1
|
Romito E, Battistella I, Plakhova V, Paplekaj A, Forastieri C, Toffolo E, Musio C, Conti L, Battaglioli E, Rusconi F. A comprehensive protocol for efficient differentiation of human NPCs into electrically competent neurons. J Neurosci Methods 2024; 410:110225. [PMID: 39053772 DOI: 10.1016/j.jneumeth.2024.110225] [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: 04/08/2024] [Revised: 05/29/2024] [Accepted: 07/19/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND The study of neurons is fundamental to unraveling the complexities of the nervous system. Primary neuronal cultures from rodents have long been a cornerstone of experimental studies, yet limitations related to their non-human nature and ethical concerns have prompted the development of alternatives. In recent years, the derivation of neurons from human-induced pluripotent stem cells (hiPSCs) has emerged as a powerful option, offering a scalable source of cells for diverse applications. Neural progenitor cells (NPCs) derived from hiPSCs can be efficiently differentiated into functional neurons, providing a platform to study human neural physiology and pathology in vitro. However, challenges persist in achieving consistent and reproducible outcomes across experimental settings. COMPARISON WITH EXISTING METHODS Our aim is to provide a step-by-step methodological protocol, augmenting existing procedures with additional instructions and parameters, to guide researchers in achieving reproducible results. METHODS AND RESULTS We outline procedures for the differentiation of hiPSC-derived NPCs into electrically competent neurons, encompassing initial cell density, morphology, maintenance, and differentiation. We also describe the analysis of specific markers for assessing neuronal phenotype, along with electrophysiological analysis to evaluate biophysical properties of neuronal excitability. Additionally, we conduct a comparative analysis of three different chemical methods-KCl, N-methyl-D-aspartate (NMDA), and bicuculline-to induce neuronal depolarization and assess their effects on the induction of both fast and slow post-translational, transcriptional, and post-transcriptional responses. CONCLUSION Our protocol provides clear instructions for generating reliable human neuronal cultures with defined electrophysiological properties to investigate neuronal differentiation and model diseases in vitro.
Collapse
Affiliation(s)
- Elena Romito
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Via Fratelli Cervi, 93, Segrate, Milan 20054, Italy
| | - Ingrid Battistella
- Department of Cellular, Computational and Integrative Biology - CIBIO, Università degli Studi di Trento, Via Sommarive, 9, Trento 38123, Italy
| | - Vera Plakhova
- Institute of Biophysics (IBF), National Research Council (CNR), Trento Unit, & LabSSAH, Bruno Kessler Foundation (FBK), Via Sommarive, 18, Trento 38123, Italy
| | - Arteda Paplekaj
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Via Fratelli Cervi, 93, Segrate, Milan 20054, Italy
| | - Chiara Forastieri
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Via Fratelli Cervi, 93, Segrate, Milan 20054, Italy
| | - Emanuela Toffolo
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Via Fratelli Cervi, 93, Segrate, Milan 20054, Italy
| | - Carlo Musio
- Institute of Biophysics (IBF), National Research Council (CNR), Trento Unit, & LabSSAH, Bruno Kessler Foundation (FBK), Via Sommarive, 18, Trento 38123, Italy
| | - Luciano Conti
- Department of Cellular, Computational and Integrative Biology - CIBIO, Università degli Studi di Trento, Via Sommarive, 9, Trento 38123, Italy.
| | - Elena Battaglioli
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Via Fratelli Cervi, 93, Segrate, Milan 20054, Italy
| | - Francesco Rusconi
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Via Fratelli Cervi, 93, Segrate, Milan 20054, Italy.
| |
Collapse
|
2
|
Seo J, Saha S, Brown ME. The past, present, and future promise of pluripotent stem cells. JOURNAL OF IMMUNOLOGY AND REGENERATIVE MEDICINE 2024; 22-23:100077. [PMID: 38706532 PMCID: PMC11065261 DOI: 10.1016/j.regen.2024.100077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2024]
Affiliation(s)
| | | | - Matthew E. Brown
- University of Wisconsin-Madison, School of Medicine and Public Health, Department of Surgery, Division of Transplantation, 600 Highland Avenue, Madison, WI, 53792, United States
| |
Collapse
|
3
|
Şen B, Balcı‐Peynircioğlu B. Cellular models in autoinflammatory disease research. Clin Transl Immunology 2024; 13:e1481. [PMID: 38213819 PMCID: PMC10784111 DOI: 10.1002/cti2.1481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/12/2023] [Accepted: 12/12/2023] [Indexed: 01/13/2024] Open
Abstract
Systemic autoinflammatory diseases are a heterogeneous group of rare genetic disorders caused by dysregulation of the innate immune system. Understanding the complex mechanisms underlying these conditions is critical for developing effective treatments. Cellular models are essential for identifying new conditions and studying their pathogenesis. Traditionally, these studies have used primary cells and cell lines of disease-relevant cell types, although newer induced pluripotent stem cell (iPSC)-based models might have unique advantages. In this review, we discuss the three cellular models used in autoinflammatory disease research, their strengths and weaknesses, and their applications to inform future research in the field.
Collapse
Affiliation(s)
- Başak Şen
- Department of Medical BiologyHacettepe University Faculty of Medicine, SıhhiyeAnkaraTurkey
| | | |
Collapse
|
4
|
Wang Y, Xia Y, Kou L, Yin S, Chi X, Li J, Sun Y, Wu J, Zhou Q, Zou W, Jin Z, Huang J, Xiong N, Wang T. Astrocyte-to-neuron reprogramming and crosstalk in the treatment of Parkinson's disease. Neurobiol Dis 2023:106224. [PMID: 37433411 DOI: 10.1016/j.nbd.2023.106224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/24/2023] [Accepted: 07/07/2023] [Indexed: 07/13/2023] Open
Abstract
Parkinson's disease (PD) is currently the fastest growing disabling neurological disorder worldwide, with motor and non-motor symptoms being its main clinical manifestations. The primary pathological features include a reduction in the number of dopaminergic neurons in the substantia nigra and decrease in dopamine levels in the nigrostriatal pathway. Existing treatments only alleviate clinical symptoms and do not stop disease progression; slowing down the loss of dopaminergic neurons and stimulating their regeneration are emerging therapies. Preclinical studies have demonstrated that transplantation of dopamine cells generated from human embryonic or induced pluripotent stem cells can restore the loss of dopamine. However, the application of cell transplantation is limited owing to ethical controversies and the restricted source of cells. Until recently, the reprogramming of astrocytes to replenish lost dopaminergic neurons has provided a promising alternative therapy for PD. In addition, repair of mitochondrial perturbations, clearance of damaged mitochondria in astrocytes, and control of astrocyte inflammation may be extensively neuroprotective and beneficial against chronic neuroinflammation in PD. Therefore, this review primarily focuses on the progress and remaining issues in astrocyte reprogramming using transcription factors (TFs) and miRNAs, as well as exploring possible new targets for treating PD by repairing astrocytic mitochondria and reducing astrocytic inflammation.
Collapse
Affiliation(s)
- Yiming Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yun Xia
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Liang Kou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Sijia Yin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaosa Chi
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jingwen Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yadi Sun
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jiawei Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qiulu Zhou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wenkai Zou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zongjie Jin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jinsha Huang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Nian Xiong
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Tao Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
5
|
In Search of the Holy Grail: Stem Cell Therapy as a Novel Treatment of Heart Failure with Preserved Ejection Fraction. Int J Mol Sci 2023; 24:ijms24054903. [PMID: 36902332 PMCID: PMC10003723 DOI: 10.3390/ijms24054903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/20/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023] Open
Abstract
Heart failure, a leading cause of hospitalizations and deaths, is a major clinical problem. In recent years, the increasing incidence of heart failure with preserved ejection fraction (HFpEF) has been observed. Despite extensive research, there is no efficient treatment for HFpEF available. However, a growing body of evidence suggests stem cell transplantation, due to its immunomodulatory effect, may decrease fibrosis and improve microcirculation and therefore, could be the first etiology-based therapy of the disease. In this review, we explain the complex pathogenesis of HFpEF, delineate the beneficial effects of stem cells in cardiovascular therapy, and summarize the current knowledge concerning cell therapy in diastolic dysfunction. Furthermore, we identify outstanding knowledge gaps that may indicate directions for future clinical studies.
Collapse
|
6
|
Giovenale AMG, Ruotolo G, Soriano AA, Turco EM, Rotundo G, Casamassa A, D’Anzi A, Vescovi AL, Rosati J. Deepening the understanding of CNVs on chromosome 15q11-13 by using hiPSCs: An overview. Front Cell Dev Biol 2023; 10:1107881. [PMID: 36684422 PMCID: PMC9852989 DOI: 10.3389/fcell.2022.1107881] [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: 11/25/2022] [Accepted: 12/16/2022] [Indexed: 01/09/2023] Open
Abstract
The human α7 neuronal nicotinic acetylcholine receptor gene (CHRNA7) is widely expressed in the central and peripheral nervous systems. This receptor is implicated in both brain development and adult neurogenesis thanks to its ability to mediate acetylcholine stimulus (Ach). Copy number variations (CNVs) of CHRNA7 gene have been identified in humans and are genetically linked to cognitive impairments associated with multiple disorders, including schizophrenia, bipolar disorder, epilepsy, Alzheimer's disease, and others. Currently, α7 receptor analysis has been commonly performed in animal models due to the impossibility of direct investigation of the living human brain. But the use of model systems has shown that there are very large differences between humans and mice when researchers must study the CNVs and, in particular, the CNV of chromosome 15q13.3 where the CHRNA7 gene is present. In fact, human beings present genomic alterations as well as the presence of genes of recent origin that are not present in other model systems as well as they show a very heterogeneous symptomatology that is associated with both their genetic background and the environment where they live. To date, the induced pluripotent stem cells, obtained from patients carrying CNV in CHRNA7 gene, are a good in vitro model for studying the association of the α7 receptor to human diseases. In this review, we will outline the current state of hiPSCs technology applications in neurological diseases caused by CNVs in CHRNA7 gene. Furthermore, we will discuss some weaknesses that emerge from the overall analysis of the published articles.
Collapse
Affiliation(s)
- Angela Maria Giada Giovenale
- Cellular Reprogramming Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy,Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Giorgia Ruotolo
- Cellular Reprogramming Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy,Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Amata Amy Soriano
- Cellular Reprogramming Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Elisa Maria Turco
- Cellular Reprogramming Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Giovannina Rotundo
- Cellular Reprogramming Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Alessia Casamassa
- Cellular Reprogramming Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Angela D’Anzi
- Cellular Reprogramming Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Angelo Luigi Vescovi
- Cellular Reprogramming Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy,Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy,*Correspondence: Jessica Rosati, ; Angelo Luigi Vescovi,
| | - Jessica Rosati
- Cellular Reprogramming Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy,*Correspondence: Jessica Rosati, ; Angelo Luigi Vescovi,
| |
Collapse
|
7
|
Generation of CD34 +CD43 + Hematopoietic Progenitors to Induce Thymocytes from Human Pluripotent Stem Cells. Cells 2022; 11:cells11244046. [PMID: 36552810 PMCID: PMC9777438 DOI: 10.3390/cells11244046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/15/2022] [Accepted: 11/18/2022] [Indexed: 12/15/2022] Open
Abstract
Immunotherapy using primary T cells has revolutionized medical care in some pathologies in recent years, but limitations associated to challenging cell genome edition, insufficient cell number production, the use of only autologous cells, and the lack of product standardization have limited its clinical use. The alternative use of T cells generated in vitro from human pluripotent stem cells (hPSCs) offers great advantages by providing a self-renewing source of T cells that can be readily genetically modified and facilitate the use of standardized universal off-the-shelf allogeneic cell products and rapid clinical access. However, despite their potential, a better understanding of the feasibility and functionality of T cells differentiated from hPSCs is necessary before moving into clinical settings. In this study, we generated human-induced pluripotent stem cells from T cells (T-iPSCs), allowing for the preservation of already recombined TCR, with the same properties as human embryonic stem cells (hESCs). Based on these cells, we differentiated, with high efficiency, hematopoietic progenitor stem cells (HPSCs) capable of self-renewal and differentiation into any cell blood type, in addition to DN3a thymic progenitors from several T-iPSC lines. In order to better comprehend the differentiation, we analyzed the transcriptomic profiles of the different cell types and demonstrated that HPSCs differentiated from hiPSCs had very similar profiles to cord blood hematopoietic stem cells (HSCs). Furthermore, differentiated T-cell progenitors had a similar profile to thymocytes at the DN3a stage of thymic lymphopoiesis. Therefore, utilizing this approach, we were able to regenerate precursors of therapeutic human T cells in order to potentially treat a wide range of diseases.
Collapse
|
8
|
He X, Liang J, Paul C, Huang W, Dutta S, Wang Y. Advances in Cellular Reprogramming-Based Approaches for Heart Regenerative Repair. Cells 2022; 11:3914. [PMID: 36497171 PMCID: PMC9740402 DOI: 10.3390/cells11233914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/18/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Continuous loss of cardiomyocytes (CMs) is one of the fundamental characteristics of many heart diseases, which eventually can lead to heart failure. Due to the limited proliferation ability of human adult CMs, treatment efficacy has been limited in terms of fully repairing damaged hearts. It has been shown that cell lineage conversion can be achieved by using cell reprogramming approaches, including human induced pluripotent stem cells (hiPSCs), providing a promising therapeutic for regenerative heart medicine. Recent studies using advanced cellular reprogramming-based techniques have also contributed some new strategies for regenerative heart repair. In this review, hiPSC-derived cell therapeutic methods are introduced, and the clinical setting challenges (maturation, engraftment, immune response, scalability, and tumorigenicity), with potential solutions, are discussed. Inspired by the iPSC reprogramming, the approaches of direct cell lineage conversion are merging, such as induced cardiomyocyte-like cells (iCMs) and induced cardiac progenitor cells (iCPCs) derived from fibroblasts, without induction of pluripotency. The studies of cellular and molecular pathways also reveal that epigenetic resetting is the essential mechanism of reprogramming and lineage conversion. Therefore, CRISPR techniques that can be repurposed for genomic or epigenetic editing become attractive approaches for cellular reprogramming. In addition, viral and non-viral delivery strategies that are utilized to achieve CM reprogramming will be introduced, and the therapeutic effects of iCMs or iCPCs on myocardial infarction will be compared. After the improvement of reprogramming efficiency by developing new techniques, reprogrammed iCPCs or iCMs will provide an alternative to hiPSC-based approaches for regenerative heart therapies, heart disease modeling, and new drug screening.
Collapse
Affiliation(s)
- Xingyu He
- Department of Pathology & Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Jialiang Liang
- Department of Pathology & Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Christian Paul
- Department of Pathology & Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Wei Huang
- Department of Pathology & Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Suchandrima Dutta
- Department of Internal Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Yigang Wang
- Department of Pathology & Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| |
Collapse
|
9
|
Arslanova A, Shafaattalab S, Ye K, Asghari P, Lin L, Kim B, Roston TM, Hove-Madsen L, Van Petegem F, Sanatani S, Moore E, Lynn F, Søndergaard M, Luo Y, Chen SRW, Tibbits GF. Using hiPSC-CMs to Examine Mechanisms of Catecholaminergic Polymorphic Ventricular Tachycardia. Curr Protoc 2021; 1:e320. [PMID: 34958715 DOI: 10.1002/cpz1.320] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Catecholaminergic polymorphic ventricular tachycardia (CPVT) is a potentially lethal inherited cardiac arrhythmia condition, triggered by physical or acute emotional stress, that predominantly expresses early in life. Gain-of-function mutations in the cardiac ryanodine receptor gene (RYR2) account for the majority of CPVT cases, causing substantial disruption of intracellular calcium (Ca2+ ) homeostasis particularly during the periods of β-adrenergic receptor stimulation. However, the highly variable penetrance, patient outcomes, and drug responses observed in clinical practice remain unexplained, even for patients with well-established founder RyR2 mutations. Therefore, investigation of the electrophysiological consequences of CPVT-causing RyR2 mutations is crucial to better understand the pathophysiology of the disease. The development of strategies for reprogramming human somatic cells to human induced pluripotent stem cells (hiPSCs) has provided a unique opportunity to study inherited arrhythmias, due to the ability of hiPSCs to differentiate down a cardiac lineage. Employment of genome editing enables generation of disease-specific cell lines from healthy and diseased patient-derived hiPSCs, which subsequently can be differentiated into cardiomyocytes. This paper describes the means for establishing an hiPSC-based model of CPVT in order to recapitulate the disease phenotype in vitro and investigate underlying pathophysiological mechanisms. The framework of this approach has the potential to contribute to disease modeling and personalized medicine using hiPSC-derived cardiomyocytes. © 2021 Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Alia Arslanova
- Cellular and Regenerative Medicine Centre, British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada.,Molecular Cardiac Physiology Group, Departments of Biomedical Physiology and Kinesiology and Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Sanam Shafaattalab
- Cellular and Regenerative Medicine Centre, British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada.,Molecular Cardiac Physiology Group, Departments of Biomedical Physiology and Kinesiology and Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Kevin Ye
- Cellular and Regenerative Medicine Centre, British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada.,Molecular Cardiac Physiology Group, Departments of Biomedical Physiology and Kinesiology and Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Parisa Asghari
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Lisa Lin
- Cellular and Regenerative Medicine Centre, British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada.,Molecular Cardiac Physiology Group, Departments of Biomedical Physiology and Kinesiology and Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - BaRun Kim
- Molecular Cardiac Physiology Group, Departments of Biomedical Physiology and Kinesiology and Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Thomas M Roston
- British Columbia Children's Hospital Heart Center, Vancouver, British Columbia, Canada
| | - Leif Hove-Madsen
- Cardiac Rhythm and Contraction Group, IIBB-CSIC, CIBERCV, IIB Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Filip Van Petegem
- Department of Biochemistry and Molecular Biology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Shubhayan Sanatani
- British Columbia Children's Hospital Heart Center, Vancouver, British Columbia, Canada
| | - Edwin Moore
- Cardiac Rhythm and Contraction Group, IIBB-CSIC, CIBERCV, IIB Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Francis Lynn
- Cellular and Regenerative Medicine Centre, British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | | | - Yonglun Luo
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - S R Wayne Chen
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada
| | - Glen F Tibbits
- Cellular and Regenerative Medicine Centre, British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada.,Molecular Cardiac Physiology Group, Departments of Biomedical Physiology and Kinesiology and Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada.,School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
10
|
Mazza R, Maher J. Prospects for Development of Induced Pluripotent Stem Cell-Derived CAR-Targeted Immunotherapies. Arch Immunol Ther Exp (Warsz) 2021; 70:2. [PMID: 34897554 PMCID: PMC8666432 DOI: 10.1007/s00005-021-00640-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 11/24/2021] [Indexed: 12/27/2022]
Abstract
Technologies required to generate induced pluripotent stem cells (iPSC) were first described 15 years ago, providing a strong impetus to the field of regenerative medicine. In parallel, immunotherapy has finally emerged as a clinically meaningful modality of cancer therapy. In particular, impressive efficacy has been achieved in patients with selected haematological malignancies using ex vivo expanded autologous T cells engineered to express chimeric antigen receptors (CARs). While solid tumours account for over 90% of human cancer, they currently are largely refractory to this therapeutic approach. Nonetheless, given the considerable innovation taking place worldwide in the CAR field, it is likely that effective solutions for common solid tumours will emerge in the near future. Such a development will create significant new challenges in the scalable delivery of these complex, costly and individualised therapies. CAR-engineered immune cell products that originate from iPSCs offer the potential to generate unlimited numbers of homogeneous, standardised cell products in which multiple defined gene modification events have been introduced to ensure safety, potency and reproducibility. Here, we review some of the emerging strategies in use to engineer CAR-expressing iPSC-derived drug products.
Collapse
Affiliation(s)
- Roberta Mazza
- Leucid Bio Ltd, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - John Maher
- Leucid Bio Ltd, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK.
- King's College London, School of Cancer and Pharmaceutical Sciences, Guy's Cancer Centre, Great Maze Pond, London, SE1 9RT, UK.
- Department of Immunology, Eastbourne Hospital, Kings Drive, Eastbourne, BN21 2UD, East Sussex, UK.
| |
Collapse
|
11
|
Wang Z, Zheng J, Pan R, Chen Y. Current status and future prospects of patient-derived induced pluripotent stem cells. Hum Cell 2021; 34:1601-1616. [PMID: 34378170 DOI: 10.1007/s13577-021-00592-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 08/06/2021] [Indexed: 12/28/2022]
Abstract
Induced pluripotent stem cells (iPSCs) are produced from adult somatic cells through reprogramming, which behave like embryonic stem cells (ESCs) but avoiding the controversial ethical issues from destruction of embryos. Since the first discovery in 2006 of four factors that are essential for maintaining the basic characteristics of ESC, global researches have rapidly improved the techniques for generating iPSCs. In this paper, we review new insights into patient-specific iPSC and summarize selected "disease-in-a-dish" examples that model the genetic and epigenetic variations of human diseases. Although more researches need to be done, studies have increasingly focused on the potential utility of iPSCs. The usability of iPSC technology is changing the fields of disease modeling and precision treatment. Aside from its potential use in regenerative cellular therapy for degenerative diseases, iPSC offers a range of new opportunities for the study of genetic human disorders, particularly, rare diseases. We believe that this rapidly moving field promises many more developments that will benefit modern medicine.
Collapse
Affiliation(s)
- Zhiqiang Wang
- Division of Medical Genetics and Genomics, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China.,Department of Genetics, Institute of Genetics, School of Medicine, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang, China
| | - Jing Zheng
- Division of Medical Genetics and Genomics, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China.,Zhejiang Provincial Key Laboratory of Genetic and Developmental Disorders, Hangzhou, 310058, Zhejiang, China
| | - Ruolang Pan
- Institute for Cell-Based Drug Development of Zhejiang Province, S-Evans Biosciences, Hangzhou, 310012, Zhejiang, China.,Key Laboratory of Cell-Based Drug and Applied Technology Development in Zhejiang Province, Hangzhou, 310012, Zhejiang, China
| | - Ye Chen
- Division of Medical Genetics and Genomics, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China. .,Department of Genetics, Institute of Genetics, School of Medicine, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang, China. .,Zhejiang Provincial Key Laboratory of Genetic and Developmental Disorders, Hangzhou, 310058, Zhejiang, China.
| |
Collapse
|
12
|
Ray A, Joshi JM, Sundaravadivelu PK, Raina K, Lenka N, Kaveeshwar V, Thummer RP. An Overview on Promising Somatic Cell Sources Utilized for the Efficient Generation of Induced Pluripotent Stem Cells. Stem Cell Rev Rep 2021; 17:1954-1974. [PMID: 34100193 DOI: 10.1007/s12015-021-10200-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2021] [Indexed: 01/19/2023]
Abstract
Human induced Pluripotent Stem Cells (iPSCs) have enormous potential in understanding developmental biology, disease modeling, drug discovery, and regenerative medicine. The initial human iPSC studies used fibroblasts as a starting cell source to reprogram them; however, it has been identified to be a less appealing somatic cell source by numerous studies due to various reasons. One of the important criteria to achieve efficient reprogramming is determining an appropriate starting somatic cell type to induce pluripotency since the cellular source has a major influence on the reprogramming efficiency, kinetics, and quality of iPSCs. Therefore, numerous groups have explored various somatic cell sources to identify the promising sources for reprogramming into iPSCs with different reprogramming factor combinations. This review provides an overview of promising easily accessible somatic cell sources isolated in non-invasive or minimally invasive manner such as keratinocytes, urine cells, and peripheral blood mononuclear cells used for the generation of human iPSCs derived from healthy and diseased subjects. Notably, iPSCs generated from one of these cell types derived from the patient will offer ethical and clinical advantages. In addition, these promising somatic cell sources have the potential to efficiently generate bona fide iPSCs with improved reprogramming efficiency and faster kinetics. This knowledge will help in establishing strategies for safe and efficient reprogramming and the generation of patient-specific iPSCs from these cell types.
Collapse
Affiliation(s)
- Arnab Ray
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Jahnavy Madhukar Joshi
- Central Research Laboratory, SDM College of Medical Sciences and Hospital, Shri Dharmasthala Manjunatheshwara University, Dharwad, 580009, Karnataka, India
| | - Pradeep Kumar Sundaravadivelu
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Khyati Raina
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Nibedita Lenka
- National Centre for Cell Science, S. P. Pune University Campus, Pune - 411007, Ganeshkhind, Maharashtra, India
| | - Vishwas Kaveeshwar
- Central Research Laboratory, SDM College of Medical Sciences and Hospital, Shri Dharmasthala Manjunatheshwara University, Dharwad, 580009, Karnataka, India.
| | - Rajkumar P Thummer
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India.
| |
Collapse
|
13
|
Modeling leukemia with pediatric acute leukemia patient-derived iPSCs. Stem Cell Res 2021; 54:102404. [PMID: 34111697 DOI: 10.1016/j.scr.2021.102404] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 05/14/2021] [Accepted: 05/19/2021] [Indexed: 11/20/2022] Open
Abstract
OBJECTIVE ediatric acute leukemia (AL) is the most common hematological malignancy in childhood. However, the limitation of clinical specimens hindered the progress of research. Therefore, new research platforms are urgently needed to establish and clarify the pathogenesis of pediatric AL, and it is necessary to try to find novel targeted therapies for the clinical use. Here, the induced pluripotent stem cells (iPSCs) derived from AL provide a reliable model for basic research. METHODS eukemia cells were sorted by flow cytometry and then reprogrammed into iPSCs by Sendai virus. Cell cycle assay was used to analyze cell proliferation. RESULTS iPS cell lines from T cell acute lymphoblastic leukemia (T-ALL) and acute myeloid leukemia (AML) cells were successfully established. The reprogramming efficiency of AML cells was much higher than that of ALL cells. Disease iPS cells switched off the expression of the disease marker genes at iPS and HPC stage. When different subtypes of AML-iPSCs were differentiated into hematopoietic progenitor cells, iPS derived from acute megakaryocytic leukemia was more readily differentiated into megakaryocyte-erythroid progenitors. Whereas, the differentiation of multipotent lymphoid progenitor (MLP) and granulocyte macrophage progenitor (GMP) were blocked. The iPS derived from acute monocyte leukemia (AMCL) also showed the differentiation of common myeloid progenitors (CMP), GMP and monocytes significantly increased but MLP differentiation was inhibited. The AML-iPSC could form teratomas and we could obverse three germ layers in vivo, indicating that the AML-iPSCs have full pluripotency. However, there were not enough blood cells in teratoma to identify the leukemia. CONCLUSIONS Our results provide a novel platform for AL research and critical insight into the difference of hematopoietic differentiation between ALL and AML.
Collapse
|
14
|
Wang L, Ye Z, Jang YY. Convergence of human pluripotent stem cell, organoid, and genome editing technologies. Exp Biol Med (Maywood) 2021; 246:861-875. [PMID: 33467883 DOI: 10.1177/1535370220985808] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The last decade has seen many exciting technological breakthroughs that greatly expanded the toolboxes for biological and biomedical research, yet few have had more impact than induced pluripotent stem cells and modern-day genome editing. These technologies are providing unprecedented opportunities to improve physiological relevance of experimental models, further our understanding of developmental processes, and develop novel therapies. One of the research areas that benefit greatly from these technological advances is the three-dimensional human organoid culture systems that resemble human tissues morphologically and physiologically. Here we summarize the development of human pluripotent stem cells and their differentiation through organoid formation. We further discuss how genetic modifications, genome editing in particular, were applied to answer basic biological and biomedical questions using organoid cultures of both somatic and pluripotent stem cell origins. Finally, we discuss the potential challenges of applying human pluripotent stem cell and organoid technologies for safety and efficiency evaluation of emerging genome editing tools.
Collapse
Affiliation(s)
- Lin Wang
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Zhaohui Ye
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Yoon-Young Jang
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, John Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
15
|
Zehravi M, Wahid M, Ashraf J. Episomal reprogramming of Duchenne muscular dystrophy patients derived CD3 + T cells towards induced pluripotent stem cells. Pak J Med Sci 2021; 37:432-438. [PMID: 33679927 PMCID: PMC7931302 DOI: 10.12669/pjms.37.2.3388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Objective To derive Duchenne muscular dystrophy patient specific induced pluripotent stem cells (iPSCs) from CD3+T cells of DMD patients using episomal reprogramming and characterization of these DMD-iPSCs using immunofluorescence to confirm their pluripotent state. Methods DMD patients were selected based upon their clinical history and examination. Peripheral blood mononuclear cells were isolated from peripheral blood of DMD patients (n=3) by density gradient centrifugation and were used to generate DMD patient specific T cells (DMD-T cells) using rhIL-2, plate bound anti CD3 antibody and T cell specific media along with specific culture conditions that promote T cell expansion. CD3+ T cells were characterized by flow cytometry and reprogrammed using episomal plasmid to generate DMD-iPSCs. These DMD-iPSCs were characterized using immunofluorescence. The study was carried out at Dow Research Institute of Biotechnology and Biomedical Sciences during August 2017- July 2018 for a period of approximately 12 months. Results The peripheral blood mononuclear cells (PBMNC) derived T cells appeared as suspended cell clumps morphologically were positive for the expression of CD3 and negative for CD31. The DMD patient specific iPSCs appeared as round, compact and flat colonies with well-defined edges morphologically and were positive for the expression of pluripotency markers OCT4, SSEA-4 and TRA-1-81 on immunofluorescence. Conclusion CD3+ T cell derived DMD-iPSCs were obtained under feeder free and xeno-free culture conditions using episomal reprogramming.
Collapse
Affiliation(s)
- Mehwish Zehravi
- Mehwish Zehravi, Dow Research Institute of Biotechnology and Biomedical Sciences, Dow University of Health Sciences, Karachi, Pakistan
| | - Mohsin Wahid
- Mohsin Wahid, Dow Research Institute of Biotechnology & Biomedical Sciences and Department of Pathology, Dow International Medical College, Dow University of Health Sciences, Karachi, Pakistan
| | - Junaid Ashraf
- Junaid Ashraf (Rtd) Department of Neurosurgery, Dow University of Health Sciences, Karachi, Pakistan
| |
Collapse
|
16
|
Kido J, Nakamura K, Era T. Role of induced pluripotent stem cells in lysosomal storage diseases. Mol Cell Neurosci 2020; 108:103540. [PMID: 32828964 DOI: 10.1016/j.mcn.2020.103540] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 08/11/2020] [Accepted: 08/14/2020] [Indexed: 12/31/2022] Open
Abstract
Lysosomal storage diseases (LSDs) are a group of metabolism inborn errors caused by defective enzymes in the lysosome, resulting in the accumulation of undegraded substrates. Many characteristic cell features have been revealed in LSDs, including abnormal autophagy and mitochondrial dysfunction. The development of induced pluripotent stem cells (iPSCs) dramatically boosted research on LSDs, particularly regarding novel opportunities to clarify the disease etiology based on the storage of macromolecules, such as sphingolipids in lysosomes. iPSCs made from LSD patients (LSD-iPSCs) have been differentiated into neurons, endothelial cells, cardiomyocytes, hepatocytes, and macrophages, with each cell type closely resembling the primary disease phenotypes, providing new tools to probe the disease pathogenesis and to test therapeutic strategies. Abnormally accumulated substrates impaired autophagy and mitochondrial and synapse functions in LSD-iPSC-derived neurons. Reducing the accumulation with the treatment of drug candidates improved LSD-iPSC-derived neuron functions. Additionally, iPSC technology can help probe the gene expressions, proteomics, and metabolomics of LSDs. Further, gene repair and the generation of new mutations in causative genes in LSD-iPSCs can be used to understand both the specific roles of causative genes and the contributions of other genetic factors to these phenotypes. Moreover, the development of iPSC-derived organoids as disease models has bridged the gap between studies using cell lines and in vivo animal models. There are some reproducibility issues in iPSC research, however, including genetic and epigenetic abnormalities, such as chromosomal abnormalities, DNA mutations, and gene modifications via methylation. In this review, we present the disease and treatment concepts gathered using selected LSD-iPSCs, discuss iPSC research limitations, and set our future research visions. Such studies are expected to further inform and generate insights into LSDs and are important in research and clinical practice.
Collapse
Affiliation(s)
- Jun Kido
- Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan; Department of Pediatrics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.
| | - Kimitoshi Nakamura
- Department of Pediatrics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Takumi Era
- Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
17
|
Drakulic D, Djurovic S, Syed YA, Trattaro S, Caporale N, Falk A, Ofir R, Heine VM, Chawner SJRA, Rodriguez-Moreno A, van den Bree MBM, Testa G, Petrakis S, Harwood AJ. Copy number variants (CNVs): a powerful tool for iPSC-based modelling of ASD. Mol Autism 2020; 11:42. [PMID: 32487215 PMCID: PMC7268297 DOI: 10.1186/s13229-020-00343-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 05/04/2020] [Indexed: 02/06/2023] Open
Abstract
Patients diagnosed with chromosome microdeletions or duplications, known as copy number variants (CNVs), present a unique opportunity to investigate the relationship between patient genotype and cell phenotype. CNVs have high genetic penetrance and give a good correlation between gene locus and patient clinical phenotype. This is especially effective for the study of patients with neurodevelopmental disorders (NDD), including those falling within the autism spectrum disorders (ASD). A key question is whether this correlation between genetics and clinical presentation at the level of the patient can be translated to the cell phenotypes arising from the neurodevelopment of patient induced pluripotent stem cells (iPSCs).Here, we examine how iPSCs derived from ASD patients with an associated CNV inform our understanding of the genetic and biological mechanisms underlying the aetiology of ASD. We consider selection of genetically characterised patient iPSCs; use of appropriate control lines; aspects of human neurocellular biology that can capture in vitro the patient clinical phenotype; and current limitations of patient iPSC-based studies. Finally, we consider how future research may be enhanced to maximise the utility of CNV patients for research of pathological mechanisms or therapeutic targets.
Collapse
Affiliation(s)
- Danijela Drakulic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11042 Belgrade, 152, Serbia
| | - Srdjan Djurovic
- Department of Medical Genetics, Oslo University Hospital, 0424, Oslo, Norway
- NORMENT, Department of Clinical Science, University of Bergen, 5007, Bergen, Norway
| | - Yasir Ahmed Syed
- Neuroscience & Mental Health Research Institute, Cardiff University, Cardiff, CF24 4HQ, UK
| | - Sebastiano Trattaro
- Laboratory of Stem Cell Epigenetics, IEO, European Institute of Oncology, IRCCS, 20146, Milan, Italy
- Department of Oncology and Hemato-oncology, University of Milan, 20122, Milan, Italy
| | - Nicolò Caporale
- Laboratory of Stem Cell Epigenetics, IEO, European Institute of Oncology, IRCCS, 20146, Milan, Italy
- Department of Oncology and Hemato-oncology, University of Milan, 20122, Milan, Italy
| | - Anna Falk
- Department of Neuroscience, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Rivka Ofir
- BGU-iPSC Core Facility, The Regenerative Medicine & Stem Cell (RMSC) Research Center, Ben Gurion University of the Negev, 84105, Beer-Sheva, Israel
| | - Vivi M Heine
- Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Child and Youth Psychiatry, Emma Children's Hospital, Amsterdam UMC, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081, Amsterdam, The Netherlands
| | - Samuel J R A Chawner
- Neuroscience & Mental Health Research Institute, Cardiff University, Cardiff, CF24 4HQ, UK
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, CF24 4HQ, UK
| | - Antonio Rodriguez-Moreno
- Department of Physiology, Anatomy and Cell Biology, University Pablo de Olavide, Ctra. de Utrera, Km 1, 41013, Seville, Spain
| | - Marianne B M van den Bree
- Neuroscience & Mental Health Research Institute, Cardiff University, Cardiff, CF24 4HQ, UK
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, CF24 4HQ, UK
| | - Giuseppe Testa
- Laboratory of Stem Cell Epigenetics, IEO, European Institute of Oncology, IRCCS, 20146, Milan, Italy
- Department of Oncology and Hemato-oncology, University of Milan, 20122, Milan, Italy
- Human Technopole, Via Cristina Belgioioso 171, 20157, Milan, Italy
| | - Spyros Petrakis
- Institute of Applied Biosciences/Centre for Research and Technology Hellas, 57001, Thessaloniki, Greece.
| | - Adrian J Harwood
- Neuroscience & Mental Health Research Institute, Cardiff University, Cardiff, CF24 4HQ, UK.
| |
Collapse
|
18
|
Bragança J, Lopes JA, Mendes-Silva L, Almeida Santos JM. Induced pluripotent stem cells, a giant leap for mankind therapeutic applications. World J Stem Cells 2019; 11:421-430. [PMID: 31396369 PMCID: PMC6682501 DOI: 10.4252/wjsc.v11.i7.421] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/22/2019] [Accepted: 06/13/2019] [Indexed: 02/06/2023] Open
Abstract
Induced pluripotent stem cells (iPSC) technology has propelled the field of stem cells biology, providing new cells to explore the molecular mechanisms of pluripotency, cancer biology and aging. A major advantage of human iPSC, compared to the pluripotent embryonic stem cells, is that they can be generated from virtually any embryonic or adult somatic cell type without destruction of human blastocysts. In addition, iPSC can be generated from somatic cells harvested from normal individuals or patients, and used as a cellular tool to unravel mechanisms of human development and to model diseases in a manner not possible before. Besides these fundamental aspects of human biology and physiology that are revealed using iPSC or iPSC-derived cells, these cells hold an immense potential for cell-based therapies, and for the discovery of new or personalized pharmacological treatments for many disorders. Here, we review some of the current challenges and concerns about iPSC technology. We introduce the potential held by iPSC for research and development of novel health-related applications. We briefly present the efforts made by the scientific and clinical communities to create the necessary guidelines and regulations to achieve the highest quality standards in the procedures for iPSC generation, characterization and long-term preservation. Finally, we present some of the audacious and pioneer clinical trials in progress with iPSC-derived cells.
Collapse
Affiliation(s)
- José Bragança
- Department of Biomedical Sciences and Medicine, University of Algarve, Faro 8005-139, Portugal
- Centre for Biomedical Research (CBMR), University of Algarve, Faro 8005-139, Portugal
- ABC - Algarve Biomedical Centre, Faro 8005-139, Portugal
| | - João André Lopes
- Department of Biomedical Sciences and Medicine, University of Algarve, Faro 8005-139, Portugal
- Centre for Biomedical Research (CBMR), University of Algarve, Faro 8005-139, Portugal
| | - Leonardo Mendes-Silva
- Department of Biomedical Sciences and Medicine, University of Algarve, Faro 8005-139, Portugal
- Centre for Biomedical Research (CBMR), University of Algarve, Faro 8005-139, Portugal
| | - João Miguel Almeida Santos
- Department of Biomedical Sciences and Medicine, University of Algarve, Faro 8005-139, Portugal
- Centre for Biomedical Research (CBMR), University of Algarve, Faro 8005-139, Portugal
| |
Collapse
|
19
|
Recent Updates on Induced Pluripotent Stem Cells in Hematological Disorders. Stem Cells Int 2019; 2019:5171032. [PMID: 31191673 PMCID: PMC6525795 DOI: 10.1155/2019/5171032] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 03/31/2019] [Indexed: 02/07/2023] Open
Abstract
Over the past decade, enormous progress has been made in the field of induced pluripotent stem cells (iPSCs). Patients' somatic cells such as skin fibroblasts or blood cells can be used to generate disease-specific pluripotent stem cells, which have unlimited proliferation and can differentiate into all cell types of the body. Human iPSCs offer great promises and opportunities for treatments of degenerative diseases and studying disease pathology and drug screening. So far, many iPSC-derived disease models have led to the discovery of novel pathological mechanisms as well as new drugs in the pipeline that have been tested in the iPSC-derived cells for efficacy and potential toxicities. Furthermore, recent advances in genome editing technology in combination with the iPSC technology have provided a versatile platform for studying stem cell biology and regenerative medicine. In this review, an overview of iPSCs, patient-specific iPSCs for disease modeling and drug screening, applications of iPSCs and genome editing technology in hematological disorders, remaining challenges, and future perspectives of iPSCs in hematological diseases will be discussed.
Collapse
|
20
|
Ardila DC, Liou JJ, Maestas D, Slepian MJ, Badowski M, Wagner WR, Harris D, Vande Geest JP. Surface Modification of Electrospun Scaffolds for Endothelialization of Tissue-Engineered Vascular Grafts Using Human Cord Blood-Derived Endothelial Cells. J Clin Med 2019; 8:E185. [PMID: 30720769 PMCID: PMC6416564 DOI: 10.3390/jcm8020185] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/22/2019] [Accepted: 02/01/2019] [Indexed: 01/09/2023] Open
Abstract
Tissue engineering has gained attention as an alternative approach for developing small diameter tissue-engineered vascular grafts intended for bypass surgery, as an option to treat coronary heart disease. To promote the formation of a healthy endothelial cell monolayer in the lumen of the graft, polycaprolactone/gelatin/fibrinogen scaffolds were developed, and the surface was modified using thermoforming and coating with collagen IV and fibronectin. Human cord blood-derived endothelial cells (hCB-ECs) were seeded onto the scaffolds and the important characteristics of a healthy endothelial cell layer were evaluated under static conditions using human umbilical vein endothelial cells as a control. We found that polycaprolactone/gelatin/fibrinogen scaffolds that were thermoformed and coated are the most suitable for endothelial cell growth. hCB-ECs can proliferate, produce endothelial nitric oxide synthase, respond to interleukin 1 beta, and reduce platelet deposition.
Collapse
Affiliation(s)
| | - Jr-Jiun Liou
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15219, USA.
| | - David Maestas
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21231, USA.
| | - Marvin J Slepian
- Sarver Heart Center, The University of Arizona, Tucson, AZ 85721, USA.
- The Arizona Center for Accelerated BioMedical Innovation, University of Arizona, Tucson, AZ 85721, USA.
- BIO5 Institute for Biocollaborative Research, The University of Arizona, Tucson, AZ 85721, USA.
- Interventional Cardiology, University of Arizona, Tucson, AZ 85721, USA.
| | - Michael Badowski
- Arizona Health Science Center Biorepository, University of Arizona, Tucson, AZ 85724, USA.
| | - William R. Wagner
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15219, USA.
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA.
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA.
| | - David Harris
- Arizona Health Science Center Biorepository, University of Arizona, Tucson, AZ 85724, USA.
- Department of Immunobiology, Arizona Health Science Center Biorepository, University of Arizona, Tucson, AZ 85724, USA.
| | - Jonathan P Vande Geest
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15219, USA.
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA.
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15219, USA.
| |
Collapse
|
21
|
Korhonen P, Pollari E, Kanninen KM, Savchenko E, Lehtonen Š, Wojciechowski S, Pomeshchik Y, Van Den Bosch L, Goldsteins G, Koistinaho J, Malm T. Long-term interleukin-33 treatment delays disease onset and alleviates astrocytic activation in a transgenic mouse model of amyotrophic lateral sclerosis. IBRO Rep 2019; 6:74-86. [PMID: 30705990 PMCID: PMC6348738 DOI: 10.1016/j.ibror.2019.01.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 01/08/2019] [Indexed: 12/12/2022] Open
Abstract
Th2-type cytokine IL-33 delayed the disease onset of female SOD1-G93 A transgenic ALS mice. IL-33 decreased the proportion of T cells in the spleens and lymph nodes of female mice. IL-33 decreased astrocytic activation in the spinal cord of female mice. Male mice were unresponsive to the treatment.
Inflammation is a prominent feature of the neuropathology of amyotrophic lateral sclerosis (ALS). Emerging evidence suggests that inflammatory cascades contributing to the disease progression are not restricted to the central nervous system (CNS) but also occur peripherally. Indeed, alterations in T cell responses and their secreted cytokines have been detected in ALS patients and in animal models of ALS. One key cytokine responsible for the shift in T cell responses is interleukin-33 (IL-33), which stimulates innate type 2 immune cells to produce a large amount of Th2 cytokines that are possibly beneficial in the recovery processes of CNS injuries. Since the levels of IL-33 have been shown to be decreased in patients affected with ALS, we sought to determine whether a long-term recombinant IL-33 treatment of a transgenic mouse model of ALS expressing G93A-superoxide dismutase 1 (SOD1-G93A) alters the disease progression and ameliorates the ALS-like disease pathology. SOD1-G93A mice were treated with intraperitoneal injections of IL-33 and effects on disease onset and inflammatory status were determined. Spinal cord (SC) neurons, astrocytes and T-cells were exposed to IL-33 to evaluate the cell specific responses to IL-33. Treatment of SOD1-G93A mice with IL-33 delayed the disease onset in female mice, decreased the proportion of CD4+ and CD8 + T cell populations in the spleen and lymph nodes, and alleviated astrocytic activation in the ventral horn of the lumbar SC. Male SOD1-G93A mice were unresponsive to the treatment. In vitro studies showed that IL-33 is most likely not acting directly on neurons and astrocytes, but rather conveying its effects through peripheral T-cells. Our results suggest that strategies directed to the peripheral immune system may have therapeutic potential in ALS. The effect of gender dimorphisms to the treatment efficacy needs to be taken into consideration when designing new therapeutic strategies for CNS diseases.
Collapse
Key Words
- ALS
- ALS, amyotrophic lateral sclerosis
- ANOVA, analysis of variance
- Arg-1, arginine-1
- Astrocyte
- CM, conditioned medium
- CNS, central nervous system
- Cytokine
- DMEM, Dulbecco’s minimum essential medium
- EAE, experimental autoimmune encephalomyelitis
- GFAP, glial fibrillary acidic protein
- HO-1, hemeoxygenase-1
- IFN-γ, interferon gamma
- IL-10, interleukin-10
- IL-1RAcP, interleukin-1 receptor accessory protein
- IL-33, interleukin-33
- IL-33R, interleukin-33 receptor
- IL-6, interleukin-6
- Iba-1, ionized calcium binding adaptor molecule-1
- Inflammation
- Interleukin-33
- MCP-1, monocyte chemoattractant protein-1
- Microglia
- NFE2L2, the gene encoding Nrf2
- Nrf2, nuclear factor (erythroid-derived 2)-like 2
- PBS, phosphate buffered saline
- RT, room temperature
- SC, spinal cord
- SD, standard deviation
- SOD1, superoxide dismutase 1
- Spinal cord
- T cell
- TG, transgenic
- TNF, tumor necrosis factor
- WT, wildtype
- fALS, familial ALS
- sALS, sporadic ALS
Collapse
Affiliation(s)
- Paula Korhonen
- A. I. Virtanen Institute for Molecular Sciences, Biocenter Kuopio, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Eveliina Pollari
- KU Leuven, University of Leuven, Department of Neurosciences, Experimental Neurology, VIB Center for Brain & Disease Research, Box 912, B-3000 Leuven, Belgium
| | - Katja M Kanninen
- A. I. Virtanen Institute for Molecular Sciences, Biocenter Kuopio, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Ekaterina Savchenko
- A. I. Virtanen Institute for Molecular Sciences, Biocenter Kuopio, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Šárka Lehtonen
- A. I. Virtanen Institute for Molecular Sciences, Biocenter Kuopio, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Sara Wojciechowski
- A. I. Virtanen Institute for Molecular Sciences, Biocenter Kuopio, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Yuriy Pomeshchik
- A. I. Virtanen Institute for Molecular Sciences, Biocenter Kuopio, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Ludo Van Den Bosch
- KU Leuven, University of Leuven, Department of Neurosciences, Experimental Neurology, VIB Center for Brain & Disease Research, Box 912, B-3000 Leuven, Belgium
| | - Gundars Goldsteins
- A. I. Virtanen Institute for Molecular Sciences, Biocenter Kuopio, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Jari Koistinaho
- A. I. Virtanen Institute for Molecular Sciences, Biocenter Kuopio, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Tarja Malm
- A. I. Virtanen Institute for Molecular Sciences, Biocenter Kuopio, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| |
Collapse
|
22
|
A New Automatic Cancer Colony Forming Units Counting Method. PATTERN RECOGNITION AND IMAGE ANALYSIS 2019. [DOI: 10.1007/978-3-030-31321-0_40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
23
|
Montel-Hagen A, Crooks GM. From pluripotent stem cells to T cells. Exp Hematol 2018; 71:24-31. [PMID: 30590093 DOI: 10.1016/j.exphem.2018.12.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 12/16/2018] [Accepted: 12/19/2018] [Indexed: 02/08/2023]
Abstract
The generation of T cells from human pluripotent stem cells (PSCs) opens a valuable experimental window into developmental hematopoiesis and raises the possibility of a new therapeutic approach for T-cell immunotherapy. After directing PSCs through mesoderm and early hematopoietic developmental stages, commitment to the T-cell lineage has been achieved by several groups using coculture with stromal cells that express a notch ligand, recapitulating the critical signals that initiate the first stages of normal T-cell differentiation in the thymus. However, positive selection and the production of mature T cells from human PSCs have been limited to date. Nonetheless, T-lineage cells have been generated from PSCs with tumor antigen specificity either through a prearranged clonal T-cell receptor (TCR) or lentiviral-mediated expression of chimeric antigen receptors. The recent development of a 3D artificial organoid model has demonstrated that PSCs can generate mature conventional T cells that are fully functional and express a diverse TCR repertoire. Introduction of a transgenic TCR at the PSC stage allows for the production of tumor-antigen-specific, mature conventional T cells. The tools of gene editing in PSCs are ideally suited to produce off-the-shelf universal products for T-cell immunotherapy. In this review, we describe the studies that have led to this exciting moment in PSC biology and discuss translation to clinical applications.
Collapse
Affiliation(s)
- Amélie Montel-Hagen
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Gay M Crooks
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA; Division of Pediatric Hematology-Oncology, Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA; Broad Stem Cell Research Center, University of California, Los Angeles, CA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA.
| |
Collapse
|
24
|
Tan X, Dai Q, Guo T, Xu J, Dai Q. Efficient generation of transgene- and feeder-free induced pluripotent stem cells from human dental mesenchymal stem cells and their chemically defined differentiation into cardiomyocytes. Biochem Biophys Res Commun 2018; 495:2490-2497. [DOI: 10.1016/j.bbrc.2017.12.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 12/01/2017] [Indexed: 02/06/2023]
|
25
|
Berridge BR, Schultze AE, Heyen JR, Searfoss GH, Sarazan RD. Technological Advances in Cardiovascular Safety Assessment Decrease Preclinical Animal Use and Improve Clinical Relevance. ILAR J 2017; 57:120-132. [PMID: 28053066 DOI: 10.1093/ilar/ilw028] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 10/09/2016] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular (CV) safety liabilities are significant concerns for drug developers and preclinical animal studies are predominately where those liabilities are characterized before patient exposures. Steady progress in technology and laboratory capabilities is enabling a more refined and informative use of animals in those studies. The application of surgically implantable and telemetered instrumentation in the acute assessment of drug effects on CV function has significantly improved historical approaches that involved anesthetized or restrained animals. More chronically instrumented animals and application of common clinical imaging assessments like echocardiography and MRI extend functional and in-life structural assessments into the repeat-dose setting. A growing portfolio of circulating CV biomarkers is allowing longitudinal and repeated measures of cardiac and vascular injury and dysfunction better informing an understanding of temporal pathogenesis and allowing earlier detection of undesirable effects. In vitro modeling systems of the past were limited by their lack of biological relevance to the in vivo human condition. Advances in stem cell technology and more complex in vitro modeling platforms are quickly creating more opportunity to supplant animals in our earliest assessments for liabilities. Continuing improvement in our capabilities in both animal and nonanimal modeling should support a steady decrease in animal use for primary liability identification and optimize the translational relevance of the animal studies we continue to do.
Collapse
Affiliation(s)
- Brian R Berridge
- Brian R. Berridge, DVM, PhD, is a Senior GSK Fellow and Head of Worldwide Animal Research Strategy at GlaxoSmithKline in King of Prussia, Pennsylvania. A. Eric Schultze, DVM, PhD, is a Senior Research Advisor-Pathologist at Lilly Research Laboratories in Indianapolis, Indiana. Jon R. Heyen, MS, is a Senior Principal Scientist at Pfizer in La Jolla, California. George H. Searfoss, MS, is a Consultant Toxicologist at Lilly Research Laboratories in Indianapolis, Indiana. R. Dustan Sarazan, DVM, PhD, is a cardiovascular consultant currently residing in Rhinelander, Wisconsin
| | - A Eric Schultze
- Brian R. Berridge, DVM, PhD, is a Senior GSK Fellow and Head of Worldwide Animal Research Strategy at GlaxoSmithKline in King of Prussia, Pennsylvania. A. Eric Schultze, DVM, PhD, is a Senior Research Advisor-Pathologist at Lilly Research Laboratories in Indianapolis, Indiana. Jon R. Heyen, MS, is a Senior Principal Scientist at Pfizer in La Jolla, California. George H. Searfoss, MS, is a Consultant Toxicologist at Lilly Research Laboratories in Indianapolis, Indiana. R. Dustan Sarazan, DVM, PhD, is a cardiovascular consultant currently residing in Rhinelander, Wisconsin
| | - Jon R Heyen
- Brian R. Berridge, DVM, PhD, is a Senior GSK Fellow and Head of Worldwide Animal Research Strategy at GlaxoSmithKline in King of Prussia, Pennsylvania. A. Eric Schultze, DVM, PhD, is a Senior Research Advisor-Pathologist at Lilly Research Laboratories in Indianapolis, Indiana. Jon R. Heyen, MS, is a Senior Principal Scientist at Pfizer in La Jolla, California. George H. Searfoss, MS, is a Consultant Toxicologist at Lilly Research Laboratories in Indianapolis, Indiana. R. Dustan Sarazan, DVM, PhD, is a cardiovascular consultant currently residing in Rhinelander, Wisconsin
| | - George H Searfoss
- Brian R. Berridge, DVM, PhD, is a Senior GSK Fellow and Head of Worldwide Animal Research Strategy at GlaxoSmithKline in King of Prussia, Pennsylvania. A. Eric Schultze, DVM, PhD, is a Senior Research Advisor-Pathologist at Lilly Research Laboratories in Indianapolis, Indiana. Jon R. Heyen, MS, is a Senior Principal Scientist at Pfizer in La Jolla, California. George H. Searfoss, MS, is a Consultant Toxicologist at Lilly Research Laboratories in Indianapolis, Indiana. R. Dustan Sarazan, DVM, PhD, is a cardiovascular consultant currently residing in Rhinelander, Wisconsin
| | - R Dustan Sarazan
- Brian R. Berridge, DVM, PhD, is a Senior GSK Fellow and Head of Worldwide Animal Research Strategy at GlaxoSmithKline in King of Prussia, Pennsylvania. A. Eric Schultze, DVM, PhD, is a Senior Research Advisor-Pathologist at Lilly Research Laboratories in Indianapolis, Indiana. Jon R. Heyen, MS, is a Senior Principal Scientist at Pfizer in La Jolla, California. George H. Searfoss, MS, is a Consultant Toxicologist at Lilly Research Laboratories in Indianapolis, Indiana. R. Dustan Sarazan, DVM, PhD, is a cardiovascular consultant currently residing in Rhinelander, Wisconsin
| |
Collapse
|
26
|
The Rapidly Evolving Concept of Whole Heart Engineering. Stem Cells Int 2017; 2017:8920940. [PMID: 29250121 PMCID: PMC5700515 DOI: 10.1155/2017/8920940] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 09/12/2017] [Indexed: 01/10/2023] Open
Abstract
Whole heart engineering represents an incredible journey with as final destination the challenging aim to solve end-stage cardiac failure with a biocompatible and living organ equivalent. Its evolution started in 2008 with rodent organs and is nowadays moving closer to clinical application thanks to scaling-up strategies to human hearts. This review will offer a comprehensive examination on the important stages to be reached for the bioengineering of the whole heart, by describing the approaches of organ decellularization, repopulation, and maturation so far applied and the novel technologies of potential interest. In addition, it will carefully address important demands that still need to be satisfied in order to move to a real clinical translation of the whole bioengineering heart concept.
Collapse
|
27
|
Parameswaran S, Krishnakumar S. Pluripotent stem cells: A therapeutic source for age-related macular degeneration. Indian J Ophthalmol 2017; 65:177-183. [PMID: 28440245 PMCID: PMC5426121 DOI: 10.4103/ijo.ijo_1026_15] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Age-related macular degeneration (AMD) leads to progressive loss of central vision in the elderly. At a cellular level, there is aging of the retinal pigment epithelial (RPE) cells, and accumulation of lipofuscin that interferes with the proper functioning of RPE which eventually leads to apoptosis. Treatment depends on the stage of the disease. Wet AMD which has neovascularization is managed by local therapies such as laser photocoagulation and photodynamic therapy and is managed with injections of antivascular endothelial growth factor-based therapy. Unlike the wet AMD, an effective therapy does not exist for dry AMD and geographic atrophy. Cell replacement therapy has shown promise. This review discusses the opportunities in the various types of cell-based therapy, their limitations, and what is possible for India.
Collapse
Affiliation(s)
- Sowmya Parameswaran
- L and T Ophthalmic Pathology, Radheshyam Kanoi Stem Cell Laboratory, Vision Research Foundation, Sankara Nethralaya, Chennai, Tamil Nadu, India
| | - Subramanian Krishnakumar
- L and T Ophthalmic Pathology, Radheshyam Kanoi Stem Cell Laboratory, Vision Research Foundation, Sankara Nethralaya, Chennai, Tamil Nadu, India
| |
Collapse
|
28
|
Stem cells in cancer therapy: opportunities and challenges. Oncotarget 2017; 8:75756-75766. [PMID: 29088907 PMCID: PMC5650462 DOI: 10.18632/oncotarget.20798] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 08/17/2017] [Indexed: 01/09/2023] Open
Abstract
Metastatic cancer cells generally cannot be eradicated using traditional surgical or chemoradiotherapeutic strategies, and disease recurrence is extremely common following treatment. On the other hand, therapies employing stem cells are showing increasing promise in the treatment of cancer. Stem cells can function as novel delivery platforms by homing to and targeting both primary and metastatic tumor foci. Stem cells engineered to stably express various cytotoxic agents decrease tumor volumes and extend survival in preclinical animal models. They have also been employed as virus and nanoparticle carriers to enhance primary therapeutic efficacies and relieve treatment side effects. Additionally, stem cells can be applied in regenerative medicine, immunotherapy, cancer stem cell-targeted therapy, and anticancer drug screening applications. However, while using stem cells to treat human cancers appears technically feasible, challenges such as treatment durability and tumorigenesis necessitate further study to improve therapeutic performance and applicability. This review focuses on recent progress toward stem cell-based cancer treatments, and summarizes treatment advantages, opportunities, and shortcomings, potentially helping to refine future trials and facilitate the translation from experimental to clinical studies.
Collapse
|
29
|
Kuo YC, Rajesh R. Guided differentiation and tissue regeneration of induced pluripotent stem cells using biomaterials. J Taiwan Inst Chem Eng 2017. [DOI: 10.1016/j.jtice.2017.04.043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
30
|
Zhu LF, Xiao M, Chen YQ, Wang LY, Luo XF, Yuan XH, Ren JH, Chen ZZ, Hu JD, Yang T. In vitro effects of reprogramming factors on the expressions of pluripotent genes and CD 34 gene in human acute promyelocytic leukemia HL-60 cells. Genomics 2017; 109:331-335. [PMID: 28533192 DOI: 10.1016/j.ygeno.2017.05.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 04/28/2017] [Accepted: 05/18/2017] [Indexed: 10/19/2022]
Abstract
OBJECTIVE Our study aims to explore the in vitro effects of reprogramming factors on the expressions of pluripotent genes and CD34 gene in HL-60 cells. METHODS According to the construction of lentiviral vector LV-OSCK of reprogramming factors (Oct-4, Sox2, Klf4, c-Myc), 293T cells were transfected to detect virus titer. The endogenous pluripotent genes (Oct4, SOX2, c-Myc and Klf4) and CD34 mRNA and protein expressions were detected by AP staining, immunofluorescence staining, qRT-PCR and flow cytometry. RESULTS Expressions of Oct4, SOX2, c-Myc and Klf4 were 0.220±0.013, 0.186±0.009, 0.287±0.015 and 0.153±0.007. These levels were significantly higher in the experimental group than the control and blank groups. CD34 protein expression in the experimental group was also discovered to be significantly higher than the other two groups. CONCLUSION The reprogramming factors could increase the expressions of pluripotent genes and CD34 gene in HL-60 cells.
Collapse
Affiliation(s)
- Liang-Fang Zhu
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou 350001, PR China
| | - Min Xiao
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou 350001, PR China
| | - Yong-Quan Chen
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou 350001, PR China
| | - Ling-Yan Wang
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou 350001, PR China
| | - Xiao-Feng Luo
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou 350001, PR China
| | - Xiao-Hong Yuan
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou 350001, PR China
| | - Jin-Hua Ren
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou 350001, PR China
| | - Zhi-Zhe Chen
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou 350001, PR China
| | - Jian-Da Hu
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou 350001, PR China
| | - Ting Yang
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou 350001, PR China.
| |
Collapse
|
31
|
Chen S, Zhang JQ, Chen JZ, Chen HX, Qiu FN, Yan ML, Chen YL, Peng CH, Tian YF, Wang YD. The over expression of long non-coding RNA ANRIL promotes epithelial-mesenchymal transition by activating the ATM-E2F1 signaling pathway in pancreatic cancer: An in vivo and in vitro study. Int J Biol Macromol 2017; 102:718-728. [PMID: 28344092 DOI: 10.1016/j.ijbiomac.2017.03.123] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 03/07/2017] [Accepted: 03/21/2017] [Indexed: 11/27/2022]
Abstract
This study aims to investigate the roles of lncRNA ANRIL in epithelial-mesenchymal transition (EMT) by regulating the ATM-E2F1 signaling pathway in pancreatic cancer (PC). PC rat models were established and ANRIL overexpression and interference plasmids were transfected. The expression of ANRIL, EMT markers (E-cadherin, N-cadherin and Vimentin) and ATM-E2F1 signaling pathway-related proteins (ATM, E2F1, INK4A, INK4B and ARF) were detected. Small molecule drugs were applied to activate and inhibit the ATM-E2F1 signaling pathway. Transwell assay and the scratch test were adopted to detect cell invasion and migration abilities. ANRIL expression in the PC cells was higher than in normal pancreatic duct epithelial cells. In the PC rat models and PC cells, ANRIL interference promoted the expressions of INK4B, INK4A, ARF and E-cadherin, while reduced N-cadherin and Vimentin expression. Over-expressed ANRIL decreased the expression of INK4B, INK4A, ARF and E-cadherin, but raised N-cadherin and Vimentin expressions. By inhibiting the ATM-E2F1 signaling pathway in PC cells, E-cadherin expression increased but N-cadherin and Vimentin expressions decreased. After ANRIL was silenced or the ATM-E2F1 signaling pathway inhibited, PC cell migration and invasion abilities were decreased. In conclusion, over-expression of lncRNA ANRIL can promote EMT of PC cells by activating the ATM-E2F1 signaling pathway.
Collapse
Affiliation(s)
- Shi Chen
- Department of Hepatobiliary Surgery, Fujian Provincial Hospital, Fujian Medical University, Fuzhou 350001, P.R. China; Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, PR China; Research Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jia-Qiang Zhang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, PR China; Research Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jiang-Zhi Chen
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, PR China; Research Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Department of Hepatobiliary Surgery, Union Hospital, Fujian Medical University, Fuzhou 350001, PR China
| | - Hui-Xing Chen
- Department of Hepatobiliary Surgery, Union Hospital, Fujian Medical University, Fuzhou 350001, PR China
| | - Fu-Nan Qiu
- Department of Hepatobiliary Surgery, Fujian Provincial Hospital, Fujian Medical University, Fuzhou 350001, P.R. China
| | - Mao-Lin Yan
- Department of Hepatobiliary Surgery, Fujian Provincial Hospital, Fujian Medical University, Fuzhou 350001, P.R. China
| | - Yan-Ling Chen
- Department of Hepatobiliary Surgery, Union Hospital, Fujian Medical University, Fuzhou 350001, PR China
| | - Cheng-Hong Peng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, PR China; Research Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yi-Feng Tian
- Department of Hepatobiliary Surgery, Fujian Provincial Hospital, Fujian Medical University, Fuzhou 350001, P.R. China.
| | - Yao-Dong Wang
- Department of Hepatobiliary Surgery, Fujian Provincial Hospital, Fujian Medical University, Fuzhou 350001, P.R. China.
| |
Collapse
|
32
|
Paes BCMF, Moço PD, Pereira CG, Porto GS, de Sousa Russo EM, Reis LCJ, Covas DT, Picanço-Castro V. Ten years of iPSC: clinical potential and advances in vitro hematopoietic differentiation. Cell Biol Toxicol 2016; 33:233-250. [PMID: 28039590 DOI: 10.1007/s10565-016-9377-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 12/18/2016] [Indexed: 01/19/2023]
Abstract
Ten years have passed since the first publication announcing the generation of induced pluripotent stem cells (iPSCs). Issues related to ethics, immune rejection, and cell availability seemed to be solved following this breakthrough. The development of iPSC technology allows advances in in vitro cell differentiation for cell therapy purpose and other clinical applications. This review provides a perspective on the iPSC potential for cell therapies, particularly for hematological applications. We discuss the advances in in vitro hematopoietic differentiation, the possibilities to employ iPSC in hematology studies, and their potential clinical application in hematologic diseases. The generation of red blood cells and functional T cells and the genome editing technology applied to mutation correction are also covered. We highlight some of the requirements and obstacles to be overcome before translating these cells from research to the clinic, for instance, iPSC variability, genotoxicity, the differentiation process, and engraftment. Also, we evaluate the patent landscape and compile the clinical trials in the field of pluripotent stem cells. Currently, we know much more about iPSC than in 2006, but there are still challenges that must be solved. A greater understanding of molecular mechanisms underlying the generation of hematopoietic stem cells is necessary to produce suitable and transplantable hematopoietic stem progenitor cells from iPSC.
Collapse
Affiliation(s)
- Bárbara Cristina Martins Fernandes Paes
- Ribeirão Preto Medical School and Center for Cell-based Therapy (CTC), University of São Paulo, São Paulo, Brazil
- Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, São Paulo, 14051-140, Brazil
| | - Pablo Diego Moço
- Ribeirão Preto Medical School and Center for Cell-based Therapy (CTC), University of São Paulo, São Paulo, Brazil
- Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, São Paulo, 14051-140, Brazil
| | - Cristiano Gonçalves Pereira
- School of Economics, Business Administration and Accounting at Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Geciane Silveira Porto
- School of Economics, Business Administration and Accounting at Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Elisa Maria de Sousa Russo
- Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, São Paulo, 14051-140, Brazil
- Ribeirão Preto Pharmaceutical Sciences School, University of São Paulo, São Paulo, Brazil
| | - Luiza Cunha Junqueira Reis
- Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, São Paulo, 14051-140, Brazil
- Ribeirão Preto Pharmaceutical Sciences School, University of São Paulo, São Paulo, Brazil
| | - Dimas Tadeu Covas
- Ribeirão Preto Medical School and Center for Cell-based Therapy (CTC), University of São Paulo, São Paulo, Brazil
- Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, São Paulo, 14051-140, Brazil
| | - Virginia Picanço-Castro
- Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, São Paulo, 14051-140, Brazil.
| |
Collapse
|
33
|
Câmara DAD, Mambelli LI, Porcacchia AS, Kerkis I. Advances and Challenges on Cancer Cells Reprogramming Using Induced Pluripotent Stem Cells Technologies. J Cancer 2016; 7:2296-2303. [PMID: 27994667 PMCID: PMC5166540 DOI: 10.7150/jca.16629] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 09/18/2016] [Indexed: 12/18/2022] Open
Abstract
Cancer cells transformation into a normal state or into a cancer cell population which is less tumorigenic than the initial one is a challenge that has been discussed during last decades and it is still far to be solved. Due to the highly heterogeneous nature of cancer cells, such transformation involves many genetic and epigenetic factors which are specific for each type of tumor. Different methods of cancer cells reprogramming have been established and can represent a possibility to obtain less tumorigenic or even normal cells. These methods are quite complex, thus a simple and efficient method of reprogramming is still required. As soon as induced pluripotent stem cells (iPSC) technology, which allowed to reprogram terminally differentiated cells into embryonic stem cells (ESC)-like, was developed, the method strongly attracted the attention of researches, opening new perspectives for stem cell (SC) personalized therapies and offering a powerful in vitro model for drug screening. This technology is also used to reprogram cancer cells, thus providing a modern platform to study cancer-related genes and the interaction between these genes and the cell environment before and after reprogramming, in order to elucidate the mechanisms of cancer initiation and progression. The present review summarizes recent advances on cancer cells reprogramming using iPSC technology and shows the progress achieved in such field.
Collapse
Affiliation(s)
- Diana Aparecida Dias Câmara
- Laboratory of Genetics, Butantan Institute
- Department of Morphology and Genetics, Universidade Federal de Sao Paulo, Sao Paulo, SP, Brazil
| | | | | | | |
Collapse
|
34
|
Yao L, Chen R, Wang P, Zhang Q, Tang H, Sun H. Generation of induced pluripotent stem cells with high efficiency from human embryonic renal cortical cells. Am J Transl Res 2016; 8:4982-4993. [PMID: 27904699 PMCID: PMC5126341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 10/11/2016] [Indexed: 06/06/2023]
Abstract
Reprogramming of somatic cells into induced pluripotent stem cells (iPSCs) emerges as a prospective therapeutic angle in regenerative medicine and a tool for drug screening. Although increasing numbers of iPSCs from different sources have been generated, there has been limited progress in yield of iPSC. Here, we show that four Yamanaka factors Oct4, Sox2, Klf4 and c-Myc can convert human embryonic renal cortical cells (hERCCs) to pluripotent stem cells with a roughly 40-fold higher reprogramming efficiency compared with that of adult human dermal fibroblasts. These iPSCs show pluripotency in vitro and in vivo, as evidenced by expression of pluripotency associated genes, differentiation into three embryonic germ layers by teratoma tests, as well as neuronal fate specification by embryoid body formation. Moreover, the four exogenous genes are effectively silenced in these iPSCs. This study highlights the use of hERCCs to generate highly functional human iPSCs which may aid the study of genetic kidney diseases and accelerate the development of cell-based regenerative therapy.
Collapse
Affiliation(s)
- Ling Yao
- The Lab of Traditional Chinese Medicine, Chongqing Medical UniversityChongqing 400016, China
| | - Ruifang Chen
- Department of Obstetrics & Gynecology, Obstetrics & Gynecology Hospital, Fudan UniversityShanghai 200011, China
| | - Pu Wang
- Department of Neurosurgery, Huashan Hospital, Fudan UniversityShanghai 200040, China
| | - Qi Zhang
- Department of Dermatology, Huashan Hospital, Fudan UniversityShanghai 200040, China
| | - Hailiang Tang
- Department of Neurosurgery, Huashan Hospital, Fudan UniversityShanghai 200040, China
| | - Huaping Sun
- Department of Radiology, Huashan Hospital, Fudan UniversityShanghai 200040, China
| |
Collapse
|
35
|
Youssef AA, Ross EG, Bolli R, Pepine CJ, Leeper NJ, Yang PC. The Promise and Challenge of Induced Pluripotent Stem Cells for Cardiovascular Applications. JACC Basic Transl Sci 2016; 1:510-523. [PMID: 28580434 PMCID: PMC5451899 DOI: 10.1016/j.jacbts.2016.06.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The recent discovery of human-induced pluripotent stem cells (iPSCs) has revolutionized the field of stem cells. iPSCs have demonstrated that biological development is not an irreversible process and that mature adult somatic cells can be induced to become pluripotent. This breakthrough is projected to advance our current understanding of many disease processes and revolutionize the approach to effective therapeutics. Despite the great promise of iPSCs, many translational challenges still remain. In this article, we review the basic concept of induction of pluripotency as a novel approach to understand cardiac regeneration, cardiovascular disease modeling and drug discovery. We critically reflect on the current results of preclinical and clinical studies using iPSCs for these applications with appropriate emphasis on the challenges facing clinical translation.
Collapse
Affiliation(s)
- Amr A Youssef
- Division of Cardiology, Ain Shams University, Cairo, Egypt and Aurora Bay Area Medical Center, Marinette, Wisconsin, USA
| | - Elsie Gyang Ross
- Division of Cardiovascular Medicine and Vascular Surgery, Stanford University, California, USA
| | - Roberto Bolli
- Division of Cardiovascular Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Carl J Pepine
- Division of Cardiovascular Medicine, University of Florida, Gainesville, Florida, USA
| | - Nicholas J Leeper
- Division of Cardiovascular Medicine and Vascular Surgery, Stanford University, California, USA
| | - Phillip C Yang
- Division of Cardiovascular Medicine, Stanford University, California, USA
| |
Collapse
|
36
|
iPS Cells-The Triumphs and Tribulations. Dent J (Basel) 2016; 4:dj4020019. [PMID: 29563461 PMCID: PMC5851259 DOI: 10.3390/dj4020019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 05/18/2016] [Accepted: 05/27/2016] [Indexed: 12/12/2022] Open
Abstract
The year 2006 will be remembered monumentally in science, particularly in the stem cell biology field, for the first instance of generation of induced pluripotent stem cells (iPSCs) from mouse embryonic/adult fibroblasts being reported by Takahashi and Yamanaka. A year later, human iPSCs (hiPSCs) were generated from adult human skin fibroblasts by using quartet of genes, Oct4, Sox2, Klf4, and c-Myc. This revolutionary technology won Yamanaka Nobel Prize in Physiology and Medicine in 2012. Like human embryonic stem cells (hESCs), iPSCs are pluripotent and have the capability for self-renewal. Moreover, complications of immune rejection for therapeutic applications would be greatly eliminated by generating iPSCs from individual patients. This has enabled their use for drug screening/discovery and disease modelling in vitro; and for immunotherapy and regenerative cellular therapies in vivo, paving paths for new therapeutics. Although this breakthrough technology has a huge potential, generation of these unusual cells is still slow, ineffectual, fraught with pitfalls, and unsafe for human use. In this review, I describe how iPSCs are being triumphantly used to lay foundation for a fully functional discipline of regenerative dentistry and medicine, alongside discussing the challenges of translating therapies into clinics. I also discuss their future implications in regenerative dentistry field.
Collapse
|
37
|
Korpusik A, Kolev M. Single injection of CD8+ T lymphocytes derived from hematopoietic stem cells - Mathematical and numerical insights. Biosystems 2016; 144:46-54. [PMID: 27095371 DOI: 10.1016/j.biosystems.2016.04.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Revised: 04/01/2016] [Accepted: 04/14/2016] [Indexed: 12/22/2022]
Abstract
Recently, hematopoietic stem cell (HSC) based therapy is being discussed as a possible treatment for HIV infection. The main advantage of this approach is that it limits the immune impairing effect of infection by introducing an independent influx of antigen-specific cytotoxic T lymphocytes (CTL). In this paper, we present a mathematical approach to predict the dynamics of HSC based therapy. We use a modification of a basic mathematical model for virus induced impairment of help to study how virus - immune system dynamics can be influenced by a single injection of CD8+ T lymphocytes derived from hematopoietic stem cells. Our mathematical and numerical results indicate that a single, large enough dose of genetically derived CTL may lead to restoration of the cellular immune response and result in long-term control of infection.
Collapse
Affiliation(s)
- Adam Korpusik
- Faculty of Technical Sciences, University of Warmia and Mazury, ul. Oczapowskiego 11, 10-719 Olsztyn, Poland.
| | - Mikhail Kolev
- Faculty of Mathematics and Computer Science, University of Warmia and Mazury, ul. Słoneczna 54, 10-710 Olsztyn, Poland.
| |
Collapse
|
38
|
Saito H, Okita K, Chang AE, Ito F. Adoptive Transfer of CD8+ T Cells Generated from Induced Pluripotent Stem Cells Triggers Regressions of Large Tumors Along with Immunological Memory. Cancer Res 2016; 76:3473-83. [PMID: 27197199 DOI: 10.1158/0008-5472.can-15-1742] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 03/29/2016] [Indexed: 12/12/2022]
Abstract
Current approaches to adoptive T-cell therapy are limited by the difficulty of obtaining sufficient numbers of T cells against targeted antigens with useful in vivo characteristics. Theoretically, this limitation could be overcome by using induced pluripotent stem cells (iPSC) that could provide an unlimited source of autologous T cells. However, the therapeutic efficacy of iPSC-derived regenerated T cells remains to be demonstrated. Here, we report the first successful reprogramming of T-cell receptor (TCR) transgenic CD8(+) T cells into pluripotency. As part of the work, we established a syngeneic mouse model for evaluating in vitro and in vivo antitumor reactivity of regenerated T cells from iPSCs bearing a rearranged TCR of known antigen specificity. Stably TCR retained T-cell-derived iPSCs differentiated into CD4(+)CD8(+) T cells that expressed CD3 and the desired TCR in vitro Stimulation of iPSC-derived CD4(+)CD8(+) T cells with the cognate antigen in the presence of IL7 and IL15 followed by expansion with IL2, IL7, and IL15 generated large numbers of less-differentiated CD8(+) T cells with antigen-specific potent cytokine production and cytolytic capacity. Furthermore, adoptively transferred iPSC-derived CD8(+) T cells escaped immune rejection, mediated effective regression of large tumors, improved survival, and established antigen-specific immunological memory. Our findings illustrate the translational potential of iPSCs to provide an unlimited number of phenotypically defined, functional, and expandable autologous antigen-specific T cells with the characteristics needed to enable in vivo effectiveness. Cancer Res; 76(12); 3473-83. ©2016 AACR.
Collapse
Affiliation(s)
- Hidehito Saito
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Keisuke Okita
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Alfred E Chang
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Fumito Ito
- Department of Surgery, University of Michigan, Ann Arbor, Michigan. Department of Surgical Oncology, Roswell Park Cancer Institute, Buffalo, New York. Center for Immunotherapy, Roswell Park Cancer Institute, Buffalo, New York.
| |
Collapse
|
39
|
Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes Afford New Opportunities in Inherited Cardiovascular Disease Modeling. Cardiol Res Pract 2016; 2016:3582380. [PMID: 27110425 PMCID: PMC4826691 DOI: 10.1155/2016/3582380] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 03/03/2016] [Indexed: 01/09/2023] Open
Abstract
Fundamental studies of molecular and cellular mechanisms of cardiovascular disease pathogenesis are required to create more effective and safer methods of their therapy. The studies can be carried out only when model systems that fully recapitulate pathological phenotype seen in patients are used. Application of laboratory animals for cardiovascular disease modeling is limited because of physiological differences with humans. Since discovery of induced pluripotency generating induced pluripotent stem cells has become a breakthrough technology in human disease modeling. In this review, we discuss a progress that has been made in modeling inherited arrhythmias and cardiomyopathies, studying molecular mechanisms of the diseases, and searching for and testing drug compounds using patient-specific induced pluripotent stem cell-derived cardiomyocytes.
Collapse
|
40
|
Tang ZH, Chen JR, Zheng J, Shi HS, Ding J, Qian XD, Zhang C, Chen JL, Wang CC, Li L, Chen JZ, Yin SK, Huang TS, Chen P, Guan MX, Wang JF. Genetic Correction of Induced Pluripotent Stem Cells From a Deaf Patient With MYO7A Mutation Results in Morphologic and Functional Recovery of the Derived Hair Cell-Like Cells. Stem Cells Transl Med 2016; 5:561-71. [PMID: 27013738 DOI: 10.5966/sctm.2015-0252] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 12/22/2015] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED The genetic correction of induced pluripotent stem cells (iPSCs) induced from somatic cells of patients with sensorineural hearing loss (caused by hereditary factors) is a promising method for its treatment. The correction of gene mutations in iPSCs could restore the normal function of cells and provide a rich source of cells for transplantation. In the present study, iPSCs were generated from a deaf patient with compound heterozygous MYO7A mutations (c.1184G>A and c.4118C>T; P-iPSCs), the asymptomatic father of the patient (MYO7A c.1184G>A mutation; CF-iPSCs), and a normal donor (MYO7A(WT/WT); C-iPSCs). One of MYO7A mutation sites (c.4118C>T) in the P-iPSCs was corrected using CRISPR/Cas9. The corrected iPSCs (CP-iPSCs) retained cell pluripotency and normal karyotypes. Hair cell-like cells induced from CP-iPSCs showed restored organization of stereocilia-like protrusions; moreover, the electrophysiological function of these cells was similar to that of cells induced from C-iPSCs and CF-iPSCs. These results might facilitate the development of iPSC-based gene therapy for genetic disorders. SIGNIFICANCE Induced pluripotent stem cells (iPSCs) were generated from a deaf patient with compound heterozygous MYO7A mutations (c.1184G>A and c.4118C>T). One of the MYO7A mutation sites (c.4118C>T) in the iPSCs was corrected using CRISPR/Cas9. The genetic correction of MYO7A mutation resulted in morphologic and functional recovery of hair cell-like cells derived from iPSCs. These findings confirm the hypothesis that MYO7A plays an important role in the assembly of stereocilia into stereociliary bundles. Thus, the present study might provide further insight into the pathogenesis of sensorineural hearing loss and facilitate the development of therapeutic strategies against monogenic disease through the genetic repair of patient-specific iPSCs.
Collapse
Affiliation(s)
- Zi-Hua Tang
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Jia-Rong Chen
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Jing Zheng
- Institute of Genetics, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Hao-Song Shi
- Department of Otorhinolaryngology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital Shanghai, People's Republic of China
| | - Jie Ding
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Xiao-Dan Qian
- The Affiliated Women's Hospital, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Cui Zhang
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Jian-Ling Chen
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Cui-Cui Wang
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Liang Li
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Jun-Zhen Chen
- Department of Otolaryngology, The Affiliated Wenling People's Hospital, Wenzhou Medical University, Wenling, Zhejiang, People's Republic of China
| | - Shan-Kai Yin
- Department of Otorhinolaryngology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital Shanghai, People's Republic of China
| | - Tao-Sheng Huang
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Ping Chen
- Departments of Cell Biology and Otolaryngology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Min-Xin Guan
- Institute of Genetics, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Jin-Fu Wang
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| |
Collapse
|
41
|
Qin Y, Hutson C, Wu X, Xu J, Carroll D. Potential Cancer Prevention and Treatment by Silencing the Killer Cell Immunoglobulin-like Receptor Gene in Natural Killer Cells Derived from Induced Pluripotent Stem Cells. ENLIVEN. JOURNAL OF STEM CELL RESEARCH & REGENERATIVE MEDICINE 2016; 3:002. [PMID: 28845462 PMCID: PMC5568640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Cancer immunosurveillance is an important host protection process, monitoring the presence of irregular cells that could potentially transform into tumor cells, effectively clearing the body of transformed tumor cells at their earliest stages, and thus maintaining regular cellular homeostasis. Natural killer (NK) cells are effector lymphocytes of the innate immune system, playing a critical role in surveillance for tumor cells, while also eliminating virally infected cells. The significance of the anti-tumor role of NK cells was recently further verified by findings that immunosuppression in most cancer patients is not perceptible until late stages. NK cells express the low-affinity Fc-activating receptor, CD16, and the inhibitory receptor, killer cell immunoglobulin-like receptor (KIR). Consequently, activation of NK cells is determined by the balance of inhibitory and activating receptor stimulation. Here, we propose establishing an induced pluripotent stem cell (iPSC)-derived NK cell line with KIR gene knockout or knockdown as a possible regimen to treat and prevent cancer. We further postulate that an optimal mixture of NK iPSCs with and without KIR gene knockout, would reach a maximum antitumor activity, with minimal side effects. We also discuss the possible advantages of KIR-knockout NK iPSCs for adoptive immunotherapy in patients with cancer.
Collapse
Affiliation(s)
- Yunlong Qin
- Poxvirus & Rabies Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging & Zoonotic Infectious Diseases, U.S. Centers for Disease Control & Prevention, 1600 Clifton Rd. NE Mailstop G-06, Atlanta GA, 30333
| | - Christina Hutson
- Poxvirus & Rabies Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging & Zoonotic Infectious Diseases, U.S. Centers for Disease Control & Prevention, 1600 Clifton Rd. NE Mailstop G-06, Atlanta GA, 30333
| | - Xianfu Wu
- Poxvirus & Rabies Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging & Zoonotic Infectious Diseases, U.S. Centers for Disease Control & Prevention, 1600 Clifton Rd. NE Mailstop G-06, Atlanta GA, 30333
| | - Jingyao Xu
- Cancer Biology Program, Georgia Cancer Center for Excellence, Department of Obstetrics and gynecology, Morehouse School of Medicine, 80 Jesse Hill Jr. Dr., Atlanta, GA 30303
| | - Darin Carroll
- Poxvirus & Rabies Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging & Zoonotic Infectious Diseases, U.S. Centers for Disease Control & Prevention, 1600 Clifton Rd. NE Mailstop G-06, Atlanta GA, 30333
| |
Collapse
|
42
|
Induced Pluripotent Stem Cell as a New Source for Cancer Immunotherapy. GENETICS RESEARCH INTERNATIONAL 2016; 2016:3451807. [PMID: 27019752 PMCID: PMC4785259 DOI: 10.1155/2016/3451807] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 01/21/2016] [Accepted: 01/24/2016] [Indexed: 12/13/2022]
Abstract
The immune system consists of cells, proteins, and other molecules that beside each other have a protective function for the host against foreign pathogens. One of the most essential features of the immune system is distinguishability between self- and non-self-cells. This function has an important role in limiting development and progression of cancer cells. In this case, the immune system can detect tumor cell as a foreign pathogen; so, it can be effective in elimination of tumors in their early phases of development. This ability of the immune system resulted in the development of a novel therapeutic field for cancer treatment using host immune components which is called cancer immunotherapy. The main purpose of cancer immunotherapy is stimulation of a strong immune response against the tumor cells that can result from expressing either the immune activator cytokines in the tumor area or gene-modified immune cells. Because of the problems of culturing and manipulating immune cells ex vivo, in recent years, embryonic stem cell (ESC) and induced pluripotent stem cell (iPSC) have been used as new sources for generation of modified immune stimulatory cells. In this paper, we reviewed some of the progressions in iPSC technology for cancer immunotherapy.
Collapse
|
43
|
Sackett SD, Brown ME, Tremmel DM, Ellis T, Burlingham WJ, Odorico JS. Modulation of human allogeneic and syngeneic pluripotent stem cells and immunological implications for transplantation. Transplant Rev (Orlando) 2016; 30:61-70. [PMID: 26970668 DOI: 10.1016/j.trre.2016.02.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 02/05/2016] [Indexed: 01/20/2023]
Abstract
Tissues derived from induced pluripotent stem cells (iPSCs) are a promising source of cells for building various regenerative medicine therapies; from simply transplanting cells to reseeding decellularized organs to reconstructing multicellular tissues. Although reprogramming strategies for producing iPSCs have improved, the clinical use of iPSCs is limited by the presence of unique human leukocyte antigen (HLA) genes, the main immunologic barrier to transplantation. In order to overcome the immunological hurdles associated with allogeneic tissues and organs, the generation of patient-histocompatible iPSCs (autologous or HLA-matched cells) provides an attractive platform for personalized medicine. However, concerns have been raised as to the fitness, safety and immunogenicity of iPSC derivatives because of variable differentiation potential of different lines and the identification of genetic and epigenetic aberrations that can occur during the reprogramming process. In addition, significant cost and regulatory barriers may deter commercialization of patient specific therapies in the short-term. Nonetheless, recent studies provide some evidence of immunological benefit for using autologous iPSCs. Yet, more studies are needed to evaluate the immunogenicity of various autologous and allogeneic human iPSC-derived cell types as well as test various methods to abrogate rejection. Here, we present perspectives of using allogeneic vs. autologous iPSCs for transplantation therapies and the advantages and disadvantages of each related to differentiation potential, immunogenicity, genetic stability and tumorigenicity. We also review the current literature on the immunogenicity of syngeneic iPSCs and discuss evidence that questions the feasibility of HLA-matched iPSC banks. Finally, we will discuss emerging methods of abrogating or reducing host immune responses to PSC derivatives.
Collapse
Affiliation(s)
- S D Sackett
- Division of Transplantation, Department of Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - M E Brown
- Division of Transplantation, Department of Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - D M Tremmel
- Division of Transplantation, Department of Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - T Ellis
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - W J Burlingham
- Division of Transplantation, Department of Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - J S Odorico
- Division of Transplantation, Department of Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
44
|
Karagiannis P, Iriguchi S, Kaneko S. Reprogramming away from the exhausted T cell state. Semin Immunol 2016; 28:35-44. [DOI: 10.1016/j.smim.2015.10.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 10/23/2015] [Accepted: 10/29/2015] [Indexed: 02/07/2023]
|
45
|
Abstract
Various types of somatic cells can be reprogrammed to induced pluripotent stem (iPS) cells. Somatic stem cells may generate iPS cells more efficiently than do differentiated cells. We show that granulosa cells exhibit characteristic of somatic stem cells and can be reprogrammed to iPS cells more efficiently or with few factors. Here, we describe generation of mouse and pig iPS cells from granulosa cells with high efficiency.
Collapse
Affiliation(s)
- Jian Mao
- State Key Laboratory of Medicinal Chemical Biology, Department of Cell Biology and Genetics, College of Life Sciences, Nankai University, 96 Weijin Road, Tianjin, 300071, China
| | - Lin Liu
- State Key Laboratory of Medicinal Chemical Biology, Department of Cell Biology and Genetics, College of Life Sciences, Nankai University, 96 Weijin Road, Tianjin, 300071, China.
| |
Collapse
|
46
|
Kim S, Kim MK, Oh D, Lee SH, Kim B. Induced Pluripotent Stem Cells as a Novel Tool in Psychiatric Research. Psychiatry Investig 2016; 13:8-17. [PMID: 26766942 PMCID: PMC4701689 DOI: 10.4306/pi.2016.13.1.8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 05/11/2015] [Accepted: 06/26/2015] [Indexed: 12/19/2022] Open
Abstract
Reprogramming of somatic cells into induced pluripotent stem cells (iPSCs) provides a valuable opportunity to study neurodevelopmental and neurodegenerative psychiatric diseases by offering an unlimited source for patient-specific neuronal and glial cells. The present review focuses on the recent advancements in modeling psychiatric disorders such as Phelan-McDermid syndrome, Timothy syndrome, Rett syndrome, schizophrenia, bipolar disorder, and dementia. The treatment effects identified in studies on iPSCs using known therapeutic compounds are also summarized in this review. Here we discuss validation of cellular models and explore iPSCs as a novel drug screening tool. Although there are several limitations associated with the current methods used to study mental disorders, using iPSCs as a model system provides the advantage of rewinding and reviewing the development and degeneration of human neural cells.
Collapse
Affiliation(s)
- Sewoong Kim
- Department of Psychiatry, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Min-Kyoung Kim
- Department of Psychiatry, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Daeyoung Oh
- Department of Psychiatry, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Sang-Hyuk Lee
- Department of Psychiatry, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Borah Kim
- Department of Psychiatry, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| |
Collapse
|
47
|
Reprogramming of Melanoma Tumor-Infiltrating Lymphocytes to Induced Pluripotent Stem Cells. Stem Cells Int 2015; 2016:8394960. [PMID: 27057178 PMCID: PMC4707343 DOI: 10.1155/2016/8394960] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 10/01/2015] [Accepted: 10/01/2015] [Indexed: 12/31/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) derived from somatic cells of patients hold great promise for autologous cell therapies. One of the possible applications of iPSCs is to use them as a cell source for producing autologous lymphocytes for cell-based therapy against cancer. Tumor-infiltrating lymphocytes (TILs) that express programmed cell death protein-1 (PD-1) are tumor-reactive T cells, and adoptive cell therapy with autologous TILs has been found to achieve durable complete response in selected patients with metastatic melanoma. Here, we describe the derivation of human iPSCs from melanoma TILs expressing high level of PD-1 by Sendai virus-mediated transduction of the four transcription factors, OCT3/4, SOX2, KLF4, and c-MYC. TIL-derived iPSCs display embryonic stem cell-like morphology, have normal karyotype, express stem cell-specific surface antigens and pluripotency-associated transcription factors, and have the capacity to differentiate in vitro and in vivo. A wide variety of T cell receptor gene rearrangement patterns in TIL-derived iPSCs confirmed the heterogeneity of T cells infiltrating melanomas. The ability to reprogram TILs containing patient-specific tumor-reactive repertoire might allow the generation of patient- and tumor-specific polyclonal T cells for cancer immunotherapy.
Collapse
|
48
|
Habela CW, Song H, Ming GL. Modeling synaptogenesis in schizophrenia and autism using human iPSC derived neurons. Mol Cell Neurosci 2015; 73:52-62. [PMID: 26655799 DOI: 10.1016/j.mcn.2015.12.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 11/17/2015] [Accepted: 12/01/2015] [Indexed: 02/08/2023] Open
Abstract
Schizophrenia (SCZ) and autism spectrum disorder (ASD) are genetically and phenotypically complex disorders of neural development. Human genetic studies, as well as studies examining structural changes at the cellular level, have converged on glutamatergic synapse formation, function, and maintenance as common pathophysiologic substrates involved in both disorders. Synapses as basic functional units of the brain are continuously modified by experience throughout life, therefore they are particularly attractive candidates for targeted therapy. Until recently we lacked a system to evaluate dynamic changes that lead to synaptic abnormalities. With the development of techniques to generate induced pluripotent stem cells (iPSCs) from patients, we are now able to study neuronal and synaptic development in cells from individual patients in the context of genetic changes conferring disease susceptibility. In this review, we discuss recent studies focusing on neural cells differentiated from SCZ and ASD patient iPSCs. These studies support a central role for glutamatergic synapse formation and function in both disorders and demonstrate that iPSC derived neurons offer a potential system for further evaluation of processes leading to synaptic dysregulation and for the design and screening of future therapies.
Collapse
Affiliation(s)
- Christa W Habela
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Hongjun Song
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Guo-Li Ming
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
49
|
Korhonen P, Kanninen KM, Lehtonen Š, Lemarchant S, Puttonen KA, Oksanen M, Dhungana H, Loppi S, Pollari E, Wojciechowski S, Kidin I, García-Berrocoso T, Giralt D, Montaner J, Koistinaho J, Malm T. Immunomodulation by interleukin-33 is protective in stroke through modulation of inflammation. Brain Behav Immun 2015; 49:322-36. [PMID: 26111431 DOI: 10.1016/j.bbi.2015.06.013] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 06/05/2015] [Accepted: 06/18/2015] [Indexed: 01/19/2023] Open
Abstract
Cerebral stroke induces massive Th1-shifted inflammation both in the brain and the periphery, contributing to the outcome of stroke. A Th1-type response is neurotoxic whereas a Th2-type response is accompanied by secretion of anti-inflammatory cytokines, such as interleukin-4 (IL-4). Interleukin-33 (IL-33) is a cytokine known to induce a shift towards the Th2-type immune response, polarize macrophages/microglia towards the M2-type, and induce production of anti-inflammatory cytokines. We found that the plasma levels of the inhibitory IL-33 receptor, sST2, are increased in human stroke and correlate with a worsened stroke outcome, suggesting an insufficient IL-33-driven Th2-type response. In mouse, peripheral administration of IL-33 reduced stroke-induced cell death and improved the sensitivity of the contralateral front paw at 5days post injury. The IL-33-treated mice had increased levels of IL-4 in the spleen and in the peri-ischemic area of the cortex. Neutralization of IL-4 by administration of an IL-4 antibody partially prevented the IL-33-mediated protection. IL-33 treatment also reduced astrocytic activation in the peri-ischemic area and increased the number of Arginase-1 immunopositive microglia/macrophages at the lesion site. In human T-cells, IL-33 treatment induced IL-4 secretion, and the conditioned media from IL-33-exposed T-cells reduced astrocytic activation. This study demonstrates that IL-33 is protective against ischemic insult by induction of IL-4 secretion and may represent a novel therapeutic approach for the treatment of stroke.
Collapse
Affiliation(s)
- Paula Korhonen
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Katja M Kanninen
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Šárka Lehtonen
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Sighild Lemarchant
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Katja A Puttonen
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Minna Oksanen
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Hiramani Dhungana
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Sanna Loppi
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Eveliina Pollari
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Sara Wojciechowski
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Iurii Kidin
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Teresa García-Berrocoso
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron, 119-129, 08035 Barcelona, Spain
| | - Dolors Giralt
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron, 119-129, 08035 Barcelona, Spain
| | - Joan Montaner
- Neurovascular Unit, Department of Neurology, Vall d'Hebron Hospital, Barcelona, Spain
| | - Jari Koistinaho
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland.
| | - Tarja Malm
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| |
Collapse
|
50
|
Abstract
Periodontitis is a chronic inflammatory disease which leads to destruction of both the soft and hard tissues of the periodontium. Tissue engineering is a therapeutic approach in regenerative medicine that aims to induce new functional tissue regeneration via the synergistic combination of cells, biomaterials, and/or growth factors. Advances in our understanding of the biology of stem cells, including embryonic stem cells and mesenchymal stem cells, have provided opportunities for periodontal tissue engineering. However, there remain a number of limitations affecting their therapeutic efficiency. Due to the considerable proliferation and differentiation capacities, recently described induced pluripotent stem cells (iPSCs) provide a new way for cell-based therapies for periodontal regeneration. This review outlines the latest status of periodontal tissue engineering and highlights the potential use of iPSCs in periodontal tissue regeneration.
Collapse
Affiliation(s)
- Mi Du
- Shandong provincial key laboratory of oral tissue regeneration, Department of Periodontology, School of Stomatology, Shandong University, No.44-1 West Wenhua Rd., Jinan, 250012 People's Republic of China
| | - Xuejing Duan
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong University, No.324 Jingwu Rd., Jinan, 250000 People's Republic of China
| | - Pishan Yang
- Shandong provincial key laboratory of oral tissue regeneration, Department of Periodontology, School of Stomatology, Shandong University, No.44-1 West Wenhua Rd., Jinan, 250012 People's Republic of China
| |
Collapse
|