1
|
Wang WD, Fan XY, Wei XQ, Chai WJ, Li FH, Gao K, Liu B, Guo SZ. Synergistic combinations of Angelica sinensis for myocardial infarction treatment: network pharmacology and quadratic optimization approach. Front Pharmacol 2024; 15:1466208. [PMID: 39717556 PMCID: PMC11663646 DOI: 10.3389/fphar.2024.1466208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 11/22/2024] [Indexed: 12/25/2024] Open
Abstract
Background and aim Angelica sinensis (Oliv.) Diels (Danggui, DG), exhibits potential in myocardial infarction (MI) treatment. However, research on its synergistic combinations for cardioprotective effects has been limited owing to inadequate approaches. Experimental procedure We identified certain phenolic acids and phthalein compounds in DG. Network pharmacology analysis and experimental validation revealed the components that protected H9c2 cells and reduced lactate dehydrogenase levels. Subsequently, a combination of computational experimental strategies and a secondary phenotypic optimization platform was employed to identify effective component combinations with synergistic interactions. The Chou-Talalay and Zero Interaction Potency (ZIP) models were utilized to quantify the synergistic relationships. The optimal combination identified, Z-Ligustide and Chlorogenic acid (Z-LIG/CGA), was evaluated for its protective effects on cardiac function and cardiomyocytes apoptosis induced by inflammatory in a mouse model of induced by left anterior descending coronary artery ligation. Flow cytometry was further utilized to detect the polarization ratio of M1/M2 macrophages and the expression of inflammatory cytokines in serum was measured, assessing the inhibition of inflammatory responses and pro-inflammatory signaling factors by Z-LIG/CGA. Key results Quadratic surface analysis revealed that the Z-LIG/CGA combination displayed synergistic cardioprotective effects (combination index value <1; ZIP value >10). In vivo, Z-LIG/CGA significantly improved cardiac function and reduced the fibrotic area in mice post-MI, surpassing the results in groups treated with Z-LIG or CGA alone. Compared to the MI group, the Z-LIG/CGA group exhibited decreased ratios of the myocardial cell apoptosis-related proteins BAX/Bcl-2 and Cleaved Caspase-3/Caspase-3 in mice. Further research revealed that Z-LIG/CGA treatment significantly increased IL-1R2 levels, significantly decreased IL-17RA levels, and inhibited the activation of p-STAT1, thereby alleviating cell apoptosis after MI. Additionally, the Z-LIG/CGA combination significantly inhibited the ratio of M1/M2 macrophages and suppressed the expression levels of pro-inflammatory cytokines IL-1β, IL-6, IL-17, and TNF-α in the serum. Conclusion and implications We successfully identified a synergistic drug combination, Z-LIG/CGA, which improves MI outcomes by inhibiting cardiomyocyte apoptosis and inflammatory damage through modulating macrophage polarization and regulating the IL-1R2/IL-17RA/STAT1 signaling pathway. This study provides a charming paradigm to explore effective drug combinations in traditional Chinese medicine and a promising treatment for MI.
Collapse
Affiliation(s)
- Wen-Di Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xin-Yi Fan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiao-Qi Wei
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Wang-Jing Chai
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Fang-He Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Kuo Gao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Bin Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- The Key Research Laboratory of “Exploring Effective Substance in Classic and Famous Prescriptions of Traditional Chinese Medicine”, The State Administration of Traditional Chinese Medicine, Beijing, China
| | - Shu-Zhen Guo
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
2
|
Szydlak R. Mesenchymal stem cells in ischemic tissue regeneration. World J Stem Cells 2023; 15:16-30. [PMID: 36909782 PMCID: PMC9993139 DOI: 10.4252/wjsc.v15.i2.16] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/10/2022] [Accepted: 01/19/2023] [Indexed: 02/21/2023] Open
Abstract
Diseases caused by ischemia are one of the leading causes of death in the world. Current therapies for treating acute myocardial infarction, ischemic stroke, and critical limb ischemia do not complete recovery. Regenerative therapies opens new therapeutic strategy in the treatment of ischemic disorders. Mesenchymal stem cells (MSCs) are the most promising option in the field of cell-based therapies, due to their secretory and immunomodulatory abilities, that contribute to ease inflammation and promote the regeneration of damaged tissues. This review presents the current knowledge of the mechanisms of action of MSCs and their therapeutic effects in the treatment of ischemic diseases, described on the basis of data from in vitro experiments and preclinical animal studies, and also summarize the effects of using these cells in clinical trial settings. Since the obtained therapeutic benefits are not always satisfactory, approaches aimed at enhancing the effect of MSCs in regenerative therapies are presented at the end.
Collapse
Affiliation(s)
- Renata Szydlak
- Chair of Medical Biochemistry, Faculty of Medicine, Jagiellonian University Medical College, Kraków 31-034, Poland
| |
Collapse
|
3
|
Kang Y, Nasr M, Guo Y, Uchida S, Weirick T, Li H, Kim J, Moore JB, Muthusamy S, Bolli R, Wysoczynski M. Administration of cardiac mesenchymal cells modulates innate immunity in the acute phase of myocardial infarction in mice. Sci Rep 2020; 10:14754. [PMID: 32901075 PMCID: PMC7479609 DOI: 10.1038/s41598-020-71580-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 08/19/2020] [Indexed: 01/07/2023] Open
Abstract
Although cardiac mesenchymal cell (CMC) therapy mitigates post-infarct cardiac dysfunction, the underlying mechanisms remain unidentified. It is acknowledged that donor cells are neither appreciably retained nor meaningfully contribute to tissue regeneration-suggesting a paracrine-mediated mechanism of action. As the immune system is inextricably linked to wound healing/remodeling in the ischemically injured heart, the reparative actions of CMCs may be attributed to their immunoregulatory properties. The current study evaluated the consequences of CMC administration on post myocardial infarction (MI) immune responses in vivo and paracrine-mediated immune cell function in vitro. CMC administration preferentially elicited the recruitment of cell types associated with innate immunity (e.g., monocytes/macrophages and neutrophils). CMC paracrine signaling assays revealed enhancement in innate immune cell chemoattraction, survival, and phagocytosis, and diminished pro-inflammatory immune cell activation; data that identifies and catalogues fundamental immunomodulatory properties of CMCs, which have broad implications regarding the mechanism of action of CMCs in cardiac repair.
Collapse
Affiliation(s)
- Yi Kang
- Diabetes and Obesity Center, University of Louisville School of Medicine, 580 South Preston St. - Rm 204B, Louisville, KY, 40202, USA
| | - Marjan Nasr
- Diabetes and Obesity Center, University of Louisville School of Medicine, 580 South Preston St. - Rm 204B, Louisville, KY, 40202, USA
| | - Yiru Guo
- Institute of Molecular Cardiology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Shizuka Uchida
- Cardiovascular Innovation Institute, Univerity of Louisville School of Medicine, Louisville, KY, USA
| | - Tyler Weirick
- Cardiovascular Innovation Institute, Univerity of Louisville School of Medicine, Louisville, KY, USA
| | - Hong Li
- Diabetes and Obesity Center, University of Louisville School of Medicine, 580 South Preston St. - Rm 204B, Louisville, KY, 40202, USA
| | - Jae Kim
- Diabetes and Obesity Center, University of Louisville School of Medicine, 580 South Preston St. - Rm 204B, Louisville, KY, 40202, USA
| | - Joseph B Moore
- Diabetes and Obesity Center, University of Louisville School of Medicine, 580 South Preston St. - Rm 204B, Louisville, KY, 40202, USA
| | - Senthilkumar Muthusamy
- Diabetes and Obesity Center, University of Louisville School of Medicine, 580 South Preston St. - Rm 204B, Louisville, KY, 40202, USA
| | - Roberto Bolli
- Institute of Molecular Cardiology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Marcin Wysoczynski
- Diabetes and Obesity Center, University of Louisville School of Medicine, 580 South Preston St. - Rm 204B, Louisville, KY, 40202, USA.
| |
Collapse
|
4
|
Hu H, Zhao Q, Liu X, Yan T. Human umbilical cord blood cells rescued traumatic brain injury-induced cardiac and neurological deficits. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:278. [PMID: 32355722 PMCID: PMC7186665 DOI: 10.21037/atm.2020.03.52] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Background Traumatic brain injury (TBI) evokes neurological deficits and induces cardiac dysfunction. Treatment with human umbilical cord blood cells (HUCBCs) represents a potential therapeutic strategy for TBI-induced neurological deficits. The present study aimed to determine whether HUCBCs could ameliorate the cardiac dysfunction and neurological deficits induced by TBI. Methods Adult male C57BL/6J mice were subjected to controlled cortical impact (CCI)-induced TBI and were treated with either HUCBCs (1×106) or phosphate-buffered saline (PBS), via tail vein injections, 3 days after TBI. Neurological and cognitive functions were subsequently evaluated at multiple time points after TBI and cardiac function was assessed by echocardiography 3 and 30 days after TBI. Brain and heart tissues were paraffin-embedded 30 days after TBI. Hematoxylin and eosin (H&E) staining was performed on brain tissue sections to calculate the brain damage volume, and Picro Sirius Red (PSR) staining was performed on heart tissue sections to evaluate myocardial fibrosis. Terminal deoxynucleotide transferase dUTP nick end labeling (TUNEL) staining was employed to assess cell apoptosis 30 days after TBI. Transforming growth factor-beta (TGF-β) and NADPH oxidase-2 (NOX2) levels were assessed to evaluate inflammation and oxidative stress levels 30 days after TBI. Results TBI elicited acute and chronic cardiac deficits, identified by decreased left ventricular ejection fraction (LVEF) and fractional shortening (LVFS) values 3 and 30 days after TBI, in addition to neurological and cognitive deficits. TBI mice treated with HUCBCs exhibited enhanced LVEF and FS values 30 days after TBI compared with untreated TBI controls. HUCBC treatment significantly improved neurological and cognitive functions and reduced cardiomyocyte apoptosis, inflammatory response, oxidative stress, and cardiac fibrosis in heart tissues 30 days after TBI. Conclusions TBI induced both neurological deficits and cardiac dysfunction in mice, which were ameliorated by HUCBC treatment. The anti-inflammatory activities of HUCBCs may contribute to these observed therapeutic effects.
Collapse
Affiliation(s)
- Haotian Hu
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma, Neurorepair, and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Qiang Zhao
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma, Neurorepair, and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Xiaoxuan Liu
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma, Neurorepair, and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Tao Yan
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma, Neurorepair, and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| |
Collapse
|
5
|
Zhang Z, Tian H, Yang C, Liu J, Zhang H, Wang J, Hu S, Sun Z, He K, Chen G. Mesenchymal Stem Cells Promote the Resolution of Cardiac Inflammation After Ischemia Reperfusion Via Enhancing Efferocytosis of Neutrophils. J Am Heart Assoc 2020; 9:e014397. [PMID: 32079474 PMCID: PMC7335576 DOI: 10.1161/jaha.119.014397] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background Neutrophils play a major role in inflammation after myocardial ischemia‐reperfusion (I/R) injury. The effects of mesenchymal stem cells (MSCs) on neutrophils in I/R are complex and not fully understood. This study was designed to investigate the effects and mechanism of MSCs on alleviating myocardial I/R injury in rats. Methods and Results MSCs induced M2 macrophages polarization in vitro and enhanced macrophage efferocytosis of apoptotic neutrophils, measured by fluorescence‐activated cell sorting analysis and immunofluorescence staining. Rats myocardial I/R were induced by transient ligation of left anterior descending coronary. Adipose‐derived MSCs or vehicle were infused at initiation (immediate after reperfusion) or peak of inflammation (24 hours after I/R). Hematoxylin and eosin, 2,3,5‐triphenyltetrazolium chloride/Evans Blue staining and immunofluorescence staining were applied within 72 hours after cell infusion. Cardiac function was assessed by echocardiography and left cardiac catheterization analysis at 28 days post‐operation. MSCs infused immediately and 24 hours later both markedly ameliorated myocardial I/R injury, and immediate infusion had more significant outcome. These improvements were associated with neutrophils infiltration, measured by fluorescence‐activated cell sorting analysis and immunofluorescence staining. When infused immediately, MSCs did not significantly change neutrophil number at 24 hours but CD11b expression was significantly higher. When infused at 24 hours, MSCs markedly decreased neutrophil number by enhanced M2 macrophage infiltration and macrophage efferocytosis of neutrophils within 72 hours. Conclusions Efferocytosis is pivotal to relieve neutrophil‐mediated I/R injury and initial the immune response for healing. MSCs infusion improves cardiac function in rats after myocardial I/R via the possible mechanism of enhancing M2 macrophages‐induced efferocytosis of apoptotic neutrophils.
Collapse
Affiliation(s)
- Zeyu Zhang
- Department of Cardiology Chinese PLA General Hospital Beijing China.,Beijing Key Laboratory of Chronic Heart Failure Precision Medicine Chinese PLA General Hospital Beijing China
| | - Hongzhen Tian
- Department of Cardiology Chinese PLA General Hospital Beijing China.,Department of Cardiology 969 Hospital of Joint Logistic Support Force of PLA Huhehaote China
| | - Chen Yang
- Department of Cardiology Chinese PLA General Hospital Beijing China
| | - Jixuan Liu
- Department of Cardiology Chinese PLA General Hospital Beijing China.,Beijing Key Laboratory of Chronic Heart Failure Precision Medicine Chinese PLA General Hospital Beijing China
| | - Huawei Zhang
- Department of Cardiology Chinese PLA General Hospital Beijing China
| | - Jinda Wang
- Department of Cardiology Chinese PLA General Hospital Beijing China
| | - Shunying Hu
- Department of Cardiology Chinese PLA General Hospital Beijing China
| | - Zhijun Sun
- Department of Cardiology Chinese PLA General Hospital Beijing China
| | - Kunlun He
- Beijing Key Laboratory of Chronic Heart Failure Precision Medicine Chinese PLA General Hospital Beijing China
| | - Guanghui Chen
- Department of Cardiology Chinese PLA General Hospital Beijing China
| |
Collapse
|
6
|
Yasuhara T, Kawauchi S, Kin K, Morimoto J, Kameda M, Sasaki T, Bonsack B, Kingsbury C, Tajiri N, Borlongan CV, Date I. Cell therapy for central nervous system disorders: Current obstacles to progress. CNS Neurosci Ther 2019; 26:595-602. [PMID: 31622035 PMCID: PMC7248543 DOI: 10.1111/cns.13247] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 09/24/2019] [Accepted: 09/29/2019] [Indexed: 12/13/2022] Open
Abstract
Cell therapy for disorders of the central nervous system has progressed to a new level of clinical application. Various clinical studies are underway for Parkinson's disease, stroke, traumatic brain injury, and various other neurological diseases. Recent biotechnological developments in cell therapy have taken advantage of the technology of induced pluripotent stem (iPS) cells. The advent of iPS cells has provided a robust stem cell donor source for neurorestoration via transplantation. Additionally, iPS cells have served as a platform for the discovery of therapeutics drugs, allowing breakthroughs in our understanding of the pathology and treatment of neurological diseases. Despite these recent advances in iPS, adult tissue‐derived mesenchymal stem cells remain the widely used donor for cell transplantation. Mesenchymal stem cells are easily isolated and amplified toward the cells' unique trophic factor‐secretion property. In this review article, the milestone achievements of cell therapy for central nervous system disorders, with equal consideration on the present translational obstacles for clinic application, are described.
Collapse
Affiliation(s)
- Takao Yasuhara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Satoshi Kawauchi
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Kyohei Kin
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Jun Morimoto
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Masahiro Kameda
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Tatsuya Sasaki
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Brooke Bonsack
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida, Tampa, FL, USA
| | - Chase Kingsbury
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida, Tampa, FL, USA
| | - Naoki Tajiri
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences and Medical School, Aichi, Japan
| | - Cesario V Borlongan
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida, Tampa, FL, USA
| | - Isao Date
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Okayama, Japan
| |
Collapse
|
7
|
Colicchia M, Jones DA, Beirne AM, Hussain M, Weeraman D, Rathod K, Veerapen J, Lowdell M, Mathur A. Umbilical cord-derived mesenchymal stromal cells in cardiovascular disease: review of preclinical and clinical data. Cytotherapy 2019; 21:1007-1018. [PMID: 31540804 DOI: 10.1016/j.jcyt.2019.04.056] [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] [Received: 11/07/2018] [Revised: 04/10/2019] [Accepted: 04/15/2019] [Indexed: 02/07/2023]
Abstract
The human umbilical cord has recently emerged as an attractive potential source of mesenchymal stromal cells (MSCs) to be adopted for use in regenerative medicine. Umbilical cord MSCs (UC-MSCs) not only share the same features of all MSCs such as multi-lineage differentiation, paracrine functions and immunomodulatory properties, they also have additional advantages, such as no need for bone marrow aspiration and higher self-renewal capacities. They can be isolated from various compartments of the umbilical cord (UC) and can be used for autologous or allogeneic purposes. In the past decade, they have been adopted in cardiovascular disease and have shown promising results mainly due to their pro-angiogenic and anti-inflammatory properties. This review offers an overview of the biological properties of UC-MSCs describing available pre-clinical and clinical data with respect to their potential therapeutic use in cardiovascular regeneration, with current challenges and future directions discussed.
Collapse
Affiliation(s)
- Martina Colicchia
- Department of Cardiology, Barts Heart Centre, St. Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Daniel A Jones
- Department of Cardiology, Barts Heart Centre, St. Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom.
| | - Anne-Marie Beirne
- Department of Cardiology, Barts Heart Centre, St. Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Mohsin Hussain
- Department of Cardiology, Barts Heart Centre, St. Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Deshan Weeraman
- Department of Cardiology, Barts Heart Centre, St. Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Krishnaraj Rathod
- Department of Cardiology, Barts Heart Centre, St. Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Jessry Veerapen
- Department of Cardiology, Barts Heart Centre, St. Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Mark Lowdell
- Department of Haematology, Royal Free Hospital and University College London, London, United Kingdom
| | - Anthony Mathur
- Department of Cardiology, Barts Heart Centre, St. Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom
| |
Collapse
|
8
|
Tuazon JP, Castelli V, Borlongan CV. Drug-like delivery methods of stem cells as biologics for stroke. Expert Opin Drug Deliv 2019; 16:823-833. [PMID: 31311344 DOI: 10.1080/17425247.2019.1645116] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Introduction: Stem cell therapy is an experimental treatment for brain disorders. Although a cellular product, stem cells can be classified as biologics based on the cells' secretion of therapeutic substances. Treatment with stem cell biologics may appeal to stroke because of the secondary cell death mechanisms, especially neuroinflammation, that are rampant from the onset and remain elevated during the progressive phase of the disease requiring multi-pronged biological targets to effectively abrogate the neurodegenerative pathology. However, the optimal delivery methods, among other logistical approaches (i.e. cell doses and timing of intervention), for stem cell therapy will need to be refined before stem cell biologics can be successfully utilized for stroke in large scale clinical trials. Areas covered: In this review, we discuss how the innate qualities of stem cells characterize them as biologics, how stem cell transplantation may be an ideal treatment for stroke, and the various routes of stem cell administration that have been employed in various preclinical and clinical investigations. Expert opinion: There is a need to optimize the delivery of stem cell biologics for stroke in order to guide the safe and effective translation of this therapy from the laboratory to the clinic.
Collapse
Affiliation(s)
- Julian P Tuazon
- a Center of Excellence for Aging & Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine , Tampa , FL , USA
| | - Vanessa Castelli
- a Center of Excellence for Aging & Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine , Tampa , FL , USA
| | - Cesar V Borlongan
- a Center of Excellence for Aging & Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine , Tampa , FL , USA
| |
Collapse
|
9
|
Zhong Z, Wu H, Zhang Q, Zhong W, Zhao P. Characteristics of T cell receptor repertoires of patients with acute myocardial infarction through high-throughput sequencing. J Transl Med 2019; 17:21. [PMID: 30634977 PMCID: PMC6330436 DOI: 10.1186/s12967-019-1768-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 01/02/2019] [Indexed: 01/19/2023] Open
Abstract
Background T cells are key regulators of immunity and one of the cells recruited in atherosclerosis and participated in various stages of the development of atherosclerosis. Characterizing T-cell receptor (TCR) repertoires is a priority of great scientific interest and potential clinical utility for the early diagnosis, risk stratification and prognostic evaluation of acute myocardial infarction (AMI). Methods The TCR repertoires in 21 subjects including 7 patients with non-ST-segment elevation myocardial infarction (NSTEMI), 6 patients with ST-segment elevation myocardial infarction (STEMI) and 8 subjects with normal coronary artery (NCA) as control were characterized by using high-throughput sequencing. Bioinformatics analysis were performed. Results Patients with NSTEMI displayed more diverse TCR sequences than NCA controls, but they had lower percentage of top 200 TCR sequences. However, no significant differences were observed between the patients with STEMI and NCA controls, but STEMI group had lower percentage of top 200 TCR sequences. T cells from patients with AMI and NCA controls showed a differential V and J gene usage, especially, significant difference was observed in frequencies of V gene (TRBV2, TRBV29-1, TRBV30 and TRBV12-3) and J gene (TRBJ2-1) usage. Furthermore, significantly differences in average overlap was observed in groups of AMI and NCA control. The results showed that patients with AMI had distinct TCR repertoires which revealed the association between cardiovascular condition and T-cell clonotypes. Conclusions Our findings revealed the differences of TCR repertoires between patients with AMI and NCA controls, which might be potential biomarkers for evaluating risk stratification or diagnosis of acute coronary syndrome. Electronic supplementary material The online version of this article (10.1186/s12967-019-1768-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhixiong Zhong
- Center for Cardiovascular Diseases, Meizhou People's Hospital (Huangtang Hospital), Meizhou Academy of Medical Sciences, Meizhou Hospital Affiliated to Sun Yat-sen University, Meizhou, 514031, People's Republic of China.,Clinical Core Laboratory, Center for Precision Medicine, Guangdong Provincial Engineering and Technology Research Center for Clinical Molecular Diagnostics and Antibody Therapeutics, Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou People's Hospital (Huangtang Hospital), Meizhou Academy of Medical Sciences, Meizhou Hospital Affiliated to Sun Yat-sen University, Meizhou, 514031, People's Republic of China.,Guangdong Provincial Engineering and Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou People's Hospital (Huangtang Hospital), Meizhou Academy of Medical Sciences, Meizhou Hospital Affiliated to Sun Yat-sen University, Meizhou, 514031, People's Republic of China.,Meizhou Municipal Engineering and Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou, 514031, People's Republic of China.,Meizhou Municipal Engineering and Technology Research Center for Molecular Diagnostics of Major Genetic Disorders, Meizhou, 514031, People's Republic of China
| | - Heming Wu
- Center for Cardiovascular Diseases, Meizhou People's Hospital (Huangtang Hospital), Meizhou Academy of Medical Sciences, Meizhou Hospital Affiliated to Sun Yat-sen University, Meizhou, 514031, People's Republic of China.,Clinical Core Laboratory, Center for Precision Medicine, Guangdong Provincial Engineering and Technology Research Center for Clinical Molecular Diagnostics and Antibody Therapeutics, Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou People's Hospital (Huangtang Hospital), Meizhou Academy of Medical Sciences, Meizhou Hospital Affiliated to Sun Yat-sen University, Meizhou, 514031, People's Republic of China.,Guangdong Provincial Engineering and Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou People's Hospital (Huangtang Hospital), Meizhou Academy of Medical Sciences, Meizhou Hospital Affiliated to Sun Yat-sen University, Meizhou, 514031, People's Republic of China.,Meizhou Municipal Engineering and Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou, 514031, People's Republic of China.,Meizhou Municipal Engineering and Technology Research Center for Molecular Diagnostics of Major Genetic Disorders, Meizhou, 514031, People's Republic of China
| | - Qifeng Zhang
- Center for Cardiovascular Diseases, Meizhou People's Hospital (Huangtang Hospital), Meizhou Academy of Medical Sciences, Meizhou Hospital Affiliated to Sun Yat-sen University, Meizhou, 514031, People's Republic of China.,Clinical Core Laboratory, Center for Precision Medicine, Guangdong Provincial Engineering and Technology Research Center for Clinical Molecular Diagnostics and Antibody Therapeutics, Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou People's Hospital (Huangtang Hospital), Meizhou Academy of Medical Sciences, Meizhou Hospital Affiliated to Sun Yat-sen University, Meizhou, 514031, People's Republic of China.,Guangdong Provincial Engineering and Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou People's Hospital (Huangtang Hospital), Meizhou Academy of Medical Sciences, Meizhou Hospital Affiliated to Sun Yat-sen University, Meizhou, 514031, People's Republic of China.,Meizhou Municipal Engineering and Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou, 514031, People's Republic of China.,Meizhou Municipal Engineering and Technology Research Center for Molecular Diagnostics of Major Genetic Disorders, Meizhou, 514031, People's Republic of China
| | - Wei Zhong
- Center for Cardiovascular Diseases, Meizhou People's Hospital (Huangtang Hospital), Meizhou Academy of Medical Sciences, Meizhou Hospital Affiliated to Sun Yat-sen University, Meizhou, 514031, People's Republic of China.,Clinical Core Laboratory, Center for Precision Medicine, Guangdong Provincial Engineering and Technology Research Center for Clinical Molecular Diagnostics and Antibody Therapeutics, Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou People's Hospital (Huangtang Hospital), Meizhou Academy of Medical Sciences, Meizhou Hospital Affiliated to Sun Yat-sen University, Meizhou, 514031, People's Republic of China.,Guangdong Provincial Engineering and Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou People's Hospital (Huangtang Hospital), Meizhou Academy of Medical Sciences, Meizhou Hospital Affiliated to Sun Yat-sen University, Meizhou, 514031, People's Republic of China.,Meizhou Municipal Engineering and Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou, 514031, People's Republic of China.,Meizhou Municipal Engineering and Technology Research Center for Molecular Diagnostics of Major Genetic Disorders, Meizhou, 514031, People's Republic of China
| | - Pingsen Zhao
- Center for Cardiovascular Diseases, Meizhou People's Hospital (Huangtang Hospital), Meizhou Academy of Medical Sciences, Meizhou Hospital Affiliated to Sun Yat-sen University, Meizhou, 514031, People's Republic of China. .,Clinical Core Laboratory, Center for Precision Medicine, Guangdong Provincial Engineering and Technology Research Center for Clinical Molecular Diagnostics and Antibody Therapeutics, Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou People's Hospital (Huangtang Hospital), Meizhou Academy of Medical Sciences, Meizhou Hospital Affiliated to Sun Yat-sen University, Meizhou, 514031, People's Republic of China. .,Guangdong Provincial Engineering and Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou People's Hospital (Huangtang Hospital), Meizhou Academy of Medical Sciences, Meizhou Hospital Affiliated to Sun Yat-sen University, Meizhou, 514031, People's Republic of China. .,Meizhou Municipal Engineering and Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou, 514031, People's Republic of China. .,Meizhou Municipal Engineering and Technology Research Center for Molecular Diagnostics of Major Genetic Disorders, Meizhou, 514031, People's Republic of China.
| |
Collapse
|
10
|
Wysoczynski M, Khan A, Bolli R. New Paradigms in Cell Therapy: Repeated Dosing, Intravenous Delivery, Immunomodulatory Actions, and New Cell Types. Circ Res 2018; 123:138-158. [PMID: 29976684 PMCID: PMC6050028 DOI: 10.1161/circresaha.118.313251] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Perhaps the most important advance in the field of cell therapy for heart disease has been the recognition that all stem/progenitor cells (both adult and embryonic) fail to engraft in the heart to a significant extent and thus work via paracrine mechanisms. This fundamental advance has led to 4 new paradigms that are discussed in this review and that may importantly shape, or even revolutionize, the future of the field: (1) repeated cell therapy, (2) intravenous cell therapy, (3) immunomodulatory actions of cell therapy, and (4) new cell types. Because virtually all of our current knowledge of cell therapy is predicated on the effects of a single cell dose, the idea that the full therapeutic effects of a cell product require repeated doses is disruptive and has far-reaching implications. For example, inadequate dosing (single-dose protocols) may be responsible, at least in part, for the borderline or disappointing results obtained to date in clinical trials; furthermore, future studies (both preclinical and clinical) may need to incorporate repeated cell administrations. Another disruptive idea, supported by emerging preclinical and clinical evidence, is that intravenously injected cells can produce beneficial effects on the heart, presumably via release of paracrine factors in extracardiac organs or endocrine factors into the systemic circulation. Intravenous administration would obviate the need for direct delivery of cells to the heart, making cell therapy simpler, cheaper, safer, more scalable, and more broadly available, even on an outpatient basis. Although the mechanism of action of cell therapy remains elusive, there is compelling in vitro evidence that transplanted cells modulate the function of various immune cell types via release of paracrine factors, such as extracellular vesicles, although in vivo evidence is still limited. Investigation of the new paradigms reviewed herein should be a top priority because it may profoundly transform cell therapy and finally make it a reality.
Collapse
Affiliation(s)
- Marcin Wysoczynski
- From the Institute of Molecular Cardiology, University of Louisville, KY
| | - Abdur Khan
- From the Institute of Molecular Cardiology, University of Louisville, KY
| | - Roberto Bolli
- From the Institute of Molecular Cardiology, University of Louisville, KY.
| |
Collapse
|
11
|
Abstract
Endothelial progenitor cells (EPCs) promote angiogenesis and play a pivotal role in endothelial repair and re-endothelialization after vascular injury. Transient receptor potential-canonical1 (TRPC1) has been recently implied to play important roles on EPC function. Here, we studied the role of TRPC1 in regulating EPC function in vivo and in vitro. EPCs were cultured from TRPC1-knockout mice and their controls. In vitro, TRPC1 knockout reduced EPC functional activities, including migration and tube formation. Additionally, calmodulin (CaM)/endothelial nitric oxide synthase (eNOS) signaling activity were downregulated after TRPC1 knockout. Administration of CaM or eNOS inhibitor ameliorated TRPC1 knockout-reduced EPC migration and tube formation. In vivo Matrigel plug assay confirmed that TRPC1 knockout decreased formation of functional blood vessels of EPCs compared with wild-type EPCs. Taken together, these data suggest that TRPC1 is a critical regulator of angiogenesis.
Collapse
|
12
|
Abstract
One out of every two men and one out of every three women greater than the age of 40 will experience an acute myocardial infarction (AMI) at some time during their lifetime. As more patients survive their AMIs, the incidence of congestive heart failure (CHF) is increasing. 6 million people in the USA have ischemic cardiomyopathies and CHF. The search for new and innovative treatments for patients with AMI and CHF has led to investigations and use of human embryonic stem cells, cardiac stem/progenitor cells, bone marrow-derived mononuclear cells and mesenchymal stem cells for treatment of these heart conditions. This paper reviews current investigations with human embryonic, cardiac, bone marrow and mesenchymal stem cells, and also stem cell paracrine factors and exosomes.
Collapse
Affiliation(s)
- Robert J Henning
- Department of Environmental & Occupational Health, College of Public Health, University of South Florida & the James A Haley Hospital, Tampa, FL 33612-3805, USA
| |
Collapse
|
13
|
Liska MG, Dela Peña I. Granulocyte-colony stimulating factor and umbilical cord blood cell transplantation: Synergistic therapies for the treatment of traumatic brain injury. Brain Circ 2017; 3:143-151. [PMID: 30276316 PMCID: PMC6057694 DOI: 10.4103/bc.bc_19_17] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 08/31/2017] [Accepted: 09/05/2017] [Indexed: 02/06/2023] Open
Abstract
Traumatic brain injury (TBI) is now characterized as a progressive, degenerative disease and continues to stand as a prevalent cause of death and disability. The pathophysiology of TBI is complex, with a variety of secondary cell death pathways occurring which may persist chronically following the initial cerebral insult. Current therapeutic options for TBI are minimal, with surgical intervention or rehabilitation therapy existing as the only viable treatments. Considering the success of stem-cell therapies in various other neurological diseases, their use has been proposed as a potential potent therapy for patients suffering TBI. Moreover, stem cells are highly amenable to adjunctive use with other therapies, providing an opportunity to overcome the inherent limitations of using a single therapeutic agent. Our research has verified this additive potential by demonstrating the efficacy of co-delivering human umbilical cord blood (hUCB) cells with granulocyte-colony stimulating factor (G-CSF) in a murine model of TBI, providing encouraging results which support the potential of this approach to treat patients suffering from TBI. These findings justify ongoing research toward uncovering the mechanisms which underlie the functional improvements exhibited by hUCB + G-CSF combination therapy, thereby facilitating its safe and effect transition into the clinic. This paper is a review article. Referred literature in this paper has been listed in the reference section. The datasets supporting the conclusions of this article are available online by searching various databases, including PubMed. Some original points in this article come from the laboratory practice in our research center and the authors’ experiences.
Collapse
Affiliation(s)
- Michael G Liska
- Center of Excellence for Aging and Brain Repair, Tampa, FL 33612, USA
| | - Ike Dela Peña
- Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, College of Pharmacy, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
14
|
Zhou N, Wang J, Li X, Zhao Y, Sun Y, Zou C. Hetrombopag, a Thrombopoietin Receptor Agonist, Protects Cardiomyocyte Survival from Oxidative Stress Damage as an Enhancer of Stem Cells. Cardiovasc Drugs Ther 2016; 30:567-577. [PMID: 27838864 DOI: 10.1007/s10557-016-6696-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE Current human umbilical cord blood stem cell therapy faces the great challenges, because the stem cells are scarce and cannot survive for a long time. Here we describe how hetrombopag, an orally-active TPO receptor agonists, enhanced ex vivo expansion of human UCB stem cells, and protected cardiac myocytes from the damage caused by oxidative stress. METHODS Ex vivo expansion of stem cells were performed in serum-free medium supplemented with rhSCF and rhFL plus hetrombopag for 7 days. The percentage and number of stem cell subsets were determined by flow cytometry. Rat cardiac myocytes, ex vivo expanded stem cells, or cardiac myocytes plus ex vivo expanded stem cells were serum starved for 24 hours, and were then subjected to H2O2, hetrombopag or both for 12 hours at the indicated concentrations. Cell viability assays, protein microarrays and western blots were then performed in each group. RESULTS Our studies first revealed that the combination of hetrombopag and rhTPO manifested additive effect on ex vivo expansion of human UCB stem cells. Besides, hetrombopag dose-dependently enhanced the beneficial effects of ex vivo expanded human UCB MNCs in increasing the survival of injured cardiomyocytes during free oxygen radical stress. CONCLUSION These data, for the first time, uncovered a novel function of non-peptide small molecular TPO receptor agonists as enhancers of stem cells in protecting cardiac myocyte survival from oxidative stress damage, which might provide a new therapeutic avenue for the treatment of oxidative stress-related cardiovascular disease. Graphical abstract ᅟ.
Collapse
Affiliation(s)
- Nannan Zhou
- Department of Geriatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, People's Republic of China
| | - Jianchun Wang
- Department of Geriatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, People's Republic of China
| | - Xiaodong Li
- Department of Geriatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, People's Republic of China
| | - Yong Zhao
- Department of Geriatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, People's Republic of China
| | - Yuanyuan Sun
- Department of Geriatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, People's Republic of China
| | - Chengwei Zou
- Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, People's Republic of China.
| |
Collapse
|
15
|
Peña ID, Borlongan CV. Translating G-CSF as an Adjunct Therapy to Stem Cell Transplantation for Stroke. Transl Stroke Res 2016; 6:421-9. [PMID: 26482176 DOI: 10.1007/s12975-015-0430-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 10/05/2015] [Accepted: 10/08/2015] [Indexed: 01/26/2023]
Abstract
Among recently investigated stroke therapies, stem cell treatment holds great promise by virtue of their putative ability to replace lost cells, promote endogenous neurogenesis,and produce behavioral and functional improvement through their "bystander effects." Translating stem cell in the clinic, however, presents a number of technical difficulties. A strategy suggested to enhance therapeutic utility of stem cells is combination therapy, i.e., co-transplantation of stem cells or adjunct treatment with pharmacological agents and substrates,which is assumed to produce more profound therapeutic benefits by circumventing limitations of individual treatments and facilitating complementary brain repair processes. We previously demonstrated enhanced functional effects of cotreatment with granulocyte-colony stimulating factor (GCSF)and human umbilical cord blood cell (hUCB) transplantation in animal models of traumatic brain injury (TBI). Here,we suggest that the aforementioned combination therapy may also produce synergistic effects in stroke. Accordingly, G-CSF treatment may reduce expression of pro-inflammatory cytokines and enhance neurogenesis rendering a receptive microenvironment for hUCB engraftment. Adjunct treatment of GCSF with hUCB may facilitate stemness maintenance and guide neural lineage commitment of hUCB cells. Moreover, regenerative mechanisms afforded by G-CSF-mobilized endogenous stem cells, secretion of growth factors by hUCB grafts and G-CSF-recruited endothelial progenitor cells(EPCs), as well as the potential graft–host integration that may promote synaptic circuitry re-establishment could altogether produce more pronounced functional improvement in stroked rats subjected to a combination G-CSF treatment and hUCB transplantation. Nevertheless, differences in pathology and repair processes underlying TBI and stroke deserve consideration when testing the effects of combinatorial G-CSF and hUCB cell transplantation for stroke treatment. Further studies are also required to determine the safety and efficacy of this intervention in both preclinical and clinical stroke studies.
Collapse
|
16
|
Henning RJ. Therapeutic angiogenesis: angiogenic growth factors for ischemic heart disease. Future Cardiol 2016; 12:585-99. [PMID: 27420190 DOI: 10.2217/fca-2016-0006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Stem cells encode vascular endothelial growth factors (VEGFs), fibroblastic growth factors (FGFs), stem cell factor, stromal cell-derived factor, platelet growth factor and angiopoietin that can contribute to myocardial vascularization. VEGFs and FGFs are the most investigated growth factors. VEGFs regulate angiogenesis and vasculogenesis. FGFs stimulate vessel cell proliferation and differentiation and are regulators of endothelial cell migration, proliferation and survival. Clinical trials of VEGF or FGF for myocardial angiogenesis have produced disparate results. The efficacy of therapeutic angiogenesis can be improved by: (1) identifying the most optimal patients; (2) increased knowledge of angiogenic factor pharmacokinetics and proper dose; (3) prolonging contact of angiogenic factors with the myocardium; (4) increasing the efficiency of VEGF or FGF gene transduction; and (5) utilizing PET or MRI to measure myocardial perfusion and perfusion reserve.
Collapse
Affiliation(s)
- Robert J Henning
- The University of South Florida and the James A. Haley Hospital, Tampa, FL 33612 USA
| |
Collapse
|
17
|
van Zuylen VL, den Haan MC, Geutskens SB, Roelofs H, Fibbe WE, Schalij MJ, Atsma DE. Post-myocardial infarct inflammation and the potential role of cell therapy. Cardiovasc Drugs Ther 2015; 29:59-73. [PMID: 25583678 DOI: 10.1007/s10557-014-6568-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Myocardial infarction triggers reparative inflammatory processes programmed to repair damaged tissue. However, often additional injury to the myocardium occurs through the course of this inflammatory process, which ultimately can lead to heart failure. The potential beneficial effects of cell therapy in treating cardiac ischemic disease, the number one cause of death worldwide, are being studied extensively, both in clinical trials using adult stem cells as well as in fundamental research on cardiac stem cells and regenerative biology. This review summarizes the current knowledge on molecular and cellular processes implicated in post-infarction inflammation and discusses the potential beneficial role cell therapy might play in this process. Due to its immunomodulatory properties, the mesenchymal stromal cell is a candidate to reverse the disease progression of the infarcted heart towards heart failure, and therefore is emphasized in this review.
Collapse
Affiliation(s)
- Vanessa-leigh van Zuylen
- Department of Cardiology, Leiden University Medical Center, P.O. Box 9600, 2300, RC, Leiden, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
18
|
Le-Buu Pham T, Nguyen TT, Thi-Van Bui A, Pham HT, Phan NK, Thi-Thu Nguyen M, Van Pham P. Preliminary evaluation of treatment efficacy of umbilical cord blood-derived mesenchymal stem cell-differentiated cardiac progenitor cells in a myocardial injury mouse model. BIOMEDICAL RESEARCH AND THERAPY 2015. [DOI: 10.7603/s40730-015-0032-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
19
|
|
20
|
Porada CD, Atala AJ, Almeida-Porada G. The hematopoietic system in the context of regenerative medicine. Methods 2015; 99:44-61. [PMID: 26319943 DOI: 10.1016/j.ymeth.2015.08.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 07/06/2015] [Accepted: 08/23/2015] [Indexed: 12/16/2022] Open
Abstract
Hematopoietic stem cells (HSC) represent the prototype stem cell within the body. Since their discovery, HSC have been the focus of intensive research, and have proven invaluable clinically to restore hematopoiesis following inadvertent radiation exposure and following radio/chemotherapy to eliminate hematologic tumors. While they were originally discovered in the bone marrow, HSC can also be isolated from umbilical cord blood and can be "mobilized" peripheral blood, making them readily available in relatively large quantities. While their ability to repopulate the entire hematopoietic system would already guarantee HSC a valuable place in regenerative medicine, the finding that hematopoietic chimerism can induce immunological tolerance to solid organs and correct autoimmune diseases has dramatically broadened their clinical utility. The demonstration that these cells, through a variety of mechanisms, can also promote repair/regeneration of non-hematopoietic tissues as diverse as liver, heart, and brain has further increased their clinical value. The goal of this review is to provide the reader with a brief glimpse into the remarkable potential HSC possess, and to highlight their tremendous value as therapeutics in regenerative medicine.
Collapse
Affiliation(s)
- Christopher D Porada
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, 391 Technology Way, Winston-Salem, NC 27157-1083, United States.
| | - Anthony J Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, 391 Technology Way, Winston-Salem, NC 27157-1083, United States.
| | - Graça Almeida-Porada
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, 391 Technology Way, Winston-Salem, NC 27157-1083, United States.
| |
Collapse
|
21
|
De La Peña I, Sanberg PR, Acosta S, Lin SZ, Borlongan CV. G-CSF as an adjunctive therapy with umbilical cord blood cell transplantation for traumatic brain injury. Cell Transplant 2015; 24:447-57. [PMID: 25646620 DOI: 10.3727/096368915x686913] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Traumatic brain injury (TBI), a major contributor to deaths and permanent disability worldwide, has been recently described as a progressive cell death process rather than an acute event. TBI pathophysiology is complicated and can be distinguished by the initial primary injury and the subsequent secondary injury that ensues days after the trauma. Therapeutic opportunities for TBI remain very limited with patients subjected to surgery or rehabilitation therapy. The efficacy of stem cell-based interventions, as well as neuroprotective agents in other neurological disorders of which pathologies overlap with TBI, indicates their potential as alternative TBI treatments. Furthermore, their therapeutic limitations may be augmented when combination therapy is pursued instead of using a single agent. Indeed, we demonstrated remarkable combined efficacy of human umbilical cord blood (hUCB) cell therapy and granulocyte-colony-stimulating factor (G-CSF) treatment in TBI models, providing essential evidence for the translation of this approach to treat TBI. Further studies are warranted to determine the mechanisms underlying therapeutic benefits exerted by hUCB + G-CSF in order to enhance its safety and efficacy in the clinic.
Collapse
Affiliation(s)
- Ike De La Peña
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | | | | | | | | |
Collapse
|
22
|
Li B, Jung HJ, Kim SM, Kim MJ, Jahng JW, Lee JH. Human periodontal ligament stem cells repair mental nerve injury. Neural Regen Res 2014; 8:2827-37. [PMID: 25206604 PMCID: PMC4146018 DOI: 10.3969/j.issn.1673-5374.2013.30.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 08/26/2013] [Indexed: 01/31/2023] Open
Abstract
Human periodontal ligament stem cells are easily accessible and can differentiate into Schwann cells. We hypothesized that human periodontal ligament stem cells can be used as an alternative source for the autologous Schwann cells in promoting the regeneration of injured peripheral nerve. To validate this hypothesis, human periodontal ligament stem cells (1 × 106) were injected into the crush-injured left mental nerve in rats. Simultaneously, autologous Schwann cells (1 × 106) and PBS were also injected as controls. Real-time reverse transcriptase polymerase chain reaction showed that at 5 days after injection, mRNA expression of low affinity nerve growth factor receptor was significantaly increased in the left trigeminal ganglion of rats with mental nerve injury. Sensory tests, histomorphometric evaluation and retrograde labeling demonstrated that at 2 and 4 weeks after injection, sensory function was significantly improved, the numbers of retrograde labeled sensory neurons and myelinated axons were significantly increased, and human periodontal ligament stem cells and autologous Schwann cells exhibited similar therapeutic effects. These findings suggest that transplantation of human periodontal ligament stem cells show a potential value in repair of mental nerve injury.
Collapse
Affiliation(s)
- Bohan Li
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Binzhou Medical College, Binzhou, Shandong Province, China
| | - Hun-Jong Jung
- Department of Occupation and Environment, Konkuk Postgraduate Medical School, Choong-Ju, Korea
| | - Soung-Min Kim
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Seoul National University, Seoul, Korea
| | - Myung-Jin Kim
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Seoul National University, Seoul, Korea
| | - Jeong Won Jahng
- Dental Research Institute, Seoul National University, Seoul, Korea
| | - Jong-Ho Lee
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Seoul National University, Seoul, Korea ; Dental Research Institute, Seoul National University, Seoul, Korea
| |
Collapse
|
23
|
Njerve IU, Solheim S, Lunde K, Hoffmann P, Arnesen H, Seljeflot I. Fractalkine levels are elevated early after PCI-treated ST-elevation myocardial infarction; no influence of autologous bone marrow derived stem cell injection. Cytokine 2014; 69:131-5. [DOI: 10.1016/j.cyto.2014.05.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 04/10/2014] [Accepted: 05/21/2014] [Indexed: 12/01/2022]
|
24
|
Henning RJ, Sanberg P, Jimenez E. Human cord blood stem cell paracrine factors activate the survival protein kinase Akt and inhibit death protein kinases JNK and p38 in injured cardiomyocytes. Cytotherapy 2014; 16:1158-68. [DOI: 10.1016/j.jcyt.2014.01.415] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 01/06/2014] [Accepted: 01/30/2014] [Indexed: 01/08/2023]
|
25
|
Ding Z, Burghoff S, Buchheiser A, Kögler G, Schrader J. Survival, integration, and differentiation of unrestricted somatic stem cells in the heart. Cell Transplant 2014; 22:15-27. [PMID: 23594819 DOI: 10.3727/096368912x640466] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Unrestricted somatic stem cells (USSCs) derived from human umbilical cord blood represent an attractive cell source to reconstitute the damaged heart. We have analyzed the cardiomyogenic potential and investigated the fate of USSCs after transplantation into rat heart in vivo. USSCs demonstrated cardiomyogenic differentiation properties characterized by the spontaneously beating activity and the robust expression of cardiac α-actinin and troponin T (cTnT) at protein and mRNA level after cocultivation with neonatal rat cardiomyocytes. To study the fate in vivo, eGFP⁺ USSCs were injected transcoronarily into immunosuppressed rats via a catheter-based technique. Nearly 80% USSCs were retained within the myocardium without altering cardiac hemodynamics. After 7 days, 20% of the transplanted cells survived in the host myocardium and showed elongated morphology with weak expression of cardiac-specific markers, while some eGFP⁺ USSCs were found to integrate into the vascular wall. After 21 days, only a small fraction of USSCs were found in the myocardium (0.13%); however, the remaining cells clearly exhibited a sarcomeric structure similar to mature cardiomyocytes. Identical results were also obtained in nude rats. In addition, we found some cells stained positively for activated caspase 3 paralleled by the massive infiltration of CD11b⁺ cells into the myocardium. In summary, USSCs can differentiate into beating cardiomyocytes by cocultivation in vitro. After coronary transplantation in vivo, however, long-term survival of differentiated USSCs was rather low despite a high initial fraction of trapped cells.
Collapse
Affiliation(s)
- Zhaoping Ding
- Institut für Herz- und Kreislaufphysiologie, Heinrich-Heine-Universität Düsseldorf, Germany
| | | | | | | | | |
Collapse
|
26
|
Saparov A, Chen CW, Beckman SA, Wang Y, Huard J. The role of antioxidation and immunomodulation in postnatal multipotent stem cell-mediated cardiac repair. Int J Mol Sci 2013; 14:16258-79. [PMID: 23924945 PMCID: PMC3759910 DOI: 10.3390/ijms140816258] [Citation(s) in RCA: 22] [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: 05/30/2013] [Revised: 07/23/2013] [Accepted: 07/30/2013] [Indexed: 12/14/2022] Open
Abstract
Oxidative stress and inflammation play major roles in the pathogenesis of coronary heart disease including myocardial infarction (MI). The pathological progression following MI is very complex and involves a number of cell populations including cells localized within the heart, as well as cells recruited from the circulation and other tissues that participate in inflammatory and reparative processes. These cells, with their secretory factors, have pleiotropic effects that depend on the stage of inflammation and regeneration. Excessive inflammation leads to enlargement of the infarction site, pathological remodeling and eventually, heart dysfunction. Stem cell therapy represents a unique and innovative approach to ameliorate oxidative stress and inflammation caused by ischemic heart disease. Consequently, it is crucial to understand the crosstalk between stem cells and other cells involved in post-MI cardiac tissue repair, especially immune cells, in order to harness the beneficial effects of the immune response following MI and further improve stem cell-mediated cardiac regeneration. This paper reviews the recent findings on the role of antioxidation and immunomodulation in postnatal multipotent stem cell-mediated cardiac repair following ischemic heart disease, particularly acute MI and focuses specifically on mesenchymal, muscle and blood-vessel-derived stem cells due to their antioxidant and immunomodulatory properties.
Collapse
Affiliation(s)
- Arman Saparov
- Nazarbayev University Research and Innovation System, Nazarbayev University, Astana 010000, Kazakhstan
- Department of Orthopedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA; E-Mails: (C.-W.C.); (S.A.B.)
- Authors to whom correspondence should be addressed; E-Mails: (A.S.); (J.H.); Tel.: +7-717-270-6140 (A.S.); +1-412-648-2798 (J.H.); Fax: +7-717-270-6054 (A.S.); +1-412-648-4066 (J.H.)
| | - Chien-Wen Chen
- Department of Orthopedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA; E-Mails: (C.-W.C.); (S.A.B.)
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA; E-Mail:
- Stem Cell Research Center, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Sarah A. Beckman
- Department of Orthopedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA; E-Mails: (C.-W.C.); (S.A.B.)
- Stem Cell Research Center, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA; E-Mail:
| | - Yadong Wang
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA; E-Mail:
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Johnny Huard
- Department of Orthopedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA; E-Mails: (C.-W.C.); (S.A.B.)
- Stem Cell Research Center, University of Pittsburgh, Pittsburgh, PA 15219, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Authors to whom correspondence should be addressed; E-Mails: (A.S.); (J.H.); Tel.: +7-717-270-6140 (A.S.); +1-412-648-2798 (J.H.); Fax: +7-717-270-6054 (A.S.); +1-412-648-4066 (J.H.)
| |
Collapse
|
27
|
Oh YB, Ahn M, Lee SM, Koh HW, Lee SH, Kim SH, Park BH. Inhibition of Janus activated kinase-3 protects against myocardial ischemia and reperfusion injury in mice. Exp Mol Med 2013; 45:e23. [PMID: 23680658 PMCID: PMC3674406 DOI: 10.1038/emm.2013.43] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Recent studies have documented that Janus-activated kinase (JAK)–signal transducer and activator of transcription (STAT) pathway can modulate the apoptotic program in a myocardial ischemia/reperfusion (I/R) model. To date, however, limited studies have examined the role of JAK3 on myocardial I/R injury. Here, we investigated the potential effects of pharmacological JAK3 inhibition with JANEX-1 in a myocardial I/R model. Mice were subjected to 45 min of ischemia followed by varying periods of reperfusion. JANEX-1 was injected 1 h before ischemia by intraperitoneal injection. Treatment with JANEX-1 significantly decreased plasma creatine kinase and lactate dehydrogenase activities, reduced infarct size, reversed I/R-induced functional deterioration of the myocardium and reduced myocardial apoptosis. Histological analysis revealed an increase in neutrophil and macrophage infiltration within the infarcted area, which was markedly reduced by JANEX-1 treatment. In parallel, in in vitro studies where neutrophils and macrophages were treated with JANEX-1 or isolated from JAK3 knockout mice, there was an impairment in the migration potential toward interleukin-8 (IL-8) and monocyte chemoattractant protein-1 (MCP-1), respectively. Of note, however, JANEX-1 did not affect the expression of IL-8 and MCP-1 in the myocardium. The pharmacological inhibition of JAK3 might represent an effective approach to reduce inflammation-mediated apoptotic damage initiated by myocardial I/R injury.
Collapse
Affiliation(s)
- Young-Bin Oh
- Department of Physiology, Chonbuk National University Medical School, Jeonju, Jeonbuk, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
28
|
Yuan C, Yu G, Yang T, Li W, Ai Q, Deng L. Enhanced therapeutic effects on acute myocardial infarction with multiple intravenous transplantation of human cord blood mononuclear cells. Int J Cardiol 2013; 168:2767-73. [PMID: 23651828 DOI: 10.1016/j.ijcard.2013.03.131] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 03/19/2013] [Accepted: 03/26/2013] [Indexed: 12/20/2022]
Abstract
BACKGROUND Cell transplantation of human cord blood mononuclear progenitor cells (HCBMCs) is a new treatment that could restore cardiac functions after acute myocardial infarction (AMI). We hypothesize that multiple doses of HCBMCs might increase homing of transplanted cells to infarcted region, and improve outcome of AMI via inhibition of ischemic-induced inflammatory responses. METHODS AND RESULTS HCBMCs were injected intravenously to rabbits that previously underwent ligation of left anterior coronary artery (LAD). Single dose of HCBMCs was intravenously delivered on the 7th day or multiple doses of HCBMCs were delivered on the 7th, 9th, 11th and 13th day after AMI. Homing of HCBMCs was determined by 5-bromodeoxyuridine (BrdU) labeling. The amount of grafted cells homed and retained in the infarcted area was significantly increased in the rabbits that received multiple doses. More viable cardiomyocytes and less collagen deposition were observed also in the group with more injections. Cardiac functions were assessed by echocardiography and hemodynamics. Multiple doses of HCBMCs showed significant benefits in preservation of cardiac functions. In infarcted myocardium, multiple transplantations of HCBMCs showed a significant increase in the myocardial level of anti-inflammatory IL-10 and a marked decrease in the level of the pro-inflammatory IL-6. CONCLUSIONS Multiple injections of HCBMCs markedly increased the amount of grafted cells, beneficially improving cardiac functions after AMI. The findings suggest that multiple doses of HCBMCs might be a novel strategy in the cell therapy for AMI.
Collapse
Affiliation(s)
- Chunju Yuan
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha 410008, PR China
| | | | | | | | | | | |
Collapse
|
29
|
Jin H, Sanberg PR, Henning RJ. Human umbilical cord blood mononuclear cell-conditioned media inhibits hypoxic-induced apoptosis in human coronary artery endothelial cells and cardiac myocytes by activation of the survival protein Akt. Cell Transplant 2013; 22:1637-50. [PMID: 23336598 DOI: 10.3727/096368912x661427] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
We have previously demonstrated in acute myocardial infarctions that human umbilical cord blood mononuclear cells (HUCBCs), which contain hematopoietic, endothelial, and mesenchymal stem cells, reduce acute myocardial infarction size by ≥50% and preserve LV contractility. We hypothesize that the beneficial effects of HUCBCs are due to secretion of biologically active factors that activate in cardiac endothelial cells and myocytes the cell survival protein Akt. We determined by protein microarrays the growth factors and anti-inflammatory cytokines secreted by HUCBCs into culture media during 12 h of hypoxia (1% O2). We then determined by Western blots the effects of cell-free media from hypoxic-conditioned HUCBCs (HUCM) on activation of the cell survival protein Akt in human coronary artery endothelial cells and cardiac myocytes in culture during 24 h of 1% O2. We also determined in separate experiments endothelial cell and myocyte apoptosis by caspase-3 and Annexin V. In the present experiments, HUCBCs secreted multiple growth factors, anti-inflammatory cytokines, and inhibitors of metalloproteinase during normoxia and hypoxia. Human cord blood cells increased the concentration in culture media of angiopoietin, hepatocyte growth factor, interleukin-4, insulin-like growth factor, placental growth factor, vascular endothelial cell growth factor, angiogenin, and stem cell factor by 100 to >10,000% during 12 h of 1% O2 (p<0.001). HUCM, which contained these biological factors, significantly increased Akt phosphorylation/activation in coronary artery endothelial cells and cardiac myocytes subjected to 24 h of 1% O2 by more than 60% (p<0.05) and increased the antiapoptotic protein Bcl-2 expression by 34-50% in comparison with endothelial cells and myocytes treated without HUCM in 1% O2(p<0.05). HUCM also significantly decreased caspase-3 activity and decreased hypoxic endothelial cell and cardiac myocyte apoptosis by more than 40% in comparison with cells cultured without HUCM (p<0.05). Inhibition of Akt activation in endothelial cells and myocytes by the sensitive and specific antagonist API-1 during 24 h of hypoxia nearly completely prevented the beneficial effects of HUCM on inhibiting caspase-3 activity and apoptosis. We conclude that HUCBCs secrete biologically active factors during hypoxia that activate survival proteins in endothelial cells and myocytes that significantly limit apoptosis.
Collapse
Affiliation(s)
- Hua Jin
- Center for Cardiovascular Research and James A. Haley VA Medical Center, Tampa, FL, USA
| | | | | |
Collapse
|
30
|
Rasmussen JG, Frøbert O, Holst-Hansen C, Kastrup J, Baandrup U, Zachar V, Fink T, Simonsen U. Comparison of human adipose-derived stem cells and bone marrow-derived stem cells in a myocardial infarction model. Cell Transplant 2012; 23:195-206. [PMID: 23211469 DOI: 10.3727/096368912x659871] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Treatment of myocardial infarction (MI) with bone marrow-derived mesenchymal stem cells and recently also adipose-derived stem cells has shown promising results. In contrast to clinical trials and their use of autologous bone marrow-derived cells from the ischemic patient, the animal MI models are often using young donors and young, often immune-compromised, recipient animals. Our objective was to compare bone marrow-derived mesenchymal stem cells with adipose-derived stem cells from an elderly ischemic patient in the treatment of MI using a fully grown non-immune-compromised rat model. Mesenchymal stem cells were isolated from adipose tissue and bone marrow and compared with respect to surface markers and proliferative capability. To compare the regenerative potential of the two stem cell populations, male Sprague-Dawley rats were randomized to receive intramyocardial injections of adipose-derived stem cells, bone marrow-derived mesenchymal stem cells, or phosphate-buffered saline 1 week following induction of MI. After 4 weeks, left ventricular ejection fraction (LVEF) was improved in the adipose-derived stem cell group, and scar wall thickness was greater compared with the saline group. Adipose-derived as well as bone marrow-derived mesenchymal stem cells prevented left ventricular end diastolic dilation. Neither of the cell groups displayed increased angiogenesis in the myocardium compared with the saline group. Adipose-derived stem cells from a human ischemic patient preserved cardiac function following MI, whereas this could not be demonstrated for bone marrow-derived mesenchymal stem cells, with only adipose-derived stem cells leading to an improvement in LVEF. Neither of the stem cell types induced myocardial angiogenesis, raising the question whether donor age and health have an effect on the efficacy of stem cells used in the treatment of MI.
Collapse
Affiliation(s)
- Jeppe Grøndahl Rasmussen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Henning RJ, Dennis S, Sawmiller D, Hunter L, Sanberg P, Miller L. Human umbilical cord blood mononuclear cells activate the survival protein Akt in cardiac myocytes and endothelial cells that limits apoptosis and necrosis during hypoxia. Transl Res 2012; 159:497-506. [PMID: 22633101 DOI: 10.1016/j.trsl.2012.02.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Revised: 02/04/2012] [Accepted: 02/06/2012] [Indexed: 12/29/2022]
Abstract
We have previously reported that human umbilical cord blood mononuclear cells (HUCBC), which contain hematopoietic, mesenchymal, and endothelial stem cells, can significantly reduce acute myocardial infarction size. To determine the mechanism whereby HUCBC increase myocyte and vascular endothelial cell survival, we treated cardiac myocytes and coronary artery endothelial cells in separate experiments with HUCBC plus culture media or culture media alone and subjected the cells to 24 h of hypoxia or normoxia. We then determined in myocytes and endothelial cells activation of the cell survival protein Akt by Western blots. We also determined in these cells apoptosis by annexin V staining and necrosis by propidium iodide staining. Thereafter, we inhibited with API, a specific and sensitive Akt inhibitor, Akt activation in myocytes and endothelial cells cultured with HUCBC during hypoxia and determined cell apoptosis and necrosis. In cells cultured without HUCBC, hypoxia only slightly activated Akt. Moreover, hypoxia increased myocyte apoptosis by ≥ 226% and necrosis by 58% in comparison with myocytes in normoxia. Hypoxic treatment of endothelial cells without HUCBC increased apoptosis by 94% and necrosis by 59%. In contrast, hypoxia did not significantly affect HUCBC. Moreover, in myocyte + HUCBC cultures in hypoxia, HUCBC induced a ≥ 135% increase in myocyte phospho-Akt. Akt activation decreased myocyte apoptosis by 76% and necrosis by 35%. In endothelial cells, HUCBC increased phospho-Akt by 116%. HUCBC also decreased endothelial cell apoptosis by 58% and necrosis by 42%. Inhibition of Akt with API in myocytes and endothelial cells cultured with HUCBC during hypoxia nearly totally prevented the HUCBC-induced decrease in apoptosis and necrosis. We conclude that HUCBC can significantly decrease hypoxia-induced myocyte and endothelial cell apoptosis and necrosis by activating Akt in these cells and in this manner HUCBC can limit myocardial ischemia and injury.
Collapse
Affiliation(s)
- Robert J Henning
- James A. Haley VA Medical Center and the University of South Florida College of Medicine, Tampa, FL, USA.
| | | | | | | | | | | |
Collapse
|
32
|
Fernandes NM, Taylor GP, Manlhiot C, McCrindle BW, Ho M, Miner SES, Atkinson A, Jaeggi ET, Nield LE. The myocardium of fetuses with endocardial fibroelastosis contains fewer B and T lymphocytes than normal control myocardium. Pediatr Cardiol 2011; 32:1088-95. [PMID: 21484220 DOI: 10.1007/s00246-011-9980-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2010] [Accepted: 03/22/2011] [Indexed: 10/15/2022]
Abstract
The observation that endocardial fibroelastosis (EFE) can result from an immune response to maternal autoantibody deposition in the fetal myocardium raises the possibility that the fetal immune system may contribute to the pathogenesis of idiopathic EFE and dilated cardiomyopathy (DCM). This study sought to characterize myocardial immune cell presence in fetuses and neonates with idiopathic EFE + DCM, in those with EFE + structural heart disease, and in normal control subjects. Paraffin tissue sections from fetuses identified from the pathology database were stained for B cell, T cell, macrophage, and general hematopoietic cell surface markers. Of the 14 fetuses included in the study, 5 had EFE + DCM, 4 had EFE + structural heart disease, and 5 were normal control fetuses. The EFE + DCM group had fewer B cells than the control group (0.15 vs. 0.44 cells/mm(2); p = 0.005). The EFE + heart disease group had both fewer B cells (0.18 vs. 0.44 cells/mm(2); p = 0.08) and T cells (0.29 vs. 0.80 cells/mm(2); p = 0.04) than the control group. The CD4/CD8 ratio was similar in the EFE + DCM and EFE + heart disease groups (1.0 vs. 0.9; p = 0.17) but higher in the EFE + DCM group than in the control group (0.9 vs. 0.3; p = 0.03). The myocardium of fetuses with EFE contains fewer B and T lymphocytes than normal control fetuses.
Collapse
Affiliation(s)
- Nisha M Fernandes
- The Hospital for Sick Children, Labatt Family Heart Centre, University of Toronto, Toronto, ON, M5G 1X8, Canada.
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Hu Y, Zhang H, Lu Y, Bai H, Xu Y, Zhu X, Zhou R, Ben J, Xu Y, Chen Q. Class A scavenger receptor attenuates myocardial infarction-induced cardiomyocyte necrosis through suppressing M1 macrophage subset polarization. Basic Res Cardiol 2011; 106:1311-28. [PMID: 21769674 DOI: 10.1007/s00395-011-0204-x] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Revised: 06/27/2011] [Accepted: 07/07/2011] [Indexed: 12/30/2022]
Abstract
Classically (M1) and alternatively (M2) activated macrophage subsets play differential roles in left ventricular remodeling after myocardial infarction (MI). The precise mechanism underlying the regulation of M1/M2 polarization during MI is unknown. We hypothesized that class A scavenger receptor (SR-A), a key modulator of inflammation, may steer macrophage polarization, which in turn influences cardiomyocytes necrosis after MI. MI was induced in wild type (WT) and SR-A deficient (SR-A(-/-)) mice by left anterior descending coronary artery ligation. Cardiac function deterioration, ventricular dilatation and fibrosis were all exacerbated in SR-A(-/-) mice following MI compared to WT littermates. Meanwhile, enhanced M1 macrophage polarization was observed in SR-A(-/-) mice, along with increased production of M1 signature cytokines including interleukin-1β (IL-1β), IL-6, and tumor necrosis factor-α (TNF-α) as demonstrated by immunohistochemistry, flow cytometry, quantitative real-time PCR, and ELISA assays. Moreover, activation of the activated apoptosis signal regulating kinase 1 (ASK1)/p38 mitogen-activated protein kinase (MAPK)/nuclear factor-κB (NF-κB) signaling pathway was markedly elevated in SR-A(-/-) animals post-MI. Most importantly, transplantation using bone marrow from SR-A(+/+) mice partially restored M1 macrophages and significantly augmented left ventricular fractional shortening in SR-A(-/-) mice. SR-A attenuated MI-induced cardiac remodeling by suppressing macrophage polarization toward a skewed M1 phenotype, reducing secretion of IL-1β, IL-6, and TNF-α, and dampening the ASK1/p38/NF-κB signaling pathway. Therefore, SR-A may exert a protective effect against MI, which may represent a new interventional target for treatment of post-infarct remodeling and subsequent heart failure.
Collapse
Affiliation(s)
- Yulong Hu
- Atherosclerosis Research Center, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing 210029, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW Stem cell therapy for cardiac disease may be facing two major problems nowadays: although vasculogenesis likely occurs as a result of cell therapy, its clinical applications are limited and significant, integrated cardiomyogenesis has not demonstratively been shown to occur, even in the experimental setting, with any other source than embryonic or other pluripotent stem cells. RECENT FINDINGS In this article, we highlight several factors that will need to be optimized if we are to achieve clinically effective cardiomyogenesis, such as the identification of optimal stem cell populations, and the ideal time and methods for cell transplantation. So far, educated attempts at achieving transplanted stem cell-induced myogenesis have largely failed outside of the embryonic stem cell realm, and we present the rationale for also considering acellular techniques, which may enhance the potential of endogenous progenitor populations. SUMMARY In today's cardiovascular field, once a cardiomyocyte is lost it is lost for good, without any form of direct therapeutic option. For these reasons, cell therapy justifies our continued attention and efforts, and may constitute the holy grail of cardiovascular therapeutics.
Collapse
|
35
|
Abstract
Myocardial infarction is the leading cause of death among people in industrialized nations. Although the heart has some ability to regenerate after infarction, myocardial restoration is inadequate. Consequently, investigators are currently exploring the use of human embryonic stem cells (hESCs), skeletal myoblasts and adult bone marrow stem cells to limit infarct size. hESCs are pluripotent cells that can regenerate myocardium in infarcted hearts, attenuate heart remodeling and contribute to left ventricle (LV) systolic force development. Since hESCs can form heart teratomas, investigators are differentiating hESCs toward cardiac progenitor cells prior to transplantation into hearts. Large quantities of hESCs cardiac progenitor cells, however, must be generated, immune rejection must be prevented and grafts must survive over the long term to significantly improve myocardial performance. Transplanted autologous skeletal myoblasts can survive in infarcted myocardium in small numbers, proliferate, differentiate into skeletal myofibers and increase the LV ejection fraction. These cells, however, do not form electromechanical connections with host cardiomyocytes. Consequently, electrical re-entry can occur and cause cardiac arrhythmias. Autologous bone marrow mononuclear cells contain hematopoietic and mesenchymal stem cells. In several meta-analyses, patients with coronary disease who received autologous bone marrow cells by intracoronary injection show significant 3.7% (range: 1.9-5.4%) increases in LV ejection fraction, decreases in LV end-systolic volume of -4.8 ml (range: -1.4 to -8.2 ml) and reductions in infarct size of 5.5% (-1.9 to -9.1%), without experiencing arrhythmias. Bone marrow cells appear to release biologically active factors that limit myocardial damage. Unfortunately, bone marrow cells from patients with chronic diseases propagate poorly and can die prematurely. Substantial challenges must be addressed and resolved to advance the use of stem cells in cardiac repair including identifying the optimal stem cell(s) that permit transplantation without requirements for host immune suppression; timing of stem cell transplantation that maximizes chemoattraction of stem cells to infarcts; and determining the optimal technique for injecting stem cells for cardiac repair. Techniques must be developed to enhance survival and propagation of stem cells in the myocardium. These studies will require close cooperation and interaction of scientists and clinicians. Cell-based cardiac repair in the 21st century will offer new hope for millions of patients worldwide with myocardial infarctions who, otherwise, would suffer from the relentless progression of heart disease to heart failure and death.
Collapse
Affiliation(s)
- Robert J Henning
- James A. Haley VA Hospital/University of South Florida College of Medicine, Tampa, FL 33612, USA.
| |
Collapse
|
36
|
Singer NG, Caplan AI. Mesenchymal stem cells: mechanisms of inflammation. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2011; 6:457-78. [PMID: 21073342 DOI: 10.1146/annurev-pathol-011110-130230] [Citation(s) in RCA: 632] [Impact Index Per Article: 45.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
In adults, human mesenchymal stem cells (hMSCs) are found in vivo at low frequency and are defined by their capacity to differentiate into bone, cartilage, and adipose tissue, depending on the stimuli and culture conditions under which they are expanded. Although MSCs were initially hypothesized to be the panacea for regenerating tissues, MSCs appear to be more important in therapeutics to regulate the immune response invoked in settings such as tissue injury, transplantation, and autoimmunity. MSCs have been used therapeutically in clinical trials and subsequently in practice to treat graft-versus-host disease following bone marrow transplantation. Reports of successful immune modulation suggest efficacy in a wide range of autoimmune conditions, such as demyelinating neurological disease (multiple sclerosis), systemic lupus erythematosus, and Crohn's disease, among others. This review provides background information about hMSCs and also describes their putative mechanisms of action in inflammation. We provide a summary of ongoing clinical trials to allow (a) full comprehension of the range of diseases in which hMSC therapy may be beneficial and (b) identification of gaps in our knowledge about the mechanisms of action of therapeutic MSCs in disease.
Collapse
Affiliation(s)
- Nora G Singer
- MetroHealth Medical Center, Case Western Reserve University, Cleveland, Ohio 44109, USA.
| | | |
Collapse
|
37
|
Geng YJ, Willerson JT. Repairing the Infarcted Heart with Stem Cells from Umbilical Cord Stroma. Cardiology 2011; 120:100-2. [DOI: 10.1159/000334241] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Accepted: 10/08/2011] [Indexed: 12/24/2022]
|
38
|
Jourdan-LeSaux C, Zhang J, Lindsey ML. Extracellular matrix roles during cardiac repair. Life Sci 2010; 87:391-400. [PMID: 20670633 PMCID: PMC2946433 DOI: 10.1016/j.lfs.2010.07.010] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2010] [Revised: 06/30/2010] [Accepted: 07/16/2010] [Indexed: 02/06/2023]
Abstract
The cardiac extracellular matrix (ECM) provides a platform for cells to maintain structure and function, which in turn maintains tissue function. In response to injury, the ECM undergoes remodeling that involves synthesis, incorporation, and degradation of matrix proteins, with the net outcome determined by the balance of these processes. The major goals of this review are a) to serve as an initial resource for students and investigators new to the cardiac ECM remodeling field, and b) to highlight a few of the key exciting avenues and methodologies that have recently been explored. While we focus on cardiac injury and responses of the left ventricle (LV), the mechanisms reviewed here have pathways in common with other wound healing models.
Collapse
Affiliation(s)
- Claude Jourdan-LeSaux
- Division of Cardiology, Department of Medicine, The University of Texas Health Science Center at San Antonio
| | - Jianhua Zhang
- Division of Cardiology, Department of Medicine, The University of Texas Health Science Center at San Antonio
| | - Merry L. Lindsey
- Division of Cardiology, Department of Medicine, The University of Texas Health Science Center at San Antonio
| |
Collapse
|
39
|
Anzalone R, Iacono ML, Corrao S, Magno F, Loria T, Cappello F, Zummo G, Farina F, La Rocca G. New Emerging Potentials for Human Wharton’s Jelly Mesenchymal Stem Cells: Immunological Features and Hepatocyte-Like Differentiative Capacity. Stem Cells Dev 2010; 19:423-38. [DOI: 10.1089/scd.2009.0299] [Citation(s) in RCA: 142] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Rita Anzalone
- Sezione di Anatomia Umana, Dipartimento di Medicina Sperimentale, Università degli Studi di Palermo, Italy
| | - Melania Lo Iacono
- Sezione di Anatomia Umana, Dipartimento di Medicina Sperimentale, Università degli Studi di Palermo, Italy
| | - Simona Corrao
- Sezione di Anatomia Umana, Dipartimento di Medicina Sperimentale, Università degli Studi di Palermo, Italy
| | - Francesca Magno
- Sezione di Anatomia Umana, Dipartimento di Medicina Sperimentale, Università degli Studi di Palermo, Italy
| | - Tiziana Loria
- Sezione di Anatomia Umana, Dipartimento di Medicina Sperimentale, Università degli Studi di Palermo, Italy
| | - Francesco Cappello
- Sezione di Anatomia Umana, Dipartimento di Medicina Sperimentale, Università degli Studi di Palermo, Italy
| | - Giovanni Zummo
- Sezione di Anatomia Umana, Dipartimento di Medicina Sperimentale, Università degli Studi di Palermo, Italy
| | - Felicia Farina
- Sezione di Anatomia Umana, Dipartimento di Medicina Sperimentale, Università degli Studi di Palermo, Italy
| | - Giampiero La Rocca
- Sezione di Anatomia Umana, Dipartimento di Medicina Sperimentale, Università degli Studi di Palermo, Italy
| |
Collapse
|