclusions Whole exome sequencing is an effective diagnostic tool for patients with dilated cardiomyopathy. With stringent classification using American College of Medical Genetics and Genomics criteria, the rate of detection of pathogenic variants is lower than previous reports. Efforts to improve adherence to these guidelines will be important to prevent erroneous misclassification of nonpathogenic variants in dilated cardiomyopathy genetic testing and inappropriate cascade screening.
Collapse
Affiliation(s)
- Jay Ramchand
- Department of Medicine Austin Health The University of Melbourne Heidelberg Victoria Australia.,Department of Cardiology Austin Health Heidelberg Victoria Australia
| | - Mathew Wallis
- Department of Genetics Austin Health Heidelberg Victoria Australia
| | - Ivan Macciocca
- Victorian Clinical Genetics Services Murdoch Children's Research Institute Royal Children's Hospital Flemington Victoria Australia
| | - Elly Lynch
- Victorian Clinical Genetics Services Murdoch Children's Research Institute Royal Children's Hospital Flemington Victoria Australia.,Melbourne Genomics Health Alliance Melbourne Victoria Australia
| | - Omar Farouque
- Department of Medicine Austin Health The University of Melbourne Heidelberg Victoria Australia.,Department of Cardiology Austin Health Heidelberg Victoria Australia
| | - Melissa Martyn
- Melbourne Genomics Health Alliance Melbourne Victoria Australia.,Department of Paediatrics University of Melbourne Parkville Victoria Australia.,Murdoch Children's Research Institute Parkville Victoria Australia
| | - Dean Phelan
- Victorian Clinical Genetics Services Murdoch Children's Research Institute Royal Children's Hospital Flemington Victoria Australia
| | - Belinda Chong
- Victorian Clinical Genetics Services Murdoch Children's Research Institute Royal Children's Hospital Flemington Victoria Australia
| | - Siobhan Lockwood
- Monash Cardiovascular Research Centre and Monash Heart Monash University and Monash Health Melbourne Australia
| | - Robert Weintraub
- Victorian Clinical Genetics Services Murdoch Children's Research Institute Royal Children's Hospital Flemington Victoria Australia
| | - Tina Thompson
- Genetic Medicine Melbourne Health Parkville Victoria Australia
| | - Alison Trainer
- Genetic Medicine Melbourne Health Parkville Victoria Australia
| | - Dominica Zentner
- Department of Cardiology Melbourne Health Parkville Victoria Australia.,Genetic Medicine Melbourne Health Parkville Victoria Australia.,Royal Melbourne Hospital Clinical School Faculty of Medicine Dentistry and Health Sciences University of Melbourne Parkville Victoria Australia
| | - Jitendra Vohra
- Department of Cardiology Melbourne Health Parkville Victoria Australia.,Genetic Medicine Melbourne Health Parkville Victoria Australia.,Royal Melbourne Hospital Clinical School Faculty of Medicine Dentistry and Health Sciences University of Melbourne Parkville Victoria Australia
| | | | - David L Hare
- Department of Medicine Austin Health The University of Melbourne Heidelberg Victoria Australia.,Department of Cardiology Austin Health Heidelberg Victoria Australia
| | - Paul James
- Genetic Medicine Melbourne Health Parkville Victoria Australia
| |
Collapse
|
185
|
Zhao Y, Rafatian N, Wang EY, Wu Q, Lai BFL, Lu RX, Savoji H, Radisic M. Towards chamber specific heart-on-a-chip for drug testing applications. Adv Drug Deliv Rev 2020; 165-166:60-76. [PMID: 31917972 PMCID: PMC7338250 DOI: 10.1016/j.addr.2019.12.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/26/2019] [Accepted: 12/30/2019] [Indexed: 02/06/2023]
Abstract
Modeling of human organs has long been a task for scientists in order to lower the costs of therapeutic development and understand the pathological onset of human disease. For decades, despite marked differences in genetics and etiology, animal models remained the norm for drug discovery and disease modeling. Innovative biofabrication techniques have facilitated the development of organ-on-a-chip technology that has great potential to complement conventional animal models. However, human organ as a whole, more specifically the human heart, is difficult to regenerate in vitro, in terms of its chamber specific orientation and its electrical functional complexity. Recent progress with the development of induced pluripotent stem cell differentiation protocols, made recapitulating the complexity of the human heart possible through the generation of cells representative of atrial & ventricular tissue, the sinoatrial node, atrioventricular node and Purkinje fibers. Current heart-on-a-chip approaches incorporate biological, electrical, mechanical, and topographical cues to facilitate tissue maturation, therefore improving the predictive power for the chamber-specific therapeutic effects targeting adult human. In this review, we will give a summary of current advances in heart-on-a-chip technology and provide a comprehensive outlook on the challenges involved in the development of human physiologically relevant heart-on-a-chip.
Collapse
Affiliation(s)
- Yimu Zhao
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada
| | - Naimeh Rafatian
- Division of Cardiology and Peter Munk Cardiac Center, University of Health Network, Toronto, Ontario M5G 2N2, Canada
| | - Erika Yan Wang
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Qinghua Wu
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Benjamin F L Lai
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Rick Xingze Lu
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Houman Savoji
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Milica Radisic
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada; Toronto General Research Institute, Toronto, Ontario M5G 2C4, Canada.
| |
Collapse
|
186
|
Poetsch MS, Guan K. iPSCs for modeling of sarcomeric cardiomyopathies. RECENT ADVANCES IN IPSC DISEASE MODELING, VOLUME 1 2020:237-273. [DOI: 10.1016/b978-0-12-822227-0.00012-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
187
|
Paik DT, Chandy M, Wu JC. Patient and Disease-Specific Induced Pluripotent Stem Cells for Discovery of Personalized Cardiovascular Drugs and Therapeutics. Pharmacol Rev 2020; 72:320-342. [PMID: 31871214 PMCID: PMC6934989 DOI: 10.1124/pr.116.013003] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Human induced pluripotent stem cells (iPSCs) have emerged as an effective platform for regenerative therapy, disease modeling, and drug discovery. iPSCs allow for the production of limitless supply of patient-specific somatic cells that enable advancement in cardiovascular precision medicine. Over the past decade, researchers have developed protocols to differentiate iPSCs to multiple cardiovascular lineages, as well as to enhance the maturity and functionality of these cells. Despite significant advances, drug therapy and discovery for cardiovascular disease have lagged behind other fields such as oncology. We speculate that this paucity of drug discovery is due to a previous lack of efficient, reproducible, and translational model systems. Notably, existing drug discovery and testing platforms rely on animal studies and clinical trials, but investigations in animal models have inherent limitations due to interspecies differences. Moreover, clinical trials are inherently flawed by assuming that all individuals with a disease will respond identically to a therapy, ignoring the genetic and epigenomic variations that define our individuality. With ever-improving differentiation and phenotyping methods, patient-specific iPSC-derived cardiovascular cells allow unprecedented opportunities to discover new drug targets and screen compounds for cardiovascular disease. Imbued with the genetic information of an individual, iPSCs will vastly improve our ability to test drugs efficiently, as well as tailor and titrate drug therapy for each patient.
Collapse
Affiliation(s)
- David T Paik
- Stanford Cardiovascular Institute, Stanford University, Stanford, California
| | - Mark Chandy
- Stanford Cardiovascular Institute, Stanford University, Stanford, California
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University, Stanford, California
| |
Collapse
|
188
|
Fatica EM, DeLeonibus GA, House A, Kodger JV, Pearce RW, Shah RR, Levi L, Sandlers Y. Barth Syndrome: Exploring Cardiac Metabolism with Induced Pluripotent Stem Cell-Derived Cardiomyocytes. Metabolites 2019; 9:E306. [PMID: 31861102 PMCID: PMC6950123 DOI: 10.3390/metabo9120306] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 12/10/2019] [Accepted: 12/12/2019] [Indexed: 12/14/2022] Open
Abstract
Barth syndrome (BTHS) is an X-linked recessive multisystem disorder caused by mutations in the TAZ gene (TAZ, G 4.5, OMIM 300394) that encodes for the acyltransferase tafazzin. This protein is highly expressed in the heart and plays a significant role in cardiolipin biosynthesis. Heart disease is the major clinical manifestation of BTHS with a high incidence in early life. Although the genetic basis of BTHS and tetralinoleoyl cardiolipin deficiency in BTHS-affected individuals are well-established, downstream metabolic changes in cardiac metabolism are still uncovered. Our study aimed to characterize TAZ-induced metabolic perturbations in the heart. Control (PGP1-TAZWT) and TAZ mutant (PGP1-TAZ517delG) iPS-CM were incubated with 13C6-glucose and 13C5-glutamine and incorporation of 13C into downstream Krebs cycle intermediates was traced. Our data reveal that TAZ517delG induces accumulation of cellular long chain acylcarnitines and overexpression of fatty acid binding protein (FABP4). We also demonstrate that TAZ517delG induces metabolic alterations in pathways related to energy production as reflected by high glucose uptake, an increase in glycolytic lactate production and a decrease in palmitate uptake. Moreover, despite mitochondrial dysfunction, in the absence of glucose and fatty acids, TAZ517delG-iPS-CM can use glutamine as a carbon source to replenish the Krebs cycle.
Collapse
Affiliation(s)
- Erica M. Fatica
- Department of Chemistry, Cleveland State University, Cleveland, OH 44115, USA; (E.M.F.); (G.A.D.); (A.H.); (J.V.K.); (R.W.P.); (R.R.S.)
| | - Gina A. DeLeonibus
- Department of Chemistry, Cleveland State University, Cleveland, OH 44115, USA; (E.M.F.); (G.A.D.); (A.H.); (J.V.K.); (R.W.P.); (R.R.S.)
| | - Alisha House
- Department of Chemistry, Cleveland State University, Cleveland, OH 44115, USA; (E.M.F.); (G.A.D.); (A.H.); (J.V.K.); (R.W.P.); (R.R.S.)
| | - Jillian V. Kodger
- Department of Chemistry, Cleveland State University, Cleveland, OH 44115, USA; (E.M.F.); (G.A.D.); (A.H.); (J.V.K.); (R.W.P.); (R.R.S.)
| | - Ryan W. Pearce
- Department of Chemistry, Cleveland State University, Cleveland, OH 44115, USA; (E.M.F.); (G.A.D.); (A.H.); (J.V.K.); (R.W.P.); (R.R.S.)
| | - Rohan R. Shah
- Department of Chemistry, Cleveland State University, Cleveland, OH 44115, USA; (E.M.F.); (G.A.D.); (A.H.); (J.V.K.); (R.W.P.); (R.R.S.)
| | - Liraz Levi
- Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA;
| | - Yana Sandlers
- Department of Chemistry, Cleveland State University, Cleveland, OH 44115, USA; (E.M.F.); (G.A.D.); (A.H.); (J.V.K.); (R.W.P.); (R.R.S.)
| |
Collapse
|
189
|
Mosqueira D, Mannhardt I, Bhagwan JR, Lis-Slimak K, Katili P, Scott E, Hassan M, Prondzynski M, Harmer SC, Tinker A, Smith JGW, Carrier L, Williams PM, Gaffney D, Eschenhagen T, Hansen A, Denning C. CRISPR/Cas9 editing in human pluripotent stem cell-cardiomyocytes highlights arrhythmias, hypocontractility, and energy depletion as potential therapeutic targets for hypertrophic cardiomyopathy. Eur Heart J 2019; 39:3879-3892. [PMID: 29741611 PMCID: PMC6234851 DOI: 10.1093/eurheartj/ehy249] [Citation(s) in RCA: 173] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 04/11/2018] [Indexed: 12/26/2022] Open
Abstract
Aims Sarcomeric gene mutations frequently underlie hypertrophic cardiomyopathy (HCM), a prevalent and complex condition leading to left ventricle thickening and heart dysfunction. We evaluated isogenic genome-edited human pluripotent stem cell-cardiomyocytes (hPSC-CM) for their validity to model, and add clarity to, HCM. Methods and results CRISPR/Cas9 editing produced 11 variants of the HCM-causing mutation c.C9123T-MYH7 [(p.R453C-β-myosin heavy chain (MHC)] in 3 independent hPSC lines. Isogenic sets were differentiated to hPSC-CMs for high-throughput, non-subjective molecular and functional assessment using 12 approaches in 2D monolayers and/or 3D engineered heart tissues. Although immature, edited hPSC-CMs exhibited the main hallmarks of HCM (hypertrophy, multi-nucleation, hypertrophic marker expression, sarcomeric disarray). Functional evaluation supported the energy depletion model due to higher metabolic respiration activity, accompanied by abnormalities in calcium handling, arrhythmias, and contraction force. Partial phenotypic rescue was achieved with ranolazine but not omecamtiv mecarbil, while RNAseq highlighted potentially novel molecular targets. Conclusion Our holistic and comprehensive approach showed that energy depletion affected core cardiomyocyte functionality. The engineered R453C-βMHC-mutation triggered compensatory responses in hPSC-CMs, causing increased ATP production and αMHC to energy-efficient βMHC switching. We showed that pharmacological rescue of arrhythmias was possible, while MHY7: MYH6 and mutant: wild-type MYH7 ratios may be diagnostic, and previously undescribed lncRNAs and gene modifiers are suggestive of new mechanisms. ![]()
Collapse
Affiliation(s)
- Diogo Mosqueira
- Department of Stem Cell Biology, Centre of Biomolecular Sciences, University of Nottingham, UK
| | - Ingra Mannhardt
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Partner Site Hamburg/Kiel/Lübeck, DZHK (German Center for Cardiovascular Research), Hamburg, Germany
| | - Jamie R Bhagwan
- Department of Stem Cell Biology, Centre of Biomolecular Sciences, University of Nottingham, UK
| | - Katarzyna Lis-Slimak
- Department of Stem Cell Biology, Centre of Biomolecular Sciences, University of Nottingham, UK
| | - Puspita Katili
- Department of Stem Cell Biology, Centre of Biomolecular Sciences, University of Nottingham, UK
| | - Elizabeth Scott
- Department of Stem Cell Biology, Centre of Biomolecular Sciences, University of Nottingham, UK
| | - Mustafa Hassan
- The Heart Centre, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Charterhouse Square, London, UK
| | - Maksymilian Prondzynski
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Partner Site Hamburg/Kiel/Lübeck, DZHK (German Center for Cardiovascular Research), Hamburg, Germany
| | - Stephen C Harmer
- The Heart Centre, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Charterhouse Square, London, UK
| | - Andrew Tinker
- The Heart Centre, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Charterhouse Square, London, UK
| | - James G W Smith
- Department of Stem Cell Biology, Centre of Biomolecular Sciences, University of Nottingham, UK
| | - Lucie Carrier
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Partner Site Hamburg/Kiel/Lübeck, DZHK (German Center for Cardiovascular Research), Hamburg, Germany
| | - Philip M Williams
- Molecular Therapeutics and Formulation. School of Pharmacy, University of Nottingham, UK
| | - Daniel Gaffney
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Thomas Eschenhagen
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Partner Site Hamburg/Kiel/Lübeck, DZHK (German Center for Cardiovascular Research), Hamburg, Germany
| | - Arne Hansen
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Partner Site Hamburg/Kiel/Lübeck, DZHK (German Center for Cardiovascular Research), Hamburg, Germany
| | - Chris Denning
- Department of Stem Cell Biology, Centre of Biomolecular Sciences, University of Nottingham, UK
| |
Collapse
|
190
|
Samak M, Hinkel R. Stem Cells in Cardiovascular Medicine: Historical Overview and Future Prospects. Cells 2019; 8:cells8121530. [PMID: 31783680 PMCID: PMC6952821 DOI: 10.3390/cells8121530] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/19/2019] [Accepted: 11/23/2019] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular diseases remain the leading cause of death in the developed world, accounting for more than 30% of all deaths. In a large proportion of these patients, acute myocardial infarction is usually the first manifestation, which might further progress to heart failure. In addition, the human heart displays a low regenerative capacity, leading to a loss of cardiomyocytes and persistent tissue scaring, which entails a morbid pathologic sequela. Novel therapeutic approaches are urgently needed. Stem cells, such as induced pluripotent stem cells or embryonic stem cells, exhibit great potential for cell-replacement therapy and an excellent tool for disease modeling, as well as pharmaceutical screening of novel drugs and their cardiac side effects. This review article covers not only the origin of stem cells but tries to summarize their translational potential, as well as potential risks and clinical translation.
Collapse
Affiliation(s)
- Mostafa Samak
- Department of Laboratory Animal Science, Leibnitz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Kellnerweg 4, 37077 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Rabea Hinkel
- Department of Laboratory Animal Science, Leibnitz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Kellnerweg 4, 37077 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| |
Collapse
|
191
|
Hajipour MJ, Mehrani M, Abbasi SH, Amin A, Kassaian SE, Garbern JC, Caracciolo G, Zanganeh S, Chitsazan M, Aghaverdi H, Shahri SMK, Ashkarran A, Raoufi M, Bauser-Heaton H, Zhang J, Muehlschlegel JD, Moore A, Lee RT, Wu JC, Serpooshan V, Mahmoudi M. Nanoscale Technologies for Prevention and Treatment of Heart Failure: Challenges and Opportunities. Chem Rev 2019; 119:11352-11390. [PMID: 31490059 PMCID: PMC7003249 DOI: 10.1021/acs.chemrev.8b00323] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The adult myocardium has a limited regenerative capacity following heart injury, and the lost cells are primarily replaced by fibrotic scar tissue. Suboptimal efficiency of current clinical therapies to resurrect the infarcted heart results in injured heart enlargement and remodeling to maintain its physiological functions. These remodeling processes ultimately leads to ischemic cardiomyopathy and heart failure (HF). Recent therapeutic approaches (e.g., regenerative and nanomedicine) have shown promise to prevent HF postmyocardial infarction in animal models. However, these preclinical, clinical, and technological advancements have yet to yield substantial enhancements in the survival rate and quality of life of patients with severe ischemic injuries. This could be attributed largely to the considerable gap in knowledge between clinicians and nanobioengineers. Development of highly effective cardiac regenerative therapies requires connecting and coordinating multiple fields, including cardiology, cellular and molecular biology, biochemistry and chemistry, and mechanical and materials sciences, among others. This review is particularly intended to bridge the knowledge gap between cardiologists and regenerative nanomedicine experts. Establishing this multidisciplinary knowledge base may help pave the way for developing novel, safer, and more effective approaches that will enable the medical community to reduce morbidity and mortality in HF patients.
Collapse
Affiliation(s)
| | - Mehdi Mehrani
- Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Ahmad Amin
- Rajaie Cardiovascular, Medical and Research Center, Iran University of Medical Science Tehran, Iran
| | | | - Jessica C. Garbern
- Department of Stem Cell and Regenerative Biology, Harvard University, Harvard Stem Cell Institute, Cambridge, Massachusetts, United States
- Department of Cardiology, Boston Children’s Hospital, Boston, Massachusetts, United States
| | - Giulio Caracciolo
- Department of Molecular Medicine, Sapienza University of Rome, V.le Regina Elena 291, 00161, Rome, Italy
| | - Steven Zanganeh
- Department of Radiology, Memorial Sloan Kettering, New York, NY 10065, United States
| | - Mitra Chitsazan
- Rajaie Cardiovascular, Medical and Research Center, Iran University of Medical Science Tehran, Iran
| | - Haniyeh Aghaverdi
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham & Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Seyed Mehdi Kamali Shahri
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham & Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Aliakbar Ashkarran
- Precision Health Program, Michigan State University, East Lansing, MI, United States
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham & Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Mohammad Raoufi
- Physical Chemistry I, Department of Chemistry and Biology & Research Center of Micro and Nanochemistry and Engineering, University of Siegen, Siegen, Germany
| | - Holly Bauser-Heaton
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Jianyi Zhang
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Jochen D. Muehlschlegel
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham & Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Anna Moore
- Precision Health Program, Michigan State University, East Lansing, MI, United States
| | - Richard T. Lee
- Department of Stem Cell and Regenerative Biology, Harvard University, Harvard Stem Cell Institute, Cambridge, Massachusetts, United States
- Department of Medicine, Division of Cardiology, Brigham and Women’s Hospital and Harvard Medical School, Cambridge, Massachusetts, United States
| | - Joseph C. Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California, United States
- Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, California, United States
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, United States
| | - Vahid Serpooshan
- Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia, United States
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Morteza Mahmoudi
- Precision Health Program, Michigan State University, East Lansing, MI, United States
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham & Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Connors Center for Women’s Health & Gender Biology, Brigham & Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
192
|
Effects of Spaceflight on Human Induced Pluripotent Stem Cell-Derived Cardiomyocyte Structure and Function. Stem Cell Reports 2019; 13:960-969. [PMID: 31708475 PMCID: PMC6915842 DOI: 10.1016/j.stemcr.2019.10.006] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 10/09/2019] [Accepted: 10/10/2019] [Indexed: 12/21/2022] Open
Abstract
With extended stays aboard the International Space Station (ISS) becoming commonplace, there is a need to better understand the effects of microgravity on cardiac function. We utilized human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) to study the effects of microgravity on cell-level cardiac function and gene expression. The hiPSC-CMs were cultured aboard the ISS for 5.5 weeks and their gene expression, structure, and functions were compared with ground control hiPSC-CMs. Exposure to microgravity on the ISS caused alterations in hiPSC-CM calcium handling. RNA-sequencing analysis demonstrated that 2,635 genes were differentially expressed among flight, post-flight, and ground control samples, including genes involved in mitochondrial metabolism. This study represents the first use of hiPSC technology to model the effects of spaceflight on human cardiomyocyte structure and function.
Collapse
|
193
|
Kussauer S, David R, Lemcke H. hiPSCs Derived Cardiac Cells for Drug and Toxicity Screening and Disease Modeling: What Micro- Electrode-Array Analyses Can Tell Us. Cells 2019; 8:E1331. [PMID: 31661896 PMCID: PMC6912416 DOI: 10.3390/cells8111331] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/22/2019] [Accepted: 10/23/2019] [Indexed: 12/19/2022] Open
Abstract
Human induced pluripotent stem cell (iPSC)-derived cardiomyocytes (CM) have been intensively used in drug development and disease modeling. Since iPSC-cardiomyocyte (CM) was first generated, their characterization has become a major focus of research. Multi-/micro-electrode array (MEA) systems provide a non-invasive user-friendly platform for detailed electrophysiological analysis of iPSC cardiomyocytes including drug testing to identify potential targets and the assessment of proarrhythmic risk. Here, we provide a systematical overview about the physiological and technical background of micro-electrode array measurements of iPSC-CM. We introduce the similarities and differences between action- and field potential and the advantages and drawbacks of MEA technology. In addition, we present current studies focusing on proarrhythmic side effects of novel and established compounds combining MEA systems and iPSC-CM. MEA technology will help to open a new gateway for novel therapies in cardiovascular diseases while reducing animal experiments at the same time.
Collapse
Affiliation(s)
- Sophie Kussauer
- Department Cardiac Surgery, Medical Center, University of Rostock, 18057 Rostock, Germany.
| | - Robert David
- Department Cardiac Surgery, Medical Center, University of Rostock, 18057 Rostock, Germany.
| | - Heiko Lemcke
- Department Cardiac Surgery, Medical Center, University of Rostock, 18057 Rostock, Germany.
| |
Collapse
|
194
|
Guo F, Sun Y, Wang X, Wang H, Wang J, Gong T, Chen X, Zhang P, Su L, Fu G, Su J, Yang S, Lai R, Jiang C, Liang P. Patient-Specific and Gene-Corrected Induced Pluripotent Stem Cell-Derived Cardiomyocytes Elucidate Single-Cell Phenotype of Short QT Syndrome. Circ Res 2019; 124:66-78. [PMID: 30582453 DOI: 10.1161/circresaha.118.313518] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
RATIONALE Short QT syndrome (SQT) is a rare but arrhythmogenic disorder featured by shortened ventricular repolarization and a propensity toward life-threatening ventricular arrhythmias and sudden cardiac death. OBJECTIVE This study aimed to investigate the single-cell mechanism of SQT using patient-specific and gene-corrected induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). METHODS AND RESULTS One SQT patient carrying missense mutation T618I in potassium voltage-gated channel subfamily H member 2 ( KCNH2) was recruited as well as 2 healthy control subjects in this study. Control and SQT patient-specific iPSCs were generated from skin fibroblasts using nonintegrated Sendai virus. The KCNH2 T618I mutation was corrected by genome editing in SQT iPSC lines to generate isogenic controls. All iPSCs were differentiated into iPSC-CMs using monolayer-based differentiation protocol. SQT iPSC-CMs exhibited abnormal action potential phenotype featured by shortened action potential duration and increased beat-beat interval variability, when compared with control and gene-corrected iPSC-CMs. Furthermore, SQT iPSC-CMs showed KCNH2 gain-of-function with increased rapid delayed rectifying potassium current (IKr) density and enhanced membrane expression. Gene expression profiling of iPSC-CMs exhibited a differential cardiac ion-channel gene expression profile of SQT. Moreover, QTc of SQT patient and action potential durations of SQT iPSC-CMs were both normalized by quinidine, indicating that quinidine is beneficial to KCNH2 T618I of SQT. Importantly, shortened action potential duration phenotype observed in SQT iPSC-CMs was effectively rescued by a short-peptide scorpion toxin BmKKx2 with a mechanism of targeting KCNH2. CONCLUSIONS We demonstrate that patient-specific and gene-corrected iPSC-CMs are able to recapitulate single-cell phenotype of SQT, which is caused by the gain-of-function mutation KCNH2 T618I. These findings will help elucidate the mechanisms underlying SQT and discover therapeutic drugs for treating the disease by using peptide toxins as lead compounds.
Collapse
Affiliation(s)
- Fengfeng Guo
- From the Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, the First Affiliated Hospital (F.G., X.W., J.W., T.G., J.S., P.L.), Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yaxun Sun
- Department of Cardiology (Y.S., C.J.), Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaochen Wang
- From the Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, the First Affiliated Hospital (F.G., X.W., J.W., T.G., J.S., P.L.), Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Translational Medicine, Zhejiang University, Hangzhou, China (F.G., X.W., J.W., J.S., P.L.)
| | - Hao Wang
- Department of Prenatal Diagnosis (Screening) Center, Hangzhou Women's Hospital (Hangzhou Maternity and Child Health Care Hospital), China (H.W.)
| | - Jue Wang
- From the Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, the First Affiliated Hospital (F.G., X.W., J.W., T.G., J.S., P.L.), Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Translational Medicine, Zhejiang University, Hangzhou, China (F.G., X.W., J.W., J.S., P.L.)
| | - Tingyu Gong
- From the Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, the First Affiliated Hospital (F.G., X.W., J.W., T.G., J.S., P.L.), Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xianzhen Chen
- Department of Dermatology and Venerology (X.C.), Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ping Zhang
- Department of Cardiology, Beijing Tsinghua Changgeng Hospital, China (P.Z.)
| | - Lan Su
- Cardiovascular Medicine Department, The First Affiliated Hospital of Wenzhou Medical University, China (L.S.)
| | - Guosheng Fu
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China (F.G., X.W., J.W., J.S., P.L.)
| | - Jun Su
- From the Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, the First Affiliated Hospital (F.G., X.W., J.W., T.G., J.S., P.L.), Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Translational Medicine, Zhejiang University, Hangzhou, China (F.G., X.W., J.W., J.S., P.L.).,Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences (S.Y., L.R.), Kunming Institute of Zoology, China
| | - Shilong Yang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences (S.Y., L.R.), Kunming Institute of Zoology, China.,Key Laboratory of Bioactive Peptides of Yunnan Province (S.Y., L.R.), Kunming Institute of Zoology, China
| | - Ren Lai
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences (S.Y., L.R.), Kunming Institute of Zoology, China.,Key Laboratory of Bioactive Peptides of Yunnan Province (S.Y., L.R.), Kunming Institute of Zoology, China
| | - Chenyang Jiang
- Department of Cardiology (Y.S., C.J.), Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ping Liang
- From the Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, the First Affiliated Hospital (F.G., X.W., J.W., T.G., J.S., P.L.), Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Translational Medicine, Zhejiang University, Hangzhou, China (F.G., X.W., J.W., J.S., P.L.)
| |
Collapse
|
195
|
Berry JL, Zhu W, Tang YL, Krishnamurthy P, Ge Y, Cooke JP, Chen Y, Garry DJ, Yang HT, Rajasekaran NS, Koch WJ, Li S, Domae K, Qin G, Cheng K, Kamp TJ, Ye L, Hu S, Ogle BM, Rogers JM, Abel ED, Davis ME, Prabhu SD, Liao R, Pu WT, Wang Y, Ping P, Bursac N, Vunjak-Novakovic G, Wu JC, Bolli R, Menasché P, Zhang J. Convergences of Life Sciences and Engineering in Understanding and Treating Heart Failure. Circ Res 2019; 124:161-169. [PMID: 30605412 DOI: 10.1161/circresaha.118.314216] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
On March 1 and 2, 2018, the National Institutes of Health 2018 Progenitor Cell Translational Consortium, Cardiovascular Bioengineering Symposium, was held at the University of Alabama at Birmingham. Convergence of life sciences and engineering to advance the understanding and treatment of heart failure was the theme of the meeting. Over 150 attendees were present, and >40 scientists presented their latest work on engineering human functional myocardium for disease modeling, drug development, and heart failure research. The scientists, engineers, and physicians in the field of cardiovascular sciences met and discussed the most recent advances in their work and proposed future strategies for overcoming the major roadblocks of cardiovascular bioengineering and therapy. Particular emphasis was given for manipulation and using of stem/progenitor cells, biomaterials, and methods to provide molecular, chemical, and mechanical cues to cells to influence their identity and fate in vitro and in vivo. Collectively, these works are profoundly impacting and progressing toward deciphering the mechanisms and developing novel treatments for left ventricular dysfunction of failing hearts. Here, we present some important perspectives that emerged from this meeting.
Collapse
Affiliation(s)
- Joel L Berry
- From the Department of Biomedical Engineering (J.L.B., W.Z., P.K., G.Q., J.M.R., J.Z.), University of Alabama at Birmingham
| | - Wuqiang Zhu
- From the Department of Biomedical Engineering (J.L.B., W.Z., P.K., G.Q., J.M.R., J.Z.), University of Alabama at Birmingham
| | - Yao Liang Tang
- Vascular Biology Center, Medical College of Georgia, Augusta University (Y.T.)
| | - Prasanna Krishnamurthy
- From the Department of Biomedical Engineering (J.L.B., W.Z., P.K., G.Q., J.M.R., J.Z.), University of Alabama at Birmingham
| | - Ying Ge
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, (Y.G., T.J.K.)
| | - John P Cooke
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, TX (J.P.C.)
| | - Yabing Chen
- Department of Pathology (Y.C., N.S.R.), University of Alabama at Birmingham
| | - Daniel J Garry
- Lillehei Heart Institute, Department of Medicine, Division of Cardiology, University of Minnesota, Minneapolis (D.J.G.)
| | - Huang-Tian Yang
- Shanghai Institutes for Biological Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS), China (H.-T.Y.)
| | | | - Walter J Koch
- Department of Pharmacology, Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (W.J.K.)
| | - Song Li
- Department of Bioengineering, University of California at Los Angeles (S.L.)
| | - Keitaro Domae
- Department of Cardiovascular Surgery, Graduate School of Medicine, Osaka University, Japan (K.D.)
| | - Gangjian Qin
- From the Department of Biomedical Engineering (J.L.B., W.Z., P.K., G.Q., J.M.R., J.Z.), University of Alabama at Birmingham
| | - Ke Cheng
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh (K.C.)
| | - Timothy J Kamp
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, (Y.G., T.J.K.)
| | - Lei Ye
- National Heart Research Institute Singapore, National Heart Centre Singapore (L.Y.)
| | - Shijun Hu
- Institute for Cardiovascular Science, Medical College of Soochow University, Suzhou, China (S.H.)
| | - Brenda M Ogle
- Department of Biomedical Engineering, University of Minnesota-Twin Cities, Minneapolis, MN (B.M.O.)
| | - Jack M Rogers
- From the Department of Biomedical Engineering (J.L.B., W.Z., P.K., G.Q., J.M.R., J.Z.), University of Alabama at Birmingham
| | - E Dale Abel
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine (E.D.A.)
| | - Michael E Davis
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech College of Engineering and Emory University School of Medicine, Atlanta (M.E.D.)
| | - Sumanth D Prabhu
- Division of Cardiovascular Disease and Comprehensive Cardiovascular Center, Department of Medicine (S.D.P.), University of Alabama at Birmingham
| | - Ronglih Liao
- Stanford Cardiovascular Institute, Department of Medicine, Stanford University School of Medicine, CA (R.L., J.C.W.)
| | - William T Pu
- Department of Cardiology, Boston Children's Hospital, MA (W.T.P.)
| | - Yibin Wang
- Department of Anesthesiology and Medicine (Y.W.), David Geffen School of Medicine, University of California, Los Angeles
| | - Peipei Ping
- Department of Physiology (P.P.), David Geffen School of Medicine, University of California, Los Angeles
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC (N.B.)
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering and Department of Medicine, Columbia University, New York City, NY (G.V.-N.)
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Department of Medicine, Stanford University School of Medicine, CA (R.L., J.C.W.)
| | - Roberto Bolli
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY (R.B.)
| | - Philippe Menasché
- Department of Cardiovascular Surgery, Hôpital Européen Georges Pompidou, Paris, France (P.M.)
| | - Jianyi Zhang
- From the Department of Biomedical Engineering (J.L.B., W.Z., P.K., G.Q., J.M.R., J.Z.), University of Alabama at Birmingham
| |
Collapse
|
196
|
Devalla HD, Passier R. Cardiac differentiation of pluripotent stem cells and implications for modeling the heart in health and disease. Sci Transl Med 2019; 10:10/435/eaah5457. [PMID: 29618562 DOI: 10.1126/scitranslmed.aah5457] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 07/15/2016] [Accepted: 06/20/2017] [Indexed: 12/21/2022]
Abstract
Cellular models comprising cardiac cell types derived from human pluripotent stem cells are valuable for studying heart development and disease. We discuss transcriptional differences that define cellular identity in the heart, current methods for generating different cardiomyocyte subtypes, and implications for disease modeling, tissue engineering, and regenerative medicine.
Collapse
Affiliation(s)
- Harsha D Devalla
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZC Leiden, Netherlands.
| | - Robert Passier
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZC Leiden, Netherlands. .,Department of Applied Stem Cell Technologies, Technical Medical Center, University of Twente, 7500 AE Enschede, Netherlands
| |
Collapse
|
197
|
Kumar N, Dougherty JA, Manring HR, Elmadbouh I, Mergaye M, Czirok A, Greta Isai D, Belevych AE, Yu L, Janssen PML, Fadda P, Gyorke S, Ackermann MA, Angelos MG, Khan M. Assessment of temporal functional changes and miRNA profiling of human iPSC-derived cardiomyocytes. Sci Rep 2019; 9:13188. [PMID: 31515494 PMCID: PMC6742647 DOI: 10.1038/s41598-019-49653-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 07/31/2019] [Indexed: 12/22/2022] Open
Abstract
Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have been developed for cardiac cell transplantation studies more than a decade ago. In order to establish the hiPSC-CM-based platform as an autologous source for cardiac repair and drug toxicity, it is vital to understand the functionality of cardiomyocytes. Therefore, the goal of this study was to assess functional physiology, ultrastructural morphology, gene expression, and microRNA (miRNA) profiling at Wk-1, Wk-2 & Wk-4 in hiPSC-CMs in vitro. Functional assessment of hiPSC-CMs was determined by multielectrode array (MEA), Ca2+ cycling and particle image velocimetry (PIV). Results demonstrated that Wk-4 cardiomyocytes showed enhanced synchronization and maturation as compared to Wk-1 & Wk-2. Furthermore, ultrastructural morphology of Wk-4 cardiomyocytes closely mimicked the non-failing (NF) adult human heart. Additionally, modulation of cardiac genes, cell cycle genes, and pluripotency markers were analyzed by real-time PCR and compared with NF human heart. Increasing expression of fatty acid oxidation enzymes at Wk-4 supported the switching to lipid metabolism. Differential regulation of 12 miRNAs was observed in Wk-1 vs Wk-4 cardiomyocytes. Overall, this study demonstrated that Wk-4 hiPSC-CMs showed improved functional, metabolic and ultrastructural maturation, which could play a crucial role in optimizing timing for cell transplantation studies and drug screening.
Collapse
Affiliation(s)
- Naresh Kumar
- Department of Emergency Medicine, Dorothy M. Davis Heart Lung and Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Julie A Dougherty
- Department of Emergency Medicine, Dorothy M. Davis Heart Lung and Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Heather R Manring
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Ibrahim Elmadbouh
- Department of Emergency Medicine, Dorothy M. Davis Heart Lung and Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Muhamad Mergaye
- Department of Emergency Medicine, Dorothy M. Davis Heart Lung and Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Andras Czirok
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Dona Greta Isai
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Andriy E Belevych
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Lianbo Yu
- Center for Biostatistics, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Paul M L Janssen
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Paolo Fadda
- Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Sandor Gyorke
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Maegen A Ackermann
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Mark G Angelos
- Department of Emergency Medicine, Dorothy M. Davis Heart Lung and Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Mahmood Khan
- Department of Emergency Medicine, Dorothy M. Davis Heart Lung and Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA. .,Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
198
|
Brodehl A, Ebbinghaus H, Deutsch MA, Gummert J, Gärtner A, Ratnavadivel S, Milting H. Human Induced Pluripotent Stem-Cell-Derived Cardiomyocytes as Models for Genetic Cardiomyopathies. Int J Mol Sci 2019; 20:ijms20184381. [PMID: 31489928 PMCID: PMC6770343 DOI: 10.3390/ijms20184381] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 08/29/2019] [Accepted: 09/03/2019] [Indexed: 12/17/2022] Open
Abstract
In the last few decades, many pathogenic or likely pathogenic genetic mutations in over hundred different genes have been described for non-ischemic, genetic cardiomyopathies. However, the functional knowledge about most of these mutations is still limited because the generation of adequate animal models is time-consuming and challenging. Therefore, human induced pluripotent stem cells (iPSCs) carrying specific cardiomyopathy-associated mutations are a promising alternative. Since the original discovery that pluripotency can be artificially induced by the expression of different transcription factors, various patient-specific-induced pluripotent stem cell lines have been generated to model non-ischemic, genetic cardiomyopathies in vitro. In this review, we describe the genetic landscape of non-ischemic, genetic cardiomyopathies and give an overview about different human iPSC lines, which have been developed for the disease modeling of inherited cardiomyopathies. We summarize different methods and protocols for the general differentiation of human iPSCs into cardiomyocytes. In addition, we describe methods and technologies to investigate functionally human iPSC-derived cardiomyocytes. Furthermore, we summarize novel genome editing approaches for the genetic manipulation of human iPSCs. This review provides an overview about the genetic landscape of inherited cardiomyopathies with a focus on iPSC technology, which might be of interest for clinicians and basic scientists interested in genetic cardiomyopathies.
Collapse
Affiliation(s)
- Andreas Brodehl
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Hans Ebbinghaus
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Marcus-André Deutsch
- Department of Thoracic and Cardiovascular Surgery, Heart and Diabetes Center NRW, University Hospital Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Jan Gummert
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
- Department of Thoracic and Cardiovascular Surgery, Heart and Diabetes Center NRW, University Hospital Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Anna Gärtner
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Sandra Ratnavadivel
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Hendrik Milting
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| |
Collapse
|
199
|
Clippinger SR, Cloonan PE, Greenberg L, Ernst M, Stump WT, Greenberg MJ. Disrupted mechanobiology links the molecular and cellular phenotypes in familial dilated cardiomyopathy. Proc Natl Acad Sci U S A 2019; 116:17831-17840. [PMID: 31427533 PMCID: PMC6731759 DOI: 10.1073/pnas.1910962116] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Familial dilated cardiomyopathy (DCM) is a leading cause of sudden cardiac death and a major indicator for heart transplant. The disease is frequently caused by mutations of sarcomeric proteins; however, it is not well understood how these molecular mutations lead to alterations in cellular organization and contractility. To address this critical gap in our knowledge, we studied the molecular and cellular consequences of a DCM mutation in troponin-T, ΔK210. We determined the molecular mechanism of ΔK210 and used computational modeling to predict that the mutation should reduce the force per sarcomere. In mutant cardiomyocytes, we found that ΔK210 not only reduces contractility but also causes cellular hypertrophy and impairs cardiomyocytes' ability to adapt to changes in substrate stiffness (e.g., heart tissue fibrosis that occurs with aging and disease). These results help link the molecular and cellular phenotypes and implicate alterations in mechanosensing as an important factor in the development of DCM.
Collapse
Affiliation(s)
- Sarah R Clippinger
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110
| | - Paige E Cloonan
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110
| | - Lina Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110
| | - Melanie Ernst
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110
| | - W Tom Stump
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110
| | - Michael J Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
200
|
Reda SM, Chandra M. Dilated cardiomyopathy mutation (R174W) in troponin T attenuates the length-mediated increase in cross-bridge recruitment and myofilament Ca 2+ sensitivity. Am J Physiol Heart Circ Physiol 2019; 317:H648-H657. [PMID: 31373515 DOI: 10.1152/ajpheart.00171.2019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Alterations in length-dependent activation (LDA) may constitute a mechanism by which cardiomyopathy mutations lead to deleterious phenotypes and compromised heart function, because LDA underlies the molecular basis by which the heart tunes myocardial force production on a beat-to-beat basis (Frank-Starling mechanism). In this study, we investigated the effect of DCM-linked mutation (R173W) in human cardiac troponin T (TnT) on myofilament LDA. R173W mutation is associated with left ventricular dilatation and systolic dysfunction and is found in multiple families. R173W mutation is in the central region (residues 80-180) of TnT, which is known to be important for myofilament cooperativity and cross-bridge (XB) recruitment. Steady-state and dynamic contractile parameters were measured in detergent-skinned guinea pig left ventricular muscle fibers reconstituted with recombinant guinea pig wild-type TnT (TnTWT) or mutant TnT (TnTR174W; guinea pig analog of human R173W mutation) at two different sarcomere lengths (SL): short (1.9 µm) and long (2.3 µm). TnTR174W decreased pCa50 (-log [Ca2+]free required for half-maximal activation) to a greater extent at long than at short SL; for example, pCa50 decreased by 0.12 pCa units at long SL and by 0.06 pCa units at short SL. Differential changes in pCa50 at short and long SL attenuated the SL-dependent increase in myofilament Ca2+ sensitivity (ΔpCa50) in TnTR174W fibers; ΔpCa50 was 0.10 units in TnTWT fibers but only 0.04 units in TnTR174W fibers. Furthermore, TnTR174W blunted the SL-dependent increase in the magnitude of XB recruitment. Our observations suggest that the R173W mutation in human cardiac TnT may impair Frank-Starling mechanism.NEW & NOTEWORTHY This work characterizes the effect of dilated cardiomyopathy mutation in cardiac troponin T (TnTR174W) on myofilament length-dependent activation. TnTR174W attenuates the length-dependent increase in cross-bridge recruitment and myofilament Ca2+ sensitivity.
Collapse
Affiliation(s)
- Sherif M Reda
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Murali Chandra
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| |
Collapse
|