1
|
Chen G, Long J, Zhang Y, Zhou X, Gao B, Qin Z, Zhu Y, Song B, Cui Z, Liu Z, Xu M, Yu Z, Song B, Zhang Z. Fascia-derived stem cells enhance fat graft retention by promoting vascularization through the HMOX1-HIF-1α pathway. Stem Cell Res Ther 2025; 16:92. [PMID: 40001185 PMCID: PMC11863534 DOI: 10.1186/s13287-025-04204-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Adipose tissue is a widely used autologous soft tissue filler in plastic surgery, particularly for volumetric restoration in cases of soft tissue deficiency. However, effectively controlling the retention rate of transplanted fat remains a major challenge. Therefore, this study aims to explore strategies to enhance fat graft retention. We isolated fascia-derived stem cells (FDSCs) from human superficial fascia and compared their gene expression profiles with those of adipose-derived stem cells (ADSCs). Through bioinformatics analysis and functional experiments, we identified significant differences in the angiogenic potential of the two cell types. Based on sequencing results, we further investigated the roles of hypoxia-inducible factor-1α (HIF-1α) and heme oxygenase-1 (HMOX1). This study highlights the critical potential of FDSCs in improving fat graft retention and promoting angiogenesis, offering new strategies for enhancing graft survival and optimizing tissue regeneration therapies. METHODS We isolated fascia-derived stem cells (FDSCs) from human superficial fascia and compared them with adipose-derived stem cells (ADSCs). RNA sequencing was performed to analyze gene expression profiles, followed by bioinformatics analysis to identify differences in angiogenic potential. Functional experiments were conducted to investigate the roles of HIF-1α and HMOX1 in angiogenesis. RESULTS RNA sequencing revealed significant gene expression differences related to angiogenesis in FDSCs. The expression levels of HMOX1, HIF-1α, and VEGFa were significantly higher in FDSCs than in ADSCs, and HMOX1 positively regulated the expression of HIF-1α and VEGFa. In vitro experiments demonstrated that FDSCs promoted angiogenesis more effectively than ADSCs. In vivo co-transplantation experiments further confirmed that FDSCs improved fat graft retention and vascularization. CONCLUSIONS We demonstrated that FDSCs can more effectively promote vascularization both in vitro and in vivo, and significantly improve graft retention, indicating their broad potential for future applications in tissue repair and regeneration.
Collapse
Affiliation(s)
- Guo Chen
- Department of Plastic and Reconstruction Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jie Long
- Department of Plastic and Reconstruction Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
- Department of Medical Cosmetic Center, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, 310006, Hangzhou, Zhejiang, China
| | - Yuge Zhang
- Department of Plastic and Reconstruction Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xuhua Zhou
- Department of Plastic and Reconstruction Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Botao Gao
- Department of Plastic and Reconstruction Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Zijin Qin
- Department of Plastic and Reconstruction Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
- Department of Craniomaxillofacial Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yuhan Zhu
- Department of Plastic and Reconstruction Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Binyu Song
- Department of Plastic and Reconstruction Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Ziwei Cui
- Department of Plastic and Reconstruction Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Zhangzi Liu
- Department of Plastic and Reconstruction Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Man Xu
- Department of Plastic and Reconstruction Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Zhou Yu
- Department of Plastic and Reconstruction Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Baoqiang Song
- Department of Plastic and Reconstruction Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Ziang Zhang
- Department of Plastic and Reconstruction Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
2
|
Groen E, Mummery CL, Yiangou L, Davis RP. Three-dimensional cardiac models: a pre-clinical testing platform. Biochem Soc Trans 2024; 52:1045-1059. [PMID: 38778769 PMCID: PMC11346450 DOI: 10.1042/bst20230444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/25/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024]
Abstract
Major advancements in human pluripotent stem cell (hPSC) technology over recent years have yielded valuable tools for cardiovascular research. Multi-cell type 3-dimensional (3D) cardiac models in particular, are providing complementary approaches to animal studies that are better representatives than simple 2-dimensional (2D) cultures of differentiated hPSCs. These human 3D cardiac models can be broadly divided into two categories; namely those generated through aggregating pre-differentiated cells and those that form self-organizing structures during their in vitro differentiation from hPSCs. These models can either replicate aspects of cardiac development or enable the examination of interactions among constituent cell types, with some of these models showing increased maturity compared with 2D systems. Both groups have already emerged as physiologically relevant pre-clinical platforms for studying heart disease mechanisms, exhibiting key functional attributes of the human heart. In this review, we describe the different cardiac organoid models derived from hPSCs, their generation methods, applications in cardiovascular disease research and use in drug screening. We also address their current limitations and challenges as pre-clinical testing platforms and propose potential improvements to enhance their efficacy in cardiac drug discovery.
Collapse
Affiliation(s)
- Eline Groen
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Christine L. Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, 2300RC Leiden, The Netherlands
| | - Loukia Yiangou
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Richard P. Davis
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, 2300RC Leiden, The Netherlands
| |
Collapse
|
3
|
Allemann MS, Lee P, Beer JH, Saeedi Saravi SS. Targeting the redox system for cardiovascular regeneration in aging. Aging Cell 2023; 22:e14020. [PMID: 37957823 PMCID: PMC10726899 DOI: 10.1111/acel.14020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 09/09/2023] [Accepted: 10/05/2023] [Indexed: 11/15/2023] Open
Abstract
Cardiovascular aging presents a formidable challenge, as the aging process can lead to reduced cardiac function and heightened susceptibility to cardiovascular diseases. Consequently, there is an escalating, unmet medical need for innovative and effective cardiovascular regeneration strategies aimed at restoring and rejuvenating aging cardiovascular tissues. Altered redox homeostasis and the accumulation of oxidative damage play a pivotal role in detrimental changes to stem cell function and cellular senescence, hampering regenerative capacity in aged cardiovascular system. A mounting body of evidence underscores the significance of targeting redox machinery to restore stem cell self-renewal and enhance their differentiation potential into youthful cardiovascular lineages. Hence, the redox machinery holds promise as a target for optimizing cardiovascular regenerative therapies. In this context, we delve into the current understanding of redox homeostasis in regulating stem cell function and reprogramming processes that impact the regenerative potential of the cardiovascular system. Furthermore, we offer insights into the recent translational and clinical implications of redox-targeting compounds aimed at enhancing current regenerative therapies for aging cardiovascular tissues.
Collapse
Affiliation(s)
- Meret Sarah Allemann
- Center for Molecular CardiologyUniversity of ZurichSchlierenSwitzerland
- Department of Internal MedicineCantonal Hospital BadenBadenSwitzerland
| | - Pratintip Lee
- Center for Molecular CardiologyUniversity of ZurichSchlierenSwitzerland
- Department of Internal MedicineCantonal Hospital BadenBadenSwitzerland
| | - Jürg H. Beer
- Center for Molecular CardiologyUniversity of ZurichSchlierenSwitzerland
- Department of Internal MedicineCantonal Hospital BadenBadenSwitzerland
| | - Seyed Soheil Saeedi Saravi
- Center for Translational and Experimental Cardiology, Department of CardiologyUniversity Hospital Zurich, University of ZurichSchlierenSwitzerland
| |
Collapse
|
4
|
Lian N, Mao X, Su Y, Wang Y, Wang Y, Wang Y, Chen H, Zhu R, Yu Y, Xie K. Hydrogen-rich medium ameliorates lipopolysaccharides-induced mitochondrial fission and dysfunction in human umbilical vein endothelial cells (HUVECs) via up-regulating HO-1 expression. Int Immunopharmacol 2022; 110:108936. [PMID: 35738091 DOI: 10.1016/j.intimp.2022.108936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/22/2022] [Accepted: 06/06/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND Sepsis is defined as life-threatening organ dysfunction caused by a dysregulated host response to infection. It has been showed that the change of mitochondrial dynamics has been proved to be one of the main causes of death in patients with severe sepsis. And hydrogen has been proved to exert its protective effects against sepsis via heme oxygenase-1 (HO-1). This study was designed to demonstrate that whether the benefit effects of hydrogen can maintain the dynamic process of mitochondrial fusion/fission to mitigate human umbilical vein endothelial cells (HUVECs) injury exposed to endotoxin through HO-1. METHODS HUVECs cells cultured with medium which contained Lipopolysaccharides (LPS), Saline, hydrogen, Mdivi-1 (a dynamin-related protein 1 [Drp1] inhibitor) or zinc protoporphyrin IX (Znpp) (a HO-1 inhibitor) were also used in the research. Cell death and apoptosis were assessed using FITC annexin V and PI. Mitochondria were stained with Mitotracker orange and observed by confocal microscope. Oxygen consumption rate was assessed by seahorse xf24 extracellular analyzer. Mitochondrial membrane potential monitored by JC-1 dye. The expressions of Drp1 and HO-1 were tested by Western blot. The co-localization of Drp1 and mitochondria was determined by immunofluorescence. RESULTS LPS caused a decrease in ATP content, mitochondrial membrane potential, and maximal respiration rate. At the same time, increased expression of Drp1 were observed in LPS-stimulated HUVECs, concomitantly with excessive mitochondrial fission. We found that hydrogen-rich medium can increase ATP content, mitochondrial membrane potential and maximal respiration rate, and decrease the expression of Drp1 in LPS-treated HUVECs. Meanwhile, hydrogen can ameliorate excessive mitochondrial fission caused by LPS. Furthermore, hydrogen-rich medium had a similar effect to Mdivi-1, a mitochondrial fission blocker. Both of them rescued the up-regulation of Drp1 and mitochondrial fission induced by LPS, then normalized mitochondrial shape after LPS stimulation. But after Znpp pretreatment, HO-1 expression was inhibited and the protective effects of hydrogen were abrogated. CONCLUSIONS Hydrogen-rich medium can alleviate the LPS-induced mitochondrial fusion/fission and dysfunction in HUVECs via HO-1 up-regulation.
Collapse
Affiliation(s)
- Naqi Lian
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, Tianjin 300052, China
| | - Xing Mao
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, Tianjin 300052, China
| | - Yanchao Su
- Department of Critical Care Medicine, General Hospital of Tianjin Medical University, Tianjin 300052, China
| | - Yanyan Wang
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, Tianjin 300052, China
| | - Yaoqi Wang
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, Tianjin 300052, China
| | - Yuzun Wang
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, Tianjin 300052, China
| | - Hongguang Chen
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, Tianjin 300052, China
| | - Ruqing Zhu
- Department of Anesthesiology, Stomatology Hospital of Tianjin Medical University, Tianjin 300070, China
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, Tianjin 300052, China
| | - Keliang Xie
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, Tianjin 300052, China; Department of Critical Care Medicine, General Hospital of Tianjin Medical University, Tianjin 300052, China.
| |
Collapse
|
5
|
Floy ME, Shabnam F, Simmons AD, Bhute VJ, Jin G, Friedrich WA, Steinberg AB, Palecek SP. Advances in Manufacturing Cardiomyocytes from Human Pluripotent Stem Cells. Annu Rev Chem Biomol Eng 2022; 13:255-278. [PMID: 35320695 DOI: 10.1146/annurev-chembioeng-092120-033922] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The emergence of human pluripotent stem cell (hPSC) technology over the past two decades has provided a source of normal and diseased human cells for a wide variety of in vitro and in vivo applications. Notably, hPSC-derived cardiomyocytes (hPSC-CMs) are widely used to model human heart development and disease and are in clinical trials for treating heart disease. The success of hPSC-CMs in these applications requires robust, scalable approaches to manufacture large numbers of safe and potent cells. Although significant advances have been made over the past decade in improving the purity and yield of hPSC-CMs and scaling the differentiation process from 2D to 3D, efforts to induce maturation phenotypes during manufacturing have been slow. Process monitoring and closed-loop manufacturing strategies are just being developed. We discuss recent advances in hPSC-CM manufacturing, including differentiation process development and scaling and downstream processes as well as separation and stabilization. Expected final online publication date for the Annual Review of Chemical and Biomolecular Engineering, Volume 13 is October 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Martha E Floy
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA; , , , , ,
| | - Fathima Shabnam
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA; , , , , ,
| | - Aaron D Simmons
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA; , , , , ,
| | - Vijesh J Bhute
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, Wisconsin, USA; , .,Department of Chemical Engineering, Imperial College London, London, United Kingdom
| | - Gyuhyung Jin
- Department of Chemical Engineering, Purdue University, West Lafayette, Indiana, USA;
| | - Will A Friedrich
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA; , , , , ,
| | - Alexandra B Steinberg
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA; , , , , ,
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA; , , , , ,
| |
Collapse
|
6
|
Abstract
Nitric oxide, studied to evaluate its role in cardiovascular physiology, has cardioprotective and therapeutic effects in cellular signaling, mitochondrial function, and in regulating inflammatory processes. Heme oxygenase (major role in catabolism of heme into biliverdin, carbon monoxide (CO), and iron) has similar effects as well. CO has been suggested as the molecule that is responsible for many of the above mentioned cytoprotective and therapeutic pathways as CO is a signaling molecule in the control of physiological functions. This is counterintuitive as toxic effects are related to its binding to hemoglobin. However, CO is normally produced in the body. Experimental evidence indicates that this toxic gas, CO, exerts cytoprotective properties related to cellular stress including the heart and is being assessed for its cytoprotective and cytotherapeutic properties. While survival of adult cardiomyocytes depends on oxidative phosphorylation (survival and resulting cardiac function is impaired by mitochondrial damage), mitochondrial biogenesis is modified by the heme oxygenase-1/CO system and can result in promotion of mitochondrial biogenesis by associating mitochondrial redox status to the redox-active transcription factors. It has been suggested that the heme oxygenase-1/CO system is important in differentiation of embryonic stem cells and maturation of cardiomyocytes which is thought to mitigate progression of degenerative cardiovascular diseases. Effects on other cardiac cells are being studied. Acute exposure to air pollution (and, therefore, CO) is associated with cardiovascular mortality, myocardial infarction, and heart failure, but changes in the endogenous heme oxygenase-1 system (and, thereby, CO) positively affect cardiovascular health. We will review the effect of CO on heart health and function in this article.
Collapse
Affiliation(s)
- Vicki L Mahan
- Department of Surgery and Pediatrics, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
7
|
Häkli M, Kreutzer J, Mäki AJ, Välimäki H, Lappi H, Huhtala H, Kallio P, Aalto-Setälä K, Pekkanen-Mattila M. Human induced pluripotent stem cell-based platform for modeling cardiac ischemia. Sci Rep 2021; 11:4153. [PMID: 33603154 PMCID: PMC7893031 DOI: 10.1038/s41598-021-83740-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 02/05/2021] [Indexed: 01/31/2023] Open
Abstract
Ischemic heart disease is a major cause of death worldwide, and the only available therapy to salvage the tissue is reperfusion, which can initially cause further damage. Many therapeutics that have been promising in animal models have failed in human trials. Thus, functional human based cardiac ischemia models are required. In this study, a human induced pluripotent stem cell derived-cardiomyocyte (hiPSC-CM)-based platform for modeling ischemia-reperfusion was developed utilizing a system enabling precise control over oxygen concentration and real-time monitoring of the oxygen dynamics as well as iPS-CM functionality. In addition, morphology and expression of hypoxia-related genes and proteins were evaluated as hiPSC-CM response to 8 or 24 h hypoxia and 24 h reoxygenation. During hypoxia, initial decrease in hiPSC-CM beating frequency was observed, after which the CMs adapted to the conditions and the beating frequency gradually increased already before reoxygenation. During reoxygenation, the beating frequency typically first surpassed the baseline before settling down to the values close the baseline. Furthermore, slowing on the field potential propagation throughout the hiPSC-CM sheet as well as increase in depolarization time and decrease in overall field potential duration were observed during hypoxia. These changes were reversed during reoxygenation. Disorganization of sarcomere structures was observed after hypoxia and reoxygenation, supported by decrease in the expression of sarcomeric proteins. Furthermore, increase in the expression of gene encoding glucose transporter 1 was observed. These findings indicate, that despite their immature phenotype, hiPSC-CMs can be utilized in modeling ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Martta Häkli
- Heart Group, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland.
| | - Joose Kreutzer
- Micro- and Nanosystems Research Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Antti-Juhana Mäki
- Micro- and Nanosystems Research Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Hannu Välimäki
- Micro- and Nanosystems Research Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Henna Lappi
- Heart Group, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland
| | - Heini Huhtala
- Faculty of Social Sciences, Tampere University, Tampere, Finland
| | - Pasi Kallio
- Micro- and Nanosystems Research Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Katriina Aalto-Setälä
- Heart Group, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland
| | - Mari Pekkanen-Mattila
- Heart Group, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland
| |
Collapse
|
8
|
Stępniewski J, Tomczyk M, Andrysiak K, Kraszewska I, Martyniak A, Langrzyk A, Kulik K, Wiśniewska E, Jeż M, Florczyk-Soluch U, Polak K, Podkalicka P, Kachamakova-Trojanowska N, Józkowicz A, Jaźwa-Kusior A, Dulak J. Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes, in Contrast to Adipose Tissue-Derived Stromal Cells, Efficiently Improve Heart Function in Murine Model of Myocardial Infarction. Biomedicines 2020; 8:biomedicines8120578. [PMID: 33297443 PMCID: PMC7762393 DOI: 10.3390/biomedicines8120578] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 11/30/2020] [Accepted: 12/05/2020] [Indexed: 12/23/2022] Open
Abstract
Cell therapies are extensively tested to restore heart function after myocardial infarction (MI). Survival of any cell type after intracardiac administration, however, may be limited due to unfavorable conditions of damaged tissue. Therefore, the aim of this study was to evaluate the therapeutic effect of adipose-derived stromal cells (ADSCs) and human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) overexpressing either the proangiogenic SDF-1α or anti-inflammatory heme oxygenase-1 (HO-1) in a murine model of MI. ADSCs and hiPSCs were transduced with lentiviral vectors encoding luciferase (Luc), GFP and either HO-1 or SDF-1α. hiPSCs were then differentiated to hiPSC-CMs using small molecules modulating the WNT pathway. Genetically modified ADSCs were firstly administered via intracardiac injection after MI induction in Nude mice. Next, ADSCs-Luc-GFP and genetically modified hiPSC-CMs were injected into the hearts of the more receptive NOD/SCID strain to compare the therapeutic effect of both cell types. Ultrasonography, performed on days 7, 14, 28 and 42, revealed a significant decrease of left ventricular ejection fraction (LVEF) in all MI-induced groups. No improvement of LVEF was observed in ADSC-treated Nude and NOD/SCID mice. In contrast, administration of hiPSC-CMs resulted in a substantial increase of LVEF, occurring between 28 and 42 days after MI, and decreased fibrosis, regardless of genetic modification. Importantly, bioluminescence analysis, as well as immunofluorescent staining, confirmed the presence of hiPSC-CMs in murine tissue. Interestingly, the luminescence signal was strongest in hearts treated with hiPSC-CMs overexpressing HO-1. Performed experiments demonstrate that hiPSC-CMs, unlike ADSCs, are effective in improving heart function after MI. Additionally, long-term evaluation of heart function seems to be crucial for proper assessment of the effect of cell administration.
Collapse
Affiliation(s)
- Jacek Stępniewski
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (K.A.); (I.K.); (A.M.); (A.L.); (K.K.); (E.W.); (M.J.); (U.F.-S.); (K.P.); (P.P.); (A.J.); (A.J.-K.)
- Correspondence: (J.S.); (J.D.); Tel.: +48-12-664-6375 (J.D.)
| | - Mateusz Tomczyk
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (K.A.); (I.K.); (A.M.); (A.L.); (K.K.); (E.W.); (M.J.); (U.F.-S.); (K.P.); (P.P.); (A.J.); (A.J.-K.)
| | - Kalina Andrysiak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (K.A.); (I.K.); (A.M.); (A.L.); (K.K.); (E.W.); (M.J.); (U.F.-S.); (K.P.); (P.P.); (A.J.); (A.J.-K.)
| | - Izabela Kraszewska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (K.A.); (I.K.); (A.M.); (A.L.); (K.K.); (E.W.); (M.J.); (U.F.-S.); (K.P.); (P.P.); (A.J.); (A.J.-K.)
| | - Alicja Martyniak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (K.A.); (I.K.); (A.M.); (A.L.); (K.K.); (E.W.); (M.J.); (U.F.-S.); (K.P.); (P.P.); (A.J.); (A.J.-K.)
| | - Agnieszka Langrzyk
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (K.A.); (I.K.); (A.M.); (A.L.); (K.K.); (E.W.); (M.J.); (U.F.-S.); (K.P.); (P.P.); (A.J.); (A.J.-K.)
- Kardio-Med Silesia, Curie-Skłodowskiej 10C, 41-800 Zabrze, Poland
| | - Klaudia Kulik
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (K.A.); (I.K.); (A.M.); (A.L.); (K.K.); (E.W.); (M.J.); (U.F.-S.); (K.P.); (P.P.); (A.J.); (A.J.-K.)
- Kardio-Med Silesia, Curie-Skłodowskiej 10C, 41-800 Zabrze, Poland
| | - Ewa Wiśniewska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (K.A.); (I.K.); (A.M.); (A.L.); (K.K.); (E.W.); (M.J.); (U.F.-S.); (K.P.); (P.P.); (A.J.); (A.J.-K.)
- Kardio-Med Silesia, Curie-Skłodowskiej 10C, 41-800 Zabrze, Poland
| | - Mateusz Jeż
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (K.A.); (I.K.); (A.M.); (A.L.); (K.K.); (E.W.); (M.J.); (U.F.-S.); (K.P.); (P.P.); (A.J.); (A.J.-K.)
| | - Urszula Florczyk-Soluch
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (K.A.); (I.K.); (A.M.); (A.L.); (K.K.); (E.W.); (M.J.); (U.F.-S.); (K.P.); (P.P.); (A.J.); (A.J.-K.)
| | - Katarzyna Polak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (K.A.); (I.K.); (A.M.); (A.L.); (K.K.); (E.W.); (M.J.); (U.F.-S.); (K.P.); (P.P.); (A.J.); (A.J.-K.)
| | - Paulina Podkalicka
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (K.A.); (I.K.); (A.M.); (A.L.); (K.K.); (E.W.); (M.J.); (U.F.-S.); (K.P.); (P.P.); (A.J.); (A.J.-K.)
| | | | - Alicja Józkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (K.A.); (I.K.); (A.M.); (A.L.); (K.K.); (E.W.); (M.J.); (U.F.-S.); (K.P.); (P.P.); (A.J.); (A.J.-K.)
| | - Agnieszka Jaźwa-Kusior
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (K.A.); (I.K.); (A.M.); (A.L.); (K.K.); (E.W.); (M.J.); (U.F.-S.); (K.P.); (P.P.); (A.J.); (A.J.-K.)
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (K.A.); (I.K.); (A.M.); (A.L.); (K.K.); (E.W.); (M.J.); (U.F.-S.); (K.P.); (P.P.); (A.J.); (A.J.-K.)
- Correspondence: (J.S.); (J.D.); Tel.: +48-12-664-6375 (J.D.)
| |
Collapse
|
9
|
Constantinou C, Miranda AMA, Chaves P, Bellahcene M, Massaia A, Cheng K, Samari S, Rothery SM, Chandler AM, Schwarz RP, Harding SE, Punjabi P, Schneider MD, Noseda M. Human pluripotent stem cell-derived cardiomyocytes as a target platform for paracrine protection by cardiac mesenchymal stromal cells. Sci Rep 2020; 10:13016. [PMID: 32747668 PMCID: PMC7400574 DOI: 10.1038/s41598-020-69495-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 05/22/2020] [Indexed: 12/14/2022] Open
Abstract
Ischemic heart disease remains the foremost cause of death globally, with survivors at risk for subsequent heart failure. Paradoxically, cell therapies to offset cardiomyocyte loss after ischemic injury improve long-term cardiac function despite a lack of durable engraftment. An evolving consensus, inferred preponderantly from non-human models, is that transplanted cells benefit the heart via early paracrine signals. Here, we tested the impact of paracrine signals on human cardiomyocytes, using human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) as the target of mouse and human cardiac mesenchymal stromal cells (cMSC) with progenitor-like features. In co-culture and conditioned medium studies, cMSCs markedly inhibited human cardiomyocyte death. Little or no protection was conferred by mouse tail tip or human skin fibroblasts. Consistent with the results of transcriptomic profiling, functional analyses showed that the cMSC secretome suppressed apoptosis and preserved cardiac mitochondrial transmembrane potential. Protection was independent of exosomes under the conditions tested. In mice, injecting cMSC-conditioned media into the infarct border zone reduced apoptotic cardiomyocytes > 70% locally. Thus, hPSC-CMs provide an auspicious, relevant human platform to investigate extracellular signals for cardiac muscle survival, substantiating human cardioprotection by cMSCs, and suggesting the cMSC secretome or its components as potential cell-free therapeutic products.
Collapse
Affiliation(s)
- Chrystalla Constantinou
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Antonio M A Miranda
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London, W12 0NN, UK
- British Heart Foundation Centre for Research Excellence and Centre for Regenerative Medicine, Imperial College London, London, W12 0NN, UK
| | - Patricia Chaves
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London, W12 0NN, UK
- British Heart Foundation Centre for Research Excellence and Centre for Regenerative Medicine, Imperial College London, London, W12 0NN, UK
| | - Mohamed Bellahcene
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London, W12 0NN, UK
- British Heart Foundation Centre for Research Excellence and Centre for Regenerative Medicine, Imperial College London, London, W12 0NN, UK
| | - Andrea Massaia
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London, W12 0NN, UK
- British Heart Foundation Centre for Research Excellence and Centre for Regenerative Medicine, Imperial College London, London, W12 0NN, UK
| | - Kevin Cheng
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Sara Samari
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London, W12 0NN, UK
- British Heart Foundation Centre for Research Excellence and Centre for Regenerative Medicine, Imperial College London, London, W12 0NN, UK
| | - Stephen M Rothery
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Anita M Chandler
- Kardia Therapeutics, Houston, TX, 77030, USA
- Department of Bioengineering, BioScience Research Collaborative, Rice University, Houston, TX, 77005, USA
| | - Richard P Schwarz
- Kardia Therapeutics, Houston, TX, 77030, USA
- CV Ventures, LLC, Blue Bell, PA, 19422, USA
| | - Sian E Harding
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London, W12 0NN, UK
- British Heart Foundation Centre for Research Excellence and Centre for Regenerative Medicine, Imperial College London, London, W12 0NN, UK
| | - Prakash Punjabi
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London, W12 0NN, UK
- British Heart Foundation Centre for Research Excellence and Centre for Regenerative Medicine, Imperial College London, London, W12 0NN, UK
- Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, W12 0HS, UK
| | - Michael D Schneider
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London, W12 0NN, UK.
- British Heart Foundation Centre for Research Excellence and Centre for Regenerative Medicine, Imperial College London, London, W12 0NN, UK.
| | - Michela Noseda
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London, W12 0NN, UK.
- British Heart Foundation Centre for Research Excellence and Centre for Regenerative Medicine, Imperial College London, London, W12 0NN, UK.
| |
Collapse
|
10
|
Selective protection of human cardiomyocytes from anthracycline cardiotoxicity by small molecule inhibitors of MAP4K4. Sci Rep 2020; 10:12060. [PMID: 32694738 PMCID: PMC7374628 DOI: 10.1038/s41598-020-68907-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 06/25/2020] [Indexed: 12/11/2022] Open
Abstract
Given the poor track record to date of animal models for creating cardioprotective drugs, human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) have been proposed as a therapeutically relevant human platform to guide target validation and cardiac drug development. Mitogen-Activated Protein Kinase Kinase Kinase Kinase-4 (MAP4K4) is an “upstream” member of the MAPK superfamily that is implicated in human cardiac muscle cell death from oxidative stress, based on gene silencing and pharmacological inhibition in hPSC-CMs. A further role for MAP4K4 was proposed in heart muscle cell death triggered by cardiotoxic anti-cancer drugs, given its reported activation in failing human hearts with doxorubicin (DOX) cardiomyopathy, and its activation acutely by DOX in cultured cardiomyocytes. Here, we report successful protection from DOX in two independent hPSC-CM lines, using two potent, highly selective MAP4K4 inhibitors. The MAP4K4 inhibitors enhanced viability and reduced apoptosis at otherwise lethal concentrations of DOX, and preserved cardiomyocyte function, as measured by spontaneous calcium transients, at sub-maximal ones. Notably, in contrast, no intereference was seen in tumor cell killing, caspase activation, or mitochondrial membrane dissipation by DOX, in human cancer cell lines. Thus, MAP4K4 is a plausible, tractable, selective therapeutic target in DOX-induced human heart muscle cell death.
Collapse
|
11
|
Modeling Cardiovascular Diseases with hiPSC-Derived Cardiomyocytes in 2D and 3D Cultures. Int J Mol Sci 2020; 21:ijms21093404. [PMID: 32403456 PMCID: PMC7246991 DOI: 10.3390/ijms21093404] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 12/15/2022] Open
Abstract
In the last decade, the generation of cardiac disease models based on human-induced pluripotent stem cells (hiPSCs) has become of common use, providing new opportunities to overcome the lack of appropriate cardiac models. Although much progress has been made toward the generation of hiPSC-derived cardiomyocytes (hiPS-CMs), several lines of evidence indicate that two-dimensional (2D) cell culturing presents significant limitations, including hiPS-CMs immaturity and the absence of interaction between different cell types and the extracellular matrix. More recently, new advances in bioengineering and co-culture systems have allowed the generation of three-dimensional (3D) constructs based on hiPSC-derived cells. Within these systems, biochemical and physical stimuli influence the maturation of hiPS-CMs, which can show structural and functional properties more similar to those present in adult cardiomyocytes. In this review, we describe the latest advances in 2D- and 3D-hiPSC technology for cardiac disease mechanisms investigation, drug development, and therapeutic studies.
Collapse
|
12
|
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: 0.8] [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
|
13
|
Golforoush P, Schneider MD. Intensive care for human hearts in pluripotent stem cell models. NPJ Regen Med 2020; 5:4. [PMID: 32194989 PMCID: PMC7060343 DOI: 10.1038/s41536-020-0090-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 02/06/2020] [Indexed: 12/12/2022] Open
Abstract
Successful drug discovery is ultimately contingent on the availability of workable, relevant, predictive model systems. Conversely, for cardiac muscle, the lack of human preclinical models to inform target validation and compound development has likely contributed to the perennial problem of clinical trial failures, despite encouraging non-human results. By contrast, human cardiomyocytes produced from pluripotent stem cell models have recently been applied to safety pharmacology, phenotypic screening, target validation and high-throughput assays, facilitating cardiac drug discovery. Here, we review the impact of human pluripotent stem cell models in cardiac drug discovery, discussing the range of applications, readouts, and disease models employed, along with the challenges and prospects to advance this fruitful mode of research further.
Collapse
Affiliation(s)
- Pelin Golforoush
- National Heart and Lung Institute, Imperial College London, London, W12 0NN UK
| | | |
Collapse
|
14
|
Okano S, Shiba Y. Therapeutic Potential of Pluripotent Stem Cells for Cardiac Repair after Myocardial Infarction. Biol Pharm Bull 2019; 42:524-530. [PMID: 30930411 DOI: 10.1248/bpb.b18-00257] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Myocardial infarction occurs as a result of acute arteriosclerotic plaque rupture in the coronary artery, triggering strong inflammatory responses. The necrotic cardiomyocytes are gradually replaced with noncontractile scar tissue that eventually manifests as heart failure. Pluripotent stem cells (PSCs) show great promise for widespread clinical applications, particularly for tissue regeneration, and are being actively studied around the world to help elucidate disease mechanisms and in the development of new drugs. Human induced PSCs also show potential for regeneration of the myocardial tissue in experiments with small animals and in in vitro studies. Although emerging evidence points to the effectiveness of these stem cell-derived cardiomyocytes in cardiac regeneration, several challenges remain before clinical application can become a reality. Here, we provide an overview of the present state of PSC-based heart regeneration and highlight the remaining hurdles, with a particular focus on graft survival, immunogenicity, posttransplant arrhythmia, maintained function, and tumor formation. Rapid progress in this field along with advances in biotechnology are expected to resolve these issues, which will require international collaboration and standardization.
Collapse
Affiliation(s)
- Satomi Okano
- Department of Regenerative Science and Medicine, Shinshu University.,Institute for Biomedical Sciences, Shinshu University
| | - Yuji Shiba
- Department of Regenerative Science and Medicine, Shinshu University.,Institute for Biomedical Sciences, Shinshu University.,Department of Cardiovascular Medicine, Shinshu University
| |
Collapse
|
15
|
HiPS-Cardiac Trilineage Cell Generation and Transplantation: a Novel Therapy for Myocardial Infarction. J Cardiovasc Transl Res 2019; 13:110-119. [PMID: 31152358 DOI: 10.1007/s12265-019-09891-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 04/29/2019] [Indexed: 12/12/2022]
Abstract
Despite primary percutaneous coronary intervention (PPCI) and the availability of optimal medications, including dual antiplatelet therapy (DAPT), most patients still experience major adverse cardiovascular events (MACEs) due to frequent recurrence of thrombotic complications and myocardial infarction (MI). MI occurs secondary to a massive loss of endothelial cells (ECs), vascular smooth muscle cells (VSMCs), and cardiomyocytes (CMs). The adult cardiovascular system gradually loses the ability to spontaneously and regularly regenerate ECs, VSMCs, and CMs. However, human cells can be induced by cytokines and growth factors to regenerate human-induced pluripotent stem cells (hiPSCs), which progress to produce cardiac trilineage cells (CTCs) such as ECs, VSMCs, and CMs, replacing lost cells and inducing myocardial repair. Nevertheless, the processes and pathways involved in hiPSC-CTC generation and their potential therapeutic effects remain unknown. Herein, we provide evidence of in vitro CTC generation, the pathways involved, in vivo transplantation, and its therapeutic effect, which may provide novel targets in regenerative medicine for the treatment of cardiovascular diseases (CVDs).
Collapse
|
16
|
Fiedler LR, Chapman K, Xie M, Maifoshie E, Jenkins M, Golforoush PA, Bellahcene M, Noseda M, Faust D, Jarvis A, Newton G, Paiva MA, Harada M, Stuckey DJ, Song W, Habib J, Narasimhan P, Aqil R, Sanmugalingam D, Yan R, Pavanello L, Sano M, Wang SC, Sampson RD, Kanayaganam S, Taffet GE, Michael LH, Entman ML, Tan TH, Harding SE, Low CMR, Tralau-Stewart C, Perrior T, Schneider MD. MAP4K4 Inhibition Promotes Survival of Human Stem Cell-Derived Cardiomyocytes and Reduces Infarct Size In Vivo. Cell Stem Cell 2019; 24:579-591.e12. [PMID: 30853557 PMCID: PMC6458995 DOI: 10.1016/j.stem.2019.01.013] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 12/24/2018] [Accepted: 01/30/2019] [Indexed: 12/17/2022]
Abstract
Heart disease is a paramount cause of global death and disability. Although cardiomyocyte death plays a causal role and its suppression would be logical, no clinical counter-measures target the responsible intracellular pathways. Therapeutic progress has been hampered by lack of preclinical human validation. Mitogen-activated protein kinase kinase kinase kinase-4 (MAP4K4) is activated in failing human hearts and relevant rodent models. Using human induced-pluripotent-stem-cell-derived cardiomyocytes (hiPSC-CMs) and MAP4K4 gene silencing, we demonstrate that death induced by oxidative stress requires MAP4K4. Consequently, we devised a small-molecule inhibitor, DMX-5804, that rescues cell survival, mitochondrial function, and calcium cycling in hiPSC-CMs. As proof of principle that drug discovery in hiPSC-CMs may predict efficacy in vivo, DMX-5804 reduces ischemia-reperfusion injury in mice by more than 50%. We implicate MAP4K4 as a well-posed target toward suppressing human cardiac cell death and highlight the utility of hiPSC-CMs in drug discovery to enhance cardiomyocyte survival.
Collapse
Affiliation(s)
- Lorna R Fiedler
- British Heart Foundation Centre of Research Excellence, National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| | - Kathryn Chapman
- Drug Discovery Centre, Department of Medicine, Imperial College London, London SW7 2AZ, UK; Department of Surgery and Cancer, Imperial College London, London W12 0NN, UK; Domainex, Chesterford Research Park, Little Chesterford, Saffron Walden, Essex CB10 1XL, UK
| | - Min Xie
- Michael E. DeBakey Heart Center, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Evie Maifoshie
- British Heart Foundation Centre of Research Excellence, National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| | - Micaela Jenkins
- British Heart Foundation Centre of Research Excellence, National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| | - Pelin Arabacilar Golforoush
- British Heart Foundation Centre of Research Excellence, National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| | - Mohamed Bellahcene
- British Heart Foundation Centre of Research Excellence, National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| | - Michela Noseda
- British Heart Foundation Centre of Research Excellence, National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| | - Dörte Faust
- British Heart Foundation Centre of Research Excellence, National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| | - Ashley Jarvis
- Domainex, Chesterford Research Park, Little Chesterford, Saffron Walden, Essex CB10 1XL, UK
| | - Gary Newton
- Domainex, Chesterford Research Park, Little Chesterford, Saffron Walden, Essex CB10 1XL, UK
| | - Marta Abreu Paiva
- British Heart Foundation Centre of Research Excellence, National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| | - Mutsuo Harada
- British Heart Foundation Centre of Research Excellence, National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| | - Daniel J Stuckey
- British Heart Foundation Centre of Research Excellence, National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| | - Weihua Song
- British Heart Foundation Centre of Research Excellence, National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| | - Josef Habib
- British Heart Foundation Centre of Research Excellence, National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| | - Priyanka Narasimhan
- Domainex, Chesterford Research Park, Little Chesterford, Saffron Walden, Essex CB10 1XL, UK
| | - Rehan Aqil
- Domainex, Chesterford Research Park, Little Chesterford, Saffron Walden, Essex CB10 1XL, UK
| | - Devika Sanmugalingam
- Domainex, Chesterford Research Park, Little Chesterford, Saffron Walden, Essex CB10 1XL, UK
| | - Robert Yan
- Domainex, Chesterford Research Park, Little Chesterford, Saffron Walden, Essex CB10 1XL, UK
| | - Lorenzo Pavanello
- Domainex, Chesterford Research Park, Little Chesterford, Saffron Walden, Essex CB10 1XL, UK
| | - Motoaki Sano
- Michael E. DeBakey Heart Center, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sam C Wang
- Michael E. DeBakey Heart Center, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Robert D Sampson
- British Heart Foundation Centre of Research Excellence, National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| | - Sunthar Kanayaganam
- British Heart Foundation Centre of Research Excellence, National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| | - George E Taffet
- Michael E. DeBakey Heart Center, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lloyd H Michael
- Michael E. DeBakey Heart Center, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mark L Entman
- Michael E. DeBakey Heart Center, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Tse-Hua Tan
- Immunology Research Center, National Health Research Institutes, Zhunan 35053, Taiwan; Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sian E Harding
- British Heart Foundation Centre of Research Excellence, National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| | - Caroline M R Low
- Drug Discovery Centre, Department of Medicine, Imperial College London, London SW7 2AZ, UK
| | | | - Trevor Perrior
- Domainex, Chesterford Research Park, Little Chesterford, Saffron Walden, Essex CB10 1XL, UK
| | - Michael D Schneider
- British Heart Foundation Centre of Research Excellence, National Heart and Lung Institute, Imperial College London, London W12 0NN, UK; Michael E. DeBakey Heart Center, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
17
|
Floresta G, Amata E, Gentile D, Romeo G, Marrazzo A, Pittalà V, Salerno L, Rescifina A. Fourfold Filtered Statistical/Computational Approach for the Identification of Imidazole Compounds as HO-1 Inhibitors from Natural Products. Mar Drugs 2019; 17:md17020113. [PMID: 30759842 PMCID: PMC6409521 DOI: 10.3390/md17020113] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 02/08/2019] [Accepted: 02/09/2019] [Indexed: 12/16/2022] Open
Abstract
Over-regulation of Heme oxygenase 1 (HO-1) has been recently identified in many types of human cancer, and in these cases, poor clinical outcomes are normally reported. Indeed, the inhibition of HO-1 is being considered as an anticancer approach. Imidazole scaffold is normally present in most of the classical HO-1 inhibitors and seems indispensable to the inhibitory activity due to its strong interaction with the Fe(II) of the heme group. In this paper, we searched for new potentially HO-1 inhibitors among three different databases: Marine Natural Products (MNP), ZINC Natural Products (ZNP) and Super Natural II (SN2). 484,527 compounds were retrieved from the databases and filtered through four statistical/computational filters (2D descriptors, 2D-QSAR pharmacophoric model, 3D-QSAR pharmacophoric model, and docking). Different imidazole-based compounds were suggested by our methodology to be potentially active in inhibiting the HO-1, and the results have been rationalized by the bioactivity of the filtered molecules reported in the literature.
Collapse
Affiliation(s)
- Giuseppe Floresta
- Department of Drug Sciences, University of Catania, V.le A. Doria, 95125 Catania, Italy.
| | - Emanuele Amata
- Department of Drug Sciences, University of Catania, V.le A. Doria, 95125 Catania, Italy.
| | - Davide Gentile
- Department of Drug Sciences, University of Catania, V.le A. Doria, 95125 Catania, Italy.
| | - Giuseppe Romeo
- Department of Drug Sciences, University of Catania, V.le A. Doria, 95125 Catania, Italy.
| | - Agostino Marrazzo
- Department of Drug Sciences, University of Catania, V.le A. Doria, 95125 Catania, Italy.
| | - Valeria Pittalà
- Department of Drug Sciences, University of Catania, V.le A. Doria, 95125 Catania, Italy.
| | - Loredana Salerno
- Department of Drug Sciences, University of Catania, V.le A. Doria, 95125 Catania, Italy.
| | - Antonio Rescifina
- Department of Drug Sciences, University of Catania, V.le A. Doria, 95125 Catania, Italy.
- Consorzio Interuniversitario Nazionale di ricerca in Metodologie e Processi Innovativi di Sintesi (C.I.N.M.P.S.), Via E. Orabona, 4, 70125 Bari, Italy.
| |
Collapse
|
18
|
Wang X, Han Z, Yu Y, Xu Z, Cai B, Yuan Y. Potential Applications of Induced Pluripotent Stem Cells for Cardiovascular Diseases. Curr Drug Targets 2018; 20:763-774. [PMID: 30539693 DOI: 10.2174/1389450120666181211164147] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/05/2018] [Accepted: 12/05/2018] [Indexed: 12/13/2022]
Abstract
Owning the high incidence and disability rate in the past decades, to be expected, cardiovascular diseases (CVDs) have become one of the leading death causes worldwide. Currently, induced pluripotent stem cells (iPSCs), with the potential to form fresh myocardium and improve the functions of damaged hearts, have been studied widely in experimental CVD therapy. Moreover, iPSC-derived cardiomyocytes (CMs), as novel disease models, play a significant role in drug screening, drug safety assessment, along with the exploration of pathological mechanisms of diseases. Furthermore, a lot of studies have been carried out to clarify the biological basis of iPSCs and its derived cells in the treatment of CVDs. Their molecular mechanisms were associated with release of paracrine factors, regulation of miRNAs, mechanical support of new tissues, activation of specific pathways and specific enzymes, etc. In addition, a few small chemical molecules and suitable biological scaffolds play positive roles in enhancing the efficiency of iPSC transplantation. This article reviews the development and limitations of iPSCs in CVD therapy, and summarizes the latest research achievements regarding the application of iPSCs in CVDs.
Collapse
Affiliation(s)
- Xiaotong Wang
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Zhenbo Han
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Ying Yu
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Zihang Xu
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Benzhi Cai
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Ye Yuan
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| |
Collapse
|
19
|
Allaway RJ, La Rosa S, Guinney J, Gosline SJC. Probing the chemical-biological relationship space with the Drug Target Explorer. J Cheminform 2018; 10:41. [PMID: 30128806 PMCID: PMC6102167 DOI: 10.1186/s13321-018-0297-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 08/16/2018] [Indexed: 01/29/2023] Open
Abstract
Modern phenotypic high-throughput screens (HTS) present several challenges including identifying the target(s) that mediate the effect seen in the screen, characterizing ‘hits’ with a polypharmacologic target profile, and contextualizing screen data within the large space of drugs and screening models. To address these challenges, we developed the Drug–Target Explorer. This tool allows users to query molecules within a database of experimentally-derived and curated compound-target interactions to identify structurally similar molecules and their targets. It enables network-based visualizations of the compound-target interaction space, and incorporates comparisons to publicly-available in vitro HTS datasets. Furthermore, users can identify molecules using a query target or set of targets. The Drug Target Explorer is a multifunctional platform for exploring chemical space as it relates to biological targets, and may be useful at several steps along the drug development pipeline including target discovery, structure–activity relationship, and lead compound identification studies.
Collapse
Affiliation(s)
- Robert J Allaway
- Sage Bionetworks, 1100 Fairview Avenue N, Seattle, WA, 98109, USA
| | | | - Justin Guinney
- Sage Bionetworks, 1100 Fairview Avenue N, Seattle, WA, 98109, USA
| | - Sara J C Gosline
- Sage Bionetworks, 1100 Fairview Avenue N, Seattle, WA, 98109, USA.
| |
Collapse
|