1
|
Kagami H, Li X. Spheroids and organoids: Their implications for oral and craniofacial tissue/organ regeneration. J Oral Biol Craniofac Res 2024; 14:540-546. [PMID: 39092136 PMCID: PMC11292544 DOI: 10.1016/j.jobcr.2024.07.002] [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: 08/19/2023] [Revised: 06/09/2024] [Accepted: 07/01/2024] [Indexed: 08/04/2024] Open
Abstract
Spheroids are spherical aggregates of cells. Normally, most of adherent cells cannot survive in suspension; however, if they adhere to each other and grow to a certain size, they can survive without attaching to the dish surface. Studies have shown that spheroid formation induces dedifferentiation and improves plasticity, proliferative capability, and differentiation capability. In particular, spontaneous spheroids represent a selective and efficient cultivation technique for somatic stem cells. Organoids are considered mini-organs composed of multiple types of cells with extracellular matrices that are maintained in three-dimensional culture. Although their culture environment is similar to that of spheroids, organoids consist of differentiated cells with fundamental tissue/organ structures similar to those of native organs. Organoids have been used for drug development, disease models, and basic biological studies. Spheroid culture has been reported for various cell types in the oral and craniofacial regions, including salivary gland epithelial cells, periodontal ligament cells, dental pulp stem cells, and oral mucosa-derived cells. For broader clinical application, it is crucial to identify treatment targets that can leverage the superior stemness of spheroids. Organoids have been developed from various organs, including taste buds, oral mucosa, teeth, and salivary glands, for basic biological studies and also with the goal to replace damaged or defective organs. The development of novel immune-tolerant cell sources is the key to the widespread clinical application of organoids in regenerative medicine. Further efforts to understand the underlying basic mechanisms of spheroids and organoids will lead to the development of safe and efficient next-generation regenerative therapies.
Collapse
Affiliation(s)
- Hideaki Kagami
- Department of Dentistry and Oral Surgery, Aichi Medical University, Aichi, Japan
| | - Xianqi Li
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Matsumoto Dental University, Shiojiri, 399-0781, Japan
- Department of Hard Tissue Research, Graduate School of Oral Medicine, Matsumoto Dental University, Shiojiri, 399-0781, Japan
| |
Collapse
|
2
|
Soares R, Lourenço DM, Mota IF, Sebastião AM, Xapelli S, Morais VA. Lineage-specific changes in mitochondrial properties during neural stem cell differentiation. Life Sci Alliance 2024; 7:e202302473. [PMID: 38664022 PMCID: PMC11045976 DOI: 10.26508/lsa.202302473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 04/06/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Neural stem cells (NSCs) reside in discrete regions of the adult mammalian brain where they can differentiate into neurons, astrocytes, and oligodendrocytes. Several studies suggest that mitochondria have a major role in regulating NSC fate. Here, we evaluated mitochondrial properties throughout NSC differentiation and in lineage-specific cells. For this, we used the neurosphere assay model to isolate, expand, and differentiate mouse subventricular zone postnatal NSCs. We found that the levels of proteins involved in mitochondrial fusion (Mitofusin [Mfn] 1 and Mfn 2) increased, whereas proteins involved in fission (dynamin-related protein 1 [DRP1]) decreased along differentiation. Importantly, changes in mitochondrial dynamics correlated with distinct patterns of mitochondrial morphology in each lineage. Particularly, we found that the number of branched and unbranched mitochondria increased during astroglial and neuronal differentiation, whereas the area occupied by mitochondrial structures significantly reduced with oligodendrocyte maturation. In addition, comparing the three lineages, neurons revealed to be the most energetically flexible, whereas astrocytes presented the highest ATP content. Our work identified putative mitochondrial targets to enhance lineage-directed differentiation of mouse subventricular zone-derived NSCs.
Collapse
Affiliation(s)
- Rita Soares
- Instituto de Medicina Molecular | João Lobo Antunes (iMM|JLA), Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Biologia Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Diogo M Lourenço
- Instituto de Medicina Molecular | João Lobo Antunes (iMM|JLA), Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Isa F Mota
- Instituto de Medicina Molecular | João Lobo Antunes (iMM|JLA), Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Biologia Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Ana M Sebastião
- Instituto de Medicina Molecular | João Lobo Antunes (iMM|JLA), Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Sara Xapelli
- Instituto de Medicina Molecular | João Lobo Antunes (iMM|JLA), Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Vanessa A Morais
- Instituto de Medicina Molecular | João Lobo Antunes (iMM|JLA), Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Biologia Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
3
|
Sharma K, Puranik N, Yadav D. Neural Stem Cell-based Regenerative Therapy: A New Approach to Diabetes Treatment. Endocr Metab Immune Disord Drug Targets 2024; 24:531-540. [PMID: 37183465 DOI: 10.2174/1871530323666230512121416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 03/14/2023] [Accepted: 03/16/2023] [Indexed: 05/16/2023]
Abstract
Diabetes mellitus (DM) is the most common metabolic disorder that occurs due to the loss, or impaired function of insulin-secreting pancreatic beta cells, which are of two types - type 1 (T1D) and type 2 (T2D). To cure DM, the replacement of the destroyed pancreatic beta cells of islet of Langerhans is the most widely practiced treatment. For this, isolating neuronal stem cells and cultivating them as a source of renewable beta cells is a significant breakthrough in medicine. The functions, growth, and gene expression of insulin-producing pancreatic beta cells and neurons are very similar in many ways. A diabetic patient's neural stem cells (obtained from the hippocampus and olfactory bulb) can be used as a replacement source of beta cells for regenerative therapy to treat diabetes. The same protocol used to create functional neurons from progenitor cells can be used to create beta cells. Recent research suggests that replacing lost pancreatic beta cells with autologous transplantation of insulin-producing neural progenitor cells may be a perfect therapeutic strategy for diabetes, allowing for a safe and normal restoration of function and a reduction in potential risks and a long-term cure.
Collapse
Affiliation(s)
- Kajal Sharma
- School of Sciences in Biotechnology, Jiwaji University, Gwalior, 474011, Madhya Pradesh, India
| | - Nidhi Puranik
- Department of Bio-logical Sciences, Bharathiar University, Tamil Nadu, India
| | - Dhananjay Yadav
- Department of Life Science, Yeungnam University, Gyeongsan, 38541, Korea
| |
Collapse
|
4
|
Reshamwala R, Oieni F, Shah M. Non-stem Cell Mediated Tissue Regeneration and Repair. Regen Med 2023. [DOI: 10.1007/978-981-19-6008-6_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
|
5
|
Epigenomic Profiling of Epithelial Ovarian Cancer Stem-Cell Differentiation Reveals GPD1 Associated Immune Suppressive Microenvironment and Poor Prognosis. Int J Mol Sci 2022; 23:ijms23095120. [PMID: 35563509 PMCID: PMC9101898 DOI: 10.3390/ijms23095120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/03/2022] [Accepted: 05/03/2022] [Indexed: 11/21/2022] Open
Abstract
Intraperitoneal metastasis is a challenging clinical scenario in epithelial ovarian cancer (EOC). As they are distinct from hematogenous metastasizing tumors, epithelial ovarian cancer cells primarily disseminate within the peritoneal cavity to form superficially invasive carcinomas. Unfavorable pharmacokinetics for peritoneal tumors and gut toxicity collectively lead to a narrow therapeutic window and therefore limit the opportunities for a favorable clinical outcome. New insights into tumor metastasis in the peritoneal microenvironment are keenly awaited to develop new therapeutic strategies. Epithelial ovarian cancer stem cell (OCSC) seeding is considered to be a critical component of the peritoneal spread. Using a unique and stepwise process of the OCSC differentiation model may provide insight into the intraperitoneal metastasis. The transcriptome and epigenome of OCSC differentiation were characterized by expression array and MethylCap-Seq. The TCGA, AOCS, and KM-Plotter databases were used to evaluate the association between survival outcomes and the methylation/expression levels of candidate genes in the EOC datasets. The STRING database was used to investigate the protein–protein interaction (PPI) for candidates and their associated genes. The infiltration level of immune cells in EOC patients and the association between clinical outcome and OCSCs differentiation genes were estimated using the TIDE and TIME2.0 algorithms. We established an EOC differentiation model using OCSCs. After an integrated transcriptomics and methylomics analysis of OCSCs differentiation, we revealed that the genes associated with earlier OCSC differentiation were better able to reflect the patient’s outcome. The OCSC differentiation genes were involved in regulating metabolism shift and the suppressive immune microenvironment. High GPD1 expression with high pro-tumorigenic immune cells (M2 macrophage, and cancer associated fibroblast) had worst survival. Moreover, we developed a methylation signature, constituted by GNPDA1, GPD1, GRASP, HOXC11, and MSLN, that may be useful for prognostic prediction in EOC. Our results revealed a novel role of epigenetic plasticity OCSC differentiation and suggested metabolic and immune intervention as a new therapeutic strategy.
Collapse
|
6
|
Regulating Endogenous Neural Stem Cell Activation to Promote Spinal Cord Injury Repair. Cells 2022; 11:cells11050846. [PMID: 35269466 PMCID: PMC8909806 DOI: 10.3390/cells11050846] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 12/12/2022] Open
Abstract
Spinal cord injury (SCI) affects millions of individuals worldwide. Currently, there is no cure, and treatment options to promote neural recovery are limited. An innovative approach to improve outcomes following SCI involves the recruitment of endogenous populations of neural stem cells (NSCs). NSCs can be isolated from the neuroaxis of the central nervous system (CNS), with brain and spinal cord populations sharing common characteristics (as well as regionally distinct phenotypes). Within the spinal cord, a number of NSC sub-populations have been identified which display unique protein expression profiles and proliferation kinetics. Collectively, the potential for NSCs to impact regenerative medicine strategies hinges on their cardinal properties, including self-renewal and multipotency (the ability to generate de novo neurons, astrocytes, and oligodendrocytes). Accordingly, endogenous NSCs could be harnessed to replace lost cells and promote structural repair following SCI. While studies exploring the efficacy of this approach continue to suggest its potential, many questions remain including those related to heterogeneity within the NSC pool, the interaction of NSCs with their environment, and the identification of factors that can enhance their response. We discuss the current state of knowledge regarding populations of endogenous spinal cord NSCs, their niche, and the factors that regulate their behavior. In an attempt to move towards the goal of enhancing neural repair, we highlight approaches that promote NSC activation following injury including the modulation of the microenvironment and parenchymal cells, pharmaceuticals, and applied electrical stimulation.
Collapse
|
7
|
Abstract
Traumatic injury of the central nervous system (CNS) is a worldwide health problem affecting millions of people. Trauma of the CNS, that is, traumatic brain injury (TBI) and spinal cord injury (SCI), lead to massive and progressive cell loss and axonal degeneration, usually with very limited regeneration. At present, there are no treatments to protect injured CNS tissue or to replace the lost tissue. Stem cells are a cell type that by definition can self-renew and give rise to multiple cell lineages. In recent years, therapies using stem and progenitor cells have shown promising effects in experimental CNS trauma, particularly in the acute-subacute stage, but also in chronic injuries. However, the therapeutic mechanisms by which transplanted cells achieve the structural and/or functional improvements are often not clear. Stem cell therapies for CNS trauma can be categorized into 2 main concepts, transplantation of exogenous neural stem cells and neural progenitor cells and recruitment of endogenous stem and progenitor cells. In this review, focusing on the advances during the last decade, we will discuss the major cell therapies, the pros and cons of these 2 concepts for TBI and SCI, and the treatment strategies we believe will be successful.
Collapse
Affiliation(s)
- Xiaofei Li
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Erik Sundström
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Corresponding author: Erik Sundström, Department of Neurobiology, Care Sciences and Society (NVS), BioClinicum J9:20, Karolinska University Hospital, S17164 Solna, Sweden.
| |
Collapse
|
8
|
Kuburich NA, den Hollander P, Deshmukh AP, Vasaikar S, Joseph R, Wicha MS, Mani SA. In Vitro Quantification of Cancer Stem Cells Using a Mammosphere Formation Assay. Methods Mol Biol 2022; 2429:509-513. [PMID: 35507185 DOI: 10.1007/978-1-0716-1979-7_35] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Cancer stem cells (CSCs) are a small subpopulation of self-renewing cancer cells that are present within tumors. In this chapter, we provide a detailed method for the quantification of CSCs in vitro through mammosphere formation.
Collapse
Affiliation(s)
- Nick A Kuburich
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Petra den Hollander
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Abhijeet P Deshmukh
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Suhas Vasaikar
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robiya Joseph
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Max S Wicha
- Forbes Institute for Cancer Discovery, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Sendurai A Mani
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
9
|
Čater M, Majdič G. In Vitro Culturing of Adult Stem Cells: The Importance of Serum and Atmospheric Oxygen. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1376:101-118. [PMID: 34426961 DOI: 10.1007/5584_2021_656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Adult stem cells are undifferentiated cells found in many different tissues in the adult human and animal body and are thought to be important for replacing damaged and dead cells during life. Due to their differentiation abilities, they have significant potential for regeneration and consequently therapeutic potential in various medical conditions. Studies on in vitro cultivation of different types of adult stem cells have shown that they have specific requirements for optimal proliferation and stemness maintenance as well as induced differentiation. The main factors affecting the success of stem cell cultivation are the composition of the growth medium, including the presence of serum, temperature, humidity, and contact with other cells and the composition of the atmosphere in which the cells grow. In this chapter, we review the literature and describe our own experience regarding the influence of the presence of fetal bovine serum in the medium and the oxygen concentration in the atmosphere on the stemness maintenance and survival of adult stem cells from various tissue sources such as adipose tissue, muscle, brain, and testicular tissue.
Collapse
Affiliation(s)
- Maša Čater
- Laboratory for Animal Genomics, Institute for Preclinical Studies, Veterinary faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Gregor Majdič
- Laboratory for Animal Genomics, Institute for Preclinical Studies, Veterinary faculty, University of Ljubljana, Ljubljana, Slovenia. .,Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia.
| |
Collapse
|
10
|
Calinescu AA, Kauss MC, Sultan Z, Al-Holou WN, O'Shea SK. Stem cells for the treatment of glioblastoma: a 20-year perspective. CNS Oncol 2021; 10:CNS73. [PMID: 34006134 PMCID: PMC8162173 DOI: 10.2217/cns-2020-0026] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma, the deadliest form of primary brain tumor, remains a disease without cure. Treatment resistance is in large part attributed to limitations in the delivery and distribution of therapeutic agents. Over the last 20 years, numerous preclinical studies have demonstrated the feasibility and efficacy of stem cells as antiglioma agents, leading to the development of trials to test these therapies in the clinic. In this review we present and analyze these studies, discuss mechanisms underlying their beneficial effect and highlight experimental progress, limitations and the emergence of promising new therapeutic avenues. We hope to increase awareness of the advantages brought by stem cells for the treatment of glioblastoma and inspire further studies that will lead to accelerated implementation of effective therapies. Glioblastoma is the deadliest and most common form of brain tumor, for which there is no cure. It is very difficult to deliver medicine to the tumor cells, because they spread out widely into the normal brain, and local blood vessels represent a barrier that most medicines cannot cross. It was shown, in many studies over the last 20 years, that stem cells are attracted toward the tumor and that they can deliver many kinds of therapeutic agents directly to brain cancer cells and shrink the tumor. In this review we analyze these studies and present new discoveries that can be used to make stem cell therapies for glioblastoma more effective to prolong the life of patients with brain tumors.
Collapse
Affiliation(s)
| | - McKenzie C Kauss
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,College of Literature Science & Arts, University of Michigan, Ann Arbor, MI 48109, USA
| | - Zain Sultan
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,College of Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Wajd N Al-Holou
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Sue K O'Shea
- Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
11
|
Functional Expression of Choline Transporters in Human Neural Stem Cells and Its Link to Cell Proliferation, Cell Viability, and Neurite Outgrowth. Cells 2021; 10:cells10020453. [PMID: 33672580 PMCID: PMC7924032 DOI: 10.3390/cells10020453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/16/2021] [Accepted: 02/18/2021] [Indexed: 01/11/2023] Open
Abstract
Choline and choline metabolites are essential for all cellular functions. They have also been reported to be crucial for neural development. In this work, we studied the functional characteristics of the choline uptake system in human neural stem cells (hNSCs). Additionally, we investigated the effect of extracellular choline uptake inhibition on the cellular activities in hNSCs. We found that the mRNAs and proteins of choline transporter-like protein 1 (CTL1) and CTL2 were expressed at high levels. Immunostaining showed that CTL1 and CTL2 were localized in the cell membrane and partly in the mitochondria, respectively. The uptake of extracellular choline was saturable and performed by a single uptake mechanism, which was Na+-independent and pH-dependent. We conclude that CTL1 is responsible for extracellular choline uptake, and CTL2 may uptake choline in the mitochondria and be involved in DNA methylation via choline oxidation. Extracellular choline uptake inhibition caused intracellular choline deficiency in hNSCs, which suppressed cell proliferation, cell viability, and neurite outgrowth. Our findings contribute to the understanding of the role of choline in neural development as well as the pathogenesis of various neurological diseases caused by choline deficiency or choline uptake impairment.
Collapse
|
12
|
Brooks AC, Henderson BJ. Systematic Review of Nicotine Exposure's Effects on Neural Stem and Progenitor Cells. Brain Sci 2021; 11:172. [PMID: 33573081 PMCID: PMC7912116 DOI: 10.3390/brainsci11020172] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 01/22/2021] [Accepted: 01/24/2021] [Indexed: 12/24/2022] Open
Abstract
While various modalities of chronic nicotine use have been associated with numerous negative consequences to human health, one possible benefit of nicotine exposure has been uncovered. The discovery of an inverse correlation between smoking and Parkinson's disease, and later Alzheimer's disease as well, motivated investigation of nicotine as a neuroprotective agent. Some studies have demonstrated that nicotine elicits improvements in cognitive function. The hippocampus, along with the subventricular zone (SVZ), is a distinct brain region that allow for ongoing postnatal neurogenesis throughout adulthood and plays a major role in certain cognitive behaviors like learning and memory. Therefore, one hypothesis underlying nicotine-induced neuroprotection is possible effects on neural stem cells and neural precursor cells. On the other hand, nicotine withdrawal frequently leads to cognitive impairments, particularly in hippocampal-dependent behaviors, possibly suggesting an impairment of hippocampal neurogenesis with nicotine exposure. This review discusses the current body of evidence on nicotine's effects on neural stem cells and neural progenitors. Changes in neural stem cell proliferation, survival, intracellular dynamics, and differentiation following acute and chronic nicotine exposure are examined.
Collapse
Affiliation(s)
- Arrin C. Brooks
- Department of Biomedical Science, Joan C Edwards School of Medicine, Marshall University, Huntington, WV 25545, USA;
| | | |
Collapse
|
13
|
Frajewicki A, Laštůvka Z, Borbélyová V, Khan S, Jandová K, Janišová K, Otáhal J, Mysliveček J, Riljak V. Perinatal hypoxic-ischemic damage: review of the current treatment possibilities. Physiol Res 2020; 69:S379-S401. [PMID: 33464921 DOI: 10.33549/physiolres.934595] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Neonatal hypoxic-ischemic encephalopathy is a disorder with heterogeneous manifestation due to asphyxia during perinatal period. It affects approximately 3-12 children per 1000 live births and cause death of 1 million neonates worldwide per year. Besides, motor disabilities, seizures, impaired muscle tone and epilepsy are few of the consequences of hypoxic-ischemic encephalopathy. Despite an extensive research effort regarding various treatment strategies, therapeutic hypothermia with intensive care unit supportive treatment remains the only approved method for neonates who have suffered from moderate to severe hypoxic-ischemic encephalopathy. However, these protocols are only partially effective given that many infants still suffer from severe brain damage. Thus, further research to systematically test promising neuroprotective treatments in combination with hypothermia is essential. In this review, we discussed the pathophysiology of hypoxic-ischemic encephalopathy and delved into different promising treatment modalities, such as melatonin and erythropoietin. However, preclinical studies and clinical trials are still needed to further elucidate the mechanisms of action of these modalities.
Collapse
Affiliation(s)
- A Frajewicki
- Institute of Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic.
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Perego MGL, Galli N, Nizzardo M, Govoni A, Taiana M, Bresolin N, Comi GP, Corti S. Current understanding of and emerging treatment options for spinal muscular atrophy with respiratory distress type 1 (SMARD1). Cell Mol Life Sci 2020; 77:3351-3367. [PMID: 32123965 PMCID: PMC11104977 DOI: 10.1007/s00018-020-03492-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 02/08/2020] [Accepted: 02/20/2020] [Indexed: 12/11/2022]
Abstract
Spinal muscular atrophy (SMA) with respiratory distress type 1 (SMARD1) is an autosomal recessive motor neuron disease that is characterized by distal and proximal muscle weakness and diaphragmatic palsy that leads to respiratory distress. Without intervention, infants with the severe form of the disease die before 2 years of age. SMARD1 is caused by mutations in the IGHMBP2 gene that determine a deficiency in the encoded IGHMBP2 protein, which plays a critical role in motor neuron survival because of its functions in mRNA processing and maturation. Although it is rare, SMARD1 is the second most common motor neuron disease of infancy, and currently, treatment is primarily supportive. No effective therapy is available for this devastating disease, although multidisciplinary care has been an essential element of the improved quality of life and life span extension in these patients in recent years. The objectives of this review are to discuss the current understanding of SMARD1 through a summary of the presently known information regarding its clinical presentation and pathogenesis and to discuss emerging therapeutic approaches. Advances in clinical care management have significantly extended the lives of individuals affected by SMARD1 and research into the molecular mechanisms that lead to the disease has identified potential strategies for intervention that target the underlying causes of SMARD1. Gene therapy via gene replacement or gene correction provides the potential for transformative therapies to halt or possibly prevent neurodegenerative disease in SMARD1 patients. The recent approval of the first gene therapy approach for SMA associated with mutations in the SMN1 gene may be a turning point for the application of this strategy for SMARD1 and other genetic neurological diseases.
Collapse
Affiliation(s)
- Martina G L Perego
- Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, University of Milan, Via Francesco Sforza 35, 20122, Milan, Italy
| | - Noemi Galli
- Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, University of Milan, Via Francesco Sforza 35, 20122, Milan, Italy
| | - Monica Nizzardo
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122, Milan, Italy
| | - Alessandra Govoni
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122, Milan, Italy
| | - Michela Taiana
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122, Milan, Italy
| | - Nereo Bresolin
- Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, University of Milan, Via Francesco Sforza 35, 20122, Milan, Italy
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122, Milan, Italy
| | - Giacomo P Comi
- Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, University of Milan, Via Francesco Sforza 35, 20122, Milan, Italy
- Neuromuscular and Rare Diseases Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122, Milan, Italy
| | - Stefania Corti
- Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, University of Milan, Via Francesco Sforza 35, 20122, Milan, Italy.
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122, Milan, Italy.
| |
Collapse
|
15
|
Purvis EM, O'Donnell JC, Chen HI, Cullen DK. Tissue Engineering and Biomaterial Strategies to Elicit Endogenous Neuronal Replacement in the Brain. Front Neurol 2020; 11:344. [PMID: 32411087 PMCID: PMC7199479 DOI: 10.3389/fneur.2020.00344] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 04/07/2020] [Indexed: 12/19/2022] Open
Abstract
Neurogenesis in the postnatal mammalian brain is known to occur in the dentate gyrus of the hippocampus and the subventricular zone. These neurogenic niches serve as endogenous sources of neural precursor cells that could potentially replace neurons that have been lost or damaged throughout the brain. As an example, manipulation of the subventricular zone to augment neurogenesis has become a popular strategy for attempting to replace neurons that have been lost due to acute brain injury or neurodegenerative disease. In this review article, we describe current experimental strategies to enhance the regenerative potential of endogenous neural precursor cell sources by enhancing cell proliferation in neurogenic regions and/or redirecting migration, including pharmacological, biomaterial, and tissue engineering strategies. In particular, we discuss a novel replacement strategy based on exogenously biofabricated "living scaffolds" that could enhance and redirect endogenous neuroblast migration from the subventricular zone to specified regions throughout the brain. This approach utilizes the first implantable, biomimetic tissue-engineered rostral migratory stream, thereby leveraging the brain's natural mechanism for sustained neuronal replacement by replicating the structure and function of the native rostral migratory stream. Across all these strategies, we discuss several challenges that need to be overcome to successfully harness endogenous neural precursor cells to promote nervous system repair and functional restoration. With further development, the diverse and innovative tissue engineering and biomaterial strategies explored in this review have the potential to facilitate functional neuronal replacement to mitigate neurological and psychiatric symptoms caused by injury, developmental disorders, or neurodegenerative disease.
Collapse
Affiliation(s)
- Erin M. Purvis
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
| | - John C. O'Donnell
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
| | - H. Isaac Chen
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
| | - D. Kacy Cullen
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
16
|
High-content imaging of 3D-cultured neural stem cells on a 384-pillar plate for the assessment of cytotoxicity. Toxicol In Vitro 2020; 65:104765. [PMID: 31923580 DOI: 10.1016/j.tiv.2020.104765] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 12/20/2019] [Accepted: 01/05/2020] [Indexed: 12/17/2022]
Abstract
The assessment of neurotoxicity has been performed traditionally with animals. However, in vivo studies are highly expensive and time-consuming, and often do not correlate to human outcomes. Thus, there is a need for cost-effective, high-throughput, highly predictive alternative in vitro test methods based on early markers of mechanisms of toxicity. High-content imaging (HCI) assays performed on three-dimensionally (3D) cultured cells could provide better understanding of the mechanism of toxicity needed to predict neurotoxicity in humans. However, current 3D cell culture systems lack the throughput required for screening neurotoxicity against a large number of chemicals. Therefore, we have developed miniature 3D neural stem cell (NSC) culture on a unique 384-pillar plate, which is complementary to conventional 384-well plates. Mitochondrial membrane impairment, intracellular glutathione level, cell membrane integrity, DNA damage, and apoptosis have been tested against 3D-cultured ReNcell VM on the 384-pillar plate with four model compounds rotenone, 4-aminopyridine, digoxin, and topotecan. The HCI assays performed in 3D-cultured ReNcell VM on the 384-pillar plates were highly robust and reproducible as indicated by the average Z' factor of 0.6 and CV values around 12%. From concentration-response curves and IC50 values, mitochondrial membrane impairment appears to be the early stage marker of cell death by the compounds.
Collapse
|
17
|
Isolation and Characterization of Neural Stem Cells from the Rat Inferior Colliculus. Stem Cells Int 2019; 2019:5831240. [PMID: 31781242 PMCID: PMC6875198 DOI: 10.1155/2019/5831240] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 08/16/2019] [Accepted: 08/27/2019] [Indexed: 12/12/2022] Open
Abstract
The inferior colliculus (IC) is a nucleus of the auditory pathway and its fourth relay station. It integrates afferent information from the superior olivary complex and the cochlear nucleus. To date, no causal therapeutic options are known for damaged neuronal structures in this area. Regenerative medicine offers a potential approach to causally treating hearing impairment. After neural stem cells had been identified in certain areas of the auditory pathway, the question arouses, whether the IC also has a neurogenic potential. Cells from the IC of postnatal day 6 rats were extracted and cultured as neurospheres. Cells in the neurospheres showed mitotic activity and positive stain of neural stem cell markers (Nestin, DCX, Atoh1, and Sox-2). In addition, single cells were differentiated into neuronal and glial cells shown by the markers β-III-tubulin, GFAP, and MBP. In summary, basic stem cell criteria could be detected and characterized in cells isolated from the IC of the rat. These findings will lead to a better understanding of the development of the auditory pathway and may also be relevant for identifying causal therapeutic approaches in the future.
Collapse
|
18
|
Polonio-Alcalá E, Rabionet M, Ruiz-Martínez S, Ciurana J, Puig T. Three-Dimensional Manufactured Supports for Breast Cancer Stem Cell Population Characterization. Curr Drug Targets 2019; 20:839-851. [DOI: 10.2174/1389450120666181122113300] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 11/03/2018] [Accepted: 11/07/2018] [Indexed: 12/23/2022]
Abstract
Breast Cancer (BC) is the most common cancer among women and the second cause of female death for cancer. When the tumor is not correctly eradicated, there is a high relapse risk and incidence of metastasis. Breast Cancer Stem Cells (BCSCs) are responsible for initiating tumors and are resistant to current anticancer therapies being in part responsible for tumor relapse and metastasis. The study of BCSCs is limited due to their low percentage within both tumors and established cell models. Hence, three-dimensional (3D) supports are presented as an interesting tool to keep the stem-like features in 3D cell culture. In this review, several 3D culture systems are discussed. Moreover, scaffolds are presented as a tool to enrich in BCSCs in order to find new specific therapeutic strategies against this malignant subpopulation. Anticancer treatments focused on BCSCs could be useful for BC patients, with particular interest in those that progress to current therapies.
Collapse
Affiliation(s)
- Emma Polonio-Alcalá
- New Therapeutic Targets Laboratory (TargetsLab), Department of Medical Sciences, Faculty of Medicine, University of Girona, Girona, Spain
| | - Marc Rabionet
- New Therapeutic Targets Laboratory (TargetsLab), Department of Medical Sciences, Faculty of Medicine, University of Girona, Girona, Spain
| | - Santiago Ruiz-Martínez
- New Therapeutic Targets Laboratory (TargetsLab), Department of Medical Sciences, Faculty of Medicine, University of Girona, Girona, Spain
| | - Joaquim Ciurana
- Product, Process and Production Engineering Research Group (GREP), Department of Mechanical Engineering and Industrial Construction, University of Girona, Girona, Spain
| | - Teresa Puig
- New Therapeutic Targets Laboratory (TargetsLab), Department of Medical Sciences, Faculty of Medicine, University of Girona, Girona, Spain
| |
Collapse
|
19
|
Deshpande K, Saatian B, Martirosian V, Lin M, Julian A, Neman J. Isolation of Neural Stem Cells from Whole Brain Tissues of Adult Mice. ACTA ACUST UNITED AC 2019; 49:e80. [DOI: 10.1002/cpsc.80] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Krutika Deshpande
- Department of Neurological Surgery, Keck School of Medicine University of Southern California Los Angeles California
| | - Behnaz Saatian
- Department of Neurological Surgery, Keck School of Medicine University of Southern California Los Angeles California
| | - Vahan Martirosian
- Department of Neurological Surgery, Keck School of Medicine University of Southern California Los Angeles California
| | - Michelle Lin
- Department of Neurological Surgery, Keck School of Medicine University of Southern California Los Angeles California
| | - Alex Julian
- Department of Neurological Surgery, Keck School of Medicine University of Southern California Los Angeles California
| | - Josh Neman
- Department of Neurological Surgery, Keck School of Medicine University of Southern California Los Angeles California
| |
Collapse
|
20
|
Shinozuka T, Takada R, Yoshida S, Yonemura S, Takada S. Wnt produced by stretched roof-plate cells is required for the promotion of cell proliferation around the central canal of the spinal cord. Development 2019; 146:146/2/dev159343. [PMID: 30651295 DOI: 10.1242/dev.159343] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 12/14/2018] [Indexed: 01/23/2023]
Abstract
Cell morphology changes dynamically during embryogenesis, and these changes create new interactions with surrounding cells, some of which are presumably mediated by intercellular signaling. However, the effects of morphological changes on intercellular signaling remain to be fully elucidated. In this study, we examined the effect of morphological changes in Wnt-producing cells on intercellular signaling in the spinal cord. After mid-gestation, roof-plate cells stretched along the dorsoventral axis in the mouse spinal cord, resulting in new contact at their tips with the ependymal cells that surround the central canal. Wnt1 and Wnt3a were produced by the stretched roof-plate cells and delivered to the cell process tip. Whereas Wnt signaling was activated in developing ependymal cells, Wnt activation in dorsal ependymal cells, which were close to the stretched roof plate, was significantly suppressed in embryos with roof plate-specific conditional knockout of Wls, which encodes a factor that is essential for Wnt secretion. Furthermore, proliferation of these cells was impaired in Wls conditional knockout mice during development and after induced spinal cord injury in adults. Therefore, morphological changes in Wnt-producing cells appear to generate new Wnt signal targets.
Collapse
Affiliation(s)
- Takuma Shinozuka
- Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan.,National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan.,Department of Basic Biology in the School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), 5-1 Higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan
| | - Ritsuko Takada
- Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan.,National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan
| | - Shosei Yoshida
- National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan.,Department of Basic Biology in the School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), 5-1 Higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan
| | - Shigenobu Yonemura
- RIKEN Center for Life Science Technologies, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.,Department of Cell Biology, Tokushima University Graduate School of Medical Science, 3-18-15, Kuramoto-cho, Tokushima 770-8503, Japan
| | - Shinji Takada
- Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan .,National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan.,Department of Basic Biology in the School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), 5-1 Higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan
| |
Collapse
|
21
|
Periostin and Integrin Signaling in Stem Cell Regulation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1132:163-176. [DOI: 10.1007/978-981-13-6657-4_16] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
22
|
Lampasona AA, Czaplinski K. Hnrnpab regulates neural cell motility through transcription of Eps8. RNA (NEW YORK, N.Y.) 2019; 25:45-59. [PMID: 30314980 PMCID: PMC6298563 DOI: 10.1261/rna.067413.118] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 10/01/2018] [Indexed: 05/05/2023]
Abstract
Cell migration requires a complicated network of structural and regulatory proteins. Changes in cellular motility can impact migration as a result of cell-type or developmental stage regulated expression of critical motility genes. Hnrnpab is a conserved RNA-binding protein found as two isoforms produced by alternative splicing. Its expression is enriched in the subventricular zone (SVZ) and the rostral migratory stream within the brain, suggesting possible support of the migration of neural progenitor cells in this region. Here we show that the migration of cells from the SVZ of developing Hnrnpab-/- mouse brains is impaired. An RNA-seq analysis to identify Hnrnpab-dependent cell motility genes led us to Eps8, and in agreement with the change in cell motility, we show that Eps8 is decreased in Hnrnpab-/- SVZ tissue. We scrutinized the motility of Hnrnpab-/- cells and confirmed that the decreases in both cell motility and Eps8 are restored by ectopically coexpressing both alternatively spliced Hnrnpab isoforms, therefore these variants are surprisingly nonredundant for cell motility. Our results support a model where both Hnrnpab isoforms work in concert to regulate Eps8 transcription in the mouse SVZ to promote the normal migration of neural cells during CNS development.
Collapse
Affiliation(s)
- Alexa A Lampasona
- Program in Molecular and Cellular Pharmacology, Stony Brook University, Stony Brook, New York 11749, USA
- Centers for Molecular Medicine, Stony Brook University, Stony Brook, New York 11749, USA
| | - Kevin Czaplinski
- Centers for Molecular Medicine, Stony Brook University, Stony Brook, New York 11749, USA
- Department of Anesthesiology, Stony Brook University, Stony Brook, New York 11749, USA
| |
Collapse
|
23
|
Azevedo-Pereira RL, Rangel B, Tovar-Moll F, Gasparetto EL, Attias M, Zaverucha-do-Valle C, Jasmin, Mendez-Otero R. Superparamagnetic iron oxide nanoparticles as a tool to track mouse neural stem cells in vivo. Mol Biol Rep 2018; 46:191-198. [PMID: 30421128 DOI: 10.1007/s11033-018-4460-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 11/01/2018] [Indexed: 12/21/2022]
Abstract
Cell transplantation offers a promising approach in many neurological disorders. Neural stem (NS) cells are potential candidates for cell therapy. The ability to track the grafted cells in the host tissue will refine this therapy. Superparamagnetic iron oxide nanoparticles (SPION) have been suggested as a feasible method, but there is no consensus about its safety. Here we investigated the feasibility of label NS cells with SPION and track by MRI after transplantation into mouse striatum with SPION cells and its therapeutic effects by grafting the cells into mouse striatum. We demonstrated that SPION-labeled NS cells display normal patterns of cellular processes including proliferation, migration, differentiation and neurosphere formation. Transmission electron microscopy reveals SPION in the cytoplasm of the cells, which was confirmed by microanalysis. Neurons and astrocytes generated from SPION-labeled NS cells were able to carry nanoparticles after 7 days under differentiation. SPION-labeled NS cells transplanted into striatum of mice were detected by magnetic resonance imaging (MRI) and microscopy 51 days later. In agreement with others reports, we demonstrated that NS cells are able to incorporate SPION in vitro without altering the stemness, and can survive and be tracked by MRI after they have been grafted into mice striatum.
Collapse
Affiliation(s)
- Ricardo Luiz Azevedo-Pereira
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Department of Neurosurgery, Stanford University, Palo Alto, CA, USA
| | - Bárbara Rangel
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Marcia Attias
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Camila Zaverucha-do-Valle
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Jasmin
- Núcleo Multidisciplinar de Pesquisa em Biologia-Duque de Caxias, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rosalia Mendez-Otero
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
24
|
Rosati J, Ferrari D, Altieri F, Tardivo S, Ricciolini C, Fusilli C, Zalfa C, Profico DC, Pinos F, Bernardini L, Torres B, Manni I, Piaggio G, Binda E, Copetti M, Lamorte G, Mazza T, Carella M, Gelati M, Valente EM, Simeone A, Vescovi AL. Establishment of stable iPS-derived human neural stem cell lines suitable for cell therapies. Cell Death Dis 2018; 9:937. [PMID: 30224709 PMCID: PMC6141489 DOI: 10.1038/s41419-018-0990-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 07/30/2018] [Accepted: 08/01/2018] [Indexed: 12/13/2022]
Abstract
Establishing specific cell lineages from human induced pluripotent stem cells (hiPSCs) is vital for cell therapy approaches in regenerative medicine, particularly for neurodegenerative disorders. While neural precursors have been induced from hiPSCs, the establishment of hiPSC-derived human neural stem cells (hiNSCs), with characteristics that match foetal hNSCs and abide by cGMP standards, thus allowing clinical applications, has not been described. We generated hiNSCs by a virus-free technique, whose properties recapitulate those of the clinical-grade hNSCs successfully used in an Amyotrophic Lateral Sclerosis (ALS) phase I clinical trial. Ex vivo, hiNSCs critically depend on exogenous mitogens for stable self-renewal and amplification and spontaneously differentiate into astrocytes, oligodendrocytes and neurons upon their removal. In the brain of immunodeficient mice, hiNSCs engraft and differentiate into neurons and glia, without tumour formation. These findings now warrant the establishment of clinical-grade, autologous and continuous hiNSC lines for clinical trials in neurological diseases such as Huntington’s, Parkinson’s and Alzheimer’s, among others.
Collapse
Affiliation(s)
- Jessica Rosati
- Cellular Reprogramming Unit, IRCCS Casa Sollievo della Sofferenza, Viale dei Cappuccini, 71013, San Giovanni Rotondo, Foggia, Italy.
| | - Daniela Ferrari
- Department of Biotechnology and Biosciences, University of Milan Bicocca, Piazza della Scienza, 220126, Milan, Italy
| | - Filomena Altieri
- Cellular Reprogramming Unit, IRCCS Casa Sollievo della Sofferenza, Viale dei Cappuccini, 71013, San Giovanni Rotondo, Foggia, Italy
| | - Silvia Tardivo
- Neurogenetics Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Claudia Ricciolini
- Stem Cell Laboratory, Cell Factory e Biobank, Terni Hospital, Via Tristano di Joannuccio 1, 05100, Terni, Italy
| | - Caterina Fusilli
- Bioinformatics Unit, IRCCS Casa Sollievo della Sofferenza, Viale dei Cappuccini, 71013, San Giovanni Rotondo, Foggia, Italy
| | - Cristina Zalfa
- Department of Biotechnology and Biosciences, University of Milan Bicocca, Piazza della Scienza, 220126, Milan, Italy
| | - Daniela C Profico
- Production Unit of Advanced Therapies (UPTA), Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), IRCCS Casa Sollievo della Sofferenza, Viale dei Cappuccini, 71013, San Giovanni Rotondo, Foggia, Italy
| | - Francesca Pinos
- Department of Biotechnology and Biosciences, University of Milan Bicocca, Piazza della Scienza, 220126, Milan, Italy
| | - Laura Bernardini
- Medical Genetics Unit, IRCCS Casa Sollievo della Sofferenza, Viale dei Cappuccini, 71013, San Giovanni Rotondo, Foggia, Italy
| | - Barbara Torres
- Medical Genetics Unit, IRCCS Casa Sollievo della Sofferenza, Viale dei Cappuccini, 71013, San Giovanni Rotondo, Foggia, Italy
| | - Isabella Manni
- Department of Research, Diagnosis and Innovative Technologies, Regina Elena National Cancer Institute, Rome, Italy
| | - Giulia Piaggio
- Department of Research, Diagnosis and Innovative Technologies, Regina Elena National Cancer Institute, Rome, Italy
| | - Elena Binda
- Cancer Stem Cells Unit (ICS), Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), IRCCS Casa Sollievo della Sofferenza, Viale dei Cappuccini, 71013, San Giovanni Rotondo, Foggia, Italy
| | - Massimiliano Copetti
- Biostatistic Unit, IRCCS Casa Sollievo della Sofferenza, Viale dei Cappuccini, 71013, San Giovanni Rotondo, Foggia, Italy
| | - Giuseppe Lamorte
- Medical Genetics Unit, IRCCS Casa Sollievo della Sofferenza, Viale dei Cappuccini, 71013, San Giovanni Rotondo, Foggia, Italy
| | - Tommaso Mazza
- Bioinformatics Unit, IRCCS Casa Sollievo della Sofferenza, Viale dei Cappuccini, 71013, San Giovanni Rotondo, Foggia, Italy
| | - Massimo Carella
- Medical Genetics Unit, IRCCS Casa Sollievo della Sofferenza, Viale dei Cappuccini, 71013, San Giovanni Rotondo, Foggia, Italy
| | - Maurizio Gelati
- Department of Biotechnology and Biosciences, University of Milan Bicocca, Piazza della Scienza, 220126, Milan, Italy.,Stem Cell Laboratory, Cell Factory e Biobank, Terni Hospital, Via Tristano di Joannuccio 1, 05100, Terni, Italy.,Production Unit of Advanced Therapies (UPTA), Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), IRCCS Casa Sollievo della Sofferenza, Viale dei Cappuccini, 71013, San Giovanni Rotondo, Foggia, Italy
| | - Enza Maria Valente
- Neurogenetics Unit, IRCCS Santa Lucia Foundation, Rome, Italy.,Department of Molecular Medicine, University of Pavia, Via Forlanini 14, 27100, Pavia, Italy
| | - Antonio Simeone
- Institute of Genetics and Biophysics Adriano Buzzati Traverso, CNR, Via P. Castellino 111, 80131, Naples, Italy.,IRCSS Neuromed, 86077, Pozzilli, Isernia, Italy
| | - Angelo L Vescovi
- Cellular Reprogramming Unit, IRCCS Casa Sollievo della Sofferenza, Viale dei Cappuccini, 71013, San Giovanni Rotondo, Foggia, Italy. .,Department of Biotechnology and Biosciences, University of Milan Bicocca, Piazza della Scienza, 220126, Milan, Italy. .,Stem Cell Laboratory, Cell Factory e Biobank, Terni Hospital, Via Tristano di Joannuccio 1, 05100, Terni, Italy.
| |
Collapse
|
25
|
Zheng C, Chen D, Zhang Y, Bai Y, Huang S, Zheng D, Liang W, She S, Peng X, Wang P, Mo X, Song Q, Lv P, Huang J, Ye RD, Wang Y. FAM19A1 is a new ligand for GPR1 that modulates neural stem-cell proliferation and differentiation. FASEB J 2018; 32:fj201800020RRR. [PMID: 29799787 DOI: 10.1096/fj.201800020rrr] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
FAM19A1 is a member of the family with sequence similarity 19 with unknown function. FAM19A1 mRNA expression is restricted to the CNS. Here, we report that FAM19A1 is a classic secretory protein, and expression levels correlate with brain development, increasing from embryonic d 12.5, peaking between postnatal d (P)1 and P7 and decreasing at wk 8. The adult hippocampus is a region of FAM19A1 high expression. Recombinant FAM19A1 suppressed the proliferation and self-renewal of neural stem cells (NSCs) and altered the lineage progression of NSCs with promoted neuron differentiation and suppressed astrocyte differentiation. Although GPCR 1 (GPR1) has been reported to be expressed in the CNS, its functions in the brain remain unclear. We identified GPR1 to be a functional receptor for FAM19A1. FAM19A1 interacted with GPR1 via the N-terminal domain (GPR1-ND), and its NSC modulatory functions required the Rho-associated protein kinase (ROCK) /ERK1/2 and ROCK/signal transducer and activator of transcription 3 signaling pathways. GPR1-ND that selectively bound to FAM19A1 neutralized the effects of FAM19A1 on NSC functions. Taken together, our results show, for the first time to our knowledge, that FAM19A1 is a novel regulatory factor of the proliferation and differentiation of NSCs, and identified a novel mechanism by which GPCR mediates the effects of FAM19A1 on NSC functions that may be important for brain development and neurogenesis. Additional exploration of the functions of FAM19A1 and GPR1 in the CNS may broaden the range of therapeutic options available for major brain disorders.-Zheng, C., Chen, D., Zhang, Y., Bai, Y., Huang, S., Zheng, D., Liang, W., She, S., Peng, X., Wang, P., Mo, X., Song, Q., Lv, P., Huang, J., Ye, R. D., Wang, Y. FAM19A1 is a new ligand for GPR1 that modulates neural stem-cell proliferation and differentiation.
Collapse
Affiliation(s)
- Can Zheng
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Key Laboratory of Medical Immunology, Ministry of Health, Peking University Health Science Center, Beijing, China
| | - Dixin Chen
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Key Laboratory of Medical Immunology, Ministry of Health, Peking University Health Science Center, Beijing, China
| | - Yan Zhang
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Key Laboratory of Medical Immunology, Ministry of Health, Peking University Health Science Center, Beijing, China
| | - Yun Bai
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Shiyang Huang
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Key Laboratory of Medical Immunology, Ministry of Health, Peking University Health Science Center, Beijing, China
| | - Danfeng Zheng
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Key Laboratory of Medical Immunology, Ministry of Health, Peking University Health Science Center, Beijing, China
| | - Weiwei Liang
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Key Laboratory of Medical Immunology, Ministry of Health, Peking University Health Science Center, Beijing, China
| | - Shaoping She
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Key Laboratory of Medical Immunology, Ministry of Health, Peking University Health Science Center, Beijing, China
| | - Xinjian Peng
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Key Laboratory of Medical Immunology, Ministry of Health, Peking University Health Science Center, Beijing, China
| | - Pingzhang Wang
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Key Laboratory of Medical Immunology, Ministry of Health, Peking University Health Science Center, Beijing, China
- Center for Human Disease Genomics, Peking University, Beijing, China
| | - Xiaoning Mo
- Center for Human Disease Genomics, Peking University, Beijing, China
| | - Quansheng Song
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Key Laboratory of Medical Immunology, Ministry of Health, Peking University Health Science Center, Beijing, China
- Center for Human Disease Genomics, Peking University, Beijing, China
| | - Ping Lv
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Key Laboratory of Medical Immunology, Ministry of Health, Peking University Health Science Center, Beijing, China
- Center for Human Disease Genomics, Peking University, Beijing, China
| | - Jing Huang
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Key Laboratory of Medical Immunology, Ministry of Health, Peking University Health Science Center, Beijing, China
- Center for Human Disease Genomics, Peking University, Beijing, China
| | - Richard D Ye
- Institute of Chinese Medical Sciences, University of Macau, Macau Special Administrative Region, China
| | - Ying Wang
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Key Laboratory of Medical Immunology, Ministry of Health, Peking University Health Science Center, Beijing, China
- Center for Human Disease Genomics, Peking University, Beijing, China
| |
Collapse
|
26
|
Mogi A, Yomoda R, Kimura S, Tsushima C, Takouda J, Sawauchi M, Maekawa T, Ohta H, Nishino S, Kurita M, Mano N, Osumi N, Moriya T. Entrainment of the Circadian Clock in Neural Stem Cells by Epidermal Growth Factor is Closely Associated with ERK1/2-mediated Induction of Multiple Clock-related Genes. Neuroscience 2018. [DOI: 10.1016/j.neuroscience.2018.02.045] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
27
|
Neurospheres from neural stem/neural progenitor cells (NSPCs) of non-hydrocephalic HTx rats produce neurons, astrocytes and multiciliated ependyma: the cerebrospinal fluid of normal and hydrocephalic rats supports such a differentiation. Cell Tissue Res 2018; 373:421-438. [PMID: 29651556 DOI: 10.1007/s00441-018-2828-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 03/14/2018] [Indexed: 01/01/2023]
Abstract
Fetal onset hydrocephalus and abnormal neurogenesis are two inseparable phenomena turned on by a cell junction pathology first affecting neural stem/progenitor cells (NSPCs) and later the multiciliated ependyma. The neurological impairment of children born with hydrocephalus is not reverted by derivative surgery. NSPCs and neurosphere (NE) grafting into the cerebrospinal fluid (CSF) of hydrocephalic fetuses thus appears as a promising therapeutic procedure. There is little information about the cell lineages actually forming the NE as they grow throughout their days in vitro (DIV). Furthermore, there is no information on how good a host the CSF is for grafted NE. Here, we use the HTx rat, a model with hereditary hydrocephalus, with the mutation expressed in about 30% of the litter (hyHTx), while the littermates develop normally (nHTx). The investigation was designed (i) to establish the nature of the cells forming 4 and 6-DIV NE grown from NSPCs collected from PN1/nHTx rats and (ii) to study the effects on these NEs of CSF collected from nHTx and hyHTx. Immunofluorescence analyses showed that 90% of cells forming 4-DIV NEs were non-committed multipotential NSPCs, while in 6-DIV NE, 40% of the NSPCs were already committed into neuronal, glial and ependymal lineages. Six-DIV NE further cultured for 3 weeks in the presence of fetal bovine serum, CSF from nHTx or CSF from hyHTx, differentiated into neurons, astrocytes and βIV-tubulin+ multiciliated ependymal cells that were joined together by adherent junctions and displayed synchronized cilia beating. This supports the possibility that ependymal cells are born from subpopulations of NSC with their own time table of differentiation. As a whole, the findings indicate that the CSF is a supportive medium to host NE and that NE grafted into the CSF have the potential to produce neurons, glia and ependyma.
Collapse
|
28
|
Luo YT, Cheng J, Feng X, He SJ, Wang YW, Huang Q. The viable circulating tumor cells with cancer stem cells feature, where is the way out? J Exp Clin Cancer Res 2018; 37:38. [PMID: 29482576 PMCID: PMC5828305 DOI: 10.1186/s13046-018-0685-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 01/19/2018] [Indexed: 02/07/2023] Open
Abstract
With cancer stem cells (CSCs) became the research hotspot, emerging studies attempt to reveal the functions of these special subsets in tumorigenesis. Although various approaches have been used in CSCs researches, only a few could really reflect or simulate the microenvironment in vivo. At present, CSCs theories are still difficult to apply for clinical remedy because CSCs subpopulations are always hard to identify and trace. Thus an ideal approach for clinicians and researchers is urgently needed. Circulating tumor cells (CTCs), as the method of noninvasive-liquid biopsy, could be detected in the peripheral blood (PB) from many tumors and even could be treated as procurators for CSCs deeper researches from patient-derived sample. However, CTCs, as a diagnostic marker, also raise much controversy over theirs clinical value. Mechanisms causing CTCs to shed from the tumor have not been fully characterized, thus it is unclear whether CTCs represent the entire makeup of cancer cells in the tumor or only a subset. The heterogeneity of CTCs also caused different clinical outcomes. To overcome these unsolved problems, recently, CTC researches are not just depend on enumerations, whereas those CTC subsets that could expand in vitro may play a pivotal role in the metastatic cascade. Here, we retrospect the CTC developmental history and discourse upon the enrichment of viable CTCs in functional assays, probe the further avenue at the crossroad.
Collapse
Affiliation(s)
- Y T Luo
- Molecular Diagnostic Laboratory of Cancer Center, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 201620, China
| | - J Cheng
- Molecular Diagnostic Laboratory of Cancer Center, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 201620, China
| | - X Feng
- Molecular Diagnostic Laboratory of Cancer Center, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 201620, China
| | - S J He
- Molecular Diagnostic Laboratory of Cancer Center, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 201620, China
| | - Y W Wang
- Molecular Diagnostic Laboratory of Cancer Center, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 201620, China
| | - Q Huang
- Molecular Diagnostic Laboratory of Cancer Center, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 201620, China.
| |
Collapse
|
29
|
Selective killing of spinal cord neural stem cells impairs locomotor recovery in a mouse model of spinal cord injury. J Neuroinflammation 2018; 15:58. [PMID: 29475438 PMCID: PMC5824446 DOI: 10.1186/s12974-018-1085-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 02/01/2018] [Indexed: 01/19/2023] Open
Abstract
Background Spinal cord injury (SCI) is a devastating condition mainly deriving from a traumatic damage of the spinal cord (SC). Immune cells and endogenous SC-neural stem cells (SC-NSCs) play a critical role in wound healing processes, although both are ineffective to completely restore tissue functioning. The role of SC-NSCs in SCI and, in particular, whether such cells can interplay with the immune response are poorly investigated issues, although mechanisms governing such interactions might open new avenues to develop novel therapeutic approaches. Methods We used two transgenic mouse lines to trace as well as to kill SC-NSCs in mice receiving SCI. We used Nestin CreERT2 mice to trace SC-NSCs descendants in the spinal cord of mice subjected to SCI. While mice carrying the suicide gene thymidine kinase (TK) along with the GFP reporter, under the control of the Nestin promoter regions (NestinTK mice) were used to label and selectively kill SC-NSCs. Results We found that SC-NSCs are capable to self-activate after SCI. In addition, a significant worsening of clinical and pathological features of SCI was observed in the NestinTK mice, upon selective ablation of SC-NSCs before the injury induction. Finally, mice lacking in SC-NSCs and receiving SCI displayed reduced levels of different neurotrophic factors in the SC and significantly higher number of M1-like myeloid cells. Conclusion Our data show that SC-NSCs undergo cell proliferation in response to traumatic spinal cord injury. Mice lacking SC-NSCs display overt microglia activation and exaggerate expression of pro-inflammatory cytokines. The absence of SC-NSCs impaired functional recovery as well as neuronal and oligodendrocyte cell survival. Collectively our data indicate that SC-NSCs can interact with microglia/macrophages modulating their activation/responses and that such interaction is importantly involved in mechanisms leading tissue recovery. Electronic supplementary material The online version of this article (10.1186/s12974-018-1085-9) contains supplementary material, which is available to authorized users.
Collapse
|
30
|
Torrente Y, Belicchi M, Pisati F, Pagano SF, Fortunato F, Sironi M, D'angelo MG, Parati EA, Scarlato G, Bresolin N. Alternative Sources of Neurons and Glia from Somatic Stem Cells. Cell Transplant 2017. [DOI: 10.3727/096020198389753] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Stem cell populations have been shown to be extremely versatile: they can generate differentiated cells specific to the tissue in which they reside and descendents that are of different germ layer origin. This raises the possibility of obtaining neuronal cells from new biological source of the same adult human subjects. In this study, we found that epidermal growth factor (EGF) and basic fibroblast growth factor (bFGF) cooperated to induce the proliferation, self-renewal, and expansion of neural stem cell-like population isolated from several newborn and adult mouse tissues: muscle and hematopoietic tissues. This population, in both primary culture and secondary expanded clones, formed spheres of undifferentiated cells that were induced to differentiate into neurons, astrocytes, and oligodendrocytes. Brain engraftment of the somatic-derived neural stem cells generated neuronal phenotypes, demonstrating the great plasticity of these cells with potential clinical application.
Collapse
Affiliation(s)
- Yvan Torrente
- IRCCS Ospedale Maggiore Policlinico, Italy
- Centro Dino Ferrari, Institute of Clinical Neurology, Milan, Italy
| | | | | | - Stefano F. Pagano
- Laboratory of Neuropharmacology, National Neurological Institute “C. Besta,” Milan, Italy
| | | | | | | | - Eugenio A. Parati
- Laboratory of Neuropharmacology, National Neurological Institute “C. Besta,” Milan, Italy
| | | | - Nereo Bresolin
- IRCCS Ospedale Maggiore Policlinico, Italy
- IRCCS Eugenio Medea, Bosisio Parini, Italy
| |
Collapse
|
31
|
Kanemura Y, Mori H, Nakagawa A, Islam MO, Kodama E, Yamamoto A, Shofuda T, Kobayashi S, Miyake J, Yamazaki T, Hirano SI, Yamasaki M, Okano H. In Vitro Screening of Exogenous Factors for Human Neural Stem/Progenitor Cell Proliferation Using Measurement of Total ATP Content in Viable Cells. Cell Transplant 2017; 14:673-682. [DOI: 10.3727/000000005783982701] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
One of the newest and most promising methods for treating intractable neuronal diseases and injures is the transplantation of ex vivo-expanded human neural stem/progenitor cells (NSPCs). Human NSPCs are selectively expanded as free-floating neurospheres in serum-free culture medium containing fibroblast growth factor 2 (FGF2) and/or epidermal growth factor (EGF); however, the culture conditions still need to be optimized for performance and cost before the method is used clinically. Here, to improve the NSPC culture method for clinical use, we used an ATP assay to screen the effects of various reagents on human NSPC proliferation. Human NSPCs responded to EGF, FGF2, and leukemia inhibitory factor (LIF) in a dose-dependent manner, and the minimum concentrations eliciting maximum effects were 10 ng/ml EGF, 10 ng/ml FGF2, and 5 ng/ml LIF. EGF and LIF were stable in culture medium without NSPCs, although FGF2 was degraded. In the presence of human NSPCs, however, FGF2 and LIF were both degraded very rapidly, to below the estimated minimum concentration on day 3, but EGF remained above the minimum concentration for 5 days. Adding supplemental doses of each growth factor during the incubation promoted human NSPC proliferation. Among other supplements, insulin and transferrin promoted human NSPC growth, but progesterone, putrescine, selenite, D-glucose, and lactate were not effective and were cytotoxic at higher concentrations. Supplementing with conditioned medium from human NSPCs significantly increased human NSPC proliferation, but using a high percentage of the medium had a negative effect. These findings suggest that human NSPC culture is regulated by a balance in the culture medium between decreasing growth factor levels and increasing positive or negative factors derived from the NSPCs. Thus, in designing culture conditions for human NSPCs, it is useful to take the individual properties of each factor into consideration.
Collapse
Affiliation(s)
- Yonehiro Kanemura
- Research Institute for Cell Engineering, National Institute of Advanced Industrial Science and Technology, 3–11–46 Nakoji, Amagasaki, Hyogo 661–0974, Japan
- Institute for Clinical Research, Osaka National Hospital, National Hospital Organization, Osaka 540–0006, Japan
| | - Hideki Mori
- Research Institute for Cell Engineering, National Institute of Advanced Industrial Science and Technology, 3–11–46 Nakoji, Amagasaki, Hyogo 661–0974, Japan
| | - Atsuyo Nakagawa
- Research Institute for Cell Engineering, National Institute of Advanced Industrial Science and Technology, 3–11–46 Nakoji, Amagasaki, Hyogo 661–0974, Japan
| | - Mohammed Omedul Islam
- Research Institute for Cell Engineering, National Institute of Advanced Industrial Science and Technology, 3–11–46 Nakoji, Amagasaki, Hyogo 661–0974, Japan
- Institute of Biomedical Research and Innovation, Kobe, Hyogo 650–0047, Japan
| | - Eri Kodama
- Research Institute for Cell Engineering, National Institute of Advanced Industrial Science and Technology, 3–11–46 Nakoji, Amagasaki, Hyogo 661–0974, Japan
| | - Atsuyo Yamamoto
- Research Institute for Cell Engineering, National Institute of Advanced Industrial Science and Technology, 3–11–46 Nakoji, Amagasaki, Hyogo 661–0974, Japan
| | - Tomoko Shofuda
- Research Institute for Cell Engineering, National Institute of Advanced Industrial Science and Technology, 3–11–46 Nakoji, Amagasaki, Hyogo 661–0974, Japan
| | - Satoshi Kobayashi
- Research Institute for Cell Engineering, National Institute of Advanced Industrial Science and Technology, 3–11–46 Nakoji, Amagasaki, Hyogo 661–0974, Japan
| | - Jun Miyake
- Research Institute for Cell Engineering, National Institute of Advanced Industrial Science and Technology, 3–11–46 Nakoji, Amagasaki, Hyogo 661–0974, Japan
| | - Tomohiko Yamazaki
- Research Institute for Cell Engineering, National Institute of Advanced Industrial Science and Technology, 3–11–46 Nakoji, Amagasaki, Hyogo 661–0974, Japan
- New Energy and Industrial Technology Development Organization, Tokyo 170–6028, Japan
| | - Shun-Ichiro Hirano
- Research Institute for Cell Engineering, National Institute of Advanced Industrial Science and Technology, 3–11–46 Nakoji, Amagasaki, Hyogo 661–0974, Japan
- Institute of Biomedical Research and Innovation, Kobe, Hyogo 650–0047, Japan
| | - Mami Yamasaki
- Institute for Clinical Research, Osaka National Hospital, National Hospital Organization, Osaka 540–0006, Japan
- Department of Neurosurgery, Osaka National Hospital, National Hospital Organization, Osaka 540–0006, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo 160–8582, Japan
| |
Collapse
|
32
|
Mizumoto H, Mizumoto K, Whiteley SJO, Shatos M, Klassen H, Young MJ. Transplantation of Human Neural Progenitor Cells to the Vitreous Cavity of the Royal College of Surgeons Rat. Cell Transplant 2017. [DOI: 10.3727/000000001783986936] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Hiroyuki Mizumoto
- Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114
| | - Keiko Mizumoto
- Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114
| | - Simon J. O. Whiteley
- Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114
| | - Marie Shatos
- Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114
| | - Henry Klassen
- CHOC Research, Children's Hospital of Orange County, Orange, CA 92868
| | - Michael J. Young
- Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114
| |
Collapse
|
33
|
Gokuladhas K, Sivapriya N, Barath M, NewComer CH. Ocular progenitor cells and current applications in regenerative medicines - Review. Genes Dis 2017; 4:88-99. [PMID: 30258910 PMCID: PMC6136601 DOI: 10.1016/j.gendis.2017.01.002] [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] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 01/31/2017] [Indexed: 12/31/2022] Open
Abstract
The recent emerging field of regenerative medicine is to present solutions for chronic diseases which cannot be sufficiently repaired by the body's own mechanisms. Stem cells are undifferentiated biological cells and have the potential to develop into many different cell types in the body during early life and growth. Self renewal and totipotency are the characteristic features of stem cells and it holds a promising result for treating various diseases like diabetic foot ulcer, heart diseases, lung diseases, Autism, Skin diseases, arthritis including eye disease. Failure of complete recovery of eye diseases and complications that follow conventional treatments have shifted search to a new form of regenerative medicine using Stem cells. The ocular progenitor cells are remarkable in stem cell biology and replenishing degenerated cells despite being present in low quantity and quiescence in our body has a high therapeutic value. In this paper we have review the applications on ocular progenitor stem cells in treatment of human eye diseases and address the strategies that have been exploited in an effort to regain visual function in the advance treatment of stem cells without any side effects and also present the significance in advance stem cell research.
Collapse
Affiliation(s)
- K Gokuladhas
- World Stem Cell Clinic India LLP (ISO 9001:2015 Certified Clinic), #6, 9th Cross Street, Kapaleeshwar Nagar, Neelankarai, Chennai 600115, India
| | - N Sivapriya
- World Stem Cell Clinic India LLP (ISO 9001:2015 Certified Clinic), #6, 9th Cross Street, Kapaleeshwar Nagar, Neelankarai, Chennai 600115, India
| | - M Barath
- World Stem Cell Clinic India LLP (ISO 9001:2015 Certified Clinic), #6, 9th Cross Street, Kapaleeshwar Nagar, Neelankarai, Chennai 600115, India
| | - Charles H NewComer
- World Stem Cell Clinic India LLP (ISO 9001:2015 Certified Clinic), #6, 9th Cross Street, Kapaleeshwar Nagar, Neelankarai, Chennai 600115, India
| |
Collapse
|
34
|
Yang XH, Ding Y, Li W, Zhang RY, Wu JL, Ling EA, Wu W, Zeng YS. Effects of electroacupuncture and the retinoid X receptor (RXR) signalling pathway on oligodendrocyte differentiation in the demyelinated spinal cord of rats. Acupunct Med 2016; 35:122-132. [PMID: 27841975 PMCID: PMC5466916 DOI: 10.1136/acupmed-2016-011134] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2016] [Indexed: 11/13/2022]
Abstract
Objectives In spinal cord demyelination, some oligodendrocyte precursor cells (OPCs) remain in the demyelinated region but have a reduced capacity to differentiate into oligodendrocytes. This study investigated whether ‘Governor Vessel’ (GV) electroacupuncture (EA) would promote the differentiation of endogenous OPCs into oligodendrocytes by activating the retinoid X receptor γ (RXR-γ)-mediated signalling pathway. Methods Adult rats were microinjected with ethidium bromide (EB) into the T10 spinal cord to establish a model of spinal cord demyelination. EB-injected rats remained untreated (EB group, n=26) or received EA treatment (EB+EA group, n=26). A control group (n=26) was also included that underwent dural exposure without EB injection. After euthanasia at 7 days (n=5 per group), 15 days (n=8 per group) or 30 days (n=13 per group), protein expression of RXR-γ in the demyelinated spinal cord was evaluated by immunohistochemistry and Western blotting. In addition, OPCs derived from rat embryonic spinal cord were cultured in vitro, and exogenous 9-cis-RA (retinoic acid) and RXR-γ antagonist HX531 were administered to determine whether RA could activate RXR-γ and promote OPC differentiation. Results EA was found to increase the numbers of both OPCs and oligodendrocytes expressing RXR-γ and RALDH2, and promote remyelination in the remyelinated spinal cord. Exogenous 9-cis-RA enhanced the differentiation of OPCs into mature oligodendrocytes by activating RXR-γ. Conclusions The results suggest that EA may activate RXR signalling to promote the differentiation of OPCs into oligodendrocytes in spinal cord demyelination.
Collapse
Affiliation(s)
- Xiao-Hua Yang
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Ying Ding
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Wen Li
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Rong-Yi Zhang
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jin-Lang Wu
- Department of Electron Microscope, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Eng-Ang Ling
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Wutian Wu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong.,State Key Laboratory of Brain and Cognitive Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, Hong Kong.,Joint Laboratory of Jinan University and The University of Hong Kong, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Yuan-Shan Zeng
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, China.,Institute of Spinal Cord Injury, Sun Yat-sen University, Guangzhou, China.,Co-innovation Center of Neuroregeneration, Jiangsu, China.,Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
35
|
The emerging role of in vitro electrophysiological methods in CNS safety pharmacology. J Pharmacol Toxicol Methods 2016; 81:47-59. [DOI: 10.1016/j.vascn.2016.03.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 03/30/2016] [Accepted: 03/30/2016] [Indexed: 01/16/2023]
|
36
|
Abstract
It is possible, by a variety of means, to isolate, propagate, and characterize engraftable clones of cells from the mammalian CNS that fulfill the operational definition of a "stem cell": self-maintaining, self-renewing, and extremely multipotent in vitro and in vivo. Even as debates flourish over how neural stem cells might best be defined, identified, represented, and manipulated, clonal cells with "stem-like" features have begun to provide valuable models for studying commitment, differentiation, and plasticity in the CNS. Furthermore, by learning in this way the basic biology of neural stem cells, and by then exploiting those inherent properties for therapeutic ends, novel and multifaceted strategies seem poised to emerge for redressing a variety of heretofore untreatable CNS dysfunctions. Stem-like cells have begun to show promise for neural cell re placement and molecular support therapy in various animal models of degenerative, developmental, and acquired CNS insult. NEUROSCIENTIST 4:408-425, 1998
Collapse
Affiliation(s)
- Evan Y. Snyder
- Departments of Neurology (Division of Neuroscience) and Pediatrics (Division of Newborn Medicine) Harvard Medical School Children's Hospital Boston, Massachusetts
| |
Collapse
|
37
|
Liu CC, Lin SP, Hsu HS, Yang SH, Lin CH, Yang MH, Hung MC, Hung SC. Suspension survival mediated by PP2A-STAT3-Col XVII determines tumour initiation and metastasis in cancer stem cells. Nat Commun 2016; 7:11798. [PMID: 27306323 PMCID: PMC4912642 DOI: 10.1038/ncomms11798] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 04/28/2016] [Indexed: 12/27/2022] Open
Abstract
Targeting tumour-initiating cells (TICs) would lead to new therapies to cure cancer. We previously demonstrated that TICs have the capacity to survive under suspension conditions, while other cells undergo anoikis. Here we show that TICs exhibit increased phosphorylation levels of S727STAT3 because of PP2A inactivation. Collagen 17 gene expression is upregulated in a STAT3-dependent manner, which also stabilizes laminin 5 and engages cells to form hemidesmosome-like junctions in response. Blocking the PP2A-S727STAT3-collagen 17 pathway inhibits the suspension survival of TICs and their ability to form tumours in mice, while activation of the same pathway increases the suspension survival and tumour-initiation capacities of bulk cancer cells. The S727STAT3 phosphorylation levels correlate with collagen 17 expression in colon tumour samples, and correlate inversely with survival. Finally, this signalling axis enhances the ability of TIC to form tumours in mouse models of malignant lung cancer pleural effusion and spontaneous colon cancer metastasis.
Collapse
Affiliation(s)
- Chen-Chi Liu
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 112, Taiwan
| | - Shih-Pei Lin
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 112, Taiwan
| | - Han-Shui Hsu
- Institute of Emergency and Critical Care Medicine, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan
- Department of Surgery, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Shung-Haur Yang
- Department of Surgery, Taipei Veterans General Hospital, Taipei 112, Taiwan
- Department of Surgery, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan
| | - Chiu-Hua Lin
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 112, Taiwan
| | - Muh-Hwa Yang
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 112, Taiwan
- Department of Oncology, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Mien-Chie Hung
- Graduate Institute of Cancer Biology, College of Medicine, Center for Molecular Medicine, China Medical University and Hospital, Taichung 404, Taiwan
- Department of Molecular and Cellular Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA
| | - Shih-Chieh Hung
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 112, Taiwan
- Department of Medical Research and Education, Stem Cell Laboratory, Taipei Veterans General Hospital, Taipei 112, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
- Department of Orthopedics, Integrative Stem Cell Center, China Medical University Hospital, Taichung 404, Taiwan
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung 404, Taiwan
| |
Collapse
|
38
|
Deshpande N, Rangarajan A. Cancer Stem Cells: Formidable Allies of Cancer. Indian J Surg Oncol 2016; 6:400-14. [PMID: 27081258 DOI: 10.1007/s13193-015-0451-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 07/24/2015] [Indexed: 12/17/2022] Open
Abstract
Cancer stem cells (CSC) represent the subpopulation of cells within a tumour showing two fundamental properties of stem cells - self-renewal (the ability to make more of their own kind) and differentiation (the ability to generate diverse cell types present within a tissue). The CSC hypothesis posits that CSCs play an important role in tumour initiation, maintenance and progression. Furthermore, owing to their intrinsic drug resistance, they remain refractory to currently used therapy, thereby contributing to tumour relapse. Thus, targeting or taming CSCs can lead to more effective cancer treatment in the coming decades. In this review, we will discuss about the origin of CSC hypothesis, evidence showing their existence, clinical relevance and translational significance.
Collapse
Affiliation(s)
- Neha Deshpande
- Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, 560065 India
| | - Annapoorni Rangarajan
- Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, 560065 India
| |
Collapse
|
39
|
Mitrousis N, Tropepe V, Hermanson O. Post-Translational Modifications of Histones in Vertebrate Neurogenesis. Front Neurosci 2015; 9:483. [PMID: 26733796 PMCID: PMC4689847 DOI: 10.3389/fnins.2015.00483] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 12/04/2015] [Indexed: 11/13/2022] Open
Abstract
The process of neurogenesis, through which the entire nervous system of an organism is formed, has attracted immense scientific attention for decades. How can a single neural stem cell give rise to astrocytes, oligodendrocytes, and neurons? Furthermore, how is a neuron led to choose between the hundreds of different neuronal subtypes that the vertebrate CNS contains? Traditionally, niche signals and transcription factors have been on the spotlight. Recent research is increasingly demonstrating that the answer may partially lie in epigenetic regulation of gene expression. In this article, we comprehensively review the role of post-translational histone modifications in neurogenesis in both the embryonic and adult CNS.
Collapse
Affiliation(s)
- Nikolaos Mitrousis
- Institute of Biomaterials and Biomedical Engineering, University of Toronto Toronto, ON, Canada
| | - Vincent Tropepe
- Department of Cell and Systems Biology, Centre for the Analysis of Genome Evolution and Function, University of Toronto Toronto, ON, Canada
| | - Ola Hermanson
- Department of Neuroscience, Karolinska Institutet Stockholm, Sweden
| |
Collapse
|
40
|
Oh M, Nör JE. The Perivascular Niche and Self-Renewal of Stem Cells. Front Physiol 2015; 6:367. [PMID: 26696901 PMCID: PMC4667083 DOI: 10.3389/fphys.2015.00367] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 11/17/2015] [Indexed: 01/09/2023] Open
Abstract
Postnatal stem cells are typically found in niches that provide signaling cues to maintain their self-renewal and multipotency. While stem cell populations may serve distinct purposes within their tissue of origin, understanding the conserved biology of stem cells and their respective niches provides insights to the behavior of these cells during homeostasis and tissue repair. Here, we discuss perivascular niches of two distinct stem cell populations (i.e., hematopoietic stem cells, mesenchymal stem cells) and explore mechanisms that sustain these stem cells postnatally. We highlight work that demonstrates the impact of cellular crosstalk to stem cell self-renewal and maintenance of functional perivascular niches. We also discuss the importance of the crosstalk within the perivascular niche to the biology of stem cells, and describe the regenerative potential of perivascular cells. We postulate that signaling events that establish and/or stabilize the perivascular niche, particularly through the modulation of self-renewing factors, are key to the long-term success of regenerated tissues.
Collapse
Affiliation(s)
- Min Oh
- Department of Cariology, Restorative Sciences, Endodontics, University of Michigan School of Dentistry Ann Arbor, MI, USA
| | - Jacques E Nör
- Department of Cariology, Restorative Sciences, Endodontics, University of Michigan School of Dentistry Ann Arbor, MI, USA ; Department of Otolaryngology, University of Michigan School of Medicine Ann Arbor, MI, USA ; Department of Biomedical Engineering, University of Michigan College of Engineering Ann Arbor, MI, USA
| |
Collapse
|
41
|
Yu YH, Narayanan G, Sankaran S, Ramasamy S, Chan SY, Lin S, Chen J, Yang H, Srivats H, Ahmed S. Purification, Visualization, and Molecular Signature of Neural Stem Cells. Stem Cells Dev 2015; 25:189-201. [PMID: 26464067 PMCID: PMC4770853 DOI: 10.1089/scd.2015.0190] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Neural stem cells (NSCs) are isolated from primary brain tissue and propagated as a heterogeneous mix of cells, including neural progenitors. To date, NSCs have not been purified in vitro to allow study of their biology and utility in regenerative medicine. In this study, we identify C1qR1 as a novel marker for NSCs and show that it can be used along with Lewis-X (LeX) to yield a highly purified population of NSCs. Using time-lapse microscopy, we are able to follow NSCs forming neurospheres, allowing their visualization. Finally, using single-cell polymerase chain reaction (PCR), we determine the molecular signature of NSCs. The single-cell PCR data suggest that along with the Notch and Shh pathways, the Hippo pathway plays an important role in NSC activity.
Collapse
Affiliation(s)
- Yuan Hong Yu
- 1 Neural Stem Cell Laboratory, Institute of Medical Biology , Singapore, Singapore
| | - Gunaseelan Narayanan
- 1 Neural Stem Cell Laboratory, Institute of Medical Biology , Singapore, Singapore
| | - Shvetha Sankaran
- 1 Neural Stem Cell Laboratory, Institute of Medical Biology , Singapore, Singapore
| | - Srinivas Ramasamy
- 1 Neural Stem Cell Laboratory, Institute of Medical Biology , Singapore, Singapore
| | - Shi Yu Chan
- 1 Neural Stem Cell Laboratory, Institute of Medical Biology , Singapore, Singapore
| | - Shuping Lin
- 1 Neural Stem Cell Laboratory, Institute of Medical Biology , Singapore, Singapore
| | - Jinmiao Chen
- 2 Bioinformatics Laboratory , Singapore Immunology Network, Singapore, Singapore
| | - Henry Yang
- 2 Bioinformatics Laboratory , Singapore Immunology Network, Singapore, Singapore
| | - Hariharan Srivats
- 1 Neural Stem Cell Laboratory, Institute of Medical Biology , Singapore, Singapore
| | - Sohail Ahmed
- 1 Neural Stem Cell Laboratory, Institute of Medical Biology , Singapore, Singapore
| |
Collapse
|
42
|
Tuladhar A, Morshead CM, Shoichet MS. Circumventing the blood–brain barrier: Local delivery of cyclosporin A stimulates stem cells in stroke-injured rat brain. J Control Release 2015; 215:1-11. [DOI: 10.1016/j.jconrel.2015.07.023] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 06/19/2015] [Accepted: 07/21/2015] [Indexed: 01/05/2023]
|
43
|
Zhao J, Sui M, Lü X, Jin D, Zhuang Z, Yan T. Electroacupuncture promotes neural stem cell proliferation and neurogenesis in the dentate gyrus of rats following stroke via upregulation of Notch1 expression. Mol Med Rep 2015; 12:6911-7. [PMID: 26328605 DOI: 10.3892/mmr.2015.4279] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 01/09/2015] [Indexed: 11/05/2022] Open
Abstract
Neural stem cells (NSCs) are important in rehabilitation following stroke. Electroacupuncture (EA) treatment has been observed to promote the recovery of neurological functions subsequent to stroke, however, the effects of EA on the proliferation and differentiation of NSCs and its potential mechanisms remain to be elucidated. In the present study, rats, in which a stroke was induced through middle cerebral artery occlusion (MCAO), were treated with EA or control manipulation for 21 days. The modified Neurological Severity score and Morris water maze tests were used to assess the neurological functions of the rats. Bromodeoxyuridine (BrdU)/glial fibrillary acidic protein (GFAP) or BrdU/neuronal marker (NeuN) double immunofluorescence staining were used to examine the proliferation and differentiation of the NSCs. Reverse transcription quantitative polymerase chain reaction (RT‑qPCR) and western blot analyses were performed to detect the expression levels of Notch1 and Hes1 in the dentate gyrus (DG) of the hippocampus of rats following MCAO. The results demonstrated that EA treatment significantly improved the neurological functional recovery of rats following stroke. A significant increase was observed in the number of BrdU+/GAFP+ and BrdU+/NeuN+ cells in the DG area in the EA‑treated rats compared with that of the control group. RT‑qPCR analysis revealed that EA treatment significantly increased the expression levels of Notch1 and Hes1, which may account for the enhanced proliferation and differentiation of NSCs. In conclusion, to the best of our knowledge, the present study was the first to demonstrate that EA treatment promoted NSC proliferation and neurogenesis in the DG area through the upregulation of Notch signaling following a stroke; therefore, EA may be a useful novel therapeutic strategy in future stroke treatment.
Collapse
Affiliation(s)
- Junhong Zhao
- Department of Rehabilitation Medicine, Sun Yat‑sen Memorial Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Minghong Sui
- Department of Rehabilitation Medicine, Sun Yat‑sen Memorial Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Xiao Lü
- Department of Rehabilitation Medicine, Sun Yat‑sen Memorial Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Dongmei Jin
- Department of Rehabilitation Medicine, Sun Yat‑sen Memorial Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Zhiqiang Zhuang
- Department of Rehabilitation Medicine, Sun Yat‑sen Memorial Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Tiebin Yan
- Department of Rehabilitation Medicine, Sun Yat‑sen Memorial Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510120, P.R. China
| |
Collapse
|
44
|
Vanoli F, Rinchetti P, Porro F, Parente V, Corti S. Clinical and molecular features and therapeutic perspectives of spinal muscular atrophy with respiratory distress type 1. J Cell Mol Med 2015; 19:2058-66. [PMID: 26095024 PMCID: PMC4568910 DOI: 10.1111/jcmm.12606] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Accepted: 04/02/2015] [Indexed: 12/13/2022] Open
Abstract
Spinal muscular atrophy with respiratory distress (SMARD1) is an autosomal recessive neuromuscular disease caused by mutations in the IGHMBP2 gene, encoding the immunoglobulin μ-binding protein 2, leading to motor neuron degeneration. It is a rare and fatal disease with an early onset in infancy in the majority of the cases. The main clinical features are muscular atrophy and diaphragmatic palsy, which requires prompt and permanent supportive ventilation. The human disease is recapitulated in the neuromuscular degeneration (nmd) mouse. No effective treatment is available yet, but novel therapeutical approaches tested on the nmd mouse, such as the use of neurotrophic factors and stem cell therapy, have shown positive effects. Gene therapy demonstrated effectiveness in SMA, being now at the stage of clinical trial in patients and therefore representing a possible treatment for SMARD1 as well. The significant advancement in understanding of both SMARD1 clinical spectrum and molecular mechanisms makes ground for a rapid translation of pre-clinical therapeutic strategies in humans.
Collapse
Affiliation(s)
- Fiammetta Vanoli
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Paola Rinchetti
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Francesca Porro
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Valeria Parente
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefania Corti
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
45
|
Ma SM, Chen LX, Lin YF, Yan H, Lv JW, Xiong M, Li J, Cheng GQ, Yang Y, Qiu ZL, Zhou WH. Periostin Promotes Neural Stem Cell Proliferation and Differentiation following Hypoxic-Ischemic Injury. PLoS One 2015; 10:e0123585. [PMID: 25894199 PMCID: PMC4404137 DOI: 10.1371/journal.pone.0123585] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 03/04/2015] [Indexed: 11/24/2022] Open
Abstract
Neural stem cell (NSC) proliferation and differentiation are required to replace neurons damaged or lost after hypoxic-ischemic events and recover brain function. Periostin (POSTN), a novel matricellular protein, plays pivotal roles in the survival, migration, and regeneration of various cell types, but its function in NSCs of neonatal rodent brain is still unknown. The purpose of this study was to investigate the role of POSTN in NSCs following hypoxia-ischemia (HI). We found that POSTN mRNA levels significantly increased in differentiating NSCs. The proliferation and differentiation of NSCs in the hippocampus is compromised in POSTN knockout mice. Moreover, NSC proliferation and differentiation into neurons and astrocytes significantly increased in cultured NSCs treated with recombinant POSTN. Consistently, injection of POSTN into neonatal hypoxic-ischemic rat brains stimulated NSC proliferation and differentiation in the subventricular and subgranular zones after 7 and 14 days of brain injury. Lastly, POSTN treatment significantly improved the spatial learning deficits of rats subjected to HI. These results suggest that POSTN significantly enhances NSC proliferation and differentiation after HI, and provides new insights into therapeutic strategies for the treatment of hypoxic-ischemic encephalopathy.
Collapse
Affiliation(s)
- Si-Min Ma
- Department of Neonatology, Children’s Hospital of Fudan University, Shanghai, China
- Key Laboratory of Neonatal Diseases, Ministry of Health, Children’s Hospital of Fudan University, Shanghai, China
| | - Long-Xia Chen
- Key Laboratory of Birth Defect, Children’s Hospital of Fudan University, Shanghai, China
- Key Laboratory of Neonatal Diseases, Ministry of Health, Children’s Hospital of Fudan University, Shanghai, China
| | - Yi-Feng Lin
- Key Laboratory of Birth Defect, Children’s Hospital of Fudan University, Shanghai, China
- Key Laboratory of Neonatal Diseases, Ministry of Health, Children’s Hospital of Fudan University, Shanghai, China
| | - Hu Yan
- Key Laboratory of Neonatal Diseases, Ministry of Health, Children’s Hospital of Fudan University, Shanghai, China
| | - Jing-Wen Lv
- Key Laboratory of Neonatal Diseases, Ministry of Health, Children’s Hospital of Fudan University, Shanghai, China
| | - Man Xiong
- Key Laboratory of Neonatal Diseases, Ministry of Health, Children’s Hospital of Fudan University, Shanghai, China
| | - Jin Li
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Guo-Qiang Cheng
- Department of Neonatology, Children’s Hospital of Fudan University, Shanghai, China
| | - Yi Yang
- Key Laboratory of Neonatal Diseases, Ministry of Health, Children’s Hospital of Fudan University, Shanghai, China
| | - Zi-Long Qiu
- Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wen-Hao Zhou
- Department of Neonatology, Children’s Hospital of Fudan University, Shanghai, China
- Key Laboratory of Neonatal Diseases, Ministry of Health, Children’s Hospital of Fudan University, Shanghai, China
- * E-mail:
| |
Collapse
|
46
|
Koutmani Y, Karalis KP. Neural stem cells respond to stress hormones: distinguishing beneficial from detrimental stress. Front Physiol 2015; 6:77. [PMID: 25814957 PMCID: PMC4356227 DOI: 10.3389/fphys.2015.00077] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 02/26/2015] [Indexed: 11/13/2022] Open
Abstract
Neural stem cells (NSCs), the progenitors of the nervous system, control distinct, position-specific functions and are critically involved in the maintenance of homeostasis in the brain. The responses of these cells to various stressful stimuli are shaped by genetic, epigenetic, and environmental factors via mechanisms that are age and developmental stage-dependent and still remain, to a great extent, elusive. Increasing evidence advocates for the beneficial impact of the stress response in various settings, complementing the extensive number of studies on the detrimental effects of stress, particularly in the developing brain. In this review, we discuss suggested mechanisms mediating both the beneficial and detrimental effects of stressors on NSC activity across the lifespan. We focus on the specific effects of secreted factors and we propose NSCs as a “sensor,” capable of distinguishing among the different stressors and adapting its functions accordingly. All the above suggest the intriguing hypothesis that NSCs are an important part of the adaptive response to stressors via direct and indirect, specific mechanisms.
Collapse
Affiliation(s)
- Yassemi Koutmani
- Center for Experimental Surgery, Clinical and Translational Research, Biomedical Research Foundation of the Academy of Athens Athens, Greece
| | - Katia P Karalis
- Center for Experimental Surgery, Clinical and Translational Research, Biomedical Research Foundation of the Academy of Athens Athens, Greece ; Endocrine Division, Children's Hospital, Harvard Medical School Boston, MA, USA
| |
Collapse
|
47
|
Castro-Garcia P, Díaz-Moreno M, Gil-Gas C, Fernández-Gómez FJ, Honrubia-Gómez P, Álvarez-Simón CB, Sánchez-Sánchez F, Cano JCC, Almeida F, Blanco V, Jordán J, Mira H, Ramírez-Castillejo C. Defects in subventricular zone pigmented epithelium-derived factor niche signaling in the senescence-accelerated mouse prone-8. FASEB J 2015; 29:1480-92. [PMID: 25636741 DOI: 10.1096/fj.13-244442] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 12/05/2014] [Indexed: 12/27/2022]
Abstract
We studied potential changes in the subventricular zone (SVZ) stem cell niche of the senescence-accelerated mouse prone-8 (SAM-P8) aging model. Bromodeoxyuridine (BrdU) assays with longtime survival revealed a lower number of label-retaining stem cells in the SAM-P8 SVZ compared with the SAM-Resistant 1 (SAM-R1) control strain. We also found that in SAM-P8 niche signaling is attenuated and the stem cell pool is less responsive to the self-renewal niche factor pigmented epithelium-derived factor (PEDF). Protein analysis demonstrated stable amounts of the PEDF ligand in the SAM-P8 SVZ niche; however, SAM-P8 stem cells present a significant expression decrease of patatin-like phospholipase domain containing 2, a receptor for PEDF (PNPLA2-PEDF) receptor, but not of laminin receptor (LR), a receptor for PEDF (LR-PEDF) receptor. We observed changes in self-renewal related genes (hairy and enhancer of split 1 (Hes1), hairy and enhancer of split 1 (Hes5), Sox2] and report that although these genes are down-regulated in SAM-P8, differentiation genes (Pax6) are up-regulated and neurogenesis is increased. Finally, sheltering mammalian telomere complexes might be also involved given a down-regulation of telomeric repeat binding factor 1 (Terf1) expression was observed in SAM-P8 at young age periods. Differences between these 2 models, SAM-P8 and SAM-R1 controls, have been previously detected at more advanced ages. We now describe alterations in the PEDF signaling pathway and stem cell self-renewal at a very young age, which could be involved in the premature senescence observed in the SAM-P8 model.
Collapse
Affiliation(s)
- Paola Castro-Garcia
- *Laboratorio de Células Madre, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, Albacete, Spain; Unidad Funcional de Investigación en Enfermedades Crónicas, Instituto de Salud Carlos III, Madrid, Spain; Grupo de Neurofarmacología, Departamento de Ciencias Médicas, Area de Genética, Facultad de Medicina de Albacete, and Instituto de Investigación en Discapacidades Neurológicas, Universidad de Castilla-La Mancha, Albacete, Spain; and Departamento Estadística, I. O. y Computación, Universidad de La Laguna, La Laguna, Canarias, Spain
| | - María Díaz-Moreno
- *Laboratorio de Células Madre, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, Albacete, Spain; Unidad Funcional de Investigación en Enfermedades Crónicas, Instituto de Salud Carlos III, Madrid, Spain; Grupo de Neurofarmacología, Departamento de Ciencias Médicas, Area de Genética, Facultad de Medicina de Albacete, and Instituto de Investigación en Discapacidades Neurológicas, Universidad de Castilla-La Mancha, Albacete, Spain; and Departamento Estadística, I. O. y Computación, Universidad de La Laguna, La Laguna, Canarias, Spain
| | - Carmen Gil-Gas
- *Laboratorio de Células Madre, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, Albacete, Spain; Unidad Funcional de Investigación en Enfermedades Crónicas, Instituto de Salud Carlos III, Madrid, Spain; Grupo de Neurofarmacología, Departamento de Ciencias Médicas, Area de Genética, Facultad de Medicina de Albacete, and Instituto de Investigación en Discapacidades Neurológicas, Universidad de Castilla-La Mancha, Albacete, Spain; and Departamento Estadística, I. O. y Computación, Universidad de La Laguna, La Laguna, Canarias, Spain
| | - Francisco J Fernández-Gómez
- *Laboratorio de Células Madre, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, Albacete, Spain; Unidad Funcional de Investigación en Enfermedades Crónicas, Instituto de Salud Carlos III, Madrid, Spain; Grupo de Neurofarmacología, Departamento de Ciencias Médicas, Area de Genética, Facultad de Medicina de Albacete, and Instituto de Investigación en Discapacidades Neurológicas, Universidad de Castilla-La Mancha, Albacete, Spain; and Departamento Estadística, I. O. y Computación, Universidad de La Laguna, La Laguna, Canarias, Spain
| | - Paloma Honrubia-Gómez
- *Laboratorio de Células Madre, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, Albacete, Spain; Unidad Funcional de Investigación en Enfermedades Crónicas, Instituto de Salud Carlos III, Madrid, Spain; Grupo de Neurofarmacología, Departamento de Ciencias Médicas, Area de Genética, Facultad de Medicina de Albacete, and Instituto de Investigación en Discapacidades Neurológicas, Universidad de Castilla-La Mancha, Albacete, Spain; and Departamento Estadística, I. O. y Computación, Universidad de La Laguna, La Laguna, Canarias, Spain
| | - Carmen Belén Álvarez-Simón
- *Laboratorio de Células Madre, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, Albacete, Spain; Unidad Funcional de Investigación en Enfermedades Crónicas, Instituto de Salud Carlos III, Madrid, Spain; Grupo de Neurofarmacología, Departamento de Ciencias Médicas, Area de Genética, Facultad de Medicina de Albacete, and Instituto de Investigación en Discapacidades Neurológicas, Universidad de Castilla-La Mancha, Albacete, Spain; and Departamento Estadística, I. O. y Computación, Universidad de La Laguna, La Laguna, Canarias, Spain
| | - Francisco Sánchez-Sánchez
- *Laboratorio de Células Madre, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, Albacete, Spain; Unidad Funcional de Investigación en Enfermedades Crónicas, Instituto de Salud Carlos III, Madrid, Spain; Grupo de Neurofarmacología, Departamento de Ciencias Médicas, Area de Genética, Facultad de Medicina de Albacete, and Instituto de Investigación en Discapacidades Neurológicas, Universidad de Castilla-La Mancha, Albacete, Spain; and Departamento Estadística, I. O. y Computación, Universidad de La Laguna, La Laguna, Canarias, Spain
| | - Juan Carlos Castillo Cano
- *Laboratorio de Células Madre, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, Albacete, Spain; Unidad Funcional de Investigación en Enfermedades Crónicas, Instituto de Salud Carlos III, Madrid, Spain; Grupo de Neurofarmacología, Departamento de Ciencias Médicas, Area de Genética, Facultad de Medicina de Albacete, and Instituto de Investigación en Discapacidades Neurológicas, Universidad de Castilla-La Mancha, Albacete, Spain; and Departamento Estadística, I. O. y Computación, Universidad de La Laguna, La Laguna, Canarias, Spain
| | - Francisco Almeida
- *Laboratorio de Células Madre, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, Albacete, Spain; Unidad Funcional de Investigación en Enfermedades Crónicas, Instituto de Salud Carlos III, Madrid, Spain; Grupo de Neurofarmacología, Departamento de Ciencias Médicas, Area de Genética, Facultad de Medicina de Albacete, and Instituto de Investigación en Discapacidades Neurológicas, Universidad de Castilla-La Mancha, Albacete, Spain; and Departamento Estadística, I. O. y Computación, Universidad de La Laguna, La Laguna, Canarias, Spain
| | - Vicente Blanco
- *Laboratorio de Células Madre, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, Albacete, Spain; Unidad Funcional de Investigación en Enfermedades Crónicas, Instituto de Salud Carlos III, Madrid, Spain; Grupo de Neurofarmacología, Departamento de Ciencias Médicas, Area de Genética, Facultad de Medicina de Albacete, and Instituto de Investigación en Discapacidades Neurológicas, Universidad de Castilla-La Mancha, Albacete, Spain; and Departamento Estadística, I. O. y Computación, Universidad de La Laguna, La Laguna, Canarias, Spain
| | - Joaquín Jordán
- *Laboratorio de Células Madre, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, Albacete, Spain; Unidad Funcional de Investigación en Enfermedades Crónicas, Instituto de Salud Carlos III, Madrid, Spain; Grupo de Neurofarmacología, Departamento de Ciencias Médicas, Area de Genética, Facultad de Medicina de Albacete, and Instituto de Investigación en Discapacidades Neurológicas, Universidad de Castilla-La Mancha, Albacete, Spain; and Departamento Estadística, I. O. y Computación, Universidad de La Laguna, La Laguna, Canarias, Spain
| | - Helena Mira
- *Laboratorio de Células Madre, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, Albacete, Spain; Unidad Funcional de Investigación en Enfermedades Crónicas, Instituto de Salud Carlos III, Madrid, Spain; Grupo de Neurofarmacología, Departamento de Ciencias Médicas, Area de Genética, Facultad de Medicina de Albacete, and Instituto de Investigación en Discapacidades Neurológicas, Universidad de Castilla-La Mancha, Albacete, Spain; and Departamento Estadística, I. O. y Computación, Universidad de La Laguna, La Laguna, Canarias, Spain
| | - Carmen Ramírez-Castillejo
- *Laboratorio de Células Madre, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, Albacete, Spain; Unidad Funcional de Investigación en Enfermedades Crónicas, Instituto de Salud Carlos III, Madrid, Spain; Grupo de Neurofarmacología, Departamento de Ciencias Médicas, Area de Genética, Facultad de Medicina de Albacete, and Instituto de Investigación en Discapacidades Neurológicas, Universidad de Castilla-La Mancha, Albacete, Spain; and Departamento Estadística, I. O. y Computación, Universidad de La Laguna, La Laguna, Canarias, Spain
| |
Collapse
|
48
|
Stem cell therapy for glaucoma: science or snake oil? Surv Ophthalmol 2014; 60:93-105. [PMID: 25132498 DOI: 10.1016/j.survophthal.2014.07.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 06/30/2014] [Accepted: 07/09/2014] [Indexed: 01/15/2023]
Abstract
In recent years there has been substantial progress in developing stem cell treatments for glaucoma. As a downstream approach that targets the underlying susceptibility of retinal ganglion and trabecular meshwork cells, stem cell therapy has the potential to both replace lost, and protect damaged, cells by secreting neurotrophic factors. A variety of sources, including embryonic cells, adult cells derived from the central nervous system, and induced pluripotent stem cells show promise as therapeutic approaches. Even though safety concerns and ethical controversies have limited clinical implementation, some institutions have already commercialized stem cell therapy and are using direct-to-consumer advertising to attract patients with glaucoma. We review the progress of stem cell therapy and its current commercial availability.
Collapse
|
49
|
Adami R, Scesa G, Bottai D. Stem cell transplantation in neurological diseases: improving effectiveness in animal models. Front Cell Dev Biol 2014; 2:17. [PMID: 25364724 PMCID: PMC4206985 DOI: 10.3389/fcell.2014.00017] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 04/22/2014] [Indexed: 12/14/2022] Open
Abstract
Neurological diseases afflict a growing proportion of the human population. There are two reasons for this: first, the average age of the population (especially in the industrialized world) is increasing, and second, the diagnostic tools to detect these pathologies are now more sophisticated and can be used on a higher percentage of the population. In many cases, neurological disease has a pharmacological treatment which, as in the case of Alzheimer's disease, Parkinson's disease, Epilepsy, and Multiple Sclerosis can reduce the symptoms and slow down the course of the disease but cannot reverse its effects or heal the patient. In the last two decades the transplantation approach, by means of stem cells of different origin, has been suggested for the treatment of neurological diseases. The choice of slightly different animal models and the differences in methods of stem cell preparation make it difficult to compare the results of transplantation experiments. Moreover, the translation of these results into clinical trials with human subjects is difficult and has so far met with little success. This review seeks to discuss the reasons for these difficulties by considering the differences between human and animal cells (including isolation, handling and transplantation) and between the human disease model and the animal disease model.
Collapse
Affiliation(s)
- Raffaella Adami
- Department of Health Science, Faculty of Medicine, University of Milan Milan, Italy
| | - Giuseppe Scesa
- Department of Health Science, Faculty of Medicine, University of Milan Milan, Italy
| | - Daniele Bottai
- Department of Health Science, Faculty of Medicine, University of Milan Milan, Italy
| |
Collapse
|
50
|
Compagnucci C, Nizzardo M, Corti S, Zanni G, Bertini E. In vitro neurogenesis: development and functional implications of iPSC technology. Cell Mol Life Sci 2014; 71:1623-39. [PMID: 24252976 PMCID: PMC11113522 DOI: 10.1007/s00018-013-1511-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 10/28/2013] [Accepted: 10/30/2013] [Indexed: 12/21/2022]
Abstract
Neurogenesis is the developmental process regulating cell proliferation of neural stem cells, determining their differentiation into glial and neuronal cells, and orchestrating their organization into finely regulated functional networks. Can this complex process be recapitulated in vitro using induced pluripotent stem cell (iPSC) technology? Can neurodevelopmental and neurodegenerative diseases be modeled using iPSCs? What is the potential of iPSC technology in neurobiology? What are the recent advances in the field of neurological diseases? Since the applications of iPSCs in neurobiology are based on the capacity to regulate in vitro differentiation of human iPSCs into different neuronal subtypes and glial cells, and the possibility of obtaining iPSC-derived neurons and glial cells is based on and hindered by our poor understanding of human embryonic development, we reviewed current knowledge on in vitro neural differentiation from a developmental and cellular biology perspective. We highlight the importance to further advance our understanding on the mechanisms controlling in vivo neurogenesis in order to efficiently guide neurogenesis in vitro for cell modeling and therapeutical applications of iPSCs technology.
Collapse
Affiliation(s)
- Claudia Compagnucci
- Unit of Neuromuscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, Department of Neurosciences, Bambino Gesù Children's Research Hospital, IRCCS, 0165, Rome, Italy,
| | | | | | | | | |
Collapse
|