1
|
Piovani G, Ferraro RM, Giliani SC. Establishment and characterization of Cri Du Chat neuronal stem cells: a novel promising resource to study the syndrome. Hum Cell 2025; 38:98. [PMID: 40343585 PMCID: PMC12064636 DOI: 10.1007/s13577-025-01230-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 05/02/2025] [Indexed: 05/11/2025]
Abstract
The Cri Du Chat (CdC) Syndrome is a rare chromosome disease condition resulting from variable size deletion occurring on the short arm of one of the chromosomes 5. This disorder, which affects one in 50,000 births, is responsible for developmental retardation, the mechanism of which has remained unexplained. TERT, SEMA5 A, CTNND2, TPPP, mapped in chromosome 5 short arm, are known to be expressed in the brain, and to play a role in the development of the nervous system, oligodentrocytes and in the regulation of glutamatergic and dopaminergic synaptic transmission. It is critical to understand how their haploinsufficiency might affect the development and presentation of the disease. In the absence of an animal model and of significant accessible, human tissue, human pluripotent stem cells (iPSC) directly reprogrammed from patient somatic cells open a new area of disease modeling as they can virtually be differentiated into any cell type. Our study reports, for the first time, the generation of neuronal stem cells (NSCs) from CdC-iPSCs line and in addition, subsequent differentiation into a heterogeneous population of neurons. Gene expression of the mentioned and single copy deleted genes was also evaluated by comparing their expression level in iPSC, NSCs and neuron lines. The present research represents the first and the most innovative approach, to create an in vitro CdC neuronal model to have a new translational framework to study the pathologic processes.
Collapse
Affiliation(s)
- Giovanna Piovani
- Department of Molecular and Translational Medicine, University of Brescia, 25123, Brescia, Italy.
- Scientific Committee of A.B.C. Associazione Bambini Cri du Chat, 50026, Florence, Italy.
| | - Rosalba Monica Ferraro
- Department of Molecular and Translational Medicine, University of Brescia, 25123, Brescia, Italy
- "A. Nocivelli" Institute for Molecular Medicine, Department of Molecular and Translational Medicine, University of Brescia, 25123, Brescia, Italy
| | - Silvia Clara Giliani
- Department of Molecular and Translational Medicine, University of Brescia, 25123, Brescia, Italy
- "A. Nocivelli" Institute for Molecular Medicine, Department of Molecular and Translational Medicine, University of Brescia, 25123, Brescia, Italy
| |
Collapse
|
2
|
Gavrilov NS, Ignatyeva NV, Medvedeva EV, Timashev PS. Articular cartilage tissue engineering using genetically modified induced pluripotent stem cell lines. GENES & CELLS 2024; 19:404-424. [DOI: 10.17816/gc633492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2025]
Abstract
Mature hyaline cartilage has a low regenerative potential and its repair remains a complex clinical and research issue. Articular cartilage injuries often contribute to the development of osteoarthritis, resulting in loss of joint function and patient disability. Surgical techniques for repairing articular surfaces, such as mosaic chondroplasty and microfracture, which are designed for small defects, cannot be used for degenerative and dystrophic cartilage lesions. Cell therapy using chondrocytes differentiated from induced pluripotent stem cells (iPSCs) is a promising approach to reconstruct articular cartilage tissue. iPSCs have high proliferative activity, which allows the harvesting of autologous cells in quantities necessary to repair a joint defect. CRISPR-Cas genome editing technology, based on the bacterial adaptive immune system, enables the genetic modification of iPSCs to obtain progenitor cells with specific characteristics and properties.
This review describes specific research papers on the combined use of iPSC and CRISPR-Cas technologies for the evaluation of cartilage regenerative medicine. Papers were evaluated for the last twelve years since CRISPR-Cas technology was introduced to the global community. CRISPR-Cas is currently being used to address therapeutic issues in articular cartilage regeneration by increasing the efficiency of chondrogenic differentiation of iPSC lines and harvesting a more homogeneous population of chondroprogenitor cells. Another approach is to remove the pro-inflammatory cytokine receptor sequence to produce inflammation-resistant cartilage. Finally, knocking out genes for components of the major histocompatibility complex allows harvesting chondrocytes that are invisible to the recipient's immune system. This kind of research contributes to personalized healthcare and can improve the quality of life of the world's population in the long term.
Collapse
|
3
|
Hassan OI, Takamiya S, Asgarihafshejani A, Fehlings MG. Bridging the gap: a translational perspective in spinal cord injury. Exp Biol Med (Maywood) 2024; 249:10266. [PMID: 39391076 PMCID: PMC11464315 DOI: 10.3389/ebm.2024.10266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 08/27/2024] [Indexed: 10/12/2024] Open
Abstract
Traumatic spinal cord injury (SCI) is a devastating and complex condition to treat with no curative options. In the past few decades, rapid advancements in our understanding of SCI pathophysiology as well as the mergence of new treatments has created more optimism. Focusing on clinical translation, this paper provides a comprehensive overview of SCI through its epidemiology, pathophysiology, currently employed management strategies, and emerging therapeutic approaches. Additionally, it emphasizes the importance of addressing the heavy quality of life (QoL) challenges faced by SCI patients and their desires, providing a basis to tailor patient-centric forms of care. Furthermore, this paper discusses the frequently encountered barriers in translation from preclinical models to clinical settings. It also seeks to summarize significant completed and ongoing SCI clinical trials focused on neuroprotective and neuroregenerative strategies. While developing a cohesive regenerative treatment strategy remains challenging, even modest improvements in sensory and motor function can offer meaningful benefits and motivation for patients coping with this highly debilitating condition.
Collapse
Affiliation(s)
- Omar Imad Hassan
- Division of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Soichiro Takamiya
- Division of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, ON, Canada
| | - Azam Asgarihafshejani
- Division of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, ON, Canada
| | - Michael G. Fehlings
- Division of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Division of Neurosurgery, Krembil Neuroscience Centre, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
- Division of Neurosurgery and Spine Program, Department of Surgery, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
4
|
Novoa J, Westra I, Steeneveld E, Neves NF, Daleman L, Asensio AB, Davis RP, Carlotti F, Freund C, Rabelink T, Meij P, Wieles B. Validating human induced pluripotent stem cell-specific quality control tests for the release of an intermediate drug product in a Good Manufacturing Practice quality system. Cytotherapy 2024; 26:1105-1117. [PMID: 38703154 DOI: 10.1016/j.jcyt.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/29/2024] [Accepted: 04/10/2024] [Indexed: 05/06/2024]
Abstract
One of the challenges in Good Manufacturing Practice (GMP)-compliant human induced pluripotent stem cell (hiPSC) production is the validation of quality control (QC) tests specific for hiPSCs, which are required for GMP batch release. This study presents a comprehensive description of the validation process for hiPSC-specific GMP-compliant QC assays; more specifically, the validation of assays to assess the potential presence of residual episomal vectors (REVs), the expression of markers of the undifferentiated state and the directed differentiation potential of hiPSCs. Critical aspects and specific acceptance criteria were formulated in a validation plan prior to assay validation. Assay specificity, sensitivity and reproducibility were tested, and the equipment used for each assay was subjected to performance qualification. A minimum input of 20 000 cells (120 ng of genomic DNA) was defined for accurate determination of the presence of REVs. Furthermore, since vector loss in hiPSC lines is a passage-dependent process, we advocate screening for REVs between passages eight and 10, as testing at earlier passages might lead to unnecessary rejection of hiPSC lines. The cutoff value for assessment of markers of the undifferentiated state was set to the expression of at least three individual markers on at least 75% of the cells. When multi-color flow cytometry panels are used, a fluorescence minus one control is advised to ensure the control for fluorescent spread. For the assay to assess the directed differentiation potential, the detection limit was set to two of three positive lineage-specific markers for each of the three individual germ layers. All of our assays proved to be reproducible and specific. Our data demonstrate that our implemented analytical procedures are suitable as QC assays for the batch release of GMP-compliant hiPSCs.
Collapse
Affiliation(s)
- Juan Novoa
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Inge Westra
- Center for Cell and Gene Therapy, Leiden University Medical Center, Leiden, the Netherlands
| | - Esther Steeneveld
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Natascha Fonseca Neves
- Center for Cell and Gene Therapy, Leiden University Medical Center, Leiden, the Netherlands
| | - Lizanne Daleman
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Albert Blanch Asensio
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands; Novo Nordisk Foundation Center for Stem Cell Medicine, Leiden University Medical Center, the Netherlands
| | - Richard P Davis
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands; Novo Nordisk Foundation Center for Stem Cell Medicine, Leiden University Medical Center, the Netherlands
| | - Françoise Carlotti
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Christian Freund
- Leiden University Medical Center hiPSC Hotel, Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ton Rabelink
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands; Novo Nordisk Foundation Center for Stem Cell Medicine, Leiden University Medical Center, the Netherlands.
| | - Pauline Meij
- Center for Cell and Gene Therapy, Leiden University Medical Center, Leiden, the Netherlands
| | - Brigitte Wieles
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
5
|
Han X, Cai C, Deng W, Shi Y, Li L, Wang C, Zhang J, Rong M, Liu J, Fang B, He H, Liu X, Deng C, He X, Cao X. Landscape of human organoids: Ideal model in clinics and research. Innovation (N Y) 2024; 5:100620. [PMID: 38706954 PMCID: PMC11066475 DOI: 10.1016/j.xinn.2024.100620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 03/29/2024] [Indexed: 05/07/2024] Open
Abstract
In the last decade, organoid research has entered a golden era, signifying a pivotal shift in the biomedical landscape. The year 2023 marked a milestone with the publication of thousands of papers in this arena, reflecting exponential growth. However, amid this burgeoning expansion, a comprehensive and accurate overview of the field has been conspicuously absent. Our review is intended to bridge this gap, providing a panoramic view of the rapidly evolving organoid landscape. We meticulously analyze the organoid field from eight distinctive vantage points, harnessing our rich experience in academic research, industrial application, and clinical practice. We present a deep exploration of the advances in organoid technology, underpinned by our long-standing involvement in this arena. Our narrative traverses the historical genesis of organoids and their transformative impact across various biomedical sectors, including oncology, toxicology, and drug development. We delve into the synergy between organoids and avant-garde technologies such as synthetic biology and single-cell omics and discuss their pivotal role in tailoring personalized medicine, enhancing high-throughput drug screening, and constructing physiologically pertinent disease models. Our comprehensive analysis and reflective discourse provide a deep dive into the existing landscape and emerging trends in organoid technology. We spotlight technological innovations, methodological evolution, and the broadening spectrum of applications, emphasizing the revolutionary influence of organoids in personalized medicine, oncology, drug discovery, and other fields. Looking ahead, we cautiously anticipate future developments in the field of organoid research, especially its potential implications for personalized patient care, new avenues of drug discovery, and clinical research. We trust that our comprehensive review will be an asset for researchers, clinicians, and patients with keen interest in personalized medical strategies. We offer a broad view of the present and prospective capabilities of organoid technology, encompassing a wide range of current and future applications. In summary, in this review we attempt a comprehensive exploration of the organoid field. We offer reflections, summaries, and projections that might be useful for current researchers and clinicians, and we hope to contribute to shaping the evolving trajectory of this dynamic and rapidly advancing field.
Collapse
Affiliation(s)
- Xinxin Han
- Organ Regeneration X Lab, Lisheng East China Institute of Biotechnology, Peking University, Jiangsu 226200, China
- Shanghai Lisheng Biotech, Shanghai 200092, China
| | - Chunhui Cai
- Shanghai Lisheng Biotech, Shanghai 200092, China
| | - Wei Deng
- LongHua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wanping South Road, Xuhui District, Shanghai 200032, China
- Department of Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
| | - Yanghua Shi
- Shanghai Lisheng Biotech, Shanghai 200092, China
| | - Lanyang Li
- Shanghai Lisheng Biotech, Shanghai 200092, China
| | - Chen Wang
- Shanghai Lisheng Biotech, Shanghai 200092, China
| | - Jian Zhang
- Shanghai Lisheng Biotech, Shanghai 200092, China
| | - Mingjie Rong
- Shanghai Lisheng Biotech, Shanghai 200092, China
| | - Jiping Liu
- Shanghai Lisheng Biotech, Shanghai 200092, China
| | - Bangjiang Fang
- LongHua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wanping South Road, Xuhui District, Shanghai 200032, China
| | - Hua He
- Department of Neurosurgery, Third Affiliated Hospital, Naval Medical University, Shanghai 200438, China
| | - Xiling Liu
- Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Academy of Forensic Science, Ministry of Justice, Shanghai 200063, China
| | - Chuxia Deng
- Cancer Center, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
- Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR 999078, China
| | - Xiao He
- CAS Key Lab for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Xin Cao
- Zhongshan Hospital Institute of Clinical Science, Fudan University Shanghai Medical College, Shanghai 200032, China
| |
Collapse
|
6
|
Roman A, Huntemer-Silveira A, Waldron MA, Khalid Z, Blake J, Parr AM, Low WC. Cell Transplantation for Repair of the Spinal Cord and Prospects for Generating Region-Specific Exogenic Neuronal Cells. Cell Transplant 2024; 33:9636897241241998. [PMID: 38590295 PMCID: PMC11005494 DOI: 10.1177/09636897241241998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 03/05/2024] [Accepted: 03/11/2024] [Indexed: 04/10/2024] Open
Abstract
Spinal cord injury (SCI) is associated with currently irreversible consequences in several functional components of the central nervous system. Despite the severity of injury, there remains no approved treatment to restore function. However, with a growing number of preclinical studies and clinical trials, cell transplantation has gained significant potential as a treatment for SCI. Researchers have identified several cell types as potential candidates for transplantation. To optimize successful functional outcomes after transplantation, one key factor concerns generating neuronal cells with regional and subtype specificity, thus calling on the developmental transcriptome patterning of spinal cord cells. A potential source of spinal cord cells for transplantation is the generation of exogenic neuronal progenitor cells via the emerging technologies of gene editing and blastocyst complementation. This review highlights the use of cell transplantation to treat SCI in the context of relevant developmental gene expression patterns useful for producing regionally specific exogenic spinal cells via in vitro differentiation and blastocyst complementation.
Collapse
Affiliation(s)
- Alex Roman
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Department of Neurosurgery, Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Anne Huntemer-Silveira
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Madison A. Waldron
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Zainab Khalid
- Department of Neurosurgery, Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Jeffrey Blake
- Department of Neurosurgery, Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Ann M. Parr
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Department of Neurosurgery, Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Walter C. Low
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Department of Neurosurgery, Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
7
|
Meng C, Huang S, Cheng T, Zhang X, Yan X. Induction of Salivary Gland-Like Tissue by Induced Pluripotent Stem Cells In Vitro. Tissue Eng Regen Med 2022; 19:389-401. [PMID: 35171451 PMCID: PMC8971325 DOI: 10.1007/s13770-021-00402-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/17/2021] [Accepted: 09/22/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND To investigate the in vitro induction of salivary gland-like tissue by ips cells in an interferon regulatory factor 6 (IRF6) overexpression and parotid conditioned medium environment. METHODS Urine-derived ips cells were isolated, identified, transfected with IRF6 and cultured in parotid conditioned medium to induce ips cells into salivary gland differentiation, morphological changes of ips cells were observed, CCK-8 was used to determine the cell proliferation efficiency and transcriptome sequencing was used to detect the expression of genes related to parotid gland formation. RESULTS Immunofluorescence staining showed that the isolated ips cells were positive for NANOG, SSEA4 and OCT4 and had embryonic-like stem cell characteristics; CCK-8 showed that there was no statistical difference in the proliferation efficiency between the IRF6+ induced group and the simple induced group after induction of ips cells into salivary glands. The results of transcriptome sequencing showed that there were a total of 643 differentially expressed genes, including 365 up-regulated genes and 278 down-regulated genes in the IRF6+ induced group compared to the blank control group, and the salivary gland related genes HAPLN1, CCL2, MSX2, ANXA1, CYP11A1, HES1 and LUM were all highly expressed in the IRF6+ induced group. CONCLUSION IRF6 promotes salivary gland differentiation in urine-derived iPSCs, and its mechanism of promoting differentiation may be that IRF6 upregulates the expression of HAPLN1, CCL2, MSX2, ANXA1, CYP11A1, HES1 and LUM to promote epithelial differentiation.
Collapse
Affiliation(s)
- Cen Meng
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Shengyuan Huang
- Department of Stomatology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Taiqi Cheng
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xue Zhang
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xing Yan
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
8
|
Chakrabarty K, Shetty R, Argulwar S, Das D, Ghosh A. Induced pluripotent stem cell-based disease modeling and prospective immune therapy for coronavirus disease 2019. Cytotherapy 2021; 24:235-248. [PMID: 34656419 PMCID: PMC8437760 DOI: 10.1016/j.jcyt.2021.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/14/2021] [Accepted: 08/14/2021] [Indexed: 11/30/2022]
Abstract
The emergence of the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic poses a never before seen challenge to human health and the economy. Considering its clinical impact, with no streamlined therapeutic strategies in sight, it is crucial to understand the infection process of SARS-CoV-2. Our limited knowledge of the mechanisms underlying SARS-CoV-2 infection impedes the development of alternative therapeutics to address the pandemic. This aspect can be addressed by modeling SARS-CoV-2 infection in the human context to facilitate drug screening and discovery. Human induced pluripotent stem cell (iPSC)-derived lung epithelial cells and organoids recapitulating the features and functionality of the alveolar cell types can serve as an in vitro human model and screening platform for SARS-CoV-2. Recent studies suggest an immune system asynchrony leading to compromised function and a decreased proportion of specific immune cell types in coronavirus disease 2019 (COVID-19) patients. Replenishing these specific immune cells may serve as useful treatment modality against SARS-CoV-2 infection. Here the authors review protocols for deriving lung epithelial cells, alveolar organoids and specific immune cell types, such as T lymphocytes and natural killer cells, from iPSCs with the aim to aid investigators in making relevant in vitro models of SARS-CoV-2 along with the possibility derive immune cell types to treat COVID-19.
Collapse
Affiliation(s)
| | - Rohit Shetty
- Cornea and Refractive Surgery, Narayana Nethralaya, Bangalore, India
| | - Shubham Argulwar
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, India
| | - Debashish Das
- Stem Cell Research Laboratory, GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, India
| | - Arkasubhra Ghosh
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, India
| |
Collapse
|
9
|
Uçkan-Çetinkaya D, Haider KH. Induced Pluripotent Stem Cells in Pediatric Research and Clinical Translation. Stem Cells 2021. [DOI: 10.1007/978-3-030-77052-5_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
10
|
New Frontiers or the Treatment of Interstitial Cystitis/Bladder Pain Syndrome - Focused on Stem Cells, Platelet-Rich Plasma, and Low-Energy Shock Wave. Int Neurourol J 2020; 24:211-221. [PMID: 33017892 PMCID: PMC7538293 DOI: 10.5213/inj.2040104.052] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 05/29/2020] [Indexed: 12/22/2022] Open
Abstract
Interstitial cystitis/bladder pain syndrome (IC/BPS), which is characterized by bladder pain and irritative voiding symptoms, is a frustrating disease without effective treatment. The cause is still largely not understood, although urothelium ischemia/hypoxia, apoptosis, denudation, and infiltration of inflammatory cells are common histopathological findings. The current uncertainty regarding the etiology and pathology of IC/BPS has a negative impact on its timely and successful treatment; therefore, the development of new treatment modalities is urgently needed. Herein, we present advances in our knowledge on this topic and review the potential application of regenerative medicine for the treatment of IC/BPS. This article provides information on the basic characteristics and clinical evidence of stem cells, platelet-rich plasma (PRP), and low-energy shock waves (LESWs) based on a literature review with a search strategy for articles related to IC/BPS, stem cells, PRP, and LESW published in MEDLINE and PubMed. Stem cells, PRP, and LESW, which modulate inflammatory processes and promote tissue repair, have been proven to improve bladder regeneration, relieve bladder pain, inhibit bladder inflammation, and increase bladder capacity in some preclinical studies. However, clinical studies are still in their infancy. Based on the mechanisms of action of stem cells, PRP, and LESW documented in many preclinical studies, the potential applications of regenerative medicine for the treatment of IC/BPS is an emerging frontier of interest. However, solid evidence from clinical studies remains to be obtained.
Collapse
|
11
|
Abstract
Derivation of induced Pluripotent Stem Cells (iPSCs) by reprogramming somatic cells to a pluripotent state has revolutionized stem cell research. Ensuing this, various groups have used genetic and non-genetic approaches to generate iPSCs from numerous cell types. However, achieving a pluripotent state in most of the reprogramming studies is marred by serious limitations such as low reprogramming efficiency and slow kinetics. These limitations are mainly due to the presence of potent barriers that exist during reprogramming when a mature cell is coaxed to achieve a pluripotent state. Several studies have revealed that intrinsic factors such as non-optimal stoichiometry of reprogramming factors, specific signaling pathways, cellular senescence, pluripotency-inhibiting transcription factors and microRNAs act as a roadblock. In addition, the epigenetic state of somatic cells and specific epigenetic modifications that occur during reprogramming also remarkably impede the generation of iPSCs. In this review, we present a comprehensive overview of the barriers that inhibit reprogramming and the understanding of which will pave the way to develop safe strategies for efficient reprogramming.
Collapse
|
12
|
Abstract
Stem cells are capable of self-renewal and differentiation into a range of cell types and promote the release of chemokines and progenitor cells necessary for tissue regeneration. Mesenchymal stem cells are multipotent progenitor cells with enhanced proliferation and differentiation capabilities and less tumorigenicity than conventional adult stem cells; these cells are also easier to acquire. Bladder dysfunction is often chronic in nature with limited treatment modalities due to its undetermined pathophysiology. Most treatments focus on symptom alleviation rather than pathognomonic changes repair. The potential of stem cell therapy for bladder dysfunction has been reported in preclinical models for stress urinary incontinence, overactive bladder, detrusor underactivity, and interstitial cystitis/bladder pain syndrome. Despite these findings, however, stem cell therapy is not yet available for clinical use. Only one pilot study on detrusor underactivity and a handful of clinical trials on stress urinary incontinence have reported the effects of stem cell treatment. This limitation may be due to stem cell function loss following ex vivo expansion, poor in vivo engraftment or survival after transplantation, or a lack of understanding of the precise mechanisms of action underlying therapeutic outcomes and in vivo behavior of stem cells administered to target organs. Efficacy comparisons with existing treatment modalities are also needed for the successful clinical application of stem cell therapies. This review describes the current status of stem cell research on treating bladder dysfunction and suggests future directions to facilitate clinical applications of this promising treatment modality, particularly for bladder dysfunction.
Collapse
|
13
|
Pareja E, Gómez-Lechón MJ, Tolosa L. Induced pluripotent stem cells for the treatment of liver diseases: challenges and perspectives from a clinical viewpoint. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:566. [PMID: 32775367 PMCID: PMC7347783 DOI: 10.21037/atm.2020.02.164] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The only curative treatment for severe end-stage liver disease (ESLD) is liver transplantation (LT) but it is limited by the shortage of organ donors. The increase of the incidence of liver disease has led to develop new therapeutic approaches such as liver cell transplantation. Current challenges that limit a wider application of this therapy include a limited cell source and the poor engraftment in the host liver of cryopreserved hepatocytes after thawing. Induced pluripotent stem cells (iPSCs) that can be differentiated into hepatocyte-like cells (HLCs) are being widely explored as an alternative to human hepatocytes because of their unlimited proliferation capacity and their potential ability to avoid the immune system. Their large-scale production could provide a new tool to produce enough HLCs for treating patients with metabolic diseases, acute liver failure (ALF), those with ESLD or patients not considered for organ transplantation. In this review we discuss current challenges for generating differentiated cells compatible with human application as well as in-depth safety evaluation. This analysis highlights the uncertainties and deficiencies that should be addressed before their clinical use but also points out the potential benefits that will produce a great impact in the field of hepatology.
Collapse
Affiliation(s)
- Eugenia Pareja
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe, Valencia, Spain.,Unidad Hepatobiliopancreáctica, Hospital Universitario Doctor Peset, Valencia, Spain
| | - M José Gómez-Lechón
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe, Valencia, Spain.,CIBERehd, ISCIII, Madrid, Spain
| | - Laia Tolosa
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| |
Collapse
|
14
|
Andrejew R, Glaser T, Oliveira-Giacomelli Á, Ribeiro D, Godoy M, Granato A, Ulrich H. Targeting Purinergic Signaling and Cell Therapy in Cardiovascular and Neurodegenerative Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1201:275-353. [PMID: 31898792 DOI: 10.1007/978-3-030-31206-0_14] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Extracellular purines exert several functions in physiological and pathophysiological mechanisms. ATP acts through P2 receptors as a neurotransmitter and neuromodulator and modulates heart contractility, while adenosine participates in neurotransmission, blood pressure, and many other mechanisms. Because of their capability to differentiate into mature cell types, they provide a unique therapeutic strategy for regenerating damaged tissue, such as in cardiovascular and neurodegenerative diseases. Purinergic signaling is pivotal for controlling stem cell differentiation and phenotype determination. Proliferation, differentiation, and apoptosis of stem cells of various origins are regulated by purinergic receptors. In this chapter, we selected neurodegenerative and cardiovascular diseases with clinical trials using cell therapy and purinergic receptor targeting. We discuss these approaches as therapeutic alternatives to neurodegenerative and cardiovascular diseases. For instance, promising results were demonstrated in the utilization of mesenchymal stem cells and bone marrow mononuclear cells in vascular regeneration. Regarding neurodegenerative diseases, in general, P2X7 and A2A receptors mostly worsen the degenerative state. Stem cell-based therapy, mainly through mesenchymal and hematopoietic stem cells, showed promising results in improving symptoms caused by neurodegeneration. We propose that purinergic receptor activity regulation combined with stem cells could enhance proliferative and differentiation rates as well as cell engraftment.
Collapse
Affiliation(s)
- Roberta Andrejew
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Talita Glaser
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Ágatha Oliveira-Giacomelli
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Deidiane Ribeiro
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Mariana Godoy
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil.,Laboratory of Neurodegenerative Diseases, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alessandro Granato
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Henning Ulrich
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
15
|
Tan DC, Roth IM, Wickremesekera AC, Davis PF, Kaye AH, Mantamadiotis T, Stylli SS, Tan ST. Therapeutic Targeting of Cancer Stem Cells in Human Glioblastoma by Manipulating the Renin-Angiotensin System. Cells 2019; 8:cells8111364. [PMID: 31683669 PMCID: PMC6912312 DOI: 10.3390/cells8111364] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 10/23/2019] [Accepted: 10/29/2019] [Indexed: 12/11/2022] Open
Abstract
Patients with glioblastoma (GB), a highly aggressive brain tumor, have a median survival of 14.6 months following neurosurgical resection and adjuvant chemoradiotherapy. Quiescent GB cancer stem cells (CSCs) invariably cause local recurrence. These GB CSCs can be identified by embryonic stem cell markers, express components of the renin-angiotensin system (RAS) and are associated with circulating CSCs. Despite the presence of circulating CSCs, GB patients rarely develop distant metastasis outside the central nervous system. This paper reviews the current literature on GB growth inhibition in relation to CSCs, circulating CSCs, the RAS and the novel therapeutic approach by repurposing drugs that target the RAS to improve overall symptom-free survival and maintain quality of life.
Collapse
Affiliation(s)
- David Ch Tan
- Department of Neurosurgery, Wellington Regional Hospital, Wellington 6021, New Zealand.
| | - Imogen M Roth
- Gillies McIndoe Research Institute, Wellington 6021, New Zealand.
| | - Agadha C Wickremesekera
- Department of Neurosurgery, Wellington Regional Hospital, Wellington 6021, New Zealand.
- Gillies McIndoe Research Institute, Wellington 6021, New Zealand.
- Department of Surgery, The University of Melbourne, Parkville, Victoria 3050, Australia.
| | - Paul F Davis
- Gillies McIndoe Research Institute, Wellington 6021, New Zealand.
| | - Andrew H Kaye
- Department of Surgery, The University of Melbourne, Parkville, Victoria 3050, Australia.
- Department of Neurosurgery, Hadassah Hebrew University Medical Centre, Jerusalem 91120, Israel.
| | - Theo Mantamadiotis
- Department of Surgery, The University of Melbourne, Parkville, Victoria 3050, Australia.
| | - Stanley S Stylli
- Department of Surgery, The University of Melbourne, Parkville, Victoria 3050, Australia.
- Department of Neurosurgery, The Royal Melbourne Hospital, Parkville, Victoria 3050, Australia.
| | - Swee T Tan
- Gillies McIndoe Research Institute, Wellington 6021, New Zealand.
- Department of Surgery, The University of Melbourne, Parkville, Victoria 3050, Australia.
- Wellington Regional Plastic, Maxillofacial & Burns Unit, Hutt Hospital, Lower Hutt 5040, New Zealand.
| |
Collapse
|
16
|
Trawczynski M, Liu G, David BT, Fessler RG. Restoring Motor Neurons in Spinal Cord Injury With Induced Pluripotent Stem Cells. Front Cell Neurosci 2019; 13:369. [PMID: 31474833 PMCID: PMC6707336 DOI: 10.3389/fncel.2019.00369] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 07/29/2019] [Indexed: 12/14/2022] Open
Abstract
Spinal cord injury (SCI) is a devastating neurological disorder that damages motor, sensory, and autonomic pathways. Recent advances in stem cell therapy have allowed for the in vitro generation of motor neurons (MNs) showing electrophysiological and synaptic activity, expression of canonical MN biomarkers, and the ability to graft into spinal lesions. Clinical translation, especially the transplantation of MN precursors in spinal lesions, has thus far been elusive because of stem cell heterogeneity and protocol variability, as well as a hostile microenvironment such as inflammation and scarring, which yield inconsistent pre-clinical results without a consensus best-practice therapeutic strategy. Induced pluripotent stem cells (iPSCs) in particular have lower ethical and immunogenic concerns than other stem cells, which could make them more clinically applicable. In this review, we focus on the differentiation of iPSCs into neural precursors, MN progenitors, mature MNs, and MN subtype fates. Previous reviews have summarized MN development and differentiation, but an up-to-date summary of technological and experimental advances holding promise for bench-to-bedside translation, especially those targeting individual MN subtypes in SCI, is currently lacking. We discuss biological mechanisms of MN lineage, recent experimental protocols and techniques for MN differentiation from iPSCs, and transplantation of neural precursors and MN lineage cells in spinal cord lesions to restore motor function. We emphasize efficient, clinically safe, and personalized strategies for the application of MN and their subtypes as therapy in spinal lesions.
Collapse
Affiliation(s)
- Matthew Trawczynski
- Department of Neurosurgery, Rush University Medical Center, Chicago, IL, United States
| | - Gele Liu
- Department of Neurosurgery, Rush University Medical Center, Chicago, IL, United States
| | - Brian T David
- Department of Neurosurgery, Rush University Medical Center, Chicago, IL, United States
| | - Richard G Fessler
- Department of Neurosurgery, Rush University Medical Center, Chicago, IL, United States
| |
Collapse
|
17
|
Dolatshad H, Tatwavedi D, Ahmed D, Tegethoff JF, Boultwood J, Pellagatti A. Application of induced pluripotent stem cell technology for the investigation of hematological disorders. Adv Biol Regul 2019; 71:19-33. [PMID: 30341008 DOI: 10.1016/j.jbior.2018.10.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 10/08/2018] [Accepted: 10/09/2018] [Indexed: 06/08/2023]
Abstract
Induced pluripotent stem cells (iPSCs) were first described over a decade ago and are currently used in various basic biology and clinical research fields. Recent advances in the field of human iPSCs have opened the way to a better understanding of the biology of human diseases. Disease-specific iPSCs provide an unparalleled opportunity to establish novel human cell-based disease models, with the potential to enhance our understanding of the molecular mechanisms underlying human malignancies, and to accelerate the identification of effective new drugs. When combined with genome editing technologies, iPSCs represent a new approach to study single or multiple disease-causing mutations and model specific diseases in vitro. In addition, genetically corrected patient-specific iPSCs could potentially be used for stem cell based therapy. Furthermore, the reprogrammed cells share patient-specific genetic background, offering a new platform to develop personalized therapy/medicine for patients. In this review we discuss the recent advances in iPSC research technology and their potential applications in hematological diseases. Somatic cell reprogramming has presented new routes for generating patient-derived iPSCs, which can be differentiated to hematopoietic stem cells and the various downstream hematopoietic lineages. iPSC technology shows promise in the modeling of both inherited and acquired hematological disorders. A direct reprogramming and differentiation strategy is able to recapitulate hematological disorder progression and capture the earliest molecular alterations that underlie the initiation of hematological malignancies.
Collapse
Affiliation(s)
- Hamid Dolatshad
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and Oxford BRC Haematology Theme, Oxford, UK
| | - Dharamveer Tatwavedi
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and Oxford BRC Haematology Theme, Oxford, UK
| | - Doaa Ahmed
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and Oxford BRC Haematology Theme, Oxford, UK; Clinical Pathology Department, Assiut University Hospitals, Faculty of Medicine, Assiut, Egypt
| | - Jana F Tegethoff
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and Oxford BRC Haematology Theme, Oxford, UK
| | - Jacqueline Boultwood
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and Oxford BRC Haematology Theme, Oxford, UK
| | - Andrea Pellagatti
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and Oxford BRC Haematology Theme, Oxford, UK.
| |
Collapse
|
18
|
Page S, Patel R, Raut S, Al-Ahmad A. Neurological diseases at the blood-brain barrier: Stemming new scientific paradigms using patient-derived induced pluripotent cells. Biochim Biophys Acta Mol Basis Dis 2018; 1866:165358. [PMID: 30593893 DOI: 10.1016/j.bbadis.2018.12.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 11/15/2018] [Accepted: 12/05/2018] [Indexed: 02/08/2023]
Abstract
The blood-brain barrier (BBB) is a component of the neurovascular unit formed by specialized brain microvascular endothelial cells (BMECs) surrounded by a specific basement membrane interacting with astrocytes, neurons, and pericytes. The BBB plays an essential function in the maintenance of brain homeostasis, by providing a physical and chemical barrier against pathogens and xenobiotics. Although the disruption of the BBB occurs with several neurological disorders, the scarcity of patient material source and lack of reliability of current in vitro models hindered our ability to model the BBB during such neurological conditions. The development of novel in vitro models based on patient-derived stem cells opened new venues in modeling the human BBB in vitro, by being more accurate than existing in vitro models, but also bringing such models closer to the in vivo setting. In addition, patient-derived models of the BBB opens the avenue to address the contribution of genetic factors commonly associated with certain neurological diseases on the BBB pathophysiology. This review provides a comprehensive understanding of the BBB, the current development of stem cell-based models in the field, the current challenges and limitations of such models.
Collapse
Affiliation(s)
- Shyanne Page
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, School of Pharmacy, Amarillo, TX, United States of America
| | - Ronak Patel
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, School of Pharmacy, Amarillo, TX, United States of America
| | - Snehal Raut
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, School of Pharmacy, Amarillo, TX, United States of America
| | - Abraham Al-Ahmad
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, School of Pharmacy, Amarillo, TX, United States of America.
| |
Collapse
|
19
|
Application of induced pluripotent stem cell transplants: Autologous or allogeneic? Life Sci 2018; 212:145-149. [DOI: 10.1016/j.lfs.2018.09.057] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 09/30/2018] [Indexed: 12/14/2022]
|
20
|
Colasuonno F, Borghi R, Niceforo A, Muzzi M, Bertini E, Di Giulio A, Moreno S, Compagnucci C. Senescence-associated ultrastructural features of long-term cultures of induced pluripotent stem cells (iPSCs). Aging (Albany NY) 2018; 9:2209-2222. [PMID: 29064821 PMCID: PMC5680563 DOI: 10.18632/aging.101309] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Accepted: 10/15/2017] [Indexed: 12/12/2022]
Abstract
Induced pluripotent stem cells (iPSCs) hold great promise for developing personalized regenerative medicine, however characterization of their biological features is still incomplete. Moreover, changes occurring in long-term cultured iPSCs have been reported, suggesting these as a model of cellular aging. For this reason, we addressed the ultrastructural characterization of iPSCs, with a focus on possible time-dependent changes, involving specific cell compartments. To this aim, we comparatively analysed cultures at different timepoints, by an innovative electron microscopic technology (FIB/SEM). We observed progressive loss of cell-to-cell contacts, associated with increased occurrence of exosomes. Mitochondria gradually increased, while acquiring an elongated shape, with well-developed cristae. Such mitochondrial maturation was accompanied by their turnover, as assessed by the presence of autophagomes (undetectable in young iPSCs), some containing recognizable mitochondria. This finding was especially frequent in middle-aged iPSCs, while being occasional in aged cells, suggesting early autophagic activation followed by a decreased efficiency of the process with culturing time. Accordingly, confocal microscopy showed age-dependent alterations to the expression and distribution of autophagic markers. Interestingly, responsivity to rapamycin, highest in young iPSCs, was almost lost in aged cells. Overall, our results strongly support long-term cultured iPSCs as a model for studying relevant aspects of cellular senescence, involving intercellular communication, energy metabolism, and autophagy.
Collapse
Affiliation(s)
- Fiorella Colasuonno
- Department of Science, LIME, University "Roma Tre", Rome 00146, Italy.,Department of Neuroscience, Unit of Neuromuscular and Neurodegenerative Diseases, Laboratory of Molecular Medicine, Bambino Gesu' Children's Research Hospital, IRCCS, Rome 00146, Italy
| | - Rossella Borghi
- Department of Science, LIME, University "Roma Tre", Rome 00146, Italy.,Department of Neuroscience, Unit of Neuromuscular and Neurodegenerative Diseases, Laboratory of Molecular Medicine, Bambino Gesu' Children's Research Hospital, IRCCS, Rome 00146, Italy
| | - Alessia Niceforo
- Department of Science, LIME, University "Roma Tre", Rome 00146, Italy.,Department of Neuroscience, Unit of Neuromuscular and Neurodegenerative Diseases, Laboratory of Molecular Medicine, Bambino Gesu' Children's Research Hospital, IRCCS, Rome 00146, Italy
| | - Maurizio Muzzi
- Department of Science, LIME, University "Roma Tre", Rome 00146, Italy
| | - Enrico Bertini
- Department of Neuroscience, Unit of Neuromuscular and Neurodegenerative Diseases, Laboratory of Molecular Medicine, Bambino Gesu' Children's Research Hospital, IRCCS, Rome 00146, Italy
| | - Andrea Di Giulio
- Department of Science, LIME, University "Roma Tre", Rome 00146, Italy
| | - Sandra Moreno
- Department of Science, LIME, University "Roma Tre", Rome 00146, Italy
| | - Claudia Compagnucci
- Department of Neuroscience, Unit of Neuromuscular and Neurodegenerative Diseases, Laboratory of Molecular Medicine, Bambino Gesu' Children's Research Hospital, IRCCS, Rome 00146, Italy
| |
Collapse
|
21
|
Zhou H, Wang B, Sun H, Xu X, Wang Y. Epigenetic Regulations in Neural Stem Cells and Neurological Diseases. Stem Cells Int 2018; 2018:6087143. [PMID: 29743892 PMCID: PMC5878882 DOI: 10.1155/2018/6087143] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 01/08/2018] [Indexed: 12/17/2022] Open
Abstract
Among the regulatory mechanisms of the renewal and differentiation of neural stem cells, recent evidences support that epigenetic modifications such as DNA methylation, histone modification, and noncoding RNAs play critical roles in the regulation on the proliferation and differentiation of neural stem cells. In this review, we discussed recent advances of DNA modifications on the regulative mechanisms of neural stem cells. Among these epigenetic modifications, DNA 5-hydroxymethylcytosine (5hmC) modification is emerging as an important modulator on the proliferation and differentiation of neural stem cells. At the same time, Ten-eleven translocation (Tet) methylcytosine dioxygenases, the rate-limiting enzyme for the 5-hydroxymethylation reaction from 5-methylcytosine to 5-hydroxymethylcytosine, play a critical role in the tumorigenesis and the proliferation and differentiation of stem cells. The functions of 5hmC and TET proteins on neural stem cells and their roles in neurological diseases are discussed.
Collapse
Affiliation(s)
- Hang Zhou
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Bin Wang
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Hao Sun
- Department of Orthopedics, Clinical Medical School, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou 225001, China
| | - Xingshun Xu
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Yongxiang Wang
- Department of Orthopedics, Clinical Medical School, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou 225001, China
| |
Collapse
|