151
|
Fleck TPK, Ayala R, Kroll J, Siepe M, Schibilsky D, Benk C, Maier S, Reineker K, Hoehn R, Humburger F, Beyersdorf F, Stiller B. Ex Vivo Allograft Perfusion for Complex Pediatric Heart Transplant Recipients. Ann Thorac Surg 2021; 112:1275-1280. [PMID: 33421388 DOI: 10.1016/j.athoracsur.2020.12.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/18/2020] [Accepted: 12/21/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND Pediatric heart transplant (HTx) recipients with congenital heart defects require complex concomitant surgical procedures with the risk of prolonging the allograft's ischemic time. Ex vivo allograft perfusion with the Organ Care System (OCS; Transmedics, Andover, MA) may improve survival of these challenging patients. METHODS In this retrospective, single-center study a consecutive series of 8 children with allografts preserved using the OCS was compared with 13 children after HTx with cold storage of the donor heart from March 2018 to March 2020. RESULTS Median recipient age in the control group was 18 months (range, 1-189) versus 155 months (range, 83-214) in the OCS group, and the baseline differences between the 2 groups were not significant. Fifty percent of the children in the OCS group had complex congenital heart defects (vs 15% of the control subjects). Median operation time during HTx in the OCS group was 616 minutes (range, 270-809) versus 329 minutes (range, 283-617). Because of the time of ex vivo allograft perfusion (265 minutes [range, 202-372]) median total ischemia time was significantly shorter in the OCS group: 78 minutes (range, 52-111) versus 222 minutes (range, 74-326). The incidence of primary graft, renal, or hepatic failure did not differ between the groups. Graft function and the occurrence of any treated rejection at follow-up revealed no significant difference between the 2 groups. One-year survival was 88% in the OCS group (vs 85%). CONCLUSIONS Ex vivo allograft perfusion enabled complex pediatric HTx, yielding outcomes as positive as those of children whose donor hearts were stored in ice-cold solution.
Collapse
Affiliation(s)
- Thilo P K Fleck
- Department of Congenital Heart Disease and Pediatric Cardiology, University Heart Center Freiburg - Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Rafael Ayala
- Department of Cardiovascular Surgery, University Heart Center Freiburg - Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Johannes Kroll
- Department of Cardiovascular Surgery, University Heart Center Freiburg - Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Matthias Siepe
- Department of Cardiovascular Surgery, University Heart Center Freiburg - Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - David Schibilsky
- Department of Cardiovascular Surgery, University Heart Center Freiburg - Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christoph Benk
- Department of Cardiovascular Surgery, University Heart Center Freiburg - Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sven Maier
- Department of Cardiovascular Surgery, University Heart Center Freiburg - Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Katja Reineker
- Department of Congenital Heart Disease and Pediatric Cardiology, University Heart Center Freiburg - Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - René Hoehn
- Department of Congenital Heart Disease and Pediatric Cardiology, University Heart Center Freiburg - Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Frank Humburger
- Deparment of Anesthesiology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Friedhelm Beyersdorf
- Department of Cardiovascular Surgery, University Heart Center Freiburg - Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Brigitte Stiller
- Department of Congenital Heart Disease and Pediatric Cardiology, University Heart Center Freiburg - Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
152
|
Karagiannis P, Yoshida Y. Making Cardiomyocytes from Pluripotent Stem Cells. Methods Mol Biol 2021; 2320:3-7. [PMID: 34302642 DOI: 10.1007/978-1-0716-1484-6_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The ability to differentiate pluripotent stem cells to cardiomyocyte lineages (PSC-CMs) has opened the door to new disease models and innovative drug and cell therapies for the heart. Nevertheless, further advances in the differentiation protocols are needed to fulfill the promise of PSC-CMs. Obstacles that remain include deriving PSC-CMs with proper electromechanical properties, coalescing them into functional tissue structures, and manipulating the genome to test the impact mutations have on arrhythmias and other heart disorders. This chapter gives a brief consideration of these challenges and outlines current methodologies that offer partial solutions.
Collapse
Affiliation(s)
- Peter Karagiannis
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Yoshinori Yoshida
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan.
| |
Collapse
|
153
|
Perucca G, de Lange C, Franchi-Abella S, Napolitano M, Riccabona M, Ključevšek D, Toso S, Herrmann J, Stafrace S, Darge K, Damasio MB, Bruno C, Woźniak MM, Lobo L, Ibe D, Smets AM, Petit P, Ording Müller LS. Surveillance of Fontan-associated liver disease: current standards and a proposal from the European Society of Paediatric Radiology Abdominal Task Force. Pediatr Radiol 2021; 51:2598-2606. [PMID: 34654967 PMCID: PMC8599216 DOI: 10.1007/s00247-021-05173-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/08/2021] [Accepted: 07/31/2021] [Indexed: 12/16/2022]
Abstract
Since Francis Fontan first introduced the eponymous technique, the Fontan procedure, this type of surgical palliation has allowed thousands of children affected by specific heart malformations to reach adulthood. Nevertheless, abdominal, thoracic, lymphatic and neurologic complications are the price that is paid by these patients. Our review focuses on Fontan-associated liver disease; the purpose is to summarize the current understanding of its physiopathology, the aim of follow-up and the specific radiologic follow-up performed in Europe. Finally, we as members of the Abdominal Task Force of the European Society of Paediatric Radiology propose a consensus-based imaging follow-up algorithm.
Collapse
Affiliation(s)
- Giulia Perucca
- Department of Pediatric Radiology, Regina Margherita Children’s Hospital, Turin, Italy
| | - Charlotte de Lange
- Department of Radiology and Clinical Physiology, Queen Silvia Children’s Hospital, Sahlgrenska University Hospital, Göteborg, Sweden
| | - Stéphanie Franchi-Abella
- Pediatric Radiology Department, Hôpital Bicêtre, Hôpitaux Universitaire Paris-Sud, Assistance Publique Hôpitaux de Paris, Le Kremlin-Bicêtre, France
| | - Marcello Napolitano
- Department of Paediatric Radiology and Neuroradiology, V. Buzzi Children’s Hospital, Milan, Italy
| | - Michael Riccabona
- Department of Radiology, Division of Pediatric Radiology, Medical University Graz and University Hospital LKH, Graz, Austria
| | - Damjana Ključevšek
- Department of Radiology, University Children’s Hospital Ljubljana, Ljubljana, Slovenia
| | - Seema Toso
- Department of Pediatric Radiology, University Hospital of Geneva, Geneva, Switzerland
| | - Jochen Herrmann
- Department of Pediatric Radiology, University Hospital Hamburg Eppendorf, Hamburg, Germany
| | - Samuel Stafrace
- Department of Diagnostic Imaging, Sidra Medicine, Doha, Qatar ,Weill Cornell Medicine, Doha, Qatar
| | - Kassa Darge
- Department of Radiology, The Children’s Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA USA
| | | | - Costanza Bruno
- Department of Radiology, Azienda Ospedaliera Universitaria Integrata Verona (AOUI), Verona, Italy
| | | | - Luisa Lobo
- Serviço de Imagiologia Geral, Hospital de Santa Maria–Centro Hospitalar Universitário Lisboa, Norte (CHULN), Lisbon, Portugal
| | - Donald Ibe
- Department of Radiology, Silhouette Diagnostic Consultants, Abuja, Nigeria
| | - Anne M. Smets
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Philippe Petit
- Aix Marseille Université, AP-HM, Equipe d’Accueil 3279 - IFR 125, Hôpital Timone Enfants, Service d’Imagerie Pédiatrique et Prénatale, Marseille, France
| | - Lil-Sofie Ording Müller
- Unit for Paediatric Radiology, Department of Radiology, Oslo University Hospital, Rikshospitalet, PB 4950 Nydalen, 0424 Oslo, Norway.
| |
Collapse
|
154
|
Sridharan D, Palaniappan A, Blackstone BN, Dougherty JA, Kumar N, Seshagiri PB, Sayed N, Powell HM, Khan M. In situ differentiation of human-induced pluripotent stem cells into functional cardiomyocytes on a coaxial PCL-gelatin nanofibrous scaffold. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 118:111354. [PMID: 33254974 PMCID: PMC7708677 DOI: 10.1016/j.msec.2020.111354] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 08/03/2020] [Accepted: 08/05/2020] [Indexed: 12/20/2022]
Abstract
Human-induced pluripotent stem cells (hiPSCs) derived cardiomyocytes (hiPSC-CMs) have been explored for cardiac regeneration and repair as well as for the development of in vitro 3D cardiac tissue models. Existing protocols for cardiac differentiation of hiPSCs utilize a 2D culture system. However, the efficiency of hiPSC differentiation to cardiomyocytes in 3D culture systems has not been extensively explored. In the present study, we investigated the efficiency of cardiac differentiation of hiPSCs to functional cardiomyocytes on 3D nanofibrous scaffolds. Coaxial polycaprolactone (PCL)-gelatin fibrous scaffolds were fabricated by electrospinning and characterized using scanning electron microscopy (SEM) and fourier transform infrared (FTIR) spectroscopy. hiPSCs were cultured and differentiated into functional cardiomyocytes on the nanofibrous scaffold and compared with 2D cultures. To assess the relative efficiencies of both the systems, SEM, immunofluorescence staining and gene expression analyses were performed. Contractions of differentiated cardiomyocytes were observed in 2D cultures after 2 weeks and in 3D cultures after 4 weeks. SEM analysis showed no significant differences in the morphology of cells differentiated on 2D versus 3D cultures. However, gene expression data showed significantly increased expression of cardiac progenitor genes (ISL-1, SIRPA) in 3D cultures and cardiomyocytes markers (TNNT, MHC6) in 2D cultures. In contrast, immunofluorescence staining showed no substantial differences in the expression of NKX-2.5 and α-sarcomeric actinin. Furthermore, uniform migration and distribution of the in situ differentiated cardiomyocytes was observed in the 3D fibrous scaffold. Overall, our study demonstrates that coaxial PCL-gelatin nanofibrous scaffolds can be used as a 3D culture platform for efficient differentiation of hiPSCs to functional cardiomyocytes.
Collapse
Affiliation(s)
- Divya Sridharan
- Department of Emergency Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Arunkumar Palaniappan
- Department of Emergency Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, USA; Centre for Biomaterials, Cellular and Molecular Theranostics, Vellore Institute of Technology, Vellore, India
| | - Britani N Blackstone
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH, USA
| | - Julie A Dougherty
- Department of Emergency Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, USA; Dorothy M. Davis Heart & Lung Research Institute, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Naresh Kumar
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
| | - Polani B Seshagiri
- Department of Molecular Reproduction Development and Genetics, Indian Institute of Science, C V Raman Road, Bangalore KA-560012, India
| | - Nazish Sayed
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Heather M Powell
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH, USA; Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA; Research Department, Shriners Hospitals for Children, Cincinnati, OH, USA
| | - Mahmood Khan
- Department of Emergency Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, USA; Department of Physiology and Cell Biology, Wexner Medical Center, The Ohio State University, Columbus, OH, USA; Dorothy M. Davis Heart & Lung Research Institute, Wexner Medical Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
155
|
de Lange C, Thrane KJ, Thomassen KS, Geier O, Nguyen B, Tomterstad A, Ording Müller LS, Thaulow E, Almaas R, Døhlen G, Suther KR, Möller T. Hepatic magnetic resonance T1-mapping and extracellular volume fraction compared to shear-wave elastography in pediatric Fontan-associated liver disease. Pediatr Radiol 2021; 51:66-76. [PMID: 33033916 PMCID: PMC7796890 DOI: 10.1007/s00247-020-04805-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 06/10/2020] [Accepted: 08/10/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Children with Fontan circulation are at risk of developing hepatic fibrosis/cirrhosis. Reliable noninvasive monitoring techniques are lacking or under development. OBJECTIVE To investigate surrogate indicators of hepatic fibrosis in adolescents with Fontan circulation by evaluating hepatic magnetic resonance (MR) T1 mapping and extracellular volume fraction measurements compared to US shear-wave elastography. MATERIALS AND METHODS We analyzed hepatic native T1 times and extracellular volume fractions with modified Look-Locker inversion recovery. Liver stiffness was analyzed with shear-wave elastography. We compared results between 45 pediatric patients ages 16.7±0.6 years with Fontan circulation and 15 healthy controls ages 19.2±1.2 years. Measurements were correlated to clinical and hemodynamic data from cardiac catheterization. RESULTS MR mapping was successful in 35/45 patients, revealing higher hepatic T1 times (774±44 ms) than in controls (632±52 ms; P<0.001) and higher extracellular volume fractions (47.4±5.0%) than in controls (34.6±3.8%; P<0.001). Liver stiffness was 1.91±0.13 m/s in patients vs. 1.20±0.10 m/s in controls (P<0.001). Native T1 times correlated with central venous pressures (r=0.5, P=0.007). Native T1 was not correlated with elastography in patients (r=0.2, P=0.1) or controls (r = -0.3, P=0.3). Extracellular volume fraction was correlated with elastography in patients (r=0.5, P=0.005) but not in controls (r=0.2, P=0.6). CONCLUSION Increased hepatic MR relaxometry and shear-wave elastography values in adolescents with Fontan circulation suggested the presence of hepatic fibrosis or congestion. Central venous pressure was related to T1 times. Changes were detected differently with MR relaxometry and elastography; thus, these techniques should not be used interchangeably in monitoring hepatic fibrosis.
Collapse
Affiliation(s)
- Charlotte de Lange
- Division of Radiology and Nuclear Medicine, Section of Paediatric Radiology, Oslo University Hospital, Oslo, Norway. .,Department of Radiology and Clinical Physiology, Queen Silvia Children's Hospital, Sahlgrenska University Hospital, Rondv 10, S-41615, Göteborg, Sweden.
| | - Karl Julius Thrane
- Division of Radiology and Nuclear Medicine, Section of Paediatric Radiology, Oslo University Hospital, Oslo, Norway
| | - Kristian S. Thomassen
- Division of Radiology and Nuclear Medicine, Section of Paediatric Radiology, Oslo University Hospital, Oslo, Norway
| | - Oliver Geier
- Department of Physics, Oslo University Hospital, Oslo, Norway
| | - Bac Nguyen
- Division of Radiology and Nuclear Medicine, Section of Paediatric Radiology, Oslo University Hospital, Oslo, Norway
| | - Anders Tomterstad
- Division of Radiology and Nuclear Medicine, Section of Paediatric Radiology, Oslo University Hospital, Oslo, Norway
| | - Lil-Sofie Ording Müller
- Division of Radiology and Nuclear Medicine, Section of Paediatric Radiology, Oslo University Hospital, Oslo, Norway
| | - Erik Thaulow
- Department of Paediatric Cardiology, Oslo University Hospital, Oslo, Norway ,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Runar Almaas
- Department of Paediatric Research and Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
| | - Gaute Døhlen
- Department of Paediatric Cardiology, Oslo University Hospital, Oslo, Norway
| | - Kathrine Rydén Suther
- Division of Radiology and Nuclear Medicine, Section of Paediatric Radiology, Oslo University Hospital, Oslo, Norway
| | - Thomas Möller
- Department of Paediatric Cardiology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
156
|
Ellis BW, Traktuev DO, Merfeld-Clauss S, Can UI, Wang M, Bergeron R, Zorlutuna P, March KL. Adipose stem cell secretome markedly improves rodent heart and human induced pluripotent stem cell-derived cardiomyocyte recovery from cardioplegic transport solution exposure. STEM CELLS (DAYTON, OHIO) 2020; 39:170-182. [PMID: 33159685 DOI: 10.1002/stem.3296] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 10/18/2020] [Indexed: 12/21/2022]
Abstract
Heart transplantation is a life-saving therapy for end-stage organ failure. Organ deterioration during transportation limits storage to 4 hours, limiting hearts available. Approaches ameliorating organ damage could increase the number of hearts acceptable for transplantation. Prior studies show that adipose-derived stem/stromal cell secretome (ASC-S) rescues tissues from postischemic damage in vivo. This study tested whether ASC-S preserved the function of mouse hearts and human induced pluripotent stem cell-derived cardiomyocytes (iCM) exposed to organ transportation and transplantation conditions. Hearts were subjected to cold University of Wisconsin (UW) cardioplegic solution ± ASC-S for 6 hours followed by analysis using the Langendorff technique. In parallel, the effects of ASC-S on the recovery of iCM from UW solution were examined when provided either during or after cold cardioplegia. Exposure of hearts and iCM to UW deteriorated contractile activity and caused cell apoptosis, worsening in iCM as a function of exposure time; these were ameliorated by augmenting with ASC-S. Silencing of superoxide dismutase 3 and catalase expression prior to secretome generation compromised the ASC-S cardiomyocyte-protective effects. In this study, a novel in vitro iCM model was developed to complement a rodent heart model in assessing efficacy of approaches to improve cardiac preservation. ASC-S displays strong cardioprotective activity on iCM either with or following cold cardioplegia. This effect is associated with ASC-S-mediated cellular clearance of reactive oxygen species. The effect of ASC-S on the temporal recovery of iCM function supports the possibility of lengthening heart storage by augmenting cardioplegic transport solution with ASC-S, expanding the pool of hearts for transplantation.
Collapse
Affiliation(s)
- Bradley W Ellis
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana, USA
| | - Dmitry O Traktuev
- Division of Cardiovascular Medicine and Center for Regenerative Medicine, University of Florida, Gainesville, Florida, USA.,Malcom Randall Veterans' Affairs Medical Center, Gainesville, Florida, USA
| | - Stephanie Merfeld-Clauss
- Division of Cardiovascular Medicine and Center for Regenerative Medicine, University of Florida, Gainesville, Florida, USA.,Malcom Randall Veterans' Affairs Medical Center, Gainesville, Florida, USA
| | - Uryan Isik Can
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana, USA
| | - Meijing Wang
- The Division of Cardiothoracic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ray Bergeron
- Division of Cardiovascular Medicine and Center for Regenerative Medicine, University of Florida, Gainesville, Florida, USA
| | - Pinar Zorlutuna
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana, USA.,Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana, USA
| | - Keith L March
- Division of Cardiovascular Medicine and Center for Regenerative Medicine, University of Florida, Gainesville, Florida, USA.,Malcom Randall Veterans' Affairs Medical Center, Gainesville, Florida, USA
| |
Collapse
|
157
|
Pawan KC, Mickey S, Rubia S, Yi H, Ge Z. Preseeding of Mesenchymal Stem Cells Increases Integration of an iPSC-Derived CM Sheet into a Cardiac Matrix. ACS Biomater Sci Eng 2020; 6:6808-6818. [PMID: 33320624 PMCID: PMC9841440 DOI: 10.1021/acsbiomaterials.0c00788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Cell sheet technology has demonstrated great promise in delivering a large amount of therapeutic cells for tissue repair, including in the myocardium. However, the lack of host integration remains one of the key challenges in using cell sheets for cardiac repair. Paracrine factors secreted by mesenchymal stem cells (MSCs) have been reported to facilitate tissue repair and regeneration in a variety of ways. It has been demonstrated that paracrine factors from MSCs could enhance scaffold recellularization and vascularization. In this study, we used an in vitro cardiac matrix mimic platform to examine the effects of hMSCs preseeding on the interactions between cell sheets and cardiac matrix. The fabricated human induced pluripotent stem cells-derived cardiomyocyte sheets were attached to a decellularized porcine myocardium slice with or without preseeding of hMSCs. The hMSCs preseeding significantly enhanced the interactions between cardiomyocyte sheets and cardiac matrix in terms of cell migration distance, cell distribution, and mature vascular and cardiomyocyte marker expressions in the matrix. Growth factor and matrix metalloproteinases array analysis suggested that hMSCs- induced vascularization and MMPs regulation are the two possible mechanisms that lead to the improved CMs and cardiac matrix interactions. Further examination of these two mechanisms will enable the development of new approaches to facilitate transplanted cells for tissue repair.
Collapse
Affiliation(s)
- KC Pawan
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Shah Mickey
- Department of Biomedical Engineering and Department of Integrated Bioscience, The University of Akron, Akron, Ohio 44325, United States
| | - Shaik Rubia
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Hong Yi
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Zhang Ge
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio 44325, United States
| |
Collapse
|
158
|
Orban M, Ulrich S, Dischl D, von Samson-Himmelstjerna P, Schramm R, Tippmann K, Hein-Rothweiler R, Strüven A, Lehner A, Braun D, Hausleiter J, Jakob A, Fischer M, Hagl C, Haas N, Massberg S, Mehilli J, Robert DP. Cardiac allograft vasculopathy: Differences of absolute and relative intimal hyperplasia in children versus adults in optical coherence tomography. Int J Cardiol 2020; 328:227-234. [PMID: 33316256 DOI: 10.1016/j.ijcard.2020.12.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/01/2020] [Accepted: 12/04/2020] [Indexed: 01/06/2023]
Abstract
BACKGROUND Intracoronary imaging enables an early detection of intimal changes. To what extend the development of absolute and relative intimal hyperplasia in intracoronary imaging differs depending on age and post-transplant time is not known. METHODS Aim of our retrospective study was to compare findings between 24 pediatric (cohort P) and 21 adult HTx patients (cohort A) using optical coherence tomography (OCT) at corresponding post-transplant intervals (≤5 years: P1 (n = 11) and A1 (n = 10); >5 and ≤ 10 years: P2 (n = 13) and A2 (n = 11),. Coronary intima thickness (IT), media thickness (MT) and intima to media ratio (I/M) were assessed per quadrant. Maximal IT >0.3 mm was considered absolute, I/M > 1 relative intimal hyperplasia. RESULTS Compared to A1, I/M was significantly higher in P1 (maximal I/M: P1: 5.41 [2.81-13.39] vs. A1: 2.30 [1.55-3.62], p = 0.005), whereas absolute IT values were comparable. In contrast, I/M was comparable between P2 and A2, but absolute IT were significantly higher in A2 (maximal IT: P2: 0.16 mm [0.11-0.25] vs. A2: 0.40 mm [0.30-0.71], p < 0.001). A2 presented with higher absolute IT (maximal: A1: 0.16 mm [0.12-0.44] vs. A2: 0.40 mm [0.30-0.71], p = 0.02) and I/M (maximal I/M A1: 2.30 [1.55-3.62] vs. A2: 3.79 [3.01-5.62], p = 0.04). CONCLUSION Our results suggest an age- and time-dependent difference in the prevalence of absolute and relative intimal hyperplasia in OCT, with an early peak in children and a progressive increase in adults.
Collapse
Affiliation(s)
- Madeleine Orban
- Department of Cardiology, Ludwig-Maximilians-University, Klinikum Großhadern, Munich, Germany; Partner Site Munich Heart Alliance, German Centre for Cardiovascular Research (DZHK), Germany.
| | - Sarah Ulrich
- Department of Pediatric Cardiology and Intensive Care Medicine, Ludwig-Maximilians-University, Klinikum Großhadern, Munich, Germany
| | - Dominic Dischl
- Department of Cardiology, Ludwig-Maximilians-University, Klinikum Großhadern, Munich, Germany
| | | | - René Schramm
- Department of Heart Surgery, Ludwig-Maximilians-University, Klinikum Großhadern, Munich, Germany
| | - Katharina Tippmann
- Department of Cardiology, Ludwig-Maximilians-University, Klinikum Großhadern, Munich, Germany
| | - Ralph Hein-Rothweiler
- Department of Cardiology, Ludwig-Maximilians-University, Klinikum Großhadern, Munich, Germany
| | - Anna Strüven
- Department of Cardiology, Ludwig-Maximilians-University, Klinikum Großhadern, Munich, Germany
| | - Anja Lehner
- Department of Pediatric Cardiology and Intensive Care Medicine, Ludwig-Maximilians-University, Klinikum Großhadern, Munich, Germany
| | - Daniel Braun
- Department of Cardiology, Ludwig-Maximilians-University, Klinikum Großhadern, Munich, Germany
| | - Jörg Hausleiter
- Department of Cardiology, Ludwig-Maximilians-University, Klinikum Großhadern, Munich, Germany; Partner Site Munich Heart Alliance, German Centre for Cardiovascular Research (DZHK), Germany
| | - Andre Jakob
- Department of Pediatric Cardiology and Intensive Care Medicine, Ludwig-Maximilians-University, Klinikum Großhadern, Munich, Germany
| | - Marcus Fischer
- Department of Pediatric Cardiology and Intensive Care Medicine, Ludwig-Maximilians-University, Klinikum Großhadern, Munich, Germany
| | - Christian Hagl
- Department of Heart Surgery, Ludwig-Maximilians-University, Klinikum Großhadern, Munich, Germany; Partner Site Munich Heart Alliance, German Centre for Cardiovascular Research (DZHK), Germany
| | - Nikolaus Haas
- Department of Pediatric Cardiology and Intensive Care Medicine, Ludwig-Maximilians-University, Klinikum Großhadern, Munich, Germany
| | - Steffen Massberg
- Department of Cardiology, Ludwig-Maximilians-University, Klinikum Großhadern, Munich, Germany; Partner Site Munich Heart Alliance, German Centre for Cardiovascular Research (DZHK), Germany
| | - Julinda Mehilli
- Department of Cardiology, Ludwig-Maximilians-University, Klinikum Großhadern, Munich, Germany; Partner Site Munich Heart Alliance, German Centre for Cardiovascular Research (DZHK), Germany
| | - Dalla Pozza Robert
- Department of Pediatric Cardiology and Intensive Care Medicine, Ludwig-Maximilians-University, Klinikum Großhadern, Munich, Germany
| |
Collapse
|
159
|
Associated Factors of Liver Disease After Fontan Operation in Relation to Ultrasound Liver Elastography. Pediatr Cardiol 2020; 41:1639-1644. [PMID: 32740670 DOI: 10.1007/s00246-020-02422-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 07/21/2020] [Indexed: 10/23/2022]
Abstract
Although the Fontan operation can improve outcomes, surviving patients still face Fontan-associated liver disease (FALD). The aim of this study was to determine the associated factors of FALD in relation to ultrasound liver elastography. A cross-sectional study was conducted for all patients on whom an ultrasound upper-abdomen and ultrasound liver elastography was performed at more than 1 year after the Fontan operation. The data consisted of the age at operation, type of Fontan operation, and laboratory data such as gamma-glutamyl transferase (GGT), aspartate aminotransferase-to-platelet ratio index (APRI), and fibrosis-4 (FIB-4) score. Cardiovascular evaluations included echocardiography and cardiac catheterization. Eighty patients with a median age of 12 years (range 5-36 years) were eligible for the study. The prevalence of FALD was 41%. For the purpose of univariate logistic regression analysis, the age at the Fontan operation, time elapsed since the Fontan operation, previous Glenn shunt, presence of fenestration, mean pulmonary artery pressure and IVC pressure post-Fontan operation, platelet count, GGT, and FIB-4 score were considered to be factors significantly associated with FALD. Following an adjustment by multivariate logistic regression analysis, age greater than 7 years at the time of Fontan procedure, time elapsed of more than 9 years since the procedure, and GGT level > 130 U/L were found to be significantly associated with FALD. Patients with these factors should be investigated for FALD.
Collapse
|
160
|
Mycophenolate Improves Brain-Gut Axis Inducing Remodeling of Gut Microbiota in DOCA-Salt Hypertensive Rats. Antioxidants (Basel) 2020; 9:antiox9121199. [PMID: 33260593 PMCID: PMC7761232 DOI: 10.3390/antiox9121199] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 11/26/2020] [Accepted: 11/27/2020] [Indexed: 12/17/2022] Open
Abstract
Microbiota is involved in the host blood pressure (BP) regulation. The immunosuppressive drug mofetil mycophenolate (MMF) ameliorates hypertension. The present study analyzed whether MMF improves dysbiosis in mineralocorticoid-induced hypertension. Male Wistar rats were assigned to three groups: untreated (CTR), deoxycorticosterone acetate (DOCA)-salt, and DOCA treated with MMF for 4 weeks. MMF treatment reduced systolic BP, improved endothelial dysfunction, and reduced oxidative stress and inflammation in aorta. A clear separation in the gut bacterial community between CTR and DOCA groups was found, whereas the cluster belonging to DOCA-MMF group was found to be intermixed. No changes were found at the phylum level among all experimental groups. MMF restored the elevation in lactate-producing bacteria found in DOCA-salt joined to an increase in the acetate-producing bacteria. MMF restored the percentage of anaerobic bacteria in the DOCA-salt group to values similar to control rats. The improvement of gut dysbiosis was associated with an enhanced colonic integrity and a decreased sympathetic drive in the gut. MMF inhibited neuroinflammation in the paraventricular nuclei in the hypothalamus. This study demonstrates for the first time that MMF reduces gut dysbiosis in DOCA-salt hypertension models. This effect seems to be related to its capacity to improve gut integrity due to reduced sympathetic drive in the gut associated with reduced brain neuroinflammation.
Collapse
|
161
|
Baghai Arassi M, Zeller G, Karcher N, Zimmermann M, Toenshoff B. The gut microbiome in solid organ transplantation. Pediatr Transplant 2020; 24:e13866. [PMID: 32997434 DOI: 10.1111/petr.13866] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/06/2020] [Accepted: 09/02/2020] [Indexed: 02/06/2023]
Abstract
Despite ground-breaking advances in allogeneic transplantation, allograft rejection and immunosuppressant-specific complications remain a major challenge in transplant medicine. Growing evidence suggests the human gut microbiome as a potential contributor to transplant outcome and patient health. After breakthrough findings in haematopoietic stem cell transplantation (HSCT), the relevance of the microbiome in solid organ transplantation (SOT) is becoming increasingly clear. Here, we review the role of the microbiome in SOT focusing on its significance for transplant-associated complications such as allograft rejection and infections, and highlight its potential impact on immunosuppressive treatment. Moreover, we shed light on the emerging role of the microbiome as a diagnostic biomarker and therapeutic target and discuss current microbial intervention strategies. In addition, this review includes some practical considerations in designing clinical microbiome trials and offers some advice for the interpretation of the resulting data. Further investigation of the gut microbiome harbours countless clinical application possibilities and holds great promise of having a lasting impact on transplant medicine.
Collapse
Affiliation(s)
- Maral Baghai Arassi
- Department of Paediatrics I, University Children's Hospital Heidelberg, Heidelberg, Germany.,Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Georg Zeller
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Nicolai Karcher
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Michael Zimmermann
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Burkhard Toenshoff
- Department of Paediatrics I, University Children's Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
162
|
Ehren R, Schijvens AM, Hackl A, Schreuder MF, Weber LT. Therapeutic drug monitoring of mycophenolate mofetil in pediatric patients: novel techniques and current opinion. Expert Opin Drug Metab Toxicol 2020; 17:201-213. [PMID: 33107768 DOI: 10.1080/17425255.2021.1843633] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Introduction: Mycophenolate mofetil (MMF) is an ester prodrug of the immunosuppressant mycophenolic acid (MPA) and is recommended and widely used for maintenance immunosuppressive therapy in solid organ and stem-cell transplantation as well as in immunological kidney diseases. MPA is a potent, reversible, noncompetitive inhibitor of the inosine monophosphate dehydrogenase (IMPDH), a crucial enzyme in the de novo purine synthesis in T- and B-lymphocytes, thereby inhibiting cell-mediated immunity and antibody formation. The use of therapeutic drug monitoring (TDM) of MMF is still controversial as outcome data of clinical trials are equivocal. Areas covered: This review covers in great depth the existing literature on TDM of MMF in the field of pediatric (kidney) transplantation. In addition, the relevance of TDM in immunological kidney diseases, in particular childhood nephrotic syndrome is highlighted. Expert opinion: TDM of MMF has the potential to optimize therapy in pediatric transplantation as well as in nephrotic syndrome. Limited sampling strategies to estimate MPA exposure increase its feasibility. Future perspectives rather encompass approaches reflecting total immunosuppressive load than single drug TDM.
Collapse
Affiliation(s)
- Rasmus Ehren
- Faculty of Medicine and University Hospital Cologne, Department of Pediatrics, University of Cologne , Cologne, Germany
| | - Anne M Schijvens
- Department of Pediatric Nephrology, Amalia Children's Hospital, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen, The Netherlands
| | - Agnes Hackl
- Faculty of Medicine and University Hospital Cologne, Department of Pediatrics, University of Cologne , Cologne, Germany
| | - Michiel F Schreuder
- Department of Pediatric Nephrology, Amalia Children's Hospital, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen, The Netherlands
| | - Lutz T Weber
- Faculty of Medicine and University Hospital Cologne, Department of Pediatrics, University of Cologne , Cologne, Germany
| |
Collapse
|
163
|
Jiang L, Liang J, Huang W, Wu Z, Paul C, Wang Y. Strategies and Challenges to Improve Cellular Programming-Based Approaches for Heart Regeneration Therapy. Int J Mol Sci 2020; 21:E7662. [PMID: 33081233 PMCID: PMC7589611 DOI: 10.3390/ijms21207662] [Citation(s) in RCA: 3] [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: 09/30/2020] [Revised: 10/13/2020] [Accepted: 10/15/2020] [Indexed: 12/29/2022] Open
Abstract
Limited adult cardiac cell proliferation after cardiovascular disease, such as heart failure, hampers regeneration, resulting in a major loss of cardiomyocytes (CMs) at the site of injury. Recent studies in cellular reprogramming approaches have provided the opportunity to improve upon previous techniques used to regenerate damaged heart. Using these approaches, new CMs can be regenerated from differentiation of iPSCs (similar to embryonic stem cells), the direct reprogramming of fibroblasts [induced cardiomyocytes (iCMs)], or induced cardiac progenitors. Although these CMs have been shown to functionally repair infarcted heart, advancements in technology are still in the early stages of development in research laboratories. In this review, reprogramming-based approaches for generating CMs are briefly introduced and reviewed, and the challenges (including low efficiency, functional maturity, and safety issues) that hinder further translation of these approaches into a clinical setting are discussed. The creative and combined optimal methods to address these challenges are also summarized, with optimism that further investigation into tissue engineering, cardiac development signaling, and epigenetic mechanisms will help to establish methods that improve cell-reprogramming approaches for heart regeneration.
Collapse
Affiliation(s)
- Lin Jiang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, OH 45267-0529, USA
| | - Jialiang Liang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, OH 45267-0529, USA
| | - Wei Huang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, OH 45267-0529, USA
| | - Zhichao Wu
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, OH 45267-0529, USA
| | - Christian Paul
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, OH 45267-0529, USA
| | - Yigang Wang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, OH 45267-0529, USA
| |
Collapse
|
164
|
Mota C, Camarero-Espinosa S, Baker MB, Wieringa P, Moroni L. Bioprinting: From Tissue and Organ Development to in Vitro Models. Chem Rev 2020; 120:10547-10607. [PMID: 32407108 PMCID: PMC7564098 DOI: 10.1021/acs.chemrev.9b00789] [Citation(s) in RCA: 142] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Indexed: 02/08/2023]
Abstract
Bioprinting techniques have been flourishing in the field of biofabrication with pronounced and exponential developments in the past years. Novel biomaterial inks used for the formation of bioinks have been developed, allowing the manufacturing of in vitro models and implants tested preclinically with a certain degree of success. Furthermore, incredible advances in cell biology, namely, in pluripotent stem cells, have also contributed to the latest milestones where more relevant tissues or organ-like constructs with a certain degree of functionality can already be obtained. These incredible strides have been possible with a multitude of multidisciplinary teams around the world, working to make bioprinted tissues and organs more relevant and functional. Yet, there is still a long way to go until these biofabricated constructs will be able to reach the clinics. In this review, we summarize the main bioprinting activities linking them to tissue and organ development and physiology. Most bioprinting approaches focus on mimicking fully matured tissues. Future bioprinting strategies might pursue earlier developmental stages of tissues and organs. The continuous convergence of the experts in the fields of material sciences, cell biology, engineering, and many other disciplines will gradually allow us to overcome the barriers identified on the demanding path toward manufacturing and adoption of tissue and organ replacements.
Collapse
Affiliation(s)
- Carlos Mota
- Department of Complex Tissue Regeneration,
MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Sandra Camarero-Espinosa
- Department of Complex Tissue Regeneration,
MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Matthew B. Baker
- Department of Complex Tissue Regeneration,
MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Paul Wieringa
- Department of Complex Tissue Regeneration,
MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Lorenzo Moroni
- Department of Complex Tissue Regeneration,
MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6211 LK Maastricht, The Netherlands
| |
Collapse
|
165
|
Nasser MI, Qi X, Zhu S, He Y, Zhao M, Guo H, Zhu P. Current situation and future of stem cells in cardiovascular medicine. Biomed Pharmacother 2020; 132:110813. [PMID: 33068940 DOI: 10.1016/j.biopha.2020.110813] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 09/22/2020] [Accepted: 09/25/2020] [Indexed: 12/21/2022] Open
Abstract
Cardiovascular disease (CVD) is one of the leading causes of death worldwide. Currently, many methods have been proposed by researchers for the prevention and treatment of CVD; among them, stem cell-based therapies are the most promising. As the cells of origin for various mature cells, stem cells have the ability to self-renew and differentiate. Stem cells have a powerful ability to regenerate biologically, self-repair, and enhance damaged functional tissues or organs. Allogeneic stem cells and somatic stem cells are two types of cells that can be used for cardiac repair. Theoretically, dilated cardiomyopathy and acute myocardial infarction can be treated with such cells. In addition, stem cell transplantation procedures, including intravenous, epicardial, cardiac, and endocardial injections, have been reported to provide significant benefits in clinical practice; however, there are still a number of issues that need further study and consideration, such as the form and quantity of transplanted cells and post-transplantation health. The goal of this analysis was to summarize the recent advances in stem cell-based therapies and their efficacy in cardiovascular regenerative medicine.
Collapse
Affiliation(s)
- M I Nasser
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510100, China. Address: 106 Zhongshan Er Road, Guangzhou, 510080, PR China
| | - Xiao Qi
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510100, China. Address: 106 Zhongshan Er Road, Guangzhou, 510080, PR China
| | - Shuoji Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510100, China. Address: 106 Zhongshan Er Road, Guangzhou, 510080, PR China
| | - Yin He
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510100, China. Address: 106 Zhongshan Er Road, Guangzhou, 510080, PR China
| | - Mingyi Zhao
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510100, China. Address: 106 Zhongshan Er Road, Guangzhou, 510080, PR China
| | - Huiming Guo
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510100, China. Address: 106 Zhongshan Er Road, Guangzhou, 510080, PR China
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510100, China. Address: 106 Zhongshan Er Road, Guangzhou, 510080, PR China.
| |
Collapse
|
166
|
Single-cell protein expression of hiPSC-derived cardiomyocytes using Single-Cell Westerns. J Mol Cell Cardiol 2020; 149:115-122. [PMID: 33010256 DOI: 10.1016/j.yjmcc.2020.09.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 09/21/2020] [Accepted: 09/25/2020] [Indexed: 11/22/2022]
Abstract
The ability to reprogram human somatic cells into human induced pluripotent stem cells (hiPSCs) has enabled researchers to generate cell types in vitro that have the potential to faithfully recapitulate patient-specific disease processes and phenotypes. hiPSC-derived cardiomyocytes (hiPSC-CMs) offer the promise of in vitro patient- and disease-specific models for drug testing and the discovery of novel therapeutic approaches for treating cardiovascular diseases. While methods to differentiate hiPSCs into cardiomyocytes have been demonstrated, the heterogeneity and immaturity of these differentiated populations have restricted their potential in reproducing human disease and the associated target cell phenotypes. These barriers may be overcome through comprehensive single-cell characterization to dissect the rich heterogeneity of hiPSC-CMs and to study the source of varying cell fates. In this study, we optimized and validated a new Single-Cell Western method to assess protein expression in hiPSC-CMs. To better understand distinct subpopulations generated from cardiomyocyte differentiations and to track populations at single-cell resolution over time, we measured and quantified the expression of cardiomyocyte subtype-specific proteins (MLC2V and MLC2A) using Single-Cell Westerns. By understanding their heterogeneity through single-cell protein expression and quantification, we may improve upon current cardiomyocyte differentiation protocols, generate hiPSC-CMs that are more representative of in vivo derived cardiomyocytes for disease modeling, and utilize hiPSC-CMs for regenerative medicine purposes. Single-Cell Westerns provide a robust platform for protein expression analysis at single-cell resolution.
Collapse
|
167
|
Dame K, Ribeiro AJ. Microengineered systems with iPSC-derived cardiac and hepatic cells to evaluate drug adverse effects. Exp Biol Med (Maywood) 2020; 246:317-331. [PMID: 32938227 PMCID: PMC7859673 DOI: 10.1177/1535370220959598] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hepatic and cardiac drug adverse effects are among the leading causes of attrition in drug development programs, in part due to predictive failures of current animal or in vitro models. Hepatocytes and cardiomyocytes differentiated from human induced pluripotent stem cells (iPSCs) hold promise for predicting clinical drug effects, given their human-specific properties and their ability to harbor genetically determined characteristics that underlie inter-individual variations in drug response. Currently, the fetal-like properties and heterogeneity of hepatocytes and cardiomyocytes differentiated from iPSCs make them physiologically different from their counterparts isolated from primary tissues and limit their use for predicting clinical drug effects. To address this hurdle, there have been ongoing advances in differentiation and maturation protocols to improve the quality and use of iPSC-differentiated lineages. Among these are in vitro hepatic and cardiac cellular microsystems that can further enhance the physiology of cultured cells, can be used to better predict drug adverse effects, and investigate drug metabolism, pharmacokinetics, and pharmacodynamics to facilitate successful drug development. In this article, we discuss how cellular microsystems can establish microenvironments for these applications and propose how they could be used for potentially controlling the differentiation of hepatocytes or cardiomyocytes. The physiological relevance of cells is enhanced in cellular microsystems by simulating properties of tissue microenvironments, such as structural dimensionality, media flow, microfluidic control of media composition, and co-cultures with interacting cell types. Recent studies demonstrated that these properties also affect iPSC differentiations and we further elaborate on how they could control differentiation efficiency in microengineered devices. In summary, we describe recent advances in the field of cellular microsystems that can control the differentiation and maturation of hepatocytes and cardiomyocytes for drug evaluation. We also propose how future research with iPSCs within engineered microenvironments could enable their differentiation for scalable evaluations of drug effects.
Collapse
Affiliation(s)
- Keri Dame
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translation Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Alexandre Js Ribeiro
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translation Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA
| |
Collapse
|
168
|
Iwamiya T, Segard BD, Matsuoka Y, Imamura T. Human cardiac fibroblasts expressing VCAM1 improve heart function in postinfarct heart failure rat models by stimulating lymphangiogenesis. PLoS One 2020; 15:e0237810. [PMID: 32936824 PMCID: PMC7494079 DOI: 10.1371/journal.pone.0237810] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 08/02/2020] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular diseases are a leading cause of death worldwide. After an ischemic injury, the myocardium undergoes severe necrosis and apoptosis, leading to a dramatic degradation of function. Numerous studies have reported that cardiac fibroblasts (CFs) play a critical role in heart function even after injury. However, CFs present heterogeneous characteristics according to their development stage (i.e., fetal or adult), and the molecular mechanisms by which they maintain heart function are not fully understood. The aim of this study is to explore the hypothesis that a specific population of CFs can repair the injured myocardium in heart failure following ischemic infarction, and lead to a significant recovery of cardiac function. Flow cytometry analysis of CFs defined two subpopulations according to their relative expression of vascular cell adhesion molecule 1 (VCAM1). Whole-transcriptome analysis described distinct profiles for these groups, with a correlation between VCAM1 expression and lymphangiogenesis-related genes up-regulation. Vascular formation assays showed a significant stimulation of lymphatic cells network complexity by VCFs. Injection of human VCAM1-expressing CFs (VCFs) in postinfarct heart failure rat models (ligation of the left anterior descending artery) led to a significant restoration of the left ventricle contraction. Over the course of the experiment, left ventricular ejection fraction and fractional shortening increased by 16.65% ± 5.64% and 10.43% ± 6.02%, respectively, in VCF-treated rats. Histological examinations revealed that VCFs efficiently mobilized the lymphatic endothelial cells into the infarcted area. In conclusion, human CFs present heterogeneous expression of VCAM1 and lymphangiogenesis-promoting factors. VCFs restore the mechanical properties of ventricular walls by mobilizing lymphatic endothelial cells into the infarct when injected into a rat heart failure model. These results suggest a role of this specific population of CFs in the homeostasis of the lymphatic system in cardiac regeneration, providing new information for the study and therapy of cardiac diseases.
Collapse
Affiliation(s)
- Takahiro Iwamiya
- Research & Development Department, Metcela Inc., Kawasaki, Kanagawa, Japan
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan
- * E-mail:
| | | | - Yuimi Matsuoka
- Research & Development Department, Metcela Inc., Kawasaki, Kanagawa, Japan
| | - Tomomi Imamura
- Research & Development Department, Metcela Inc., Kawasaki, Kanagawa, Japan
| |
Collapse
|
169
|
Kumar N, Sridharan D, Palaniappan A, Dougherty JA, Czirok A, Isai DG, Mergaye M, Angelos MG, Powell HM, Khan M. Scalable Biomimetic Coaxial Aligned Nanofiber Cardiac Patch: A Potential Model for "Clinical Trials in a Dish". Front Bioeng Biotechnol 2020; 8:567842. [PMID: 33042968 PMCID: PMC7525187 DOI: 10.3389/fbioe.2020.567842] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 08/13/2020] [Indexed: 12/12/2022] Open
Abstract
Recent advances in cardiac tissue engineering have shown that human induced-pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) cultured in a three-dimensional (3D) micro-environment exhibit superior physiological characteristics compared with their two-dimensional (2D) counterparts. These 3D cultured hiPSC-CMs have been used for drug testing as well as cardiac repair applications. However, the fabrication of a cardiac scaffold with optimal biomechanical properties and high biocompatibility remains a challenge. In our study, we fabricated an aligned polycaprolactone (PCL)-Gelatin coaxial nanofiber patch using electrospinning. The structural, chemical, and mechanical properties of the patch were assessed by scanning electron microscopy (SEM), immunocytochemistry (ICC), Fourier-transform infrared spectroscopy (FTIR)-spectroscopy, and tensile testing. hiPSC-CMs were cultured on the aligned coaxial patch for 2 weeks and their viability [lactate dehydrogenase (LDH assay)], morphology (SEM, ICC), and functionality [calcium cycling, multielectrode array (MEA)] were assessed. Furthermore, particle image velocimetry (PIV) and MEA were used to evaluate the cardiotoxicity and physiological functionality of the cells in response to cardiac drugs. Nanofibers patches were comprised of highly aligned core-shell fibers with an average diameter of 578 ± 184 nm. Acellular coaxial patches were significantly stiffer than gelatin alone with an ultimate tensile strength of 0.780 ± 0.098 MPa, but exhibited gelatin-like biocompatibility. Furthermore, hiPSC-CMs cultured on the surface of these aligned coaxial patches (3D cultures) were elongated and rod-shaped with well-organized sarcomeres, as observed by the expression of cardiac troponin-T and α-sarcomeric actinin. Additionally, hiPSC-CMs cultured on these coaxial patches formed a functional syncytium evidenced by the expression of connexin-43 (Cx-43) and synchronous calcium transients. Moreover, MEA analysis showed that the hiPSC-CMs cultured on aligned patches showed an improved response to cardiac drugs like Isoproterenol (ISO), Verapamil (VER), and E4031, compared to the corresponding 2D cultures. Overall, our results demonstrated that an aligned, coaxial 3D cardiac patch can be used for culturing of hiPSC-CMs. These biomimetic cardiac patches could further be used as a potential 3D in vitro model for "clinical trials in a dish" and for in vivo cardiac repair applications for treating myocardial infarction.
Collapse
Affiliation(s)
- Naresh Kumar
- Department of Emergency Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Divya Sridharan
- Department of Emergency Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Arunkumar Palaniappan
- Department of Emergency Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Centre for Biomaterials, Cellular and Molecular Theranostics, Vellore Institute of Technology, Vellore, India
| | - Julie A. Dougherty
- Department of Emergency Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Andras Czirok
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Dona Greta Isai
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, United States
| | - Muhamad Mergaye
- Department of Emergency Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Mark G. Angelos
- Department of Emergency Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Heather M. Powell
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, United States
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH, United States
- Research Department, Shriners Hospitals for Children, Cincinnati, OH, United States
| | - Mahmood Khan
- Department of Emergency Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
170
|
Taşdemir D, Aksoy N. Weight Gain, Energy Intake, Energy Expenditure, and Immunosuppressive Therapy in Kidney Transplant Recipients. Prog Transplant 2020; 30:322-328. [PMID: 32930038 DOI: 10.1177/1526924820958150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Weight gain after kidney transplantation is a common health problem. The factors in weight gain after kidney transplant include many factors such as age, ethnicity, gender, change in lifestyle (eg, kilocalorie intake and physical activity level), and immunosuppressive therapy. RESEARCH QUESTIONS This study aimed to evaluate the relationship between weight gain and energy intake in dietary, energy expenditure in physical activity, and immunosuppressive therapy in kidney transplant recipients. DESIGN This prospective, observational study included 51 participants who underwent kidney transplant, during 6 months from the start of the study. Anthropometric measurements were performed at first week, third- and sixth-month follow-ups of transplant recipients. Participants also completed 3-day "Dietary Record Form" and the "Physical Activity Record Form" at each follow-up. Simple frequency, analysis of variance analysis, and correlation analysis were used for data analysis. RESULTS Weight gain in sixth month follow-up compared to baseline value was positively related to energy intake in first week (r = 0.59), third month (r = 0.75), and sixth month (r = 0.67) follow-ups, and energy expenditure in first week (r = 0.35) and sixth month (r = 0.34) follow-ups. However, weight gain was negatively related to mycophenolate mofetil dose (mg/d) in sixth month (r = -0.31) follow-up (P < .05). DISCUSSION The results of this study provide an opportunity to reflect and discuss on modifiable risk factors such as energy intake and energy expenditure that affect weight gain posttransplantation in participants. It also examines the relationship between immunosuppressive therapy. Additionally, these results can be effective in designing interventions and managing risk factors to achieve weight management goals.
Collapse
Affiliation(s)
- Deniz Taşdemir
- Faculty of Nursing, Department of Surgical Nursing, 37502Akdeniz University, Antalya, Turkey
| | - Nilgün Aksoy
- Faculty of Nursing, Department of Surgical Nursing, 37502Akdeniz University, Antalya, Turkey
| |
Collapse
|
171
|
Lippi M, Stadiotti I, Pompilio G, Sommariva E. Human Cell Modeling for Cardiovascular Diseases. Int J Mol Sci 2020; 21:E6388. [PMID: 32887493 PMCID: PMC7503257 DOI: 10.3390/ijms21176388] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 08/28/2020] [Accepted: 08/31/2020] [Indexed: 11/17/2022] Open
Abstract
The availability of appropriate and reliable in vitro cell models recapitulating human cardiovascular diseases has been the aim of numerous researchers, in order to retrace pathologic phenotypes, elucidate molecular mechanisms, and discover therapies using simple and reproducible techniques. In the past years, several human cell types have been utilized for these goals, including heterologous systems, cardiovascular and non-cardiovascular primary cells, and embryonic stem cells. The introduction of induced pluripotent stem cells and their differentiation potential brought new prospects for large-scale cardiovascular experiments, bypassing ethical concerns of embryonic stem cells and providing an advanced tool for disease modeling, diagnosis, and therapy. Each model has its advantages and disadvantages in terms of accessibility, maintenance, throughput, physiological relevance, recapitulation of the disease. A higher level of complexity in diseases modeling has been achieved with multicellular co-cultures. Furthermore, the important progresses reached by bioengineering during the last years, together with the opportunities given by pluripotent stem cells, have allowed the generation of increasingly advanced in vitro three-dimensional tissue-like constructs mimicking in vivo physiology. This review provides an overview of the main cell models used in cardiovascular research, highlighting the pros and cons of each, and describing examples of practical applications in disease modeling.
Collapse
Affiliation(s)
- Melania Lippi
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.L.); (I.S.); (G.P.)
| | - Ilaria Stadiotti
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.L.); (I.S.); (G.P.)
| | - Giulio Pompilio
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.L.); (I.S.); (G.P.)
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy
| | - Elena Sommariva
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.L.); (I.S.); (G.P.)
| |
Collapse
|
172
|
Khoury M, McCrindle BW. The Rationale, Indications, Safety, and Use of Statins in the Pediatric Population. Can J Cardiol 2020; 36:1372-1383. [PMID: 32735868 DOI: 10.1016/j.cjca.2020.03.041] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/16/2020] [Accepted: 03/26/2020] [Indexed: 10/24/2022] Open
Abstract
Together with heart-healthy lifestyle habits, statins serve as the cornerstone of primary and secondary prevention of atherosclerotic cardiovascular disease in adults. Several conditions, most notably familial hypercholesterolemia (FH), cause early dyslipidemia and vascular disease, contributing to the development and progression of atherosclerosis from childhood and increased cardiovascular risk. In recent decades, studies increasingly have evaluated the safety and efficacy of statins in such high-risk youth. The strongest evidence for pediatric statin use is for the heterozygous FH population, whereby statin use has been shown to lower low-density lipoprotein cholesterol effectively, slow the progression of atherosclerosis and vascular dysfunction, and significantly reduce cardiovascular risk in early adulthood. Numerous meta-analyses and Cochrane reviews have demonstrated that attributed adverse effects, including liver toxicity, myositis, and rhabdomyolysis, occur no more frequently in youth receiving statins than placebos, with no impact on growth or development. However, further studies evaluating the long-term safety of pediatric statin use are required. In the current review, we summarize the pediatric experience of statin use to date, focusing on its utility for FH, Kawasaki disease, post-heart transplantation, and other at-risk populations. Current guidelines and indications for use are summarized, and the short- and medium-term safety experience is reviewed. Finally, a clinical approach to the indications, initiation, and monitoring of statins in youth is provided.
Collapse
Affiliation(s)
- Michael Khoury
- Division of Pediatric Cardiology, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada.
| | - Brian W McCrindle
- Labatt Family Heart Center, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
173
|
Sivaraj S, Chan A, Pasini E, Chen E, Lawendy B, Verna E, Watt K, Bhat M. Enteric dysbiosis in liver and kidney transplant recipients: a systematic review. Transpl Int 2020; 33:1163-1176. [PMID: 32640109 DOI: 10.1111/tri.13696] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/10/2020] [Accepted: 07/03/2020] [Indexed: 12/17/2022]
Abstract
Several factors mediate intestinal microbiome (IM) alterations in transplant recipients, including immunosuppressive (IS) and antimicrobial drugs. Studies on the structure and function of the IM in the post-transplant scenario and its role in the development of metabolic abnormalities, infection, and cancer are limited. We conducted a systematic review to study the taxonomic changes in liver (LT) and kidney (KT) transplantation, and their potential contribution to post-transplant complications. The review also includes pre-transplant taxa, which may play a critical role in microbial alterations post-transplant. Two reviewers independently screened articles, and assessed risk of bias. The review identified 13 clinical studies, which focused on adult kidney and liver transplant recipients. Patient characteristics and methodologies varied widely between studies. Ten studies reported increased an abundance of opportunistic pathogens (Enterobacteriaceae, Enterococcaceae, Fusobacteriaceae, and Streptococcaceae) followed by butyrate-producing bacteria (Lachnospiraceae and Ruminococcaceae) in nine studies in post-transplant conditions. The current evidence is mostly based on observational data and studies with no proof of causality. Therefore, further studies exploring the bacterial gene functions rather than taxonomic changes alone are in demand to better understand the potential contribution of the IM in post-transplant complications.
Collapse
Affiliation(s)
- Saranya Sivaraj
- Multi Organ Transplant Program, University Health Network, Toronto, ON, Canada
| | - Anita Chan
- Multi Organ Transplant Program, University Health Network, Toronto, ON, Canada
| | - Elisa Pasini
- Multi Organ Transplant Program, University Health Network, Toronto, ON, Canada
| | - Emily Chen
- Multi Organ Transplant Program, University Health Network, Toronto, ON, Canada
| | - Bishoy Lawendy
- Multi Organ Transplant Program, University Health Network, Toronto, ON, Canada
| | - Elizabeth Verna
- Division of Digestive and Liver Diseases, Center for Liver Disease and Transplantation, Columbia University Irving Medical Center, New York, NY, USA
| | - Kymberly Watt
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Mamatha Bhat
- Multi Organ Transplant Program, University Health Network, Toronto, ON, Canada.,Division of Gastroenterology and Hepatology, University Health Network and University of Toronto, Toronto, ON, Canada
| |
Collapse
|
174
|
Chen CC, Wong TY, Chin TY, Lee WH, Kuo CY, Hsu YC. Systems biology approach to exploring the effect of cyclic stretching on cardiac cell physiology. Aging (Albany NY) 2020; 12:16035-16045. [PMID: 32759460 PMCID: PMC7485730 DOI: 10.18632/aging.103465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 05/27/2020] [Indexed: 01/10/2023]
Abstract
Although mechanical forces are involved in pressure-overloaded cardiomyopathy, their effects on gene transcription profiles are not fully understood. Here, we used next-generation sequencing (NGS) to investigate changes in genomic profiles after cyclic mechanical stretching of human cardiomyocytes. We found that 85, 87, 32, 29, and 28 genes were differentially expressed after 1, 4, 12, 24, and 48 hours of stretching. Furthermore, 10 of the 29 genes that were up-regulated and 11 of the 28 that were down-regulated after 24 h showed the same changes after 48 h. We then examined expression of the genes that encode serpin family E member 1 (SERPINE1), DNA-binding protein inhibitor 1 (ID1), DNA-binding protein inhibitor 3 (ID3), and CCL2, a cytokine that acts as chemotactic factor in monocytes, in an RT-PCR experiment. The same changes were observed for all four genes after all cyclic stretching durations, confirming the NGS results. Taken together, these findings suggest that cyclical stretching can alter cardiac cell physiology by activating cardiac cell metabolism and impacting cholesterol biosynthesis signaling.
Collapse
Affiliation(s)
- Chien-Cheng Chen
- Department of Cardiology, Show Chwan Memorial Hospital, Changhua, Taiwan
| | - Tzyy-Yue Wong
- International Center for Wound Repair and Regeneration National Cheng Kung University, Tainan, Taiwan
| | - Tzu-Yun Chin
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan
| | - Wen-Hsien Lee
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Internal Medicine, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chan-Yen Kuo
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| | - Yi-Chiung Hsu
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan
| |
Collapse
|
175
|
Fontan-associated liver disease: pathophysiology, investigations, predictors of severity and management. Eur J Gastroenterol Hepatol 2020; 32:907-915. [PMID: 31851099 DOI: 10.1097/meg.0000000000001641] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cardiac hepatopathy is the liver injury resulting from congestion and ischaemia associated with acute or chronic heart failure. The improved longevity of adults with operated congenital heart disease who develop heart failure as an increasingly late event makes this form of liver injury increasingly clinically relevant. Patients with congenital heart disease with a single ventricle anomaly, who require creation of a Fontan circulation, are particularly vulnerable as they have elevated venous filling pressures with chronic liver congestion. Progression to liver fibrosis and eventually cirrhosis may occur, with its associated risks of liver failure and hepatocellular carcinoma. This risk likely increases over the patient's lifetime, related to the duration post-surgical repair and reflects the chronicity of congestion. Liver biopsy is rarely performed due to a higher risk of complications in the setting of elevated venous pressures, and the frequent use of anticoagulation. Non-invasive methods of liver assessment are poorly validated and different factors require consideration compared to other chronic liver diseases. This review discusses the current understanding of cardiac hepatopathy in congenital heart disease patients with a Fontan circulation. This entity has recently been called Fontan Associated Liver Disease in the literature, with the term useful in recognising that the pathophysiology is incompletely understood, and that long-standing venous pressure elevation and hypoxaemia are presumed to play an additional significant role in the pathogenesis of the liver injury.
Collapse
|
176
|
Williams B, Löbel W, Finklea F, Halloin C, Ritzenhoff K, Manstein F, Mohammadi S, Hashemi M, Zweigerdt R, Lipke E, Cremaschi S. Prediction of Human Induced Pluripotent Stem Cell Cardiac Differentiation Outcome by Multifactorial Process Modeling. Front Bioeng Biotechnol 2020; 8:851. [PMID: 32793579 PMCID: PMC7390976 DOI: 10.3389/fbioe.2020.00851] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/02/2020] [Indexed: 12/12/2022] Open
Abstract
Human cardiomyocytes (CMs) have potential for use in therapeutic cell therapy and high-throughput drug screening. Because of the inability to expand adult CMs, their large-scale production from human pluripotent stem cells (hPSC) has been suggested. Significant improvements have been made in understanding directed differentiation processes of CMs from hPSCs and their suspension culture-based production at chemically defined conditions. However, optimization experiments are costly, time-consuming, and highly variable, leading to challenges in developing reliable and consistent protocols for the generation of large CM numbers at high purity. This study examined the ability of data-driven modeling with machine learning for identifying key experimental conditions and predicting final CM content using data collected during hPSC-cardiac differentiation in advanced stirred tank bioreactors (STBRs). Through feature selection, we identified process conditions, features, and patterns that are the most influential on and predictive of the CM content at the process endpoint, on differentiation day 10 (dd10). Process-related features were extracted from experimental data collected from 58 differentiation experiments by feature engineering. These features included data continuously collected online by the bioreactor system, such as dissolved oxygen concentration and pH patterns, as well as offline determined data, including the cell density, cell aggregate size, and nutrient concentrations. The selected features were used as inputs to construct models to classify the resulting CM content as being "sufficient" or "insufficient" regarding pre-defined thresholds. The models built using random forests and Gaussian process modeling predicted insufficient CM content for a differentiation process with 90% accuracy and precision on dd7 of the protocol and with 85% accuracy and 82% precision at a substantially earlier stage: dd5. These models provide insight into potential key factors affecting hPSC cardiac differentiation to aid in selecting future experimental conditions and can predict the final CM content at earlier process timepoints, providing cost and time savings. This study suggests that data-driven models and machine learning techniques can be employed using existing data for understanding and improving production of a specific cell type, which is potentially applicable to other lineages and critical for realization of their therapeutic applications.
Collapse
Affiliation(s)
- Bianca Williams
- Department of Chemical Engineering, Auburn University, Auburn, AL, United States
| | - Wiebke Löbel
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hanover, Germany
| | - Ferdous Finklea
- Department of Chemical Engineering, Auburn University, Auburn, AL, United States
| | - Caroline Halloin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hanover, Germany
| | - Katharina Ritzenhoff
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hanover, Germany
| | - Felix Manstein
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hanover, Germany
| | - Samira Mohammadi
- Department of Chemical Engineering, Auburn University, Auburn, AL, United States
| | | | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hanover, Germany
| | - Elizabeth Lipke
- Department of Chemical Engineering, Auburn University, Auburn, AL, United States
| | - Selen Cremaschi
- Department of Chemical Engineering, Auburn University, Auburn, AL, United States
| |
Collapse
|
177
|
Than PA, Brubaker AL, Ebel NH, Profita EL, Esquivel CO. The Emerging Need for Combined Heart and Liver Transplantation in Congenital Heart Disease. CURRENT TRANSPLANTATION REPORTS 2020. [DOI: 10.1007/s40472-020-00286-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
178
|
Monaco J, Khanna A, Khazanie P. Transplant and mechanical circulatory support in patients with adult congenital heart disease. Heart Fail Rev 2020; 25:671-683. [PMID: 32472522 PMCID: PMC7811764 DOI: 10.1007/s10741-020-09976-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Advances in surgery and pediatric care over the past decades have achieved improved survival for children born with congenital heart disease (CHD) and have produced a large, growing population of patients with adult congenital heart disease (ACHD). Heart failure has emerged as the leading cause of death and a major cause of morbidity among the ACHD population, while as little evidence supports the efficacy of guideline-directed medical therapies in this population. It is increasingly important that clinicians caring for these patients understand how to utilize mechanical circulatory support (MCS) in ACHD. In this review, we summarize the data on transplantation and MCS in the ACHD-heart failure population and provide a framework for how ACHD patients may benefit from advanced heart failure therapies like transplantation and MCS.
Collapse
Affiliation(s)
- James Monaco
- Colorado University Hospital, University of Colorado Anschutz Medical Center, Aurora, CO, USA.
| | - Amber Khanna
- Colorado University Hospital, University of Colorado Anschutz Medical Center, Aurora, CO, USA
| | - Prateeti Khazanie
- Colorado University Hospital, University of Colorado Anschutz Medical Center, Aurora, CO, USA
| |
Collapse
|
179
|
Garekar S, Meeran T, Patel V, Patil S, Dhake S, Mali S, Mhatre A, Bind D, Gaur A, Sinha S, Shetty V, Sabnis K, Soni B, Malankar D, Mulay A. Early experience with pediatric cardiac transplantation in a limited resource setting. Ann Pediatr Cardiol 2020; 13:220-226. [PMID: 32863657 PMCID: PMC7437633 DOI: 10.4103/apc.apc_105_19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 03/23/2020] [Accepted: 05/21/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Pediatric heart transplantation is a now a well-established and standard treatment option for end stage heart failure for various conditions in children. Due to logistic issues, it is not an option for in most pediatric cardiac centres in the third world. AIM We sought to describe our early experience in the current era in India. METHODS This is a short term retrospective chart review of pediatric patients who underwent heart transplantation at our centre. Mean/Median with standard deviation /range was used to present data. RESULTS Twenty patients underwent orthotopic heart transplant between January 2016 and June 2019. The median age at transplant was 12.4years (range 3.3 to 17.3 years). The median weight was 23.2kg (range 10-80kg). The mean donor/recipient weight ratio was 1.62± 0.84. The mean ICU stay was 12.1days. The mean follow up post transplant was 2.03± 0.97years (range 10 days-3.57years). The 1 month and the 1 year survival was 100%. Biopsies were positive for significant rejection in 7 patients (35%). At the time of last follow-up, 3 patients (15%) had expired. The major post transplant morbidities were mechanical circulatory support (n=3), hypertension with seizure complex (n=3), post transplant lympho-proliferative disorder (n=1), pseudocyst of pancreas (n=1), coronary allograft vasculopathy (n=3) and systemic hypertension (n=7). All surviving patients (n=17) were asymptomatic at last follow up. CONCLUSION The results suggest acceptable short term outcomes in Indian pediatric patients can be achieved after heart transplantation in the current era. Significant rejection episodes and coronary allograft vasculopathy need careful follow up.
Collapse
Affiliation(s)
- Swati Garekar
- Division of Pediatric Cardiology, Fortis Hospital, Mumbai, Maharashtra, India
| | - Talha Meeran
- Division of Advanced Heart Failure, Cardiac Transplant and Pulmonary Hypertension, Fortis Hospital, Mumbai, Maharashtra, India
| | - Vinay Patel
- Division of Pediatric Cardiology, Fortis Hospital, Mumbai, Maharashtra, India
| | - Sachin Patil
- Division of Pediatric Anesthesiology and Intensive Care, Fortis Hospital, Mumbai, Maharashtra, India
| | - Shyam Dhake
- Division of Pediatric Anesthesiology and Intensive Care, Fortis Hospital, Mumbai, Maharashtra, India
| | - Shivaji Mali
- Division of Pediatric Anesthesiology and Intensive Care, Fortis Hospital, Mumbai, Maharashtra, India
| | - Amit Mhatre
- Division of Intensive Care, Fortis Hospital, Mumbai, Maharashtra, India
| | - Dilip Bind
- Division of Intensive Care, Fortis Hospital, Mumbai, Maharashtra, India
| | - Ashish Gaur
- Division of Cardiothoracic Surgery, Fortis Hospital, Mumbai, Maharashtra, India
| | - Sandeep Sinha
- Division of Cardiothoracic Surgery, Fortis Hospital, Mumbai, Maharashtra, India
| | - Vijay Shetty
- Division of Anesthesiology, Fortis Hospita, Mumbai, Maharashtra, India
| | - Kirtis Sabnis
- Division of Infectious Diseases, Fortis Hospital, Mumbai, Maharashtra, India
| | - Bharat Soni
- Division of Pediatric Cardiothoracic Surgery, Fortis Hospital, Mumbai, Maharashtra, India
| | - Dhananjay Malankar
- Division of Pediatric Cardiothoracic Surgery, Fortis Hospital, Mumbai, Maharashtra, India
| | - Anvay Mulay
- Division of Cardiothoracic Surgery, Fortis Hospital, Mumbai, Maharashtra, India
| |
Collapse
|
180
|
Parrotta EI, Lucchino V, Scaramuzzino L, Scalise S, Cuda G. Modeling Cardiac Disease Mechanisms Using Induced Pluripotent Stem Cell-Derived Cardiomyocytes: Progress, Promises and Challenges. Int J Mol Sci 2020; 21:E4354. [PMID: 32575374 PMCID: PMC7352327 DOI: 10.3390/ijms21124354] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/12/2020] [Accepted: 06/15/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVDs) are a class of disorders affecting the heart or blood vessels. Despite progress in clinical research and therapy, CVDs still represent the leading cause of mortality and morbidity worldwide. The hallmarks of cardiac diseases include heart dysfunction and cardiomyocyte death, inflammation, fibrosis, scar tissue, hyperplasia, hypertrophy, and abnormal ventricular remodeling. The loss of cardiomyocytes is an irreversible process that leads to fibrosis and scar formation, which, in turn, induce heart failure with progressive and dramatic consequences. Both genetic and environmental factors pathologically contribute to the development of CVDs, but the precise causes that trigger cardiac diseases and their progression are still largely unknown. The lack of reliable human model systems for such diseases has hampered the unraveling of the underlying molecular mechanisms and cellular processes involved in heart diseases at their initial stage and during their progression. Over the past decade, significant scientific advances in the field of stem cell biology have literally revolutionized the study of human disease in vitro. Remarkably, the possibility to generate disease-relevant cell types from induced pluripotent stem cells (iPSCs) has developed into an unprecedented and powerful opportunity to achieve the long-standing ambition to investigate human diseases at a cellular level, uncovering their molecular mechanisms, and finally to translate bench discoveries into potential new therapeutic strategies. This review provides an update on previous and current research in the field of iPSC-driven cardiovascular disease modeling, with the aim of underlining the potential of stem-cell biology-based approaches in the elucidation of the pathophysiology of these life-threatening diseases.
Collapse
|
181
|
Li K, Wang C, Zhao Z, Wu Z, Wu Z, Tian X, Xiao Y, Li Z, Wang Y. A comparison for the effects of raw, smoked, and smoked and brined areca nut extracts on the immune and inflammatory responses in the Kunming mice. J Food Biochem 2020; 44:e13319. [PMID: 32537741 DOI: 10.1111/jfbc.13319] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 05/07/2020] [Accepted: 05/19/2020] [Indexed: 12/16/2022]
Abstract
Chewing of areca nuts is quite popular in various regions worldwide. Previous studies have demonstrated the pharmacological and toxicological effects of fresh areca nuts. However, processed areca nuts, which are popular in the Hunan province of China, have not been extensively studied for its biological effect. This study aimed at investigating the impact of the acrea nut extracts (ANE) prepared from the raw material, the semi-product, and the final product on the immune system and inflammation-related markers in the Kunming mice. The mice were assigned to seven different groups and administered different ANE at two concentrations (1X and 5X) for four weeks. Total body weight gain and organ coefficient of the liver, spleen, and kidney, as well as the immune system and inflammation-related markers were evaluated. The results revealed that processed areca nuts have a much milder effect on the mice immune system and some inflammatory markers than fresh areca nut in the Kunming mice. PRACTICAL APPLICATIONS: Chewing various forms of areca nuts is popular in China, Southeast Asia, and other regions. People from Hunan, China prefer to chew a processed areca nut, which has rarely been studied. This manuscript explores the effects of three kinds of areca nut extracts on the immune system- and inflammation-related indicators in Kunming mice. The obtained results revealed that processed areca nuts had significantly milder effects than the raw nut/nut extract, particularly on the body weight, immune responses, and inflammatory markers. The results of the present study provide some new directions for the areca nut industry and raise public awareness for the undesirable effects of areca nuts.
Collapse
Affiliation(s)
- Ke Li
- College of Food Science and Technology, Hunan Agricultural University, Changsha, China.,Hunan Province Key Laboratory of Food Science and Biotechnology, Changsha, China.,National Engineering Center of Plant Functional Components Utilization, Changsha, China
| | - Chuanhua Wang
- College of Food Science and Technology, Hunan Agricultural University, Changsha, China
| | - Ziwei Zhao
- College of Food Science and Technology, Hunan Agricultural University, Changsha, China.,Hunan Province Key Laboratory of Food Science and Biotechnology, Changsha, China.,National Engineering Center of Plant Functional Components Utilization, Changsha, China
| | - Zhongqin Wu
- College of Food Science and Technology, Hunan Agricultural University, Changsha, China.,Hunan Province Key Laboratory of Food Science and Biotechnology, Changsha, China.,National Engineering Center of Plant Functional Components Utilization, Changsha, China
| | - Zhongkun Wu
- College of Food Science and Technology, Hunan Agricultural University, Changsha, China.,Hunan Province Key Laboratory of Food Science and Biotechnology, Changsha, China.,National Engineering Center of Plant Functional Components Utilization, Changsha, China
| | - Xing Tian
- College of Food Science and Technology, Hunan Agricultural University, Changsha, China.,Hunan Province Key Laboratory of Food Science and Biotechnology, Changsha, China.,National Engineering Center of Plant Functional Components Utilization, Changsha, China.,College of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Yu Xiao
- College of Food Science and Technology, Hunan Agricultural University, Changsha, China.,Hunan Province Key Laboratory of Food Science and Biotechnology, Changsha, China.,National Engineering Center of Plant Functional Components Utilization, Changsha, China
| | - Zongjun Li
- College of Food Science and Technology, Hunan Agricultural University, Changsha, China.,Hunan Province Key Laboratory of Food Science and Biotechnology, Changsha, China.,National Engineering Center of Plant Functional Components Utilization, Changsha, China
| | - Yuanliang Wang
- College of Food Science and Technology, Hunan Agricultural University, Changsha, China.,Hunan Province Key Laboratory of Food Science and Biotechnology, Changsha, China.,National Engineering Center of Plant Functional Components Utilization, Changsha, China
| |
Collapse
|
182
|
Quantitative cardiac magnetic resonance T2 imaging offers ability to non-invasively predict acute allograft rejection in children. Cardiol Young 2020; 30:852-859. [PMID: 32456723 PMCID: PMC7654096 DOI: 10.1017/s104795112000116x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Monitoring for acute allograft rejection improves outcomes after cardiac transplantation. Endomyocardial biopsy is the gold standard test defining rejection, but carries risk and has limitations. Cardiac magnetic resonance T2 mapping may be able to predict rejection in adults, but has not been studied in children. Our aim was to evaluate T2 mapping in identifying paediatric cardiac transplant patients with acute rejection. METHODS Eleven paediatric transplant patients presenting 18 times were prospectively enrolled for non-contrast cardiac magnetic resonance at 1.5 T followed by endomyocardial biopsy. Imaging included volumetry, flow, and T2 mapping. Regions of interest were manually selected on the T2 maps using the middle-third technique in the left ventricular septal and lateral wall in a short-axis and four-chamber slice. Mean and maximum T2 values were compared with Student's t-tests analysis. RESULTS Five cases of acute rejection were identified in three patients, including two cases of grade 2R on biopsy and three cases of negative biopsy treated for clinical symptoms attributed to rejection (new arrhythmia, decreased exercise capacity). A monotonic trend between increasing T2 values and higher biopsy grades was observed: grade 0R T2 53.4 ± 3 ms, grade 1R T2 54.5 ms ± 3 ms, grade 2R T2 61.3 ± 1 ms. The five rejection cases had significantly higher mean T2 values compared to cases without rejection (58.3 ± 4 ms versus 53 ± 2 ms, p = 0.001). CONCLUSIONS Cardiac magnetic resonance with quantitative T2 mapping may offer a non-invasive method for screening paediatric cardiac transplant patients for acute allograft rejection. More data are needed to understand the relationship between T2 and rejection in children.
Collapse
|
183
|
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: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [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
|
184
|
Abstract
Ischemic heart disease (IHD) is one of the most common cardiovascular diseases and is the leading cause of death worldwide. Stem cell therapy is a promising strategy to promote cardiac regeneration and myocardial function recovery. Recently, the generation of human induced pluripotent cells (hiPSCs) and their differentiation into cardiomyocytes and vascular cells offer an unprecedented opportunity for the IHD treatment. This review briefly summarizes hiPSCs and their differentiation, and presents the recent advances in hiPSC injection, engineered cardiac patch fabrication, and the application of hiPSC derived extracellular vesicle. Current challenges and further perspectives are also discussed to understand current risks and concerns, identify potential solutions, and direct future clinical trials and applications.
Collapse
|
185
|
Spitaleri G, Farrero Torres M, Sabatino M, Potena L. The pharmaceutical management of cardiac allograft vasculopathy after heart transplantation. Expert Opin Pharmacother 2020; 21:1367-1376. [PMID: 32401066 DOI: 10.1080/14656566.2020.1753698] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
INTRODUCTION Cardiac allograft vasculopathy (CAV) is a major limitation to long-term survival after heart transplantation. Its peculiar pathophysiology involves multifactorial pathways including immune-mediated and metabolic risk factors, which are associated with the development of specific pathological lesions. The often diffuse and chronic nature of the disease reduces the effectiveness of revascularization procedures, and pharmacological prevention of the disease is the sole therapeutic approach with some proven efficacy. AREAS COVERED In this article, after briefly outlining the risk factors for CAV, the authors revise the potential pharmacological approaches that may reduce the burden of CAV. While several therapies have shown convincing efficacy in terms of CAV prevention diagnosed by coronary imaging, very few have been reported to improve prognosis with any meaningful level of evidence. EXPERT OPINION The authors believe that a customizable approach is necessary for clinical practice given the currently available evidence. Furthermore, it is important, in the future, to address the glaring therapeutic gap of an effective treatment against donor-specific antibodies, whose effect on endothelial injury is currently one of the major mechanisms of CAV development and for which no pharmacological treatment is currently available.
Collapse
Affiliation(s)
- Giosafat Spitaleri
- Heart Failure and Heart Transplant Unit, Cardiovascular Institute, Hospital Clínic , Barcelona, Spain
| | - Marta Farrero Torres
- Heart Failure and Heart Transplant Unit, Cardiovascular Institute, Hospital Clínic , Barcelona, Spain
| | - Mario Sabatino
- Heart Failure and Heart Transplant Program, Bologna Academic Hospital , Bologna, Italy
| | - Luciano Potena
- Heart Failure and Heart Transplant Program, Bologna Academic Hospital , Bologna, Italy
| |
Collapse
|
186
|
Blumenrath SH, Lee BY, Low L, Prithviraj R, Tagle D. Tackling rare diseases: Clinical trials on chips. Exp Biol Med (Maywood) 2020; 245:1155-1162. [PMID: 32397761 DOI: 10.1177/1535370220924743] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
IMPACT STATEMENT Designing and conducting clinical trials are extremely difficult in rare diseases. Adapting tissue chips for rare disease therapy development is pivotal in assuring that treatments are available, especially for severe diseases that are difficult to treat. Thus far, the NCATS-led National Institutes of Health (NIH) Tissue Chip program has focused on deploying the technology towards in vitro tools for safety and efficacy assessments of therapeutics. However, exploring the feasibility and best possible approach to expanding this focus towards the development phase of therapeutics is critical to moving the field of CToCs forward and increasing confidence with the use of tissue chips. The working group of stakeholders and experts convened by NCATS and the Drug Information Association (DIA) addresses important questions related to disease setting, test agents, study design, data collection, benefit/risk, and stakeholder engagement-exploring both current and future best use cases and important prerequisites for progress in this area.
Collapse
Affiliation(s)
| | - Bo Y Lee
- National Institutes of Health, Bethesda, MD 20892, USA
| | - Lucie Low
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Danilo Tagle
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
187
|
Egbe AC, Miranda WR, Veldtman GR, Graham RP, Kamath PS. Hepatic Venous Pressure Gradient in Fontan Physiology Has Limited Diagnostic and Prognostic Significance. CJC Open 2020; 2:360-364. [PMID: 32995721 PMCID: PMC7499375 DOI: 10.1016/j.cjco.2020.04.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 04/23/2020] [Indexed: 01/06/2023] Open
Abstract
Background Hepatic venous pressure gradient (HVPG) is measure of portal pressure and a prognostic tool in patients with viral and alcoholic cirrhosis; its utility is unknown in patients with Fontan-associated liver disease (FALD). Limited data suggest that patients with FALD have normal HVPG. On the basis of the available data, we hypothesized that there would be no association between HVPG, liver disease severity, and transplant-free survival in FALD. Methods A retrospective study of Fontan patients who had liver biopsy and HVPG assessment at Mayo Clinic was performed. HVPG was calculated as wedged HVP minus free HVP; liver disease severity was measured by histologic assessment of fibrosis and standard clinical liver disease risk scores. Results Of 56 patients (aged 28 ± 7 years), the mean Fontan pressure was 16 ± 4 and the mean HVPG was 1.4 ± 0.3 mm Hg (range, 0-3). Perisinusoidal fibrosis and periportal fibrosis were present in 56 (100%) and 54 (94%) patients, respectively; 18 (32%) met criteria for cirrhosis. There was no correlation between HVPG and degree of hepatic fibrosis. Similarly, there was no correlation between HVPG and any clinical liver disease risk score. Six (11%) patients died and 2 (4%) underwent heart transplantation during follow-up; HVPG was not associated with transplant-free survival. Conclusions HVPG is not elevated in FALD even in the setting of cirrhosis and does not correlate with liver disease severity or clinical outcomes. These results suggest the limited diagnostic and prognostic role of HVPG in the management of FALD and highlight the potential pitfalls of using HVPG in this population.
Collapse
Affiliation(s)
- Alexander C. Egbe
- Department of Cardiovascular Medicine, Mayo Clinic Rochester, Rochester, Minnesota, USA
- Corresponding author: Dr Alexander C. Egbe, Mayo Clinic and Foundation, 200 First Street SW, Rochester, Minnesota 55905, USA. Tel.: +1-507-284-2520; fax: +1-507-266-0103.
| | - William R. Miranda
- Department of Cardiovascular Medicine, Mayo Clinic Rochester, Rochester, Minnesota, USA
| | - Gruschen R. Veldtman
- Department of Cardiology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Rondell P. Graham
- Department of Laboratory Medicine and Pathology, Mayo Clinic Rochester, Rochester, Minnesota, USA
| | - Patrick S. Kamath
- Divison of Gastroenterology and Hepatology, Mayo Clinic Rochester, Rochester, Minnesota, USA
| |
Collapse
|
188
|
Mallah SI, Atallah B, Moustafa F, Naguib M, El Hajj S, Bader F, Mehra MR. Evidence-based pharmacotherapy for prevention and management of cardiac allograft vasculopathy. Prog Cardiovasc Dis 2020; 63:194-209. [DOI: 10.1016/j.pcad.2020.03.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 03/17/2020] [Indexed: 01/08/2023]
|
189
|
Sepulveda M, Pirozzolo I, Alegre ML. Impact of the microbiota on solid organ transplant rejection. Curr Opin Organ Transplant 2020; 24:679-686. [PMID: 31577594 DOI: 10.1097/mot.0000000000000702] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW The microbiota in mammalian hosts can affect maturation and function of the immune system and has been associated with health and disease. We will review new findings on how this dynamic environmental factor impacts alloimmunity and therapy in transplant hosts. RECENT FINDINGS The microbiota changes after transplantation and immunosuppressive therapy. New data indicate that different microbial community structures have distinct impact on graft outcome, from promoting, to inhibiting or being neutral to transplant survival. In addition, we will address reciprocal interactions between the microbiota and immunosuppressive drugs, as well as the suitability of the microbiota as a predictive biomarker and its utility as adjunct therapy in transplantation. SUMMARY Advances in microbiome sequencing and wider availability of gnotobiotic facilities are enabling mechanistic investigations into the commensal communities and pathways that modulate allograft outcome, responsiveness to immunosuppression and side effects of drugs. A better understanding of the functions of the microbiota may help mitigate drug toxicity, predict drug dosage and dampen alloimmunity in transplant patients.
Collapse
|
190
|
Quality of Life in Adults With Congenital Heart Disease: Function Over Form. Can J Cardiol 2020; 37:186-187. [PMID: 32339650 DOI: 10.1016/j.cjca.2020.04.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 11/21/2022] Open
|
191
|
Martewicz S, Magnussen M, Elvassore N. Beyond Family: Modeling Non-hereditary Heart Diseases With Human Pluripotent Stem Cell-Derived Cardiomyocytes. Front Physiol 2020; 11:384. [PMID: 32390874 PMCID: PMC7188911 DOI: 10.3389/fphys.2020.00384] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 03/30/2020] [Indexed: 12/23/2022] Open
Abstract
Non-genetic cardiac pathologies develop as an aftermath of extracellular stress-conditions. Nevertheless, the response to pathological stimuli depends deeply on intracellular factors such as physiological state and complex genetic backgrounds. Without a thorough characterization of their in vitro phenotype, modeling of maladaptive hypertrophy, ischemia and reperfusion injury or diabetes in human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) has been more challenging than hereditary diseases with defined molecular causes. In past years, greater insights into hPSC-CM in vitro physiology and advancements in technological solutions and culture protocols have generated cell types displaying stress-responsive phenotypes reminiscent of in vivo pathological events, unlocking their application as a reductionist model of human cardiomyocytes, if not the adult human myocardium. Here, we provide an overview of the available literature of pathology models for cardiac non-genetic conditions employing healthy (or asymptomatic) hPSC-CMs. In terms of numbers of published articles, these models are significantly lagging behind monogenic diseases, which misrepresents the incidence of heart disease causes in the human population.
Collapse
Affiliation(s)
- Sebastian Martewicz
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai, China
| | - Michael Magnussen
- Stem Cells & Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Nicola Elvassore
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai, China.,Stem Cells & Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, London, United Kingdom.,Venetian Institute of Molecular Medicine, Padua, Italy.,Department of Industrial Engineering, University of Padova, Padua, Italy
| |
Collapse
|
192
|
Ing RJ, Mclennan D, Twite MD, DiMaria M. Anesthetic Considerations for Fontan-Associated Liver Disease and the Failing Fontan Circuit. J Cardiothorac Vasc Anesth 2020; 34:2224-2233. [PMID: 32249074 DOI: 10.1053/j.jvca.2020.02.040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 02/19/2020] [Accepted: 02/23/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Richard J Ing
- Department of Anesthesiology, Children's Hospital Colorado, Anschutz Medical Campus, Aurora, CO; School of Medicine, University of Colorado, Aurora, CO.
| | - Daniel Mclennan
- Stead Family Children's Hospital, University of Iowa, Iowa City, IA
| | - Mark D Twite
- Department of Anesthesiology, Children's Hospital Colorado, Anschutz Medical Campus, Aurora, CO; School of Medicine, University of Colorado, Aurora, CO
| | - Michael DiMaria
- Department of Anesthesiology, Children's Hospital Colorado, Anschutz Medical Campus, Aurora, CO; School of Medicine, University of Colorado, Aurora, CO
| |
Collapse
|
193
|
Chu AJ, Zhao EJ, Chiao M, Lim CJ. Co-culture of induced pluripotent stem cells with cardiomyocytes is sufficient to promote their differentiation into cardiomyocytes. PLoS One 2020; 15:e0230966. [PMID: 32243463 PMCID: PMC7122760 DOI: 10.1371/journal.pone.0230966] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 03/12/2020] [Indexed: 12/22/2022] Open
Abstract
Various types of stem cells and non-stem cells have been shown to differentiate or transdifferentiate into cardiomyocytes by way of co-culture with appropriate inducer cells. However, there is a limited demonstration of a co-culture induction system utilizing stem cell-derived cardiomyocytes as a stimulatory source for cardiac reprogramming (of stem cells or otherwise). In this study, we utilized an inductive co-culture method to show that previously differentiated induced pluripotent stem (iPS) cell-derived cardiomyocytes (iCMs), when co-cultivated with iPS cells, constituted a sufficient stimulatory system to induce cardiac differentiation. To enable tracking of both cell populations, we utilized GFP-labeled iPS cells and non-labeled iCMs pre-differentiated using inhibitors of GSK and Wnt signaling. Successful differentiation was assessed by the exhibition of spontaneous self-contractions, structural organization of α-actinin labeled sarcomeres, and expression of cardiac specific markers cTnT and α-actinin. We found that iCM-iPS cell-cell contact was essential for inductive differentiation, and this required overlaying already adherent iPS cells with iCMs. Importantly, this process was achieved without the exogenous addition of pathway inhibitors and morphogens, suggesting that 'older' iCMs serve as an adequate stimulatory source capable of recapitulating the necessary culture environment for cardiac differentiation.
Collapse
Affiliation(s)
- Axel J. Chu
- School of Biomedical Engineering, The University of British Columbia, Vancouver, B.C., Canada
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, B.C., Canada
| | - Eric Jiahua Zhao
- School of Biomedical Engineering, The University of British Columbia, Vancouver, B.C., Canada
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, B.C., Canada
| | - Mu Chiao
- School of Biomedical Engineering, The University of British Columbia, Vancouver, B.C., Canada
- Department of Mechanical Engineering, The University of British Columbia, Vancouver, B.C., Canada
- * E-mail: (CJL); (MC)
| | - Chinten James Lim
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, B.C., Canada
- Department of Pediatrics, The University of British Columbia, Vancouver, B.C., Canada
- * E-mail: (CJL); (MC)
| |
Collapse
|
194
|
Hu X, Ouyang S, Xie Y, Gong Z, Du J. Characterizing the gut microbiota in patients with chronic kidney disease. Postgrad Med 2020; 132:495-505. [PMID: 32241215 DOI: 10.1080/00325481.2020.1744335] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Objectives: Emerging evidence suggests that gut microbiota dysbiosis plays a critical role in chronic kidney disease (CKD). However, the relationship between altered gut microbiome profiles and disease severity remains unclear. In this study, we sought to characterize the gut microbiota in CKD patients compared to healthy controls, and to explore potential relationships between gut microbiota composition and disease severity. Methods: Fecal samples were collected from 95 patients at different stages of CKD (non-dialysis patients from stage 1 to 5) and 20 healthy controls. Bacterial DNA was extracted for 16S ribosomal DNA sequencing targeting the V3-V4 region. The diversity and relative abundance of gut microbiota were analyzed as outcome indicators. Results: Differences were observed in the microbial composition and diversity of fecal samples from CKD patients and healthy controls. Specifically, disease severity was found to alter gut microbiota composition. Compared to that in healthy controls, CKD patients showed an increased abundance of Proteobacteria and decreased Synergistetes, most notably in disease stage 5. Lower levels of butyrate-producing bacteria and higher levels of potential pathogens were also detected in CKD patients. Further, Pyramidobacter and Prevotellaceae_UCG-001 were significantly decreased in the CKD1 group compared with healthy controls. Notably, nine microbial genera, including Escherichia-Shigella, Parabacteroides, Roseburia, rectale_group, Ruminococcaceae_NK4A214_group, Prevotellaceae_UCG.001, Hungatella, Intestinimonas, and Pyramidobacter, identified using a random forest model, distinguished between patients with CKD and healthy controls with high accuracy. Functional analysis also revealed that fatty acid and inositol phosphate metabolism were enriched in the CKD group, while aminoacyl-tRNA biosynthesis, oxidative phosphorylation, phenylalanine, tyrosine, and tryptophan biosynthesis, thiamine metabolism, pantothenate, and CoA biosynthesis, as well as valine, leucine, and isoleucine biosynthesis were enriched in healthy controls. Conclusion: Gut microbiota composition and function are associated with CKD severity. And, specific gut microbes are potentially helpful for CKD early diagnosis and prognosis monitoring.
Collapse
Affiliation(s)
- Xiaofang Hu
- Department of Pharmacy, Xiangya Hospital, Central South University , Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University , Changsha, Hunan, China.,The Hunan Institute of Pharmacy Practice and Clinical Research, Xiangya Hospital , Changsha, Hunan, China
| | - Shaxi Ouyang
- Department of Nephrology, Hunan Provincial People's Hospital, the First-affiliated Hospital of Hunan Normal University , Changsha, Hunan, China
| | - Yuhong Xie
- Department of Pharmacy, Xiangya Hospital, Central South University , Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University , Changsha, Hunan, China.,The Hunan Institute of Pharmacy Practice and Clinical Research, Xiangya Hospital , Changsha, Hunan, China
| | - Zhicheng Gong
- Department of Pharmacy, Xiangya Hospital, Central South University , Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University , Changsha, Hunan, China.,The Hunan Institute of Pharmacy Practice and Clinical Research, Xiangya Hospital , Changsha, Hunan, China
| | - Jie Du
- Department of Pharmacy, Xiangya Hospital, Central South University , Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University , Changsha, Hunan, China.,The Hunan Institute of Pharmacy Practice and Clinical Research, Xiangya Hospital , Changsha, Hunan, China
| |
Collapse
|
195
|
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
|
196
|
Rasmussen ML, Taneja N, Neininger AC, Wang L, Robertson GL, Riffle SN, Shi L, Knollmann BC, Burnette DT, Gama V. MCL-1 Inhibition by Selective BH3 Mimetics Disrupts Mitochondrial Dynamics Causing Loss of Viability and Functionality of Human Cardiomyocytes. iScience 2020; 23:101015. [PMID: 32283523 PMCID: PMC7155208 DOI: 10.1016/j.isci.2020.101015] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 02/25/2020] [Accepted: 03/24/2020] [Indexed: 12/17/2022] Open
Abstract
MCL-1 is a well-characterized inhibitor of cell death that has also been shown to be a regulator of mitochondrial dynamics in human pluripotent stem cells. We used cardiomyocytes derived from human-induced pluripotent stem cells (hiPSC-CMs) to uncover whether MCL-1 is crucial for cardiac function and survival. Inhibition of MCL-1 by BH3 mimetics resulted in the disruption of mitochondrial morphology and dynamics as well as disorganization of the actin cytoskeleton. Interfering with MCL-1 function affects the homeostatic proximity of DRP-1 and MCL-1 at the outer mitochondrial membrane, resulting in decreased functionality of hiPSC-CMs. Cardiomyocytes display abnormal cardiac performance even after caspase inhibition, supporting a nonapoptotic activity of MCL-1 in hiPSC-CMs. BH3 mimetics targeting MCL-1 are promising anti-tumor therapeutics. Progression toward using BCL-2 family inhibitors, especially targeting MCL-1, depends on understanding its canonical function not only in preventing apoptosis but also in the maintenance of mitochondrial dynamics and function. BH3 mimetics targeting MCL-1 disrupt the mitochondrial network of human iPSC-CMs The BH3-mimetic-mediated effects on mitochondrial dynamics are DRP-1-dependent Targeting MCL-1 affects the survival and function of human cardiomyocytes Human iPSC-derived cardiomyocytes can be used to reveal toxicity of MCL-1 inhibitors
Collapse
Affiliation(s)
- Megan L Rasmussen
- Department of Cell & Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Nilay Taneja
- Department of Cell & Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Abigail C Neininger
- Department of Cell & Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Lili Wang
- Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Center for Arrhythmia Research and Therapeutics, Department of Medicine, Nashville, TN 37232, USA
| | - Gabriella L Robertson
- Department of Cell & Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Stellan N Riffle
- Department of Cell & Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Linzheng Shi
- Department of Cell & Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Bjorn C Knollmann
- Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Center for Arrhythmia Research and Therapeutics, Department of Medicine, Nashville, TN 37232, USA
| | - Dylan T Burnette
- Department of Cell & Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Ingram Cancer Center, Vanderbilt University, Nashville, TN 37232, USA
| | - Vivian Gama
- Department of Cell & Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Ingram Cancer Center, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
197
|
Gintant G, Traebert M. The roles of human induced pluripotent stem cell-derived cardiomyocytes in drug discovery: managing in vitro safety study expectations. Expert Opin Drug Discov 2020; 15:719-729. [PMID: 32129680 DOI: 10.1080/17460441.2020.1736549] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Human induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) preparations are increasingly employed in in vitro cardiac safety studies to support candidate drug selection and regulatory submissions. The value of hiPSC-CM-based approaches depends on their ability to recapitulate the cellular mechanisms responsible for cardiotoxicity as well as overall assay characteristics (thus defining model performance). Different expectations at different drug development stages define the utility of these human-derived models. AREAS COVERED Herein, the authors review the importance of understanding the functional characteristics of the evolving spectrum of simpler (2D) and more complex (co-cultures, 3D constructs, and engineered tissues) human-derived cardiac preparations, and how their performance may be evaluated based on analytical sensitivity, variability, and reproducibility in order to correctly match preparations with expectations of different safety assays. The need for consensus clinical examples of electrophysiologic, contractile, and structural cardiotoxicities essential for benchmarking human-derived models is also discussed. EXPERT OPINION It is helpful (but not essential) that hiPSC-CMs preparations fully recapitulate pharmacological responses of native adult human ventricular myocytes when evaluating cardiotoxicity in vitro. Further calibration and model standardization (aligning concordance with clinical findings) are necessary to understand the role of hiPSC-CMs in guiding cardiotoxicity assessments in early drug discovery efforts.
Collapse
Affiliation(s)
- Gary Gintant
- Department of Integrative Pharmacology (ZR13), AP-9A-LL, AbbVie Inc. , North Chicago, IL, USA
| | - Martin Traebert
- Novartis Institutes for Biomedical Research , Safety Pharmacology, Basel, Switzerland
| |
Collapse
|
198
|
Liew LC, Ho BX, Soh BS. Mending a broken heart: current strategies and limitations of cell-based therapy. Stem Cell Res Ther 2020; 11:138. [PMID: 32216837 PMCID: PMC7098097 DOI: 10.1186/s13287-020-01648-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/02/2020] [Accepted: 03/10/2020] [Indexed: 12/16/2022] Open
Abstract
The versatility of pluripotent stem cells, attributable to their unlimited self-renewal capacity and plasticity, has sparked a considerable interest for potential application in regenerative medicine. Over the past decade, the concept of replenishing the lost cardiomyocytes, the crux of the matter in ischemic heart disease, with pluripotent stem cell-derived cardiomyocytes (PSC-CM) has been validated with promising pre-clinical results. Nevertheless, clinical translation was hemmed in by limitations such as immature cardiac properties, long-term engraftment, graft-associated arrhythmias, immunogenicity, and risk of tumorigenicity. The continuous progress of stem cell-based cardiac therapy, incorporated with tissue engineering strategies and delivery of cardio-protective exosomes, provides an optimistic outlook on the development of curative treatment for heart failure. This review provides an overview and current status of stem cell-based therapy for heart regeneration, with particular focus on the use of PSC-CM. In addition, we also highlight the associated challenges in clinical application and discuss the potential strategies in developing successful cardiac-regenerative therapy.
Collapse
Affiliation(s)
- Lee Chuen Liew
- Disease Modeling and Therapeutics Laboratory, A*STAR Institute of Molecular and Cell Biology, 61 Biopolis Drive Proteos, Singapore, 138673, Singapore
| | - Beatrice Xuan Ho
- Disease Modeling and Therapeutics Laboratory, A*STAR Institute of Molecular and Cell Biology, 61 Biopolis Drive Proteos, Singapore, 138673, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore, 117543, Singapore
| | - Boon-Seng Soh
- Disease Modeling and Therapeutics Laboratory, A*STAR Institute of Molecular and Cell Biology, 61 Biopolis Drive Proteos, Singapore, 138673, Singapore. .,Department of Biological Sciences, National University of Singapore, Singapore, 117543, Singapore. .,Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
| |
Collapse
|
199
|
Hu X, Du J, Xie Y, Huang Q, Xiao Y, Chen J, Yan S, Gong Z, Ouyang S. Fecal microbiota characteristics of Chinese patients with primary IgA nephropathy: a cross-sectional study. BMC Nephrol 2020; 21:97. [PMID: 32169051 PMCID: PMC7071635 DOI: 10.1186/s12882-020-01741-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 02/26/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Growing evidence has shown that the gut-renal connection and gut microbiota dysbiosis play a critical role in immunoglobulin A nephropathy (IgAN). However, the fecal microbiome profile in Chinese patients with IgAN remains unknown. A cross-sectional study was designed for the first time to investigate the fecal microbiota compositions in patients with primary IgAN in China and to evaluate the relationship between the fecal microbiome and IgAN clinical presentation. METHODS Fecal samples were collected from 17 IgAN patients and 18 age-, sex-, and body mass index-matched healthy controls, and bacterial DNA was extracted for 16S ribosomal RNA gene sequencing targeting the V3-V4 region. RESULTS Fecal samples from the IgAN patients and healthy controls showed differences in gut microbiota community richness and compositions. Compared to the healthy controls, IgAN patients at the phylum level had an increased abundance of Fusobacteria, but a decreased abundance of Synergistetes. The significantly increased genera in the IgAN group were Escherichia-Shigella, Hungatella, and Eggerthella, all of which possess pathogenic potential. Furthermore, the genus Escherichia-Shigella was negatively associated with the estimated glomerular filtration rate (eGFR) but was positively associated with the urinary albumin-to-creatinine ratio (uACR). However, the genus rectale_group was present in the IgAN group with a low abundance and was negatively associated with the uACR. Functional analysis disclosed that infection-related pathways were enriched in the IgAN group. CONCLUSIONS We demonstrate that gut microbiota dysbiosis occurs in patients with IgAN, and that changes in gut bacterial populations are closely related to IgAN clinical features, suggesting that certain specific gut microbiota may be a potential therapeutic target for IgAN.
Collapse
Affiliation(s)
- Xiaofang Hu
- Department of Pharmacy, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Kaifu District, Changsha, 410008, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Kaifu District, Changsha, 410008, Hunan, China
| | - Jie Du
- Department of Pharmacy, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Kaifu District, Changsha, 410008, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Kaifu District, Changsha, 410008, Hunan, China
| | - Yuhong Xie
- Department of Pharmacy, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Kaifu District, Changsha, 410008, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Kaifu District, Changsha, 410008, Hunan, China
| | - Qiong Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Kaifu District, Changsha, 410008, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Kaifu District, Changsha, 410008, Hunan, China
| | - Yi Xiao
- Department of Pharmacy, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Kaifu District, Changsha, 410008, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Kaifu District, Changsha, 410008, Hunan, China
| | - Juan Chen
- Department of Nephrology, Hunan Provincial People's Hospital, The first-affiliated hospital of Hunan normal university, No. 61 Jie-fang West Road, Fu-Rong District, Changsha, 410005, Hunan, China
| | - Siyuan Yan
- Department of Nephrology, Hunan Provincial People's Hospital, The first-affiliated hospital of Hunan normal university, No. 61 Jie-fang West Road, Fu-Rong District, Changsha, 410005, Hunan, China
| | - Zhicheng Gong
- Department of Pharmacy, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Kaifu District, Changsha, 410008, Hunan, China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Kaifu District, Changsha, 410008, Hunan, China.
| | - Shaxi Ouyang
- Department of Nephrology, Hunan Provincial People's Hospital, The first-affiliated hospital of Hunan normal university, No. 61 Jie-fang West Road, Fu-Rong District, Changsha, 410005, Hunan, China.
| |
Collapse
|
200
|
Ruth ND, Drury NE, Bennett J, Kelly DA. Cardiac and Liver Disease in Children: Implications for Management Before and After Liver Transplantation. Liver Transpl 2020; 26:437-449. [PMID: 31872564 DOI: 10.1002/lt.25666] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 10/16/2019] [Indexed: 02/06/2023]
Abstract
There is close interaction between the functions of the liver and heart affecting the presentation, diagnosis, and outcome of acute and chronic cardiac and liver disease. Conditions affecting both organ systems should be considered when proposing transplantation because the interaction between cardiac disease and liver disease has implications for diagnosis, management, selection for transplantation, and, ultimately, for longterm outcomes after liver transplantation (LT). The combination of cardiac and liver disease is well recognized in adults but is less appreciated in pediatric patients. The focus of this review is to describe conditions affecting both the liver and heart and how they affect selection and management of LT in the pediatric population.
Collapse
Affiliation(s)
- Nicola D Ruth
- Liver Unit, Birmingham Women's & Children's Hospital, Birmingham, United Kingdom.,Institute of Infection and Immunity, University of Birmingham, Birmingham, United Kingdom
| | - Nigel E Drury
- Department of Paediatric Cardiac Surgery, Birmingham Women's & Children's Hospital, Birmingham, United Kingdom.,Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - James Bennett
- Department of Anaesthesia, Birmingham Women's & Children's Hospital, Birmingham, United Kingdom.,Department of Anaesthesia, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| | - Deirdre A Kelly
- Liver Unit, Birmingham Women's & Children's Hospital, Birmingham, United Kingdom.,Institute of Infection and Immunity, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|