1
|
Lin D, Yang H, Liang X, Yang M, Zhao Y. The involvement of mitochondria in erythrocyte pathology and diseases: from mechanisms to therapeutic strategies. Clin Exp Med 2025; 25:144. [PMID: 40343592 PMCID: PMC12064630 DOI: 10.1007/s10238-024-01555-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 12/31/2024] [Indexed: 05/11/2025]
Abstract
Erythrocytes, as the predominant cellular components within the bloodstream, are crucial for the maintenance of physiological health. Mitochondria, known as cellular powerhouses and metabolic regulators, play a critical role in the maturation of the erythroid lineage. The absence of mitochondria in red blood cells upon completing their maturation process is a defining characteristic of their development. Dysregulation of mitochondrial metabolism has been associated with the onset and progression of various diseases. Mitochondrial metabolic disorders, along with the involvement of mitochondria in the induction of oxidative stress and the activation of immune responses, significantly contribute to the pathogenesis of diverse hematologic disorders, particularly in sickle cell disease. This review offers a comprehensive overview of the role of mitochondria in disorders related to abnormal erythropoiesis, immune responses, and hemolysis, as well as evaluating potential therapeutic strategies that target mitochondria. Ultimately, we emphasize the necessity for future research to elucidate the involvement of mitochondria in red blood cell disorders, which may inform the development of novel diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Dier Lin
- Department of Anesthesiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, People's Republic of China
| | - Hongjun Yang
- Department of Transfusion, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, People's Republic of China
| | - Xiaoxue Liang
- Department of Medical Laboratory, Chengdu Qingbaijiang District People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Mengjiao Yang
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, People's Republic of China
- Graduate School of Comprehensive Human Science, University of Tsukuba, Tsukuba, Japan
| | - Yangyang Zhao
- Department of Transfusion, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, People's Republic of China.
| |
Collapse
|
2
|
Zhang S, Su S. Mitochondrial antigens: their presentation and related diseases. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1509-1511. [PMID: 39971878 DOI: 10.1007/s11427-024-2781-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 11/20/2024] [Indexed: 02/21/2025]
Affiliation(s)
- Shiyang Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Shicheng Su
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
- Department of Infectious Diseases, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China.
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
- Biotherapy Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
| |
Collapse
|
3
|
Deuse T, Schrepfer S. Progress and challenges in developing allogeneic cell therapies. Cell Stem Cell 2025; 32:513-528. [PMID: 40185072 DOI: 10.1016/j.stem.2025.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/28/2025] [Accepted: 03/05/2025] [Indexed: 04/07/2025]
Abstract
The new era of cell therapeutics has started with autologous products to avoid immune rejection. However, therapeutics derived from allogeneic cells could be scaled and made available for a much larger patient population if immune rejection could reliably be overcome. In this review, we outline gene engineering concepts aimed at generating immune-evasive cells. First, we summarize the current state of allogeneic immune cell therapies, and second, we compile the still limited data for allogeneic cell replacement therapies. We emphasize the advances in this fast-developing field and provide an optimistic outlook for future allogeneic cell therapies.
Collapse
Affiliation(s)
- Tobias Deuse
- Department of Surgery, Division of Cardiothoracic Surgery, Transplant and Stem Cell Immunobiology (TSI)-Lab, University of California, San Francisco, San Francisco, CA, USA
| | - Sonja Schrepfer
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
4
|
Shou X, Chen C, Ying H, Liu Z, Zeng L, Li Q, Lei L, Mao B, Zhang W, Cui S, Shi L. Biomimetic MOF Nanocarrier-Mediated Synergistic Delivery of Mitochondria and Anti-Inflammatory miRNA to Alleviate Acute Lung Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2416594. [PMID: 39999302 PMCID: PMC12021094 DOI: 10.1002/advs.202416594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/11/2025] [Indexed: 02/27/2025]
Abstract
Acute lung injury (ALI) is a clinically critical disease characterized by overwhelming inflammatory response and significant tissue damage with no specific treatment available currently. As a key player in the pathogenesis of ALI, macrophages are aberrantly activated and polarize toward the pro-inflammatory phenotypes, leading to overzealous inflammation and lung injury. Mitochondria is recognized as a crucial signaling hub governing macrophage function and polarization, deregulation of which is causatively related with defective metabolism of macrophages, deregulated inflammation, and hence ALI. Herein, an inflammation-responsive, biomimetic metal-organic framework (MOF) nanoplatform, termed a127/mito@ZIF@Ma is developed, which is sophistically designed for synergistic delivery of macrophage-derived mitochondria and anti-inflammatory miRNA-127 antagonist to resume pulmonary macrophages homeostasis and alleviate lung inflammation and injury. Notably, macrophage membrane encapsulation conferred the biomimetic MOF with enhanced transport efficacy both in vitro and in vivo. Therefore, the administration of the nanoparticles accordingly conferred a profound protection of mice against lung inflammation and injury induced by either bacterial or viral infection with unnoticeable tissue toxicity. The study thus devises a novel MOF-based nanosystem that integrates mitochondria transplantation and miRNA therapeutics, which may open a new avenue for treating ALI and relevant critical diseases.
Collapse
Affiliation(s)
- Xin Shou
- Key lab of Artificial Organs and Computational MedicineInstitute of Translational MedicineZhejiang Shuren UniversityHangzhouZhejiang310015China
| | - Changjiang Chen
- Department of ImmunologyNanjing University of Chinese MedicineNanjingJiangsu210023China
| | - Hangjie Ying
- Department of Experiment CenterZhejiang Cancer HospitalHangzhou Institute of Medicine (HIM)Chinese Academy of SciencesHangzhouZhejiang310022China
| | - Zhiyun Liu
- Key lab of Artificial Organs and Computational MedicineInstitute of Translational MedicineZhejiang Shuren UniversityHangzhouZhejiang310015China
| | - Lingyao Zeng
- Key lab of Artificial Organs and Computational MedicineInstitute of Translational MedicineZhejiang Shuren UniversityHangzhouZhejiang310015China
| | - Qiujie Li
- Key lab of Artificial Organs and Computational MedicineInstitute of Translational MedicineZhejiang Shuren UniversityHangzhouZhejiang310015China
| | - Lanjie Lei
- Key lab of Artificial Organs and Computational MedicineInstitute of Translational MedicineZhejiang Shuren UniversityHangzhouZhejiang310015China
| | - Bingyong Mao
- State Key Laboratory of Food Science and ResourcesJiangnan UniversityWuxiJiangsu214122China
| | - Wei Zhang
- Department of ImmunologyNanjing University of Chinese MedicineNanjingJiangsu210023China
| | - Shumao Cui
- State Key Laboratory of Food Science and ResourcesJiangnan UniversityWuxiJiangsu214122China
| | - Liyun Shi
- Key lab of Artificial Organs and Computational MedicineInstitute of Translational MedicineZhejiang Shuren UniversityHangzhouZhejiang310015China
| |
Collapse
|
5
|
Thetchinamoorthy K, Jarczak J, Kieszek P, Wierzbicka D, Ratajczak J, Kucia M, Ratajczak MZ. Very small embryonic-like stem cells (VSELs) on the way for potential applications in regenerative medicine. Front Bioeng Biotechnol 2025; 13:1564964. [PMID: 40124247 PMCID: PMC11926153 DOI: 10.3389/fbioe.2025.1564964] [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: 01/22/2025] [Accepted: 02/17/2025] [Indexed: 03/25/2025] Open
Abstract
Evidence has accumulated that adult tissues contain a population of early development stem cells capable of differentiating across germ layers into various types of cells. Our group purified these rare cells, naming them very small embryonic-like stem cells (VSELs). With their broad differentiation potential, VSELs have emerged as a new candidate population for clinical applications. This advancement is now possible due to our recent development of a model for ex vivo expansion of these rare cells. Importantly, no evidence suggests that VSELs, isolated from adult tissues, can form teratomas. In this review paper, we update current research on these cells reported in our laboratory as well as in those of several independent investigators.
Collapse
Affiliation(s)
| | - Justyna Jarczak
- Laboratory of Regenerative Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Patrycja Kieszek
- Laboratory of Regenerative Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Diana Wierzbicka
- Laboratory of Regenerative Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Janina Ratajczak
- Stem Cell Institute at Graham Brown Cancer Center, University of Louisville, Louisville, CO, United States
| | - Magdalena Kucia
- Laboratory of Regenerative Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Mariusz Z. Ratajczak
- Laboratory of Regenerative Medicine, Medical University of Warsaw, Warsaw, Poland
- Stem Cell Institute at Graham Brown Cancer Center, University of Louisville, Louisville, CO, United States
| |
Collapse
|
6
|
Hadzimustafic N, D’Elia A, Shamoun V, Haykal S. Human-Induced Pluripotent Stem Cells in Plastic and Reconstructive Surgery. Int J Mol Sci 2024; 25:1863. [PMID: 38339142 PMCID: PMC10855589 DOI: 10.3390/ijms25031863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/25/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024] Open
Abstract
A hallmark of plastic and reconstructive surgery is restoring form and function. Historically, tissue procured from healthy portions of a patient's body has been used to fill defects, but this is limited by tissue availability. Human-induced pluripotent stem cells (hiPSCs) are stem cells derived from the de-differentiation of mature somatic cells. hiPSCs are of particular interest in plastic surgery as they have the capacity to be re-differentiated into more mature cells, and cultured to grow tissues. This review aims to evaluate the applications of hiPSCs in the plastic surgery context, with a focus on recent advances and limitations. The use of hiPSCs and non-human iPSCs has been researched in the context of skin, nerve, vasculature, skeletal muscle, cartilage, and bone regeneration. hiPSCs offer a future for regenerated autologous skin grafts, flaps comprised of various tissue types, and whole functional units such as the face and limbs. Also, they can be used to model diseases affecting tissues of interest in plastic surgery, such as skin cancers, epidermolysis bullosa, and scleroderma. Tumorigenicity, immunogenicity and pragmatism still pose significant limitations. Further research is required to identify appropriate somatic origin and induction techniques to harness the epigenetic memory of hiPSCs or identify methods to manipulate epigenetic memory.
Collapse
Affiliation(s)
- Nina Hadzimustafic
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; (N.H.); (A.D.); (V.S.)
| | - Andrew D’Elia
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; (N.H.); (A.D.); (V.S.)
| | - Valentina Shamoun
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; (N.H.); (A.D.); (V.S.)
| | - Siba Haykal
- Department of Plastic and Reconstructive Surgery, University Health Network, Toronto, ON M5G 2C4, Canada
| |
Collapse
|
7
|
Gravina A, Tediashvili G, Zheng Y, Iwabuchi KA, Peyrot SM, Roodsari SZ, Gargiulo L, Kaneko S, Osawa M, Schrepfer S, Deuse T. Synthetic immune checkpoint engagers protect HLA-deficient iPSCs and derivatives from innate immune cell cytotoxicity. Cell Stem Cell 2023; 30:1538-1548.e4. [PMID: 37922880 DOI: 10.1016/j.stem.2023.10.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 08/23/2023] [Accepted: 10/04/2023] [Indexed: 11/07/2023]
Abstract
Immune rejection of allogeneic cell therapeutics remains a major problem for immuno-oncology and regenerative medicine. Allogeneic cell products so far have inferior persistence and efficacy when compared with autologous alternatives. Engineering of hypoimmune cells may greatly improve their therapeutic benefit. We present a new class of agonistic immune checkpoint engagers that protect human leukocyte antigen (HLA)-depleted induced pluripotent stem cell-derived endothelial cells (iECs) from innate immune cells. Engagers with agonistic functionality to their inhibitory receptors TIM3 and SIRPα effectively protect engineered iECs from natural killer (NK) cell and macrophage killing. The SIRPα engager can be combined with truncated CD64 to generate fully immune evasive iECs capable of escaping allogeneic cellular and immunoglobulin G (IgG) antibody-mediated rejection. Synthetic immune checkpoint engagers have high target specificity and lack retrograde signaling in the engineered cells. This modular design allows for the exploitation of more inhibitory immune pathways for immune evasion and could contribute to the advancement of allogeneic cell therapeutics.
Collapse
Affiliation(s)
- Alessia Gravina
- Transplant and Stem Cell Immunobiology (TSI)-Lab, Department of Surgery, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Grigol Tediashvili
- Transplant and Stem Cell Immunobiology (TSI)-Lab, Department of Surgery, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Yueting Zheng
- Shinobi Therapeutics, 2 Tower Place, South San Francisco, CA 94080, USA
| | - Kumiko A Iwabuchi
- Shinobi Therapeutics, 2 Tower Place, South San Francisco, CA 94080, USA
| | - Sara M Peyrot
- Shinobi Therapeutics, 2 Tower Place, South San Francisco, CA 94080, USA
| | - Susan Z Roodsari
- Shinobi Therapeutics, 2 Tower Place, South San Francisco, CA 94080, USA
| | - Lauren Gargiulo
- Shinobi Therapeutics, 2 Tower Place, South San Francisco, CA 94080, USA
| | - Shin Kaneko
- Laboratory of Regenerative Immunotherapy, Department of Cell Growth and Differentiation, Center for iPS cell Research, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Mitsujiro Osawa
- Shinobi Therapeutics, Med-Pharm Collaboration Building 46-29, Yoshida-Shimo-Adachi-Cho, Sakyo-Ku, Kyoto, Japan
| | - Sonja Schrepfer
- Transplant and Stem Cell Immunobiology (TSI)-Lab, Department of Surgery, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Tobias Deuse
- Transplant and Stem Cell Immunobiology (TSI)-Lab, Department of Surgery, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA.
| |
Collapse
|
8
|
Valantine HA. Applying Genomics to Unravel Health Disparities in Organ Transplantation: Paul I. Terasaki State-of-the-art Lecture; American Transplant Congress 2021. Transplantation 2023; 107:1258-1264. [PMID: 36584376 DOI: 10.1097/tp.0000000000004456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
An extensive body of research about team science provides empirical evidence that diverse teams outperform homogenous teams in creating more innovative solutions to complex problems. At the core of diverse and inclusive teams is a rich diversity of perspectives, experiences, and backgrounds that invite new questions and broaden the scope of research. Diverse perspectives are especially relevant for biomedicine, which seeks to find solutions for challenging problems affecting the human condition. It is essential that diversity and inclusion in biomedicine is prioritized as a key driver of innovation, both through the people who conduct the research and the science itself. Key questions have been articulated as important drivers for funding research: (1) Who is doing the science and who is building the tools? (2) What science and technology is being done and how? and (3) Who has access to the knowledge and benefits of scientific innovation? I will briefly review the empirical evidence supporting diversity as a powerful enhancer of the quality and outputs of research and clinical care. I offer my own research as a case study of incorporating a framework of diversity, equity, and inclusion into research that uses new emerging genomic tools for earlier and more precise diagnosis of organ transplant rejection. I will demonstrate how these same tools hold great promise for accelerating the discovery of hitherto unexplored mechanisms that drive the poor outcomes for African ancestry organ transplant recipients, which in turn will identify new diagnostics and therapeutic targets that benefit transplant recipients across all ancestries.
Collapse
|
9
|
Cha Y, Park TY, Leblanc P, Kim KS. Current Status and Future Perspectives on Stem Cell-Based Therapies for Parkinson's Disease. J Mov Disord 2023; 16:22-41. [PMID: 36628428 PMCID: PMC9978267 DOI: 10.14802/jmd.22141] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/15/2022] [Accepted: 10/29/2022] [Indexed: 01/12/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder after Alzheimer's disease, affecting 1%-2% of the population over the age of 65. As the population ages, it is anticipated that the burden on society will significantly escalate. Although symptom reduction by currently available pharmacological and/or surgical treatments improves the quality of life of many PD patients, there are no treatments that can slow down, halt, or reverse disease progression. Because the loss of a specific cell type, midbrain dopamine neurons in the substantia nigra, is the main cause of motor dysfunction in PD, it is considered a promising target for cell replacement therapy. Indeed, numerous preclinical and clinical studies using fetal cell transplantation have provided proof of concept that cell replacement therapy may be a viable therapeutic approach for PD. However, the use of human fetal cells remains fraught with controversy due to fundamental ethical, practical, and clinical limitations. Groundbreaking work on human pluripotent stem cells (hPSCs), including human embryonic stem cells and human induced pluripotent stem cells, coupled with extensive basic research in the stem cell field offers promising potential for hPSC-based cell replacement to become a realistic treatment regimen for PD once several major issues can be successfully addressed. In this review, we will discuss the prospects and challenges of hPSC-based cell therapy for PD.
Collapse
Affiliation(s)
- Young Cha
- Department of Psychiatry and Molecular Neurobiology Laboratory, McLean Hospital and Program in Neuroscience, Harvard Medical School, Belmont, MA, USA
| | - Tae-Yoon Park
- Department of Psychiatry and Molecular Neurobiology Laboratory, McLean Hospital and Program in Neuroscience, Harvard Medical School, Belmont, MA, USA
| | - Pierre Leblanc
- Department of Psychiatry and Molecular Neurobiology Laboratory, McLean Hospital and Program in Neuroscience, Harvard Medical School, Belmont, MA, USA
| | - Kwang-Soo Kim
- Department of Psychiatry and Molecular Neurobiology Laboratory, McLean Hospital and Program in Neuroscience, Harvard Medical School, Belmont, MA, USA
| |
Collapse
|
10
|
Gullapalli VK, Zarbin MA. New Prospects for Retinal Pigment Epithelium Transplantation. Asia Pac J Ophthalmol (Phila) 2022; 11:302-313. [PMID: 36041145 DOI: 10.1097/apo.0000000000000521] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/28/2022] [Indexed: 11/26/2022] Open
Abstract
ABSTRACT Retinal pigment epithelium (RPE) transplants rescue photoreceptors in selected animal models of retinal degenerative disease. Early clinical studies of RPE transplants as treatment for age-related macular degeneration (AMD) included autologous and allogeneic transplants of RPE suspensions and RPE sheets for atrophic and neovascular complications of AMD. Subsequent studies explored autologous RPE-Bruch membrane-choroid transplants in patients with neovascular AMD with occasional marked visual benefit, which establishes a rationale for RPE transplants in late-stage AMD. More recent work has involved transplantation of autologous and allogeneic stem cell-derived RPE for patients with AMD and those with Stargardt disease. These early-stage clinical trials have employed RPE suspensions and RPE monolayers on biocompatible scaffolds. Safety has been well documented, but evidence of efficacy is variable. Current research involves development of better scaffolds, improved modulation of immune surveillance, and modification of the extracellular milieu to improve RPE survival and integration with host retina.
Collapse
Affiliation(s)
| | - Marco A Zarbin
- Iinstitute of Ophthalmology and visual Science, Rutgers-New Jersey Medical School, Rutgers University, Newark, NJ, US
| |
Collapse
|
11
|
The Use of Trichostatin A during Pluripotent Stem Cell Generation Does Not Affect MHC Expression Level. Stem Cells Int 2022; 2022:9346767. [PMID: 35371264 PMCID: PMC8967593 DOI: 10.1155/2022/9346767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 01/02/2022] [Accepted: 01/28/2022] [Indexed: 11/17/2022] Open
Abstract
Pluripotent stem cells (PSCs) are considered as a potent tool for use in regenerative medicine. Highly efficient generation of PSCs through chromatin modulators such as trichostatin A (TSA) might change their MHC molecule expression profile. The efficiency of PSC generation and their immunogenicity is major obstacles for clinical use. Hence, we aim to investigate whether the use of TSA during PSC generation affects MHC expression level. Three PSC lines were generated by iPSCs, NT-ESCs, and IVF-ESCs' reprogramming methods from B6D2F1 mouse embryonic fibroblast cells. Established PSC lines were characterized by alkaline phosphatase assay (ALP) and immunocytochemistry. Their chromosome fidelity was checked by karyotyping. The expression level of pluripotent genes (oct4, nanog, sox2, klf4), HDACs (hdac1, hdac2, and hdac3), and immune-related genes (including Qa-1, Qa-2, H2kb, H2kd, H2db, H2db, CIITA, H2-IE-βb, H2-IE-βd) in iPSC and ESC lines were assessed by real-time PCR analysis. The presence of MHC molecules on the surface of pluripotent stem cells was also checked by flow cytometry technique. Significant increase of pluripotency markers, oct4, nanog, sox2, and klf4, was observed in 100 nM TSA-treated samples. 100 nM TSA induced significant upregulation of H2db in generated iPSCs. H2-IE-βd was remarkably downregulated in 50 and 100 nM TSA-treated iPSC lines. The expression level of other immune-related genes was not greatly affected by TSA in iPSC and NT-ESC lines. It is concluded that the use of short-term and low concentration of TSA during reprogramming in PSC generation procedure significantly increases PSC generation efficiency, but do not affect the MHC expression in established cell lines, which is in the benefit of cell transplantation in regenerative medicine.
Collapse
|
12
|
Defective chromatin architectures in embryonic stem cells derived from somatic cell nuclear transfer impair their differentiation potentials. Cell Death Dis 2021; 12:1085. [PMID: 34785659 PMCID: PMC8595669 DOI: 10.1038/s41419-021-04384-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 10/26/2021] [Accepted: 10/29/2021] [Indexed: 11/08/2022]
Abstract
Nuclear transfer embryonic stem cells (ntESCs) hold enormous promise for individual-specific regenerative medicine. However, the chromatin states of ntESCs remain poorly characterized. In this study, we employed ATAC-seq and Hi-C techniques to explore the chromatin accessibility and three-dimensional (3D) genome organization of ntESCs. The results show that the chromatin accessibility and genome structures of somatic cells are re-arranged to ESC-like states overall in ntESCs, including compartments, topologically associating domains (TADs) and chromatin loops. However, compared to fertilized ESCs (fESCs), ntESCs show some abnormal openness and structures that have not been reprogrammed completely, which impair the differentiation potential of ntESCs. The histone modification H3K9me3 may be involved in abnormal structures in ntESCs, including incorrect compartment switches and incomplete TAD rebuilding. Moreover, ntESCs and iPSCs show high similarity in 3D genome structures, while a few differences are detected due to different somatic cell origins and reprogramming mechanisms. Through systematic analyses, our study provides a global view of chromatin accessibility and 3D genome organization in ntESCs, which can further facilitate the understanding of the similarities and differences between ntESCs and fESCs.
Collapse
|
13
|
Sercel AJ, Carlson NM, Patananan AN, Teitell MA. Mitochondrial DNA Dynamics in Reprogramming to Pluripotency. Trends Cell Biol 2021; 31:311-323. [PMID: 33422359 PMCID: PMC7954944 DOI: 10.1016/j.tcb.2020.12.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/09/2020] [Accepted: 12/11/2020] [Indexed: 12/20/2022]
Abstract
Mammalian cells, with the exception of erythrocytes, harbor mitochondria, which are organelles that provide energy, intermediate metabolites, and additional activities to sustain cell viability, replication, and function. Mitochondria contain multiple copies of a circular genome called mitochondrial DNA (mtDNA), whose individual sequences are rarely identical (homoplasmy) because of inherited or sporadic mutations that result in multiple mtDNA genotypes (heteroplasmy). Here, we examine potential mechanisms for maintenance or shifts in heteroplasmy that occur in induced pluripotent stem cells (iPSCs) generated by cellular reprogramming, and further discuss manipulations that can alter heteroplasmy to impact stem and differentiated cell performance. This additional insight will assist in developing more robust iPSC-based models of disease and differentiated cell therapies.
Collapse
Affiliation(s)
- Alexander J Sercel
- Molecular Biology Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, USA 90095
| | - Natasha M Carlson
- Department of Biology, California State University Northridge, CA, USA 91330; Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA 90095
| | - Alexander N Patananan
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA 90095
| | - Michael A Teitell
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA 90095; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA 90095; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, USA 90095; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA 90095; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research University of California, Los Angeles, Los Angeles, CA, USA 90095; Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA 90095; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA 90095.
| |
Collapse
|
14
|
Nato G, Corti A, Parmigiani E, Jachetti E, Lecis D, Colombo MP, Delia D, Buffo A, Magrassi L. Immune-tolerance to human iPS-derived neural progenitors xenografted into the immature cerebellum is overridden by species-specific differences in differentiation timing. Sci Rep 2021; 11:651. [PMID: 33436685 PMCID: PMC7803978 DOI: 10.1038/s41598-020-79502-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 12/09/2020] [Indexed: 01/20/2023] Open
Abstract
We xeno-transplanted human neural precursor cells derived from induced pluripotent stem cells into the cerebellum and brainstem of mice and rats during prenatal development or the first postnatal week. The transplants survived and started to differentiate up to 1 month after birth when they were rejected by both species. Extended survival and differentiation of the same cells were obtained only when they were transplanted in NOD-SCID mice. Transplants of human neural precursor cells mixed with the same cells after partial in vitro differentiation or with a cellular extract obtained from adult rat cerebellum increased survival of the xeno-graft beyond one month. These findings are consistent with the hypothesis that the slower pace of differentiation of human neural precursors compared to that of rodents restricts induction of immune-tolerance to human antigens expressed before completion of maturation of the immune system. With further maturation the transplanted neural precursors expressed more mature antigens before the graft were rejected. Supplementation of the immature cells suspensions with more mature antigens may help to induce immune-tolerance for those antigens expressed only later by the engrafted cells.
Collapse
Affiliation(s)
- Giulia Nato
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, Via Cherasco 15, Torino, Italy.,Neuroscience Institute Cavalieri Ottolenghi (NICO), 10043, Orbassano, Torino, Italy
| | - Alessandro Corti
- Department of Research, Fondazione IRCCS Istituto Nazionale Tumori, Milano, Via Amadeo 42, 20133, Milano, Italy
| | - Elena Parmigiani
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, Via Cherasco 15, Torino, Italy.,Neuroscience Institute Cavalieri Ottolenghi (NICO), 10043, Orbassano, Torino, Italy
| | - Elena Jachetti
- Department of Research, Fondazione IRCCS Istituto Nazionale Tumori, Milano, Via Amadeo 42, 20133, Milano, Italy
| | - Daniele Lecis
- Department of Research, Fondazione IRCCS Istituto Nazionale Tumori, Milano, Via Amadeo 42, 20133, Milano, Italy
| | - Mario Paolo Colombo
- Department of Research, Fondazione IRCCS Istituto Nazionale Tumori, Milano, Via Amadeo 42, 20133, Milano, Italy
| | - Domenico Delia
- Department of Research, Fondazione IRCCS Istituto Nazionale Tumori, Milano, Via Amadeo 42, 20133, Milano, Italy.,IFOM, FIRC Institute of Molecular Oncology, Via Adamello 16, 20139, Milano, Italy
| | - Annalisa Buffo
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, Via Cherasco 15, Torino, Italy.,Neuroscience Institute Cavalieri Ottolenghi (NICO), 10043, Orbassano, Torino, Italy
| | - Lorenzo Magrassi
- Neurosurgery, Department of Clinical, Surgical, Diagnostic and Pediatric Science, University of Pavia, Foundation IRCCS Policlinico San Matteo, Pavia, Italy. .,Istituto Di Genetica Molecolare IGM-CNR, via Abbiategrasso 207, 27100, Pavia, Italy.
| |
Collapse
|
15
|
Muckom RJ, Sampayo RG, Johnson HJ, Schaffer DV. Advanced Materials to Enhance Central Nervous System Tissue Modeling and Cell Therapy. ADVANCED FUNCTIONAL MATERIALS 2020; 30:2002931. [PMID: 33510596 PMCID: PMC7840150 DOI: 10.1002/adfm.202002931] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Indexed: 05/04/2023]
Abstract
The progressively deeper understanding of mechanisms underlying stem cell fate decisions has enabled parallel advances in basic biology-such as the generation of organoid models that can further one's basic understanding of human development and disease-and in clinical translation-including stem cell based therapies to treat human disease. Both of these applications rely on tight control of the stem cell microenvironment to properly modulate cell fate, and materials that can be engineered to interface with cells in a controlled and tunable manner have therefore emerged as valuable tools for guiding stem cell growth and differentiation. With a focus on the central nervous system (CNS), a broad range of material solutions that have been engineered to overcome various hurdles in constructing advanced organoid models and developing effective stem cell therapeutics is reviewed. Finally, regulatory aspects of combined material-cell approaches for CNS therapies are considered.
Collapse
Affiliation(s)
- Riya J Muckom
- Department of Chemical and Biomolecular Engineering, UC Berkeley, Berkeley, CA 94704, USA
| | - Rocío G Sampayo
- Department of Chemical and Biomolecular Engineering, UC Berkeley, Berkeley, CA 94704, USA
| | - Hunter J Johnson
- Department of Bioengineering, UC Berkeley, Berkeley, CA 94704, USA
| | - David V Schaffer
- Department of Chemical and Biomolecular Engineering, UC Berkeley, Berkeley, CA 94704, USA
| |
Collapse
|
16
|
Hu X, Kueppers ST, Kooreman NG, Gravina A, Wang D, Tediashvili G, Schlickeiser S, Frentsch M, Nikolaou C, Thiel A, Marcus S, Fuchs S, Velden J, Reichenspurner H, Volk HD, Deuse T, Schrepfer S. The H-Y Antigen in Embryonic Stem Cells Causes Rejection in Syngeneic Female Recipients. Stem Cells Dev 2020; 29:1179-1189. [PMID: 32723003 DOI: 10.1089/scd.2019.0299] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Pluripotent stem cells are promising candidates for cell-based regenerative therapies. To avoid rejection of transplanted cells, several approaches are being pursued to reduce immunogenicity of the cells or modulate the recipient's immune response. These include gene editing to reduce the antigenicity of cell products, immunosuppression of the host, or using major histocompatibility complex-matched cells from cell banks. In this context, we have investigated the antigenicity of H-Y antigens, a class of minor histocompatibility antigens encoded by the Y chromosome, to assess whether the gender of the donor affects the cell's antigenicity. In a murine transplant model, we show that the H-Y antigen in undifferentiated embryonic stem cells (ESCs), as well as ESC-derived endothelial cells, provokes T- and B cell responses in female recipients.
Collapse
Affiliation(s)
- Xiaomeng Hu
- Transplant and Stem Cell Immunobiology Lab, Department of Surgery, University of California, San Francisco, California, USA.,Cardiovascular Research Center Hamburg (CVRC) and DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Luebeck, Hamburg, Germany.,University Heart & Vascular Center Hamburg, Hamburg, Germany
| | - Simon T Kueppers
- Transplant and Stem Cell Immunobiology Lab, Department of Surgery, University of California, San Francisco, California, USA.,Cardiovascular Research Center Hamburg (CVRC) and DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Luebeck, Hamburg, Germany.,University Heart & Vascular Center Hamburg, Hamburg, Germany
| | - Nigel G Kooreman
- Stanford Cardiovascular Institute, Stanford University, Stanford, California, USA.,Department of Medicine, Stanford University, Stanford, California, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, USA.,Department of Vascular Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Alessia Gravina
- Transplant and Stem Cell Immunobiology Lab, Department of Surgery, University of California, San Francisco, California, USA.,Cardiovascular Research Center Hamburg (CVRC) and DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Luebeck, Hamburg, Germany
| | - Dong Wang
- Transplant and Stem Cell Immunobiology Lab, Department of Surgery, University of California, San Francisco, California, USA.,Cardiovascular Research Center Hamburg (CVRC) and DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Luebeck, Hamburg, Germany.,University Heart & Vascular Center Hamburg, Hamburg, Germany
| | - Grigol Tediashvili
- Transplant and Stem Cell Immunobiology Lab, Department of Surgery, University of California, San Francisco, California, USA.,Cardiovascular Research Center Hamburg (CVRC) and DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Luebeck, Hamburg, Germany.,University Heart & Vascular Center Hamburg, Hamburg, Germany
| | - Stephan Schlickeiser
- BIH-Center for Regenerative Therapies (BCRT), Charité University Medicine and Berlin Institute of Health (BIH), Berlin, Germany.,Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, BIH, Berlin, Germany
| | - Marco Frentsch
- BIH-Center for Regenerative Therapies (BCRT), Charité University Medicine and Berlin Institute of Health (BIH), Berlin, Germany
| | - Christos Nikolaou
- BIH-Center for Regenerative Therapies (BCRT), Charité University Medicine and Berlin Institute of Health (BIH), Berlin, Germany
| | - Andreas Thiel
- BIH-Center for Regenerative Therapies (BCRT), Charité University Medicine and Berlin Institute of Health (BIH), Berlin, Germany
| | - Sivan Marcus
- Transplant and Stem Cell Immunobiology Lab, Department of Surgery, University of California, San Francisco, California, USA
| | - Sigrid Fuchs
- Institute of Human Genetics, University Medical Center Hamburg, Hamburg, Germany
| | - Joachim Velden
- Evotec AG, Histopathology and In Vivo Pharmacology, Hamburg, Germany
| | - Hermann Reichenspurner
- Cardiovascular Research Center Hamburg (CVRC) and DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Luebeck, Hamburg, Germany.,University Heart & Vascular Center Hamburg, Hamburg, Germany
| | - Hans-Dieter Volk
- BIH-Center for Regenerative Therapies (BCRT), Charité University Medicine and Berlin Institute of Health (BIH), Berlin, Germany.,Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, BIH, Berlin, Germany
| | - Tobias Deuse
- Transplant and Stem Cell Immunobiology Lab, Department of Surgery, University of California, San Francisco, California, USA
| | - Sonja Schrepfer
- Transplant and Stem Cell Immunobiology Lab, Department of Surgery, University of California, San Francisco, California, USA.,Cardiovascular Research Center Hamburg (CVRC) and DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Luebeck, Hamburg, Germany.,University Heart & Vascular Center Hamburg, Hamburg, Germany
| |
Collapse
|
17
|
Desgres M, Menasché P. Clinical Translation of Pluripotent Stem Cell Therapies: Challenges and Considerations. Cell Stem Cell 2020; 25:594-606. [PMID: 31703770 DOI: 10.1016/j.stem.2019.10.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Although the clinical outcomes of cell therapy trials have not met initial expectations, emerging evidence suggests that injury-mediated tissue damage might benefit from the delivery of cells or their secreted products. Pluripotent stem cells (PSCs) are promising cell sources primarily because of their capacity to generate stage- and lineage-specific differentiated derivatives. However, they carry inherent challenges for safe and efficacious clinical translation. This Review describes completed or ongoing trials of PSCs, discusses their potential mechanisms of action, and considers how to address the challenges required for them to become a major therapy, using heart repair as a case study.
Collapse
Affiliation(s)
- Manon Desgres
- Université de Paris, PARCC, INSERM, 75015 Paris, France
| | - Philippe Menasché
- Université de Paris, PARCC, INSERM, 75015 Paris, France; Department of Cardiovascular Surgery, Hôpital Européen Georges Pompidou 20, rue Leblanc, 75015 Paris, France.
| |
Collapse
|
18
|
Affiliation(s)
- David A Prentice
- From the Advisory Board for the Midwest Stem Cell Therapy Center, University of Kansas Medical Center, Kansas City; and Charlotte Lozier Institute, Arlington, VA
| |
Collapse
|
19
|
Khacho M, Harris R, Slack RS. Mitochondria as central regulators of neural stem cell fate and cognitive function. Nat Rev Neurosci 2019; 20:34-48. [PMID: 30464208 DOI: 10.1038/s41583-018-0091-3] [Citation(s) in RCA: 259] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Emerging evidence now indicates that mitochondria are central regulators of neural stem cell (NSC) fate decisions and are crucial for both neurodevelopment and adult neurogenesis, which in turn contribute to cognitive processes in the mature brain. Inherited mutations and accumulated damage to mitochondria over the course of ageing serve as key factors underlying cognitive defects in neurodevelopmental disorders and neurodegenerative diseases, respectively. In this Review, we explore the recent findings that implicate mitochondria as crucial regulators of NSC function and cognition. In this respect, mitochondria may serve as targets for stem-cell-based therapies and interventions for cognitive defects.
Collapse
Affiliation(s)
- Mireille Khacho
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology (OISB), University of Ottawa, Ottawa, Ontario, Canada
| | - Richard Harris
- Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, Ottawa, Ontario, Canada
| | - Ruth S Slack
- Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, Ottawa, Ontario, Canada.
| |
Collapse
|
20
|
Deuse T, Hu X, Agbor-Enoh S, Koch M, Spitzer MH, Gravina A, Alawi M, Marishta A, Peters B, Kosaloglu-Yalcin Z, Yang Y, Rajalingam R, Wang D, Nashan B, Kiefmann R, Reichenspurner H, Valantine H, Weissman IL, Schrepfer S. De novo mutations in mitochondrial DNA of iPSCs produce immunogenic neoepitopes in mice and humans. Nat Biotechnol 2019; 37:1137-1144. [PMID: 31427818 DOI: 10.1038/s41587-019-0227-7] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 07/16/2019] [Indexed: 12/12/2022]
Abstract
The utility of autologous induced pluripotent stem cell (iPSC) therapies for tissue regeneration depends on reliable production of immunologically silent functional iPSC derivatives. However, rejection of autologous iPSC-derived cells has been reported, although the mechanism underlying rejection is largely unknown. We hypothesized that de novo mutations in mitochondrial DNA (mtDNA), which has far less reliable repair mechanisms than chromosomal DNA, might produce neoantigens capable of eliciting immune recognition and rejection. Here we present evidence in mice and humans that nonsynonymous mtDNA mutations can arise and become enriched during reprogramming to the iPSC stage, long-term culture and differentiation into target cells. These mtDNA mutations encode neoantigens that provoke an immune response that is highly specific and dependent on the host major histocompatibility complex genotype. Our results reveal that autologous iPSCs and their derivatives are not inherently immunologically inert for autologous transplantation and suggest that iPSC-derived products should be screened for mtDNA mutations.
Collapse
Affiliation(s)
- Tobias Deuse
- Department of Surgery, Division of Cardiothoracic Surgery, Transplant and Stem Cell Immunobiology Lab, University of California, San Francisco, San Francisco, CA, USA
| | - Xiaomeng Hu
- Department of Surgery, Division of Cardiothoracic Surgery, Transplant and Stem Cell Immunobiology Lab, University of California, San Francisco, San Francisco, CA, USA
- Department of Cardiovascular Surgery, University Heart Center Hamburg, Hamburg, Germany
- Cardiovascular Research Center Hamburg and German Center for Cardiovascular Research, partner site Hamburg/Kiel/Luebeck, Hamburg, Germany
| | - Sean Agbor-Enoh
- Division of Pulmonary and Critical Care Medicine, The Johns Hopkins School of Medicine, Baltimore, MD, USA
- Laboratory of Transplantation Genomics, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Martina Koch
- Department of Hepatobiliary and Transplant Surgery, University Medical Center Hamburg-Eppendorf, University Transplant Center, Hamburg, Germany
| | - Matthew H Spitzer
- Departments of Otolaryngology, Head and Neck Surgery and Microbiology and Immunology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Alessia Gravina
- Department of Surgery, Division of Cardiothoracic Surgery, Transplant and Stem Cell Immunobiology Lab, University of California, San Francisco, San Francisco, CA, USA
- Cardiovascular Research Center Hamburg and German Center for Cardiovascular Research, partner site Hamburg/Kiel/Luebeck, Hamburg, Germany
| | - Malik Alawi
- Bioinformatics Core, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Argit Marishta
- Laboratory of Transplantation Genomics, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Bjoern Peters
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Zeynep Kosaloglu-Yalcin
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Yanqin Yang
- Laboratory of Transplantation Genomics, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Raja Rajalingam
- Immunogenetics and Transplantation Laboratory, Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Dong Wang
- Department of Surgery, Division of Cardiothoracic Surgery, Transplant and Stem Cell Immunobiology Lab, University of California, San Francisco, San Francisco, CA, USA
- Department of Cardiovascular Surgery, University Heart Center Hamburg, Hamburg, Germany
- Cardiovascular Research Center Hamburg and German Center for Cardiovascular Research, partner site Hamburg/Kiel/Luebeck, Hamburg, Germany
| | - Bjoern Nashan
- Department of Hepatobiliary and Transplant Surgery, University Medical Center Hamburg-Eppendorf, University Transplant Center, Hamburg, Germany
| | - Rainer Kiefmann
- Department of Anaesthesia, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hermann Reichenspurner
- Department of Cardiovascular Surgery, University Heart Center Hamburg, Hamburg, Germany
- Cardiovascular Research Center Hamburg and German Center for Cardiovascular Research, partner site Hamburg/Kiel/Luebeck, Hamburg, Germany
| | - Hannah Valantine
- Laboratory of Transplantation Genomics, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Irving L Weissman
- Department of Developmental Biology, Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Sonja Schrepfer
- Department of Surgery, Division of Cardiothoracic Surgery, Transplant and Stem Cell Immunobiology Lab, University of California, San Francisco, San Francisco, CA, USA.
- Department of Cardiovascular Surgery, University Heart Center Hamburg, Hamburg, Germany.
- Cardiovascular Research Center Hamburg and German Center for Cardiovascular Research, partner site Hamburg/Kiel/Luebeck, Hamburg, Germany.
- Sana Biotechnology Inc., South San Francisco, CA, USA.
| |
Collapse
|
21
|
Abstract
Aging is accompanied by a time-dependent progressive deterioration of multiple factors of the cellular system. The past several decades have witnessed major leaps in our understanding of the biological mechanisms of aging using dietary, genetic, pharmacological, and physical interventions. Metabolic processes, including nutrient sensing pathways and mitochondrial function, have emerged as prominent regulators of aging. Mitochondria have been considered to play a key role largely due to their production of reactive oxygen species (ROS), resulting in DNA damage that accumulates over time and ultimately causes cellular failure. This theory, known as the mitochondrial free radical theory of aging (MFRTA), was favored by the aging field, but increasing inconsistent evidence has led to criticism and rejection of this idea. However, MFRTA should not be hastily rejected in its entirety because we now understand that ROS is not simply an undesired toxic metabolic byproduct, but also an important signaling molecule that is vital to cellular fitness. Notably, mitochondrial function, a term traditionally referred to bioenergetics and apoptosis, has since expanded considerably. It encompasses numerous other key biological processes, including the following: (i) complex metabolic processes, (ii) intracellular and endocrine signaling/communication, and (iii) immunity/inflammation. Here, we will discuss shortcomings of previous concepts regarding mitochondria in aging and their emerging roles based on recent advances. We will also discuss how the mitochondrial genome integrates with major theories on the evolution of aging. [BMB Reports 2019; 52(1): 13-23].
Collapse
Affiliation(s)
- Jyung Mean Son
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Changhan Lee
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089; USC Norris Comprehensive Cancer Center, Los Angeles, CA 90089, USA; Biomedical Science, Graduate School, Ajou University, Suwon 16499, Korea
| |
Collapse
|
22
|
Jun SM, Park M, Lee JY, Jung S, Lee JE, Shim SH, Song H, Lee DR. Single cell-derived clonally expanded mesenchymal progenitor cells from somatic cell nuclear transfer-derived pluripotent stem cells ameliorate the endometrial function in the uterus of a murine model with Asherman's syndrome. Cell Prolif 2019; 52:e12597. [PMID: 30896075 PMCID: PMC6536448 DOI: 10.1111/cpr.12597] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 01/20/2019] [Accepted: 02/12/2019] [Indexed: 12/24/2022] Open
Abstract
Objectives Because primary mesenchymal progenitor cells (adult‐MPCs) have various functions that depend on the tissue origin and donor, de novo MPCs from human pluripotent stem cells (hPSCs) would be required in regenerative medicine. However, the characteristics and function of MPCs derived from reprogrammed hPSCs have not been well studied. Thus, we show that functional MPCs can be successfully established from a single cell‐derived clonal expansion following MPC derivation from somatic cell nuclear transfer‐derived (SCNT)‐hPSCs, and these cells can serve as therapeutic contributors in an animal model of Asherman's syndrome (AS). Materials and methods We developed single cell‐derived clonal expansion following MPC derivation from SCNT‐hPSCs to offer a pure population and a higher biological activity. Additionally, we investigated the therapeutic effects of SCNT‐hPSC‐MPCs in model mice of Asherman's syndrome (AS), which is characterized by synechiae or fibrosis with endometrial injury. Results Their humoral effects in proliferating host cells encouraged angiogenesis and decreased pro‐inflammatory factors via a host‐dependent mechanism, resulting in reduction in AS. We also addressed that cellular activities such as the cell proliferation and population doubling of SCNT‐hPSC‐MPCs resemble those of human embryonic stem cell‐derived MPCs (hESC‐MPCs) and are much higher than those of adult‐MPCs. Conclusions Somatic cell nuclear transfer‐derived‐hPSCs‐MPCs could be an advanced therapeutic strategy for specific diseases in the field of regenerative medicine.
Collapse
Affiliation(s)
- Sung-Min Jun
- CHA Advanced Research Institute, Seongnam, Korea
| | - Mira Park
- Department of Biomedical Science, CHA University, Seongnam, Korea
| | - Ji Yoon Lee
- Department of Biomedical Science, CHA University, Seongnam, Korea
| | | | | | - Sung Han Shim
- Department of Biomedical Science, CHA University, Seongnam, Korea
| | - Haengseok Song
- Department of Biomedical Science, CHA University, Seongnam, Korea
| | - Dong Ryul Lee
- CHA Advanced Research Institute, Seongnam, Korea.,Department of Biomedical Science, CHA University, Seongnam, Korea
| |
Collapse
|
23
|
Moon J, Roh S. Expression of polo-like kinase 1 in pre-implantation stage murine somatic cell nuclear transfer embryos. J Vet Sci 2019; 20:2-9. [PMID: 30481982 PMCID: PMC6351765 DOI: 10.4142/jvs.2019.20.1.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 10/23/2018] [Accepted: 11/06/2018] [Indexed: 11/23/2022] Open
Abstract
Somatic cell nuclear transfer (SCNT) has various applications in research, as well as in the medical field and animal husbandry. However, the efficiency of SCNT is low and the accurate mechanism of SCNT in murine embryo development is unreported. In general, the developmental rate of SCNT murine embryos is lower than in vivo counterparts. In previous studies, polo-like kinase 1 (Plk1) was reported to be a crucial element in cell division including centrosome maturation, cytokinesis, and spindle formation. In an initial series of experiments in this study, BI2536, a Plk1 inhibitor, was treated to in vivo-fertilized embryos and the embryos failed to develop beyond the 2-cell stage. This confirmed previous findings that Plk1 is crucial for the first mitotic division of murine embryos. Next, we investigated Plk1's localization and intensity by immunofluorescence analysis. In contrast to normally developed embryos, SCNT murine embryos that failed to develop exhibited two types of Plk1 expressions; a low Plk1 expression pattern and ectopic expression of Plk1. The results show that Plk1 has a critical role in SCNT murine embryos. In conclusion, this study demonstrated that the SCNT murine embryos fail to develop beyond the 2-cell stage, and the embryos show abnormal Plk1 expression patterns, which may one of the main causes of developmental failure of early SCNT murine embryos.
Collapse
Affiliation(s)
- Jeonghyeon Moon
- Cellular Reprogramming and Embryo Biotechnology Laboratory, Dental Research Institute, BK21 PLUS Dental Life Science, Seoul National University School of Dentistry, Seoul 08826, Korea
| | - Sangho Roh
- Cellular Reprogramming and Embryo Biotechnology Laboratory, Dental Research Institute, BK21 PLUS Dental Life Science, Seoul National University School of Dentistry, Seoul 08826, Korea
| |
Collapse
|
24
|
Abstract
Results from at least 20 independent laboratories indicate that adult tissues contain rare, early-development stem cells known as very small embryonic-like stem cells (VSELs), which can differentiate into cells from more than one germ layer. Further research on these cells may provide a path forward to application of these cells in regenerative medicine that perhaps may solve several problems inherent in the use of controversial embryonic stem cells (ESCs) and somehow problematic induced pluripotent stem cells (iPSCs).
Collapse
Affiliation(s)
- Mariusz Z. Ratajczak
- Stem Cell Institute, 500 South Floyd Street, James Graham Brown Cancer Center, University Louisville, Louisville, 40202, Kentucky, USA
- Department of Regenerative Medicine, Warsaw Medical University, Warsaw, Poland
| | - Janina Ratajczak
- Stem Cell Institute, 500 South Floyd Street, James Graham Brown Cancer Center, University Louisville, Louisville, 40202, Kentucky, USA
| | - Magda Kucia
- Stem Cell Institute, 500 South Floyd Street, James Graham Brown Cancer Center, University Louisville, Louisville, 40202, Kentucky, USA
- Department of Regenerative Medicine, Warsaw Medical University, Warsaw, Poland
| |
Collapse
|
25
|
Abstract
Successful cloning of monkeys, the first non-human primate species, by somatic cell nuclear transfer (SCNT) attracted worldwide attention earlier this year. Remarkably, it has taken more than 20 years since the cloning of Dolly the sheep in 1997 to achieve this feat. This success was largely due to recent understanding of epigenetic barriers that impede SCNT-mediated reprogramming and the establishment of key methods to overcome these barriers, which also allowed efficient derivation of human pluripotent stem cells for cell therapy. Here, we summarize recent advances in SCNT technology and its potential applications for both reproductive and therapeutic cloning.
Collapse
Affiliation(s)
- Shogo Matoba
- RIKEN Bioresource Research Center, Tsukuba, Ibaraki 305-0074, Japan; Cooperative Division of Veterinary Sciences, Tokyo University of Agriculture and Technology, Fuchu, Tokyo 183-8509, Japan.
| | - Yi Zhang
- Howard Hughes Medical Institute; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Boston, MA 02115, USA.
| |
Collapse
|
26
|
Manipulating cell fate while confronting reproducibility concerns. Biochem Pharmacol 2018; 151:144-156. [DOI: 10.1016/j.bcp.2018.01.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 01/04/2018] [Indexed: 12/13/2022]
|
27
|
Ni J, Sun Y, Liu Z. The Potential of Stem Cells and Stem Cell-Derived Exosomes in Treating Cardiovascular Diseases. J Cardiovasc Transl Res 2018. [PMID: 29525884 DOI: 10.1007/s12265-018-9799-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In recent years, the cardiac protective mechanisms of stem cells have become a research focus. Increasing evidence has suggested that stem cells release vesicles, including exosomes and micro-vesicles. The content of these vesicles relies on an extracellular stimulus, and active ingredients are extensively being studied. Previous studies have confirmed that stem cell-derived exosomes have a cardiac protective function similar to that of stem cells, and promote angiogenesis, decrease apoptosis, and respond to stress. Compared to stem cells, exosomes are more stable without aneuploidy and immune rejection, and may be a promising and effective therapy for cardiovascular diseases. In this review, the biological functions and molecular mechanisms of stem cells and stem cell-derived exosomes are discussed.
Collapse
Affiliation(s)
- Jing Ni
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai, China.,Pan-Vascular Research Institute, Heart, Lung, and Blood Center, Tongji University School of Medicine, Shanghai, China
| | - Yuxi Sun
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai, China.,Pan-Vascular Research Institute, Heart, Lung, and Blood Center, Tongji University School of Medicine, Shanghai, China
| | - Zheng Liu
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai, China. .,Pan-Vascular Research Institute, Heart, Lung, and Blood Center, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
28
|
Garreta E, Sanchez S, Lajara J, Montserrat N, Belmonte JCI. Roadblocks in the Path of iPSC to the Clinic. CURRENT TRANSPLANTATION REPORTS 2018; 5:14-18. [PMID: 29564204 PMCID: PMC5843691 DOI: 10.1007/s40472-018-0177-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Purpose of Review The goal of this paper is to highlight the major challenges in the translation of human pluripotent stem cells into a clinical setting. Recent Findings Innate features from human induced pluripotent stem cells (hiPSCs) positioned these patient-specific cells as an unprecedented cell source for regenerative medicine applications. Immunogenicity of differentiated iPSCs requires more research towards the definition of common criteria for the evaluation of innate and host immune responses as well as in the generation of standardized protocols for iPSC generation and differentiation. The coming years will resolve ongoing clinical trials using both human embryonic stem cells (hESCs) and hiPSCs providing exciting information for the optimization of potential clinical applications of stem cell therapies. Summary Rapid advances in the field of iPSCs generated high expectations in the field of regenerative medicine. Understanding therapeutic applications of iPSCs certainly needs further investigation on autologous/allogenic iPSC transplantation.
Collapse
Affiliation(s)
- Elena Garreta
- Pluripotent stem cells and activation of endogenous tissue programs for organ regeneration (PR Lab), Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona, Spain
| | - Sonia Sanchez
- Universidad Católica San Antonio de Murcia (UCAM), Campus de los Jerónimos, 135 Guadalupe, 30107 Murcia, Spain
| | - Jeronimo Lajara
- Universidad Católica San Antonio de Murcia (UCAM), Campus de los Jerónimos, 135 Guadalupe, 30107 Murcia, Spain
| | - Nuria Montserrat
- Pluripotent stem cells and activation of endogenous tissue programs for organ regeneration (PR Lab), Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona, Spain
- Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Juan Carlos Izpisua Belmonte
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037 USA
| |
Collapse
|
29
|
Chaterji S, Ahn EH, Kim DH. CRISPR Genome Engineering for Human Pluripotent Stem Cell Research. Theranostics 2017; 7:4445-4469. [PMID: 29158838 PMCID: PMC5695142 DOI: 10.7150/thno.18456] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 08/24/2017] [Indexed: 12/13/2022] Open
Abstract
The emergence of targeted and efficient genome editing technologies, such as repurposed bacterial programmable nucleases (e.g., CRISPR-Cas systems), has abetted the development of cell engineering approaches. Lessons learned from the development of RNA-interference (RNA-i) therapies can spur the translation of genome editing, such as those enabling the translation of human pluripotent stem cell engineering. In this review, we discuss the opportunities and the challenges of repurposing bacterial nucleases for genome editing, while appreciating their roles, primarily at the epigenomic granularity. First, we discuss the evolution of high-precision, genome editing technologies, highlighting CRISPR-Cas9. They exist in the form of programmable nucleases, engineered with sequence-specific localizing domains, and with the ability to revolutionize human stem cell technologies through precision targeting with greater on-target activities. Next, we highlight the major challenges that need to be met prior to bench-to-bedside translation, often learning from the path-to-clinic of complementary technologies, such as RNA-i. Finally, we suggest potential bioinformatics developments and CRISPR delivery vehicles that can be deployed to circumvent some of the challenges confronting genome editing technologies en route to the clinic.
Collapse
|
30
|
Precise immune tolerance for hPSC derivatives in clinical application. Cell Immunol 2017; 326:15-23. [PMID: 28866278 DOI: 10.1016/j.cellimm.2017.08.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 08/03/2017] [Accepted: 08/04/2017] [Indexed: 11/22/2022]
Abstract
Human pluripotent stem cells (hPSCs) promise a foreseeing future for regeneration medicine and cell replacement therapy with their abilities to produce almost any types of somatic cells of the body. The complicated immunogenicity of hPSC derivatives and context dependent responses in variable transplantations greatly hurdle the practical application of hPSCs in clinic. Especially for applications of hPSCs, induction of immune tolerance at the same time increases the risks of tumorigenesis. Over the past few years, thanks to the progress in immunology and practices in organ transplantation, endeavors on exploring strategies to induce long term protection of allogeneic transplants have shed light on overcoming this barrier. Novel genetic engineering techniques also allow to precisely cradle the immune response of transplantation. Here we reviewed the current understanding on immunogenicity, and efforts have been attempted on inducing immune tolerance for hPSC derivatives, with extra focus on modifying the graft cells. We also glimpse on employing cutting-edge genome editing technologies for this purpose, which will potentially endow hPSC derivatives with the nature of wide spectrum drugs for therapy.
Collapse
|
31
|
Guenther SPW, Schrepfer S, Reichenspurner H, Deuse T. Myokardiale Regeneration. ZEITSCHRIFT FUR HERZ THORAX UND GEFASSCHIRURGIE 2017. [DOI: 10.1007/s00398-016-0113-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
32
|
You can go your own way: State regulation of oocyte donation in California and New York. BIOSOCIETIES 2016. [DOI: 10.1057/s41292-016-0026-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
33
|
Huang J, Zhang H, Yao J, Qin G, Wang F, Wang X, Luo A, Zheng Q, Cao C, Zhao J. BIX-01294 increases pig cloning efficiency by improving epigenetic reprogramming of somatic cell nuclei. Reproduction 2016; 151:39-49. [PMID: 26604326 DOI: 10.1530/rep-15-0460] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Accumulating evidence suggests that faulty epigenetic reprogramming leads to the abnormal development of cloned embryos and results in the low success rates observed in all mammals produced through somatic cell nuclear transfer (SCNT). The aberrant methylation status of H3K9me and H3K9me2 has been reported in cloned mouse embryos. To explore the role of H3K9me2 and H3K9me in the porcine somatic cell nuclear reprogramming, BIX-01294, known as a specific inhibitor of G9A (histone-lysine methyltransferase of H3K9), was used to treat the nuclear-transferred (NT) oocytes for 14-16 h after activation. The results showed that the developmental competence of porcine SCNT embryos was significantly enhanced both in vitro (blastocyst rate 16.4% vs 23.2%, P<0.05) and in vivo (cloning rate 1.59% vs 2.96%) after 50 nm BIX-01294 treatment. BIX-01294 treatment significantly decreased the levels of H3K9me2 and H3K9me at the 2- and 4-cell stages, which are associated with embryo genetic activation, and increased the transcriptional expression of the pluripotency genes SOX2, NANOG and OCT4 in cloned blastocysts. Furthermore, the histone acetylation levels of H3K9, H4K8 and H4K12 in cloned embryos were decreased after BIX-01294 treatment. However, co-treatment of activated NT oocytes with BIX-01294 and Scriptaid rescued donor nuclear chromatin from decreased histone acetylation of H4K8 that resulted from exposure to BIX-01294 only and consequently improved the preimplantation development of SCNT embryos (blastocyst formation rates of 23.7% vs 21.5%). These results indicated that treatment with BIX-01294 enhanced the developmental competence of porcine SCNT embryos through improvements in epigenetic reprogramming and gene expression.
Collapse
Affiliation(s)
- Jiaojiao Huang
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, ChinaUniversity of Chinese Academy of SciencesBeijing 100049, ChinaCollege of Life SciencesCapital Normal University, 105 Xisanhuan North Road, Haidian District, Beijing 100048, China State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, ChinaUniversity of Chinese Academy of SciencesBeijing 100049, ChinaCollege of Life SciencesCapital Normal University, 105 Xisanhuan North Road, Haidian District, Beijing 100048, China
| | - Hongyong Zhang
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, ChinaUniversity of Chinese Academy of SciencesBeijing 100049, ChinaCollege of Life SciencesCapital Normal University, 105 Xisanhuan North Road, Haidian District, Beijing 100048, China State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, ChinaUniversity of Chinese Academy of SciencesBeijing 100049, ChinaCollege of Life SciencesCapital Normal University, 105 Xisanhuan North Road, Haidian District, Beijing 100048, China
| | - Jing Yao
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, ChinaUniversity of Chinese Academy of SciencesBeijing 100049, ChinaCollege of Life SciencesCapital Normal University, 105 Xisanhuan North Road, Haidian District, Beijing 100048, China
| | - Guosong Qin
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, ChinaUniversity of Chinese Academy of SciencesBeijing 100049, ChinaCollege of Life SciencesCapital Normal University, 105 Xisanhuan North Road, Haidian District, Beijing 100048, China
| | - Feng Wang
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, ChinaUniversity of Chinese Academy of SciencesBeijing 100049, ChinaCollege of Life SciencesCapital Normal University, 105 Xisanhuan North Road, Haidian District, Beijing 100048, China
| | - Xianlong Wang
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, ChinaUniversity of Chinese Academy of SciencesBeijing 100049, ChinaCollege of Life SciencesCapital Normal University, 105 Xisanhuan North Road, Haidian District, Beijing 100048, China
| | - Ailing Luo
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, ChinaUniversity of Chinese Academy of SciencesBeijing 100049, ChinaCollege of Life SciencesCapital Normal University, 105 Xisanhuan North Road, Haidian District, Beijing 100048, China
| | - Qiantao Zheng
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, ChinaUniversity of Chinese Academy of SciencesBeijing 100049, ChinaCollege of Life SciencesCapital Normal University, 105 Xisanhuan North Road, Haidian District, Beijing 100048, China State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, ChinaUniversity of Chinese Academy of SciencesBeijing 100049, ChinaCollege of Life SciencesCapital Normal University, 105 Xisanhuan North Road, Haidian District, Beijing 100048, China
| | - Chunwei Cao
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, ChinaUniversity of Chinese Academy of SciencesBeijing 100049, ChinaCollege of Life SciencesCapital Normal University, 105 Xisanhuan North Road, Haidian District, Beijing 100048, China State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, ChinaUniversity of Chinese Academy of SciencesBeijing 100049, ChinaCollege of Life SciencesCapital Normal University, 105 Xisanhuan North Road, Haidian District, Beijing 100048, China
| | - Jianguo Zhao
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, ChinaUniversity of Chinese Academy of SciencesBeijing 100049, ChinaCollege of Life SciencesCapital Normal University, 105 Xisanhuan North Road, Haidian District, Beijing 100048, China
| |
Collapse
|
34
|
Sloan DB, Fields PD, Havird JC. Mitonuclear linkage disequilibrium in human populations. Proc Biol Sci 2016; 282:rspb.2015.1704. [PMID: 26378221 DOI: 10.1098/rspb.2015.1704] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
There is extensive evidence from model systems that disrupting associations between co-adapted mitochondrial and nuclear genotypes can lead to deleterious and even lethal consequences. While it is tempting to extrapolate from these observations and make inferences about the human-health effects of altering mitonuclear associations, the importance of such associations may vary greatly among species, depending on population genetics, demographic history and other factors. Remarkably, despite the extensive study of human population genetics, the statistical associations between nuclear and mitochondrial alleles remain largely uninvestigated. We analysed published population genomic data to test for signatures of historical selection to maintain mitonuclear associations, particularly those involving nuclear genes that encode mitochondrial-localized proteins (N-mt genes). We found that significant mitonuclear linkage disequilibrium (LD) exists throughout the human genome, but these associations were generally weak, which is consistent with the paucity of population genetic structure in humans. Although mitonuclear LD varied among genomic regions (with especially high levels on the X chromosome), N-mt genes were statistically indistinguishable from background levels, suggesting that selection on mitonuclear epistasis has not preferentially maintained associations involving this set of loci at a species-wide level. We discuss these findings in the context of the ongoing debate over mitochondrial replacement therapy.
Collapse
Affiliation(s)
- Daniel B Sloan
- Department of Biology, Colorado State University, Fort Collins, CO 80523, USA
| | - Peter D Fields
- Zoological Institute, University of Basel, Vesalgasse 1, Basel, 4051, Switzerland
| | - Justin C Havird
- Department of Biology, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
35
|
Abstract
The ability to reprogram somatic cells into induced pluripotent stem cells (iPSCs) using defined factors provides new tools for biomedical research. However, some iPSC clones display tumorigenic and immunogenic potential, thus raising concerns about their utility and safety in the clinical setting. Furthermore, variability in iPSC differentiation potential has also been described. Here we discuss whether these therapeutic obstacles are specific to transcription-factor-mediated reprogramming or inherent to every cellular reprogramming method. Finally, we address whether a better understanding of the mechanism underlying the reprogramming process might improve the fidelity of reprogramming and, therefore, the iPSC quality.
Collapse
Affiliation(s)
- Natalia Tapia
- Institute of Biomedicine of Valencia, Spanish National Research Council, Jaime Roig 11, 46010 Valencia, Spain.
| | - Hans R Schöler
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany; Medical Faculty, University of Münster, Domagkstraße 3, 48149 Münster, Germany.
| |
Collapse
|
36
|
Ebert AD, Diecke S, Chen IY, Wu JC. Reprogramming and transdifferentiation for cardiovascular development and regenerative medicine: where do we stand? EMBO Mol Med 2016; 7:1090-103. [PMID: 26183451 PMCID: PMC4568945 DOI: 10.15252/emmm.201504395] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Heart disease remains a leading cause of mortality and a major worldwide healthcare burden. Recent advances in stem cell biology have made it feasible to derive large quantities of cardiomyocytes for disease modeling, drug development, and regenerative medicine. The discoveries of reprogramming and transdifferentiation as novel biological processes have significantly contributed to this paradigm. This review surveys the means by which reprogramming and transdifferentiation can be employed to generate induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) and induced cardiomyocytes (iCMs). The application of these patient-specific cardiomyocytes for both in vitro disease modeling and in vivo therapies for various cardiovascular diseases will also be discussed. We propose that, with additional refinement, human disease-specific cardiomyocytes will allow us to significantly advance the understanding of cardiovascular disease mechanisms and accelerate the development of novel therapeutic options.
Collapse
Affiliation(s)
- Antje D Ebert
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, CA, USA Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Sebastian Diecke
- Max Delbrück Center, Berlin, Germany Berlin Institute of Health, Berlin, Germany
| | - Ian Y Chen
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, CA, USA Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, CA, USA Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
37
|
Cherry C, Thompson B, Saptarshi N, Wu J, Hoh J. 2016: A 'Mitochondria' Odyssey. Trends Mol Med 2016; 22:391-403. [PMID: 27151392 DOI: 10.1016/j.molmed.2016.03.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 03/21/2016] [Accepted: 03/22/2016] [Indexed: 12/16/2022]
Abstract
The integration of the many roles of mitochondria in cellular function and the contribution of mitochondrial dysfunction to disease are major areas of research. Within this realm, the roles of mitochondria in immune defense, epigenetics, and stem cell (SC) development have recently come into the spotlight. With new understanding, mitochondria may bring together these seemingly unrelated fields, a crucial process in treatment and prevention for various diseases. In this review we describe novel findings in these three arenas, discussing the significance of the interplay between mitochondria and the cell nucleus in response to environmental cues. While we optimistically anticipate that further research in these areas can have a profound impact on disease management, we also bring forth some of the key questions and challenges that remain.
Collapse
Affiliation(s)
- Catherine Cherry
- School of Medicine, Departments of Environmental Health Science and Ophthalmology, Yale University, New Haven, CT, USA
| | - Brian Thompson
- School of Medicine, Departments of Environmental Health Science and Ophthalmology, Yale University, New Haven, CT, USA
| | - Neil Saptarshi
- School of Medicine, Departments of Environmental Health Science and Ophthalmology, Yale University, New Haven, CT, USA
| | - Jianyu Wu
- School of Medicine, Departments of Environmental Health Science and Ophthalmology, Yale University, New Haven, CT, USA
| | - Josephine Hoh
- School of Medicine, Departments of Environmental Health Science and Ophthalmology, Yale University, New Haven, CT, USA.
| |
Collapse
|
38
|
Mitochondria in pluripotent stem cells: stemness regulators and disease targets. Curr Opin Genet Dev 2016; 38:1-7. [PMID: 26953561 DOI: 10.1016/j.gde.2016.02.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 02/01/2016] [Accepted: 02/05/2016] [Indexed: 11/23/2022]
Abstract
Beyond their canonical role in efficient ATP production through oxidative metabolism, mitochondria are increasingly recognized as critical in defining stem cell function and fate. Implicating a fundamental interplay within the epigenetics of eukaryotic cell systems, the integrity of mitochondria is found vital across the developmental/differentiation spectrum from securing pluripotency maintenance to informing organotypic decisions. This overview will discuss recent progress on examining the plasticity of mitochondria in enabling the execution of programming and reprogramming regimens, as well as the application of nuclear reprogramming and somatic cell nuclear transfer as rescue techniques to generate genetically and functionally corrected pluripotent stem cells from patients with mitochondrial DNA-based disease.
Collapse
|
39
|
Abdelwahid E, Kalvelyte A, Stulpinas A, de Carvalho KAT, Guarita-Souza LC, Foldes G. Stem cell death and survival in heart regeneration and repair. Apoptosis 2016; 21:252-68. [PMID: 26687129 PMCID: PMC5200890 DOI: 10.1007/s10495-015-1203-4] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cardiovascular diseases are major causes of mortality and morbidity. Cardiomyocyte apoptosis disrupts cardiac function and leads to cardiac decompensation and terminal heart failure. Delineating the regulatory signaling pathways that orchestrate cell survival in the heart has significant therapeutic implications. Cardiac tissue has limited capacity to regenerate and repair. Stem cell therapy is a successful approach for repairing and regenerating ischemic cardiac tissue; however, transplanted cells display very high death percentage, a problem that affects success of tissue regeneration. Stem cells display multipotency or pluripotency and undergo self-renewal, however these events are negatively influenced by upregulation of cell death machinery that induces the significant decrease in survival and differentiation signals upon cardiovascular injury. While efforts to identify cell types and molecular pathways that promote cardiac tissue regeneration have been productive, studies that focus on blocking the extensive cell death after transplantation are limited. The control of cell death includes multiple networks rather than one crucial pathway, which underlies the challenge of identifying the interaction between various cellular and biochemical components. This review is aimed at exploiting the molecular mechanisms by which stem cells resist death signals to develop into mature and healthy cardiac cells. Specifically, we focus on a number of factors that control death and survival of stem cells upon transplantation and ultimately affect cardiac regeneration. We also discuss potential survival enhancing strategies and how they could be meaningful in the design of targeted therapies that improve cardiac function.
Collapse
Affiliation(s)
- Eltyeb Abdelwahid
- Feinberg School of Medicine, Feinberg Cardiovascular Research Institute, Northwestern University, 303 E. Chicago Ave., Tarry 14-725, Chicago, IL, 60611, USA.
| | - Audrone Kalvelyte
- Department of Molecular Cell Biology, Vilnius University Institute of Biochemistry, Vilnius, Lithuania
| | - Aurimas Stulpinas
- Department of Molecular Cell Biology, Vilnius University Institute of Biochemistry, Vilnius, Lithuania
| | - Katherine Athayde Teixeira de Carvalho
- Cell Therapy and Biotechnology in Regenerative Medicine Research Group, Pequeno Príncipe Faculty, Pelé Pequeno Príncipe Institute, Curitiba, Paraná, 80250-200, Brazil
| | - Luiz Cesar Guarita-Souza
- Experimental Laboratory of Institute of Biological and Health Sciences of Pontifical Catholic University of Parana, Curitiba, Paraná, 80215-901, Brazil
| | - Gabor Foldes
- National Heart and Lung Institute, Imperial College London, Imperial Centre for Experimental and Translational Medicine, Du Cane Road, London, W12 0NN, UK
| |
Collapse
|
40
|
Dynamic stem cell states: naive to primed pluripotency in rodents and humans. Nat Rev Mol Cell Biol 2016; 17:155-69. [PMID: 26860365 DOI: 10.1038/nrm.2015.28] [Citation(s) in RCA: 447] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The molecular mechanisms and signalling pathways that regulate the in vitro preservation of distinct pluripotent stem cell configurations, and their induction in somatic cells by direct reprogramming, constitute a highly exciting area of research. In this Review, we integrate recent discoveries related to isolating unique naive and primed pluripotent stem cell states with altered functional and molecular characteristics, and from different species. We provide an overview of the pathways underlying pluripotent state transitions and interconversion in vitro and in vivo. We conclude by highlighting unresolved key questions, future directions and potential novel applications of such dynamic pluripotent cell states.
Collapse
|
41
|
Krzywanski DM, Moellering DR, Westbrook DG, Dunham-Snary KJ, Brown J, Bray AW, Feeley KP, Sammy MJ, Smith MR, Schurr TG, Vita JA, Ambalavanan N, Calhoun D, Dell'Italia L, Ballinger SW. Endothelial Cell Bioenergetics and Mitochondrial DNA Damage Differ in Humans Having African or West Eurasian Maternal Ancestry. ACTA ACUST UNITED AC 2016; 9:26-36. [PMID: 26787433 DOI: 10.1161/circgenetics.115.001308] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 01/13/2016] [Indexed: 01/09/2023]
Abstract
BACKGROUND We hypothesized that endothelial cells having distinct mitochondrial genetic backgrounds would show variation in mitochondrial function and oxidative stress markers concordant with known differential cardiovascular disease susceptibilities. To test this hypothesis, mitochondrial bioenergetics were determined in endothelial cells from healthy individuals with African versus European maternal ancestries. METHODS AND RESULTS Bioenergetics and mitochondrial DNA (mtDNA) damage were assessed in single-donor human umbilical vein endothelial cells belonging to mtDNA haplogroups H and L, representing West Eurasian and African maternal ancestries, respectively. Human umbilical vein endothelial cells from haplogroup L used less oxygen for ATP production and had increased levels of mtDNA damage compared with those in haplogroup H. Differences in bioenergetic capacity were also observed in that human umbilical vein endothelial cells belonging to haplogroup L had decreased maximal bioenergetic capacities compared with haplogroup H. Analysis of peripheral blood mononuclear cells from age-matched healthy controls with West Eurasian or African maternal ancestries showed that haplogroups sharing an A to G mtDNA mutation at nucleotide pair 10398 had increased mtDNA damage compared with those lacking this mutation. Further study of angiographically proven patients with coronary artery disease and age-matched healthy controls revealed that mtDNA damage was associated with vascular function and remodeling and that age of disease onset was later in individuals from haplogroups lacking the A to G mutation at nucleotide pair 10398. CONCLUSIONS Differences in mitochondrial bioenergetics and mtDNA damage associated with maternal ancestry may contribute to endothelial dysfunction and vascular disease.
Collapse
Affiliation(s)
- David M Krzywanski
- From the Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport (D.M.K.); Department of Nutrition Sciences (D.R.M.), Center for Free Radical Biology and Medicine (D.R.M., D.G.W., K.J.D.-S., J.B., A.W.B., K.P.F., M.J.S., M.R.S., L.D., S.W.B.), Division of Molecular and Cellular Pathology, Department of Pathology (D.G.W., J.B., A.W.B., K.P.F., M.J.S., M.R.S., S.W.B.), Department of Pediatrics (N.A.), Department of Medicine (D.C., L.D.), University of Alabama at Birmingham; Department of Medicine, Queen's University, Kingston, Ontario, Canada (K.J.D.-S.); Department of Anthropology, University of Pennsylvania, Philadelphia (T.G.S.); and Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (J.A.V.)
| | - Douglas R Moellering
- From the Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport (D.M.K.); Department of Nutrition Sciences (D.R.M.), Center for Free Radical Biology and Medicine (D.R.M., D.G.W., K.J.D.-S., J.B., A.W.B., K.P.F., M.J.S., M.R.S., L.D., S.W.B.), Division of Molecular and Cellular Pathology, Department of Pathology (D.G.W., J.B., A.W.B., K.P.F., M.J.S., M.R.S., S.W.B.), Department of Pediatrics (N.A.), Department of Medicine (D.C., L.D.), University of Alabama at Birmingham; Department of Medicine, Queen's University, Kingston, Ontario, Canada (K.J.D.-S.); Department of Anthropology, University of Pennsylvania, Philadelphia (T.G.S.); and Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (J.A.V.)
| | - David G Westbrook
- From the Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport (D.M.K.); Department of Nutrition Sciences (D.R.M.), Center for Free Radical Biology and Medicine (D.R.M., D.G.W., K.J.D.-S., J.B., A.W.B., K.P.F., M.J.S., M.R.S., L.D., S.W.B.), Division of Molecular and Cellular Pathology, Department of Pathology (D.G.W., J.B., A.W.B., K.P.F., M.J.S., M.R.S., S.W.B.), Department of Pediatrics (N.A.), Department of Medicine (D.C., L.D.), University of Alabama at Birmingham; Department of Medicine, Queen's University, Kingston, Ontario, Canada (K.J.D.-S.); Department of Anthropology, University of Pennsylvania, Philadelphia (T.G.S.); and Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (J.A.V.)
| | - Kimberly J Dunham-Snary
- From the Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport (D.M.K.); Department of Nutrition Sciences (D.R.M.), Center for Free Radical Biology and Medicine (D.R.M., D.G.W., K.J.D.-S., J.B., A.W.B., K.P.F., M.J.S., M.R.S., L.D., S.W.B.), Division of Molecular and Cellular Pathology, Department of Pathology (D.G.W., J.B., A.W.B., K.P.F., M.J.S., M.R.S., S.W.B.), Department of Pediatrics (N.A.), Department of Medicine (D.C., L.D.), University of Alabama at Birmingham; Department of Medicine, Queen's University, Kingston, Ontario, Canada (K.J.D.-S.); Department of Anthropology, University of Pennsylvania, Philadelphia (T.G.S.); and Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (J.A.V.)
| | - Jamelle Brown
- From the Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport (D.M.K.); Department of Nutrition Sciences (D.R.M.), Center for Free Radical Biology and Medicine (D.R.M., D.G.W., K.J.D.-S., J.B., A.W.B., K.P.F., M.J.S., M.R.S., L.D., S.W.B.), Division of Molecular and Cellular Pathology, Department of Pathology (D.G.W., J.B., A.W.B., K.P.F., M.J.S., M.R.S., S.W.B.), Department of Pediatrics (N.A.), Department of Medicine (D.C., L.D.), University of Alabama at Birmingham; Department of Medicine, Queen's University, Kingston, Ontario, Canada (K.J.D.-S.); Department of Anthropology, University of Pennsylvania, Philadelphia (T.G.S.); and Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (J.A.V.)
| | - Alexander W Bray
- From the Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport (D.M.K.); Department of Nutrition Sciences (D.R.M.), Center for Free Radical Biology and Medicine (D.R.M., D.G.W., K.J.D.-S., J.B., A.W.B., K.P.F., M.J.S., M.R.S., L.D., S.W.B.), Division of Molecular and Cellular Pathology, Department of Pathology (D.G.W., J.B., A.W.B., K.P.F., M.J.S., M.R.S., S.W.B.), Department of Pediatrics (N.A.), Department of Medicine (D.C., L.D.), University of Alabama at Birmingham; Department of Medicine, Queen's University, Kingston, Ontario, Canada (K.J.D.-S.); Department of Anthropology, University of Pennsylvania, Philadelphia (T.G.S.); and Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (J.A.V.)
| | - Kyle P Feeley
- From the Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport (D.M.K.); Department of Nutrition Sciences (D.R.M.), Center for Free Radical Biology and Medicine (D.R.M., D.G.W., K.J.D.-S., J.B., A.W.B., K.P.F., M.J.S., M.R.S., L.D., S.W.B.), Division of Molecular and Cellular Pathology, Department of Pathology (D.G.W., J.B., A.W.B., K.P.F., M.J.S., M.R.S., S.W.B.), Department of Pediatrics (N.A.), Department of Medicine (D.C., L.D.), University of Alabama at Birmingham; Department of Medicine, Queen's University, Kingston, Ontario, Canada (K.J.D.-S.); Department of Anthropology, University of Pennsylvania, Philadelphia (T.G.S.); and Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (J.A.V.)
| | - Melissa J Sammy
- From the Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport (D.M.K.); Department of Nutrition Sciences (D.R.M.), Center for Free Radical Biology and Medicine (D.R.M., D.G.W., K.J.D.-S., J.B., A.W.B., K.P.F., M.J.S., M.R.S., L.D., S.W.B.), Division of Molecular and Cellular Pathology, Department of Pathology (D.G.W., J.B., A.W.B., K.P.F., M.J.S., M.R.S., S.W.B.), Department of Pediatrics (N.A.), Department of Medicine (D.C., L.D.), University of Alabama at Birmingham; Department of Medicine, Queen's University, Kingston, Ontario, Canada (K.J.D.-S.); Department of Anthropology, University of Pennsylvania, Philadelphia (T.G.S.); and Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (J.A.V.)
| | - Matthew R Smith
- From the Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport (D.M.K.); Department of Nutrition Sciences (D.R.M.), Center for Free Radical Biology and Medicine (D.R.M., D.G.W., K.J.D.-S., J.B., A.W.B., K.P.F., M.J.S., M.R.S., L.D., S.W.B.), Division of Molecular and Cellular Pathology, Department of Pathology (D.G.W., J.B., A.W.B., K.P.F., M.J.S., M.R.S., S.W.B.), Department of Pediatrics (N.A.), Department of Medicine (D.C., L.D.), University of Alabama at Birmingham; Department of Medicine, Queen's University, Kingston, Ontario, Canada (K.J.D.-S.); Department of Anthropology, University of Pennsylvania, Philadelphia (T.G.S.); and Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (J.A.V.)
| | - Theodore G Schurr
- From the Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport (D.M.K.); Department of Nutrition Sciences (D.R.M.), Center for Free Radical Biology and Medicine (D.R.M., D.G.W., K.J.D.-S., J.B., A.W.B., K.P.F., M.J.S., M.R.S., L.D., S.W.B.), Division of Molecular and Cellular Pathology, Department of Pathology (D.G.W., J.B., A.W.B., K.P.F., M.J.S., M.R.S., S.W.B.), Department of Pediatrics (N.A.), Department of Medicine (D.C., L.D.), University of Alabama at Birmingham; Department of Medicine, Queen's University, Kingston, Ontario, Canada (K.J.D.-S.); Department of Anthropology, University of Pennsylvania, Philadelphia (T.G.S.); and Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (J.A.V.)
| | - Joseph A Vita
- From the Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport (D.M.K.); Department of Nutrition Sciences (D.R.M.), Center for Free Radical Biology and Medicine (D.R.M., D.G.W., K.J.D.-S., J.B., A.W.B., K.P.F., M.J.S., M.R.S., L.D., S.W.B.), Division of Molecular and Cellular Pathology, Department of Pathology (D.G.W., J.B., A.W.B., K.P.F., M.J.S., M.R.S., S.W.B.), Department of Pediatrics (N.A.), Department of Medicine (D.C., L.D.), University of Alabama at Birmingham; Department of Medicine, Queen's University, Kingston, Ontario, Canada (K.J.D.-S.); Department of Anthropology, University of Pennsylvania, Philadelphia (T.G.S.); and Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (J.A.V.)
| | - Namasivayam Ambalavanan
- From the Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport (D.M.K.); Department of Nutrition Sciences (D.R.M.), Center for Free Radical Biology and Medicine (D.R.M., D.G.W., K.J.D.-S., J.B., A.W.B., K.P.F., M.J.S., M.R.S., L.D., S.W.B.), Division of Molecular and Cellular Pathology, Department of Pathology (D.G.W., J.B., A.W.B., K.P.F., M.J.S., M.R.S., S.W.B.), Department of Pediatrics (N.A.), Department of Medicine (D.C., L.D.), University of Alabama at Birmingham; Department of Medicine, Queen's University, Kingston, Ontario, Canada (K.J.D.-S.); Department of Anthropology, University of Pennsylvania, Philadelphia (T.G.S.); and Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (J.A.V.)
| | - David Calhoun
- From the Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport (D.M.K.); Department of Nutrition Sciences (D.R.M.), Center for Free Radical Biology and Medicine (D.R.M., D.G.W., K.J.D.-S., J.B., A.W.B., K.P.F., M.J.S., M.R.S., L.D., S.W.B.), Division of Molecular and Cellular Pathology, Department of Pathology (D.G.W., J.B., A.W.B., K.P.F., M.J.S., M.R.S., S.W.B.), Department of Pediatrics (N.A.), Department of Medicine (D.C., L.D.), University of Alabama at Birmingham; Department of Medicine, Queen's University, Kingston, Ontario, Canada (K.J.D.-S.); Department of Anthropology, University of Pennsylvania, Philadelphia (T.G.S.); and Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (J.A.V.)
| | - Louis Dell'Italia
- From the Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport (D.M.K.); Department of Nutrition Sciences (D.R.M.), Center for Free Radical Biology and Medicine (D.R.M., D.G.W., K.J.D.-S., J.B., A.W.B., K.P.F., M.J.S., M.R.S., L.D., S.W.B.), Division of Molecular and Cellular Pathology, Department of Pathology (D.G.W., J.B., A.W.B., K.P.F., M.J.S., M.R.S., S.W.B.), Department of Pediatrics (N.A.), Department of Medicine (D.C., L.D.), University of Alabama at Birmingham; Department of Medicine, Queen's University, Kingston, Ontario, Canada (K.J.D.-S.); Department of Anthropology, University of Pennsylvania, Philadelphia (T.G.S.); and Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (J.A.V.)
| | - Scott W Ballinger
- From the Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport (D.M.K.); Department of Nutrition Sciences (D.R.M.), Center for Free Radical Biology and Medicine (D.R.M., D.G.W., K.J.D.-S., J.B., A.W.B., K.P.F., M.J.S., M.R.S., L.D., S.W.B.), Division of Molecular and Cellular Pathology, Department of Pathology (D.G.W., J.B., A.W.B., K.P.F., M.J.S., M.R.S., S.W.B.), Department of Pediatrics (N.A.), Department of Medicine (D.C., L.D.), University of Alabama at Birmingham; Department of Medicine, Queen's University, Kingston, Ontario, Canada (K.J.D.-S.); Department of Anthropology, University of Pennsylvania, Philadelphia (T.G.S.); and Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (J.A.V.).
| |
Collapse
|
42
|
Understanding Stem Cell Immunogenicity in Therapeutic Applications. Trends Immunol 2015; 37:5-16. [PMID: 26687737 DOI: 10.1016/j.it.2015.11.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 11/11/2015] [Accepted: 11/13/2015] [Indexed: 12/14/2022]
Abstract
Stem cells and their differentiated progeny offer great hope for treating disease by providing an unlimited source of cells for repairing or replacing damaged tissue. Initial studies suggested that, unlike 'normal' transplants, specific characteristics of stem cells enabled them to avoid immune attack. However, recent findings have revealed that the immunogenicity of stem cells may have been underestimated. Here, we review the current understanding of the mechanisms of immune recognition associated with stem cell immunogenicity, and discuss the relevance of reprogramming and differentiation strategies used to generate cells or tissue from stem cells for implantation in eliciting an immune response. We examine the effectiveness of current strategies for minimising immune attack in light of our experience in the transplantation field and, in this context, outline important challenges moving forward.
Collapse
|
43
|
Breckwoldt K, Weinberger F, Eschenhagen T. Heart regeneration. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:1749-59. [PMID: 26597703 DOI: 10.1016/j.bbamcr.2015.11.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 11/06/2015] [Accepted: 11/12/2015] [Indexed: 01/14/2023]
Abstract
Regenerating an injured heart holds great promise for millions of patients suffering from heart diseases. Since the human heart has very limited regenerative capacity, this is a challenging task. Numerous strategies aiming to improve heart function have been developed. In this review we focus on approaches intending to replace damaged heart muscle by new cardiomyocytes. Different strategies for the production of cardiomyocytes from human embryonic stem cells or human induced pluripotent stem cells, by direct reprogramming and induction of cardiomyocyte proliferation are discussed regarding their therapeutic potential and respective advantages and disadvantages. Furthermore, different methods for the transplantation of pluripotent stem cell-derived cardiomyocytes are described and their clinical perspectives are discussed. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.
Collapse
Affiliation(s)
- Kaja Breckwoldt
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany
| | - Florian Weinberger
- Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA
| | - Thomas Eschenhagen
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany.
| |
Collapse
|
44
|
Dunham-Snary KJ, Ballinger SW. GENETICS. Mitochondrial-nuclear DNA mismatch matters. Science 2015; 349:1449-50. [PMID: 26404813 DOI: 10.1126/science.aac5271] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
| | - Scott W Ballinger
- Department of Pathology, Division of Molecular and Cellular Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA. Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
45
|
Yamada M, Byrne J, Egli D. From cloned frogs to patient matched stem cells: induced pluripotency or somatic cell nuclear transfer? Curr Opin Genet Dev 2015; 34:29-34. [PMID: 26282611 DOI: 10.1016/j.gde.2015.06.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 06/04/2015] [Accepted: 06/16/2015] [Indexed: 01/24/2023]
Abstract
Nuclear transfer has seen a remarkable comeback in the past few years. Three groups have independently reported the derivation of stem cell lines by somatic cell nuclear transfer, from either adult, neonatal or fetal cells. Though the ability of human oocytes to reprogram somatic cells to stem cells had long been anticipated, success did not arrive on a straightforward path. Little was known about human oocyte biology, and nuclear transfer protocols developed in animals required key changes to become effective with human eggs. By overcoming these challenges, human nuclear transfer research has contributed to a greater understanding of oocyte biology, provided a point of reference for the comparison of induced pluripotent stem cells, and delivered a method for the generation of personalized stem cells with therapeutic potential.
Collapse
Affiliation(s)
- Mitsutoshi Yamada
- The New York Stem Cell Foundation Research Institute, New York, NY 10032, USA
| | - James Byrne
- The Eli and Edythe Broad Center of Regenerative Medicine & Regenerative Medicine, CA 90095, USA
| | - Dieter Egli
- The New York Stem Cell Foundation Research Institute, New York, NY 10032, USA; Naomi Berrie Diabetes Center, Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY, USA.
| |
Collapse
|
46
|
Neofytou E, O'Brien CG, Couture LA, Wu JC. Hurdles to clinical translation of human induced pluripotent stem cells. J Clin Invest 2015; 125:2551-7. [PMID: 26132109 DOI: 10.1172/jci80575] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Human pluripotent stem cells are known to have the capacity to renew indefinitely, being intrinsically able to differentiate into many different cell types. These characteristics have generated tremendous enthusiasm about the potential applications of these cells in regenerative medicine. However, major challenges remain with the development and testing of novel experimental stem cell therapeutics in the field. In this Review, we focus on the nature of the preclinical challenges and discuss potential solutions that could help overcome them. Furthermore, we discuss the use of allogeneic versus autologous stem cell products, including a review of their respective advantages and disadvantages, major clinical requirements, quality standards, time lines, and costs of clinical grade development.
Collapse
|
47
|
Gemmell N, Wolff JN. Mitochondrial replacement therapy: Cautiously replace the master manipulator. Bioessays 2015; 37:584-5. [PMID: 25728033 DOI: 10.1002/bies.201500008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Revised: 02/09/2015] [Accepted: 02/10/2015] [Indexed: 11/10/2022]
Affiliation(s)
- Neil Gemmell
- Allan Wilson Centre, Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Jonci N Wolff
- School of Biological Sciences, Monash University, Clayton, Australia
| |
Collapse
|
48
|
Simonson OE, Domogatskaya A, Volchkov P, Rodin S. The safety of human pluripotent stem cells in clinical treatment. Ann Med 2015; 47:370-80. [PMID: 26140342 DOI: 10.3109/07853890.2015.1051579] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) have practically unlimited proliferation potential and a capability to differentiate into any cell type in the human body. Since the first derivation in 1998, they have been an attractive source of cells for regenerative medicine. Numerous ethical, technological, and regulatory complications have been hampering hPSC use in clinical applications. Human embryonic stem cells (ESCs), parthenogenetic human ESCs, human nuclear transfer ESCs, and induced pluripotent stem cells are four types of hPSCs that are different in many clinically relevant features such as propensity to epigenetic abnormalities, generation methods, and ability for development of autologous cell lines. Propensity to genetic mutations and tumorigenicity are common features of all pluripotent cells that complicate hPSC-based therapies. Several recent advances in methods of derivation, culturing, and monitoring of hPSCs have addressed many ethical concerns and technological challenges in development of clinical-grade hPSC lines. Generation of banks of such lines may be useful to minimize immune rejection of hPSC-derived allografts. In this review, we discuss different sources of hPSCs available at the moment, various safety risks associated with them, and possible solutions for successful use of hPSCs in the clinic. We also discuss ongoing clinical trials of hPSC-based treatments.
Collapse
Affiliation(s)
- Oscar E Simonson
- a Division of Cardiothoracic Surgery and Anesthesiology, Department of Molecular Medicine and Surgery , Karolinska Institutet, Karolinska University Hospital , 171 77 Stockholm , Sweden
| | | | | | | |
Collapse
|