51
|
Fernandez-Muñoz B, Garcia-Delgado AB, Arribas-Arribas B, Sanchez-Pernaute R. Human Neural Stem Cells for Cell-Based Medicinal Products. Cells 2021; 10:2377. [PMID: 34572024 PMCID: PMC8469920 DOI: 10.3390/cells10092377] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 12/15/2022] Open
Abstract
Neural stem cells represent an attractive tool for the development of regenerative therapies and are being tested in clinical trials for several neurological disorders. Human neural stem cells can be isolated from the central nervous system or can be derived in vitro from pluripotent stem cells. Embryonic sources are ethically controversial and other sources are less well characterized and/or inefficient. Recently, isolation of NSC from the cerebrospinal fluid of patients with spina bifida and with intracerebroventricular hemorrhage has been reported. Direct reprogramming may become another alternative if genetic and phenotypic stability of the reprogrammed cells is ensured. Here, we discuss the advantages and disadvantages of available sources of neural stem cells for the production of cell-based therapies for clinical applications. We review available safety and efficacy clinical data and discuss scalability and quality control considerations for manufacturing clinical grade cell products for successful clinical application.
Collapse
Affiliation(s)
- Beatriz Fernandez-Muñoz
- Cellular Reprogramming and Production Unit, Andalusian Network for the Design and Translation of Advanced Therapies, 41092 Sevilla, Spain; (A.B.G.-D.); (B.A.-A.)
| | - Ana Belen Garcia-Delgado
- Cellular Reprogramming and Production Unit, Andalusian Network for the Design and Translation of Advanced Therapies, 41092 Sevilla, Spain; (A.B.G.-D.); (B.A.-A.)
| | - Blanca Arribas-Arribas
- Cellular Reprogramming and Production Unit, Andalusian Network for the Design and Translation of Advanced Therapies, 41092 Sevilla, Spain; (A.B.G.-D.); (B.A.-A.)
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Sevilla, 41012 Sevilla, Spain
| | - Rosario Sanchez-Pernaute
- Cellular Reprogramming and Production Unit, Andalusian Network for the Design and Translation of Advanced Therapies, 41092 Sevilla, Spain; (A.B.G.-D.); (B.A.-A.)
| |
Collapse
|
52
|
Xu X, Shen D, Gao Y, Zhou Q, Ni Y, Meng H, Shi H, Le W, Chen S, Chen S. A perspective on therapies for amyotrophic lateral sclerosis: can disease progression be curbed? Transl Neurodegener 2021; 10:29. [PMID: 34372914 PMCID: PMC8353789 DOI: 10.1186/s40035-021-00250-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 07/09/2021] [Indexed: 01/17/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease involving both upper and lower motor neurons, leading to paralysis and eventually death. Symptomatic treatments such as inhibition of salivation, alleviation of muscle cramps, and relief of spasticity and pain still play an important role in enhancing the quality of life. To date, riluzole and edaravone are the only two drugs approved by the Food and Drug Administration for the treatment of ALS in a few countries. While there is adequate consensus on the modest efficacy of riluzole, there are still open questions concerning the efficacy of edaravone in slowing the disease progression. Therefore, identification of novel therapeutic strategies is urgently needed. Impaired autophagic process plays a critical role in ALS pathogenesis. In this review, we focus on therapies modulating autophagy in the context of ALS. Furthermore, stem cell therapies, gene therapies, and newly-developed biomaterials have great potentials in alleviating neurodegeneration, which might halt the disease progression. In this review, we will summarize the current and prospective therapies for ALS.
Collapse
Affiliation(s)
- Xiaojiao Xu
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China.,Institute of Neurology, Sichuan Academy of Medical Sciences-Sichuan Provincial Hospital, Chengdu, 610031, China
| | - Dingding Shen
- Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200020, China
| | - Yining Gao
- Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200020, China
| | - Qinming Zhou
- Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200020, China
| | - You Ni
- Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200020, China
| | - Huanyu Meng
- Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200020, China
| | - Hongqin Shi
- Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200020, China.,Department of Neurology, Xinrui Hospital, Wuxi, 214028, China
| | - Weidong Le
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China. .,Institute of Neurology, Sichuan Academy of Medical Sciences-Sichuan Provincial Hospital, Chengdu, 610031, China. .,Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China.
| | - Shengdi Chen
- Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200020, China.
| | - Sheng Chen
- Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200020, China.
| |
Collapse
|
53
|
Aishwarya L, Arun D, Kannan S. Stem cells as a potential therapeutic option for treating neurodegenerative diseases. Curr Stem Cell Res Ther 2021; 17:590-605. [PMID: 35135464 DOI: 10.2174/1574888x16666210810105136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 05/27/2021] [Accepted: 06/01/2021] [Indexed: 11/22/2022]
Abstract
In future, neurodegenerative diseases will take over cancer's place and become the major cause of death in the world, especially in developed countries. Advancements in the medical field and its facilities have led to an increase in the old age population, and thus contributing to the increase in number of people suffering from neurodegenerative diseases. Economically it is of a great burden to society and the affected family. No current treatment aims to replace, protect, and regenerate lost neurons; instead, it alleviates the symptoms, extends the life span by a few months and creates severe side effects. Moreover, people who are affected are physically dependent for performing their basic activities, which makes their life miserable. There is an urgent need for therapy that could be able to overcome the deficits of conventional therapy for neurodegenerative diseases. Stem cells, the unspecialized cells with the properties of self-renewing and potency to differentiate into various cells types can become a potent therapeutic option for neurodegenerative diseases. Stem cells have been widely used in clinical trials to evaluate their potential in curing different types of ailments. In this review, we discuss the various types of stem cells and their potential use in the treatment of neurodegenerative disease based on published preclinical and clinical studies.
Collapse
Affiliation(s)
- Aishwarya L
- Department of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai-600 116. India
| | - Dharmarajan Arun
- Department of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai-600 116. India
| | - Suresh Kannan
- Department of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai-600 116. India
| |
Collapse
|
54
|
Global Transcriptional Analyses of the Wnt-Induced Development of Neural Stem Cells from Human Pluripotent Stem Cells. Int J Mol Sci 2021; 22:ijms22147473. [PMID: 34299091 PMCID: PMC8308016 DOI: 10.3390/ijms22147473] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 07/02/2021] [Accepted: 07/07/2021] [Indexed: 12/28/2022] Open
Abstract
The differentiation of human pluripotent stem cells (hPSCs) to neural stem cells (NSCs) is the key initial event in neurogenesis and is thought to be dependent on the family of Wnt growth factors, their receptors and signaling proteins. The delineation of the transcriptional pathways that mediate Wnt-induced hPSCs to NSCs differentiation is vital for understanding the global genomic mechanisms of the development of NSCs and, potentially, the creation of new protocols in regenerative medicine. To understand the genomic mechanism of Wnt signaling during NSCs development, we treated hPSCs with Wnt activator (CHIR-99021) and leukemia inhibitory factor (LIF) in a chemically defined medium (N2B27) to induce NSCs, referred to as CLNSCs. The CLNSCs were subcultured for more than 40 passages in vitro; were positive for AP staining; expressed neural progenitor markers such as NESTIN, PAX6, SOX2, and SOX1; and were able to differentiate into three neural lineage cells: neurons, astrocytes, and oligodendrocytes in vitro. Our transcriptome analyses revealed that the Wnt and Hedgehog signaling pathways regulate hPSCs cell fate decisions for neural lineages and maintain the self-renewal of CLNSCs. One interesting network could be the deregulation of the Wnt/β-catenin signaling pathway in CLNSCs via the downregulation of c-MYC, which may promote exit from pluripotency and neural differentiation. The Wnt-induced spinal markers HOXA1-4, HOXA7, HOXB1-4, and HOXC4 were increased, however, the brain markers FOXG1 and OTX2, were absent in the CLNSCs, indicating that CLNSCs have partial spinal cord properties. Finally, a CLNSC simple culture condition, when applied to hPSCs, supports the generation of NSCs, and provides a new and efficient cell model with which to untangle the mechanisms during neurogenesis.
Collapse
|
55
|
Abstract
Due to the ability to differentiate into variety of cell types, mesenchymal stem cells (MSCs) hold promise as source in cell-based therapy for treating injured tissue and degenerative diseases. The potential use of MSCs to replace or repair damaged tissues may depend on the efficient differentiation protocols to derive specialized cells without any negative side effects. Identification of appropriate cues that support the lineage-specific differentiation of stem cells is critical for tissue healing and cellular therapy. Recently, a number of stimuli have been utilized to direct the differentiation of stem cells. Biochemical stimuli such as small molecule, growth factor and miRNA have been traditionally used to regulate the fate of stem cells. In recent years, many studies have reported that biophysical stimuli including cyclic mechanical strain, fluid shear stress, microgravity, electrical stimulation, matrix stiffness and topography can also be sensed by stem cells through mechanical receptors, thus affecting the stem cell behaviors including their differentiation potential. In this paper, we review all the most recent literature on the application of biochemical and biophysical cues on regulating MSC differentiation. An extensive literature search was done using electronic database (Medline/Pubmed). Although there are still some challenges that need to be taken into consideration before translating these methods into clinics, biochemical and biophysical stimulation appears to be an attractive method to manipulate the lineage commitment of MSCs.
Collapse
|
56
|
van Eijk RPA, Kliest T, van den Berg LH. Current trends in the clinical trial landscape for amyotrophic lateral sclerosis. Curr Opin Neurol 2021; 33:655-661. [PMID: 32796282 DOI: 10.1097/wco.0000000000000861] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW To review the current developments in the design and conduct of clinical trials for amyotrophic lateral sclerosis (ALS), illustrated by a critical appraisal of ClinicalTrials.gov. RECENT FINDINGS In total, 63 clinical trials were included in the analysis, of which 13 phase 1, 35 phase 2 and 15 phase 3. Virtually all phase 3 clinical trials can be classified as randomized, placebo controlled, whereas this is only true for 57% of the phase 2 clinical trials. There are promising developments in the routes of drug administration, eligibility criteria, efficacy endpoints and overall trial design. Some of these innovative approaches may, however, not fulfil clinical trial guidelines or regulatory requirements. This could delay the development of effective therapy or hamper our ability to determine whether a treatment is truly (in)effective. The initiation of trial consortia comprising patient organizations, academia, industry and funding bodies may significantly strengthen the future clinical trial landscape for ALS. SUMMARY The ALS clinical trial landscape is currently highly active with several promising innovative developments and therapeutic options. By further refinement of evidence-based guidelines, and alignment of our current endeavours, we may soon be able to positively impact the lives of people living with ALS.
Collapse
Affiliation(s)
- Ruben P A van Eijk
- Department of Neurology, UMC Utrecht Brain Centre.,Biostatistics & Research Support, Julius Centre for Health Sciences and Primary Care, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Tessa Kliest
- Department of Neurology, UMC Utrecht Brain Centre
| | | |
Collapse
|
57
|
Looking backward to move forward: a meta-analysis of stem cell therapy in amyotrophic lateral sclerosis. NPJ Regen Med 2021; 6:20. [PMID: 33795700 PMCID: PMC8016966 DOI: 10.1038/s41536-021-00131-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 03/03/2021] [Indexed: 12/11/2022] Open
Abstract
Transplantation of several types of stem cells (SC) for the treatment of amyotrophic lateral sclerosis (ALS) has been evaluated in numerous Phase I/II clinical trials with inconclusive results. Here, we conducted a meta-analysis to systematically assess the outcome of SC therapy trials which report the evolution of each patient before and after cell administration. In this way, we aimed to determine the effect of the SC intervention despite individual heterogeneity in disease progression. We identified 670 references by electronic search and 90 full-text studies were evaluated according to the eligibility criteria. Eleven studies were included comprising 220 cell-treated patients who received mesenchymal (M) SC (n = 152), neural (N) SC (n = 57), or mononuclear cells (MNC: CD34, CD117, and CD133 positive cells) (n = 11). Our analyses indicate that whereas intrathecal injection of mesenchymal stromal cells appears to have a transient positive effect on clinical progression, as measured by the ALS functional rating score, there was a worsening of respiratory function measured by forced vital capacity after all interventions. Based on current evidence, we conclude that optimal cell product and route of administration need to be determined in properly controlled preclinical models before further advancing into ALS patients. In addition, in-depth understanding of disease mechanisms in subsets of patients will help tailoring SC therapy to specific targets and increase the likelihood of improving outcomes.
Collapse
|
58
|
Ahani-Nahayati M, Shariati A, Mahmoodi M, Olegovna Zekiy A, Javidi K, Shamlou S, Mousakhani A, Zamani M, Hassanzadeh A. Stem cell in neurodegenerative disorders; an emerging strategy. Int J Dev Neurosci 2021; 81:291-311. [PMID: 33650716 DOI: 10.1002/jdn.10101] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/13/2021] [Accepted: 02/24/2021] [Indexed: 01/28/2023] Open
Abstract
Neurodegenerative disorders are a diversity of disorders, surrounding Alzheimer's (AD), Parkinson's (PD), Huntington's diseases (HD), and amyotrophic lateral sclerosis (ALS) accompanied by some other less common diseases generally characterized by either developed deterioration of central or peripheral nervous system structurally or functionally. Today, with the viewpoint of an increasingly aging society, the number of patients with neurodegenerative diseases and sociomedical burdens will spread intensely. During the last decade, stem cell technology has attracted great attention for treating neurodegenerative diseases worldwide because of its unique attributes. As acknowledged, there are several categories of stem cells being able to proliferate and differentiate into various cellular lineages, highlighting their significance in the context of regenerative medicine. In preclinical models, stem cell therapy using mesenchymal stem/stromal cells (MSCs), hematopoietic stem cells (HSCs), and neural progenitor or stem cells (NPCs or NSCs) along with pluripotent stem cells (PSCs)-derived neuronal cells could elicit desired therapeutic effects, enabling functional deficit rescue partially. Regardless of the noteworthy progress in our scientific awareness and understanding of stem cell biology, there still exist various challenges to defeat. In the present review, we provide a summary of the therapeutic potential of stem cells and discuss the current status and prospect of stem cell strategy in neurodegenerative diseases, in particular, AD, PD, ALS, and HD.
Collapse
Affiliation(s)
- Milad Ahani-Nahayati
- Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Ali Shariati
- Stem Cell Research Center, Tehran University of Medical Science, Tehran, Iran
| | - Mahnaz Mahmoodi
- Department of Biology, School of Basic Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Angelina Olegovna Zekiy
- Department of Prosthetic Dentistry, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Kamran Javidi
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran.,Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Somayeh Shamlou
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Akbar Mousakhani
- Department of Plant Sciences, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Majid Zamani
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Ali Hassanzadeh
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Neurosciences Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
59
|
Abstract
Background: Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease of upper and lower motor neurons with high burden on society. Despite tremendous efforts over the last several decades, there is still no definite cure for ALS. Up to now, only two disease-modifying agents, riluzole and edaravone, are approved by U.S. Food and Drug Administration (FDA) for ALS treatment, which only modestly improves survival and disease progression. Major challenging issues to find an effective therapy are heterogeneity in the pathogenesis and genetic variability of ALS. As such, stem cell therapy has been recently a focus of both preclinical and clinical investigations of ALS. This is because stem cells have multifaceted features that can potentially target multiple pathogenic mechanisms in ALS even though its underlying mechanisms are not completely elucidated. Methods & Results: Here, we will have an overview of stem cell therapy in ALS, including their therapeutic mechanisms, the results of recent clinical trials as well as ongoing clinical trials. In addition, we will further discuss complications and limitations of stem cell therapy in ALS. Conclusion: The determination of whether stem cells offer a viable treatment strategy for ALS rests on well-designed and appropriately powered future clinical trials. Randomized, double-blinded, and sham-controlled studies would be valuable.
Collapse
Affiliation(s)
- Goun Je
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA USA
| | - Kiandokht Keyhanian
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA USA
| | - Mehdi Ghasemi
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA USA
| |
Collapse
|
60
|
Kim JH, Ko IK, Jeon MJ, Kim I, Vanschaayk MM, Atala A, Yoo JJ. Pelvic floor muscle function recovery using biofabricated tissue constructs with neuromuscular junctions. Acta Biomater 2021; 121:237-249. [PMID: 33321220 DOI: 10.1016/j.actbio.2020.12.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 12/06/2020] [Accepted: 12/08/2020] [Indexed: 01/01/2023]
Abstract
Damages in pelvic floor muscles often cause dysfunction of the entire pelvic urogenital system, which is clinically challenging. A bioengineered skeletal muscle construct that mimics structural and functional characteristics of native skeletal muscle could provide a therapeutic option to restore normal muscle function. However, most of the current bioengineered muscle constructs are unable to provide timely innervation necessary for successful grafting and functional recovery. We previously have demonstrated that post-synaptic acetylcholine receptors (AChR) clusters can be pre-formed on cultured skeletal muscle myofibers with agrin treatment and suggested that implantation of AChR clusters containing myofibers could accelerate innervation and recovery of muscle function. In this study, we develop a 3-dimensional (3D) bioprinted human skeletal muscle construct, consisting of multi-layers bundles with aligned and AChR clusters pre-formed human myofibers, and investigate the effect of pre-formed AChR clusters in bioprinted skeletal muscle constructs and innervation efficiency in vivo. Agrin treatment successfully pre-formed functional AChR clusters on the bioprinted muscle constructs in vitro that increased neuromuscular junction (NMJ) formation in vivo in a transposed nerve implantation model in rats. In a rat model of pelvic floor muscle injury, implantation of skeletal muscle constructs containing the pre-formed AChR clusters resulted in functional muscle reconstruction with accelerated construct innervation. This approach may provide a therapeutic solution to the many challenges associated with pelvic floor reconstruction resulting from the lack of suitable bioengineered tissue for efficient innervation and muscle function restoration.
Collapse
|
61
|
Abstract
Traumatic injuries are a leading cause of death and disability in both military and civilian populations. Given the complexity and diversity of traumatic injuries, novel and individualized treatment strategies are required to optimize outcomes. Cellular therapies have potential benefit for the treatment of acute or chronic injuries, and various cell-based pharmaceuticals are currently being tested in preclinical studies or in clinical trials. Cellular therapeutics may have the ability to complement existing therapies, especially in restoring organ function lost due to tissue disruption, prolonged hypoxia or inflammatory damage. In this article we highlight the current status and discuss future directions of cellular therapies for the treatment of traumatic injury. Both published research and ongoing clinical trials are discussed here.
Collapse
|
62
|
Leyton-Jaimes MF, Ivert P, Hoeber J, Han Y, Feiler A, Zhou C, Pankratova S, Shoshan-Barmatz V, Israelson A, Kozlova EN. Empty mesoporous silica particles significantly delay disease progression and extend survival in a mouse model of ALS. Sci Rep 2020; 10:20675. [PMID: 33244084 PMCID: PMC7691331 DOI: 10.1038/s41598-020-77578-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 11/11/2020] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating incurable neurological disorder characterized by motor neuron (MN) death and muscle dysfunction leading to mean survival time after diagnosis of only 2-5 years. A potential ALS treatment is to delay the loss of MNs and disease progression by the delivery of trophic factors. Previously, we demonstrated that implanted mesoporous silica nanoparticles (MSPs) loaded with trophic factor peptide mimetics support survival and induce differentiation of co-implanted embryonic stem cell (ESC)-derived MNs. Here, we investigate whether MSP loaded with peptide mimetics of ciliary neurotrophic factor (Cintrofin), glial-derived neurotrophic factor (Gliafin), and vascular endothelial growth factor (Vefin1) injected into the cervical spinal cord of mutant SOD1 mice affect disease progression and extend survival. We also transplanted boundary cap neural crest stem cells (bNCSCs) which have been shown previously to have a positive effect on MN survival in vitro and in vivo. We show that mimetic-loaded MSPs and bNCSCs significantly delay disease progression and increase survival of mutant SOD1 mice, and also that empty particles significantly improve the condition of ALS mice. Our results suggest that intraspinal delivery of MSPs is a potential therapeutic approach for the treatment of ALS.
Collapse
Affiliation(s)
- Marcel F Leyton-Jaimes
- Department of Physiology and Cell Biology, Zlotowski Center for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O.B. 653, 84105, Beer Sheva, Israel
| | - Patrik Ivert
- Department of Neuroscience, Regenerative Neurobiology, Uppsala University Biomedical Center, Box 593, 751 24, Uppsala, Sweden
| | - Jan Hoeber
- Department of Neuroscience, Regenerative Neurobiology, Uppsala University Biomedical Center, Box 593, 751 24, Uppsala, Sweden.,Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Box 815, 751 08, Uppsala, Sweden
| | - Yilin Han
- Department of Neuroscience, Regenerative Neurobiology, Uppsala University Biomedical Center, Box 593, 751 24, Uppsala, Sweden
| | - Adam Feiler
- Nanologica AB, Forskargatan 20G, 151 36, Södertälje, Sweden.,Chemistry Department, KTH, Royal Institute of Technology, 100 44, Stockholm, Sweden
| | - Chunfang Zhou
- Chemistry Department, KTH, Royal Institute of Technology, 100 44, Stockholm, Sweden
| | - Stanislava Pankratova
- Laboratory of Neural Plasticity, Department of Neuroscience, University of Copenhagen, 2200, Copenhagen, Denmark.,Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, 1870, Frederiksberg C, Denmark
| | - Varda Shoshan-Barmatz
- Department of Life Sciences, The National Institute for Biotechnology in the Negev Ltd, Ben-Gurion University of the Negev, 84105, Beer Sheva, Israel
| | - Adrian Israelson
- Department of Physiology and Cell Biology, Zlotowski Center for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O.B. 653, 84105, Beer Sheva, Israel.
| | - Elena N Kozlova
- Department of Neuroscience, Regenerative Neurobiology, Uppsala University Biomedical Center, Box 593, 751 24, Uppsala, Sweden.
| |
Collapse
|
63
|
Portnow J, Badie B, Suzette Blanchard M, Kilpatrick J, Tirughana R, Metz M, Mi S, Tran V, Ressler J, D'Apuzzo M, Aboody KS, Synold TW. Feasibility of intracerebrally administering multiple doses of genetically modified neural stem cells to locally produce chemotherapy in glioma patients. Cancer Gene Ther 2020; 28:294-306. [PMID: 32895489 PMCID: PMC8843788 DOI: 10.1038/s41417-020-00219-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 08/04/2020] [Accepted: 08/19/2020] [Indexed: 12/13/2022]
Abstract
Neural stem cells (NSCs) are tumor tropic and can be genetically modified to produce anti-cancer therapies locally in the brain. In a prior first-in-human study we demonstrated that a single dose of intracerebrally administered allogeneic NSCs, which were retrovirally transduced to express cytosine deaminase (CD), tracked to glioma sites and converted oral 5-fluorocytosine (5-FC) to 5-fluorouracil (5-FU). The next step in the clinical development of this NSC-based anti-cancer strategy was to assess the feasibility of administering multiple intracerebral doses of CD-expressing NSCs (CD-NSCs) in patients with recurrent high grade gliomas. CD-NSCs were given every 2 weeks using an indwelling brain catheter, followed each time by a 7-day course of oral 5-FC (and leucovorin in the final patient cohort). Fifteen evaluable patients received a median of 4 (range 2–10) intracerebral CD-NSC doses; doses were escalated from 50 x 106 to 150 x 106 CD-NSCs. Neuropharmacokinetic data confirmed that CD-NSCs continuously produced 5-FU in the brain during the course of 5-FC. There were no clinical signs of immunogenicity, and only three patients developed anti-NSC antibodies. Our results suggest intracerebral administration of serial doses of CD-NSCs is safe and feasible and identified a recommended dose for phase II testing of 150 x 106 CD-NSCs.
Collapse
Affiliation(s)
- Jana Portnow
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA, 91010, USA.
| | - Behnam Badie
- Department of Surgery, Division of Neurosurgery, City of Hope Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - M Suzette Blanchard
- Department of Computational and Quantitative Medicine, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Julie Kilpatrick
- Department of Clinical Research, City of Hope Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - Revathiswari Tirughana
- Department of Developmental and Stem Cell Biology, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA.,Office of IND Development and Regulatory Affairs, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Marianne Metz
- Department of Developmental and Stem Cell Biology, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Shu Mi
- Department of Cancer Biology, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Vivi Tran
- Department of Cancer Biology, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Julie Ressler
- Department of Diagnostic Radiology, City of Hope Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - Massimo D'Apuzzo
- Department of Pathology, City of Hope Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - Karen S Aboody
- Department of Developmental and Stem Cell Biology, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Timothy W Synold
- Department of Cancer Biology, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| |
Collapse
|
64
|
Moy A. Creating Catholic Regenerative Medicine Organizations in a Secular Biotechnology Field: A Physician-Scientist Experience. LINACRE QUARTERLY 2020; 87:218-222. [PMID: 32549639 DOI: 10.1177/0024363919890941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
One aspect of the progressive secularization of biotechnology is the use of the by-products from abortion and the use of human embryos. These morally illicit cells and tissue create a significant moral and economic challenge for Catholics at different stages of their career. A practicing Catholic physician or scientific professional will face the dilemma of how to reconcile their Catholic identity with their profession. While the Catechism is clear on what actions Catholics should not pursue, there has been less religious guidance on what activities Catholics should proactively pursue in their professional life to advance the Catholic culture. This essay will examine these themes through the lens of a true story of the author's experience in starting Catholic for-profit and nonprofit biotechnology organizations. Summary Abortion and the destruction of human embryos create a moral dilemma for Catholics at different stages of a physician or scientist's career. A practicing Catholic physician or scientist must reconcile their Catholic identity with their profession. While there is little professional guidance on how to advance the culture, Jesus says that one must take up the cross and direct their God-given gifts towards His name. The only way to succeed and thrive in a secular healthcare environment is to emulate Jesus by putting aside their own self-interest; pray for courage against ridicule; accept risk; and pursue scientific and medical excellence.
Collapse
Affiliation(s)
- Alan Moy
- Cellular Engineering Technologies Inc., Coralville, IA, USA.,John Paul II Medical Research Institute, Coralville, IA, USA
| |
Collapse
|
65
|
Klimenkov IV, Sudakov NP, Pastukhov MV, Kositsyn NS. The Phenomenon of Compensatory Cell Proliferation in Olfactory Epithelium in Fish Caused by Prolonged Exposure to Natural Odorants. Sci Rep 2020; 10:8908. [PMID: 32483178 PMCID: PMC7264137 DOI: 10.1038/s41598-020-65854-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 05/05/2020] [Indexed: 12/13/2022] Open
Abstract
It was previously shown that activation of the processes of neurogenesis in the olfactory epithelium (OE) can be caused after intranasal administration of toxic or neurotrophic factors, after axon transection, or as a result of bulbectomy. Our study showed for the first time that a significant increase in olfactory cell renewal can also occur in animals due to periodic chemostimulation with natural odorants (amino acids and peptides) for 15 days. Using electron and laser confocal microscopy in fish (Paracottus knerii (Cottidae), Dybowski, 1874) from Lake Baikal, we showed that periodic stimulation of aquatic organisms with a water-soluble mixture of amino acids and peptides causes stress in OE, which leads to programmed death cells and compensatory intensification of their renewal. We estimated the level of reactive oxygen species, number of functionally active mitochondria, intensity of apoptosis processes, and mitosis activity of cells in the OE of fish in the control group and after periodic natural odorants exposure. This study showed that new stem cells are activated during enhanced odor stimulation and subsequent degenerative changes in the cells of the sensory apparatus. Those new activated stem cells are located in previously proliferatively inactive regions of OE that become involved in compensatory processes for the formation of new cells.
Collapse
Affiliation(s)
- Igor V Klimenkov
- Limnological Institute, Siberian Branch, Russian Academy of Sciences, 3 Ulan-Batorskaya St., Irkutsk, 664033, Russia. .,Irkutsk State University, 1 Karl Marx St., Irkutsk, 664003, Russia.
| | - Nikolay P Sudakov
- Limnological Institute, Siberian Branch, Russian Academy of Sciences, 3 Ulan-Batorskaya St., Irkutsk, 664033, Russia
| | - Mikhail V Pastukhov
- Vinogradov Institute of Geochemistry, Siberian Branch, Russian Academy of Sciences, 1a Favorsky St., Irkutsk, 664033, Russia
| | - Nikolay S Kositsyn
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 5a Butlerova St., Moscow, 117485, Russia
| |
Collapse
|
66
|
Ueda T, Inden M, Ito T, Kurita H, Hozumi I. Characteristics and Therapeutic Potential of Dental Pulp Stem Cells on Neurodegenerative Diseases. Front Neurosci 2020; 14:407. [PMID: 32457568 PMCID: PMC7222959 DOI: 10.3389/fnins.2020.00407] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 04/03/2020] [Indexed: 12/13/2022] Open
Abstract
To evaluate the therapeutic potential of stem cells for neurodegenerative diseases, emphasis should be placed on clarifying the characteristics of the various types of stem cells. Among stem cells, dental pulp stem cells (DPSCs) are a cell population that is rich in cell proliferation and multipotency. It has been reported that transplantation of DPSCs has protective effects against models of neurodegenerative diseases. The protective effects are not only through differentiation into the target cell type for the disease but are also related to trophic factors released from DPSCs. Recently, it has been reported that serum-free culture supernatant of dental pulp stem cell-conditioned medium (DPCM) contains various trophic factors and cytokines and that DPCM is effective for models of neurodegenerative diseases such as Alzheimer’s disease (AD), Parkinson’s disease (PD), and Amyotrophic Lateral Sclerosis (ALS). Moreover, the use of stem cells from human exfoliated deciduous teeth (SHEDs) has been considered. SHEDs are derived from deciduous teeth that have been disposed of as medical waste. SHEDs have higher differentiation capacity and proliferation ability than DPSCs. In addition, the serum-free culture supernatant of SHEDs (SHED-CM) contains more trophic factors, cytokines, and biometals than DPCM and also promotes neuroprotection. The neuroprotective effect of DPSCs, including those from deciduous teeth, will be used as the seeds of therapeutic drugs for neurodegenerative diseases. SHEDs will be used for further cell therapy of neurodegenerative diseases in the future. In this paper, we focused on the characteristics of DPSCs and their potential for neurodegenerative diseases.
Collapse
Affiliation(s)
- Tomoyuki Ueda
- Laboratory of Medical Therapeutics and Molecular Therapeutics, Gifu Pharmaceutical University, Gifu, Japan
| | - Masatoshi Inden
- Laboratory of Medical Therapeutics and Molecular Therapeutics, Gifu Pharmaceutical University, Gifu, Japan
| | - Taisei Ito
- Laboratory of Medical Therapeutics and Molecular Therapeutics, Gifu Pharmaceutical University, Gifu, Japan
| | - Hisaka Kurita
- Laboratory of Medical Therapeutics and Molecular Therapeutics, Gifu Pharmaceutical University, Gifu, Japan
| | - Isao Hozumi
- Laboratory of Medical Therapeutics and Molecular Therapeutics, Gifu Pharmaceutical University, Gifu, Japan
| |
Collapse
|
67
|
Pernia C, Tobe BTD, O'Donnell R, Snyder EY. The Evolution of Stem Cells, Disease Modeling, and Drug Discovery for Neurological Disorders. Stem Cells Dev 2020; 29:1131-1141. [PMID: 32024446 DOI: 10.1089/scd.2019.0217] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Human neurological disorders are among the most challenging areas of translational research. The difficulty of acquiring human neural samples or specific representative animal models has necessitated a multifaceted approach to understanding disease pathology and drug discovery. The dedifferentiation of somatic cells to human induced pluripotent stem cells (hiPSCs) for the generation of neural derivatives has broadened the capability of biomedical research to study human cell types in neurological disorders. The initial zeal for the potential of hiPSCs for immediate biomedical breakthroughs has evolved to more reasonable expectations. Over the past decade, hiPSC technology has demonstrated the capacity to successfully establish "disease in a dish" models of complex neurological disorders and to identify possible novel therapeutics. However, as hiPSCs are used more broadly, an increased understanding of the limitations of hiPSC studies is becoming more evident. In this study, we review the challenges of studying neurological disorders, the current limitations of stem cell-based disease modeling, and the degrees to which hiPSC studies to date have demonstrated the capacity to fill essential gaps in neurological research.
Collapse
Affiliation(s)
- Cameron Pernia
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, California, USA
| | - Brian T D Tobe
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, California, USA.,Department of Psychiatry, Veterans Administration Medical Center, La Jolla, California, USA
| | - Ryan O'Donnell
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, California, USA
| | - Evan Y Snyder
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, California, USA
| |
Collapse
|
68
|
Marchini A, Favoino C, Gelain F. Multi-Functionalized Self-Assembling Peptides as Reproducible 3D Cell Culture Systems Enabling Differentiation and Survival of Various Human Neural Stem Cell Lines. Front Neurosci 2020; 14:413. [PMID: 32431590 PMCID: PMC7214803 DOI: 10.3389/fnins.2020.00413] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/06/2020] [Indexed: 12/13/2022] Open
Abstract
Neural stem cells-based therapies have shown great potential for central nervous system regeneration, with three-dimensional (3D) culture systems representing a key technique for tissue engineering applications, as well as disease modeling and drug screenings. Self-assembling peptides (SAPs), providing biomimetic synthetic micro-environments regulating cellular functionality and tissue repair, constitute a suitable tool for the production of complex tissue-like structures in vitro. However, one of the most important drawbacks in 3D cultures, obtained via animal-derived substrates and serum-rich media, is the reproducibility and tunability of a standardized methodology capable to coax neural differentiation of different human cell lines. In this work we cultured four distinct human neural stem cell (hNSC) lines in 3D synthetic multifunctionalized hydrogel (named HYDROSAP) for up to 6 weeks. Three-dimensional cultures of differentiating hNSCs exhibited a progressive differentiation and maturation over time. All hNSCs-derived neurons in 3D culture system exhibited randomly organized entangled networks with increasing expression of GABAergic and glutamatergic phenotypes and presence of cholinergic ones. Oligodendrocytes formed insulating myelin sheaths positive for myelin basic protein (MBP). In summary, results demonstrated a successfully standardized and reproducible 3D cell culture system for hNSC differentiation and maturation in serum-free conditions useful for future therapies.
Collapse
Affiliation(s)
- Amanda Marchini
- Tissue Engineering Unit, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies-ISBReMIT, Fondazione IRCSS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.,Center for Nanomedicine and Tissue Engineering, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Chiara Favoino
- Center for Nanomedicine and Tissue Engineering, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy.,Department of Biotechnology and Biosciences, University of Milan-Bicocca, Milan, Italy
| | - Fabrizio Gelain
- Tissue Engineering Unit, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies-ISBReMIT, Fondazione IRCSS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.,Center for Nanomedicine and Tissue Engineering, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| |
Collapse
|
69
|
Disease-modifying therapies in amyotrophic lateral sclerosis. Neuropharmacology 2020; 167:107986. [DOI: 10.1016/j.neuropharm.2020.107986] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 01/21/2020] [Accepted: 01/31/2020] [Indexed: 02/08/2023]
|
70
|
Lentiviral Vector Induced Modeling of High-Grade Spinal Cord Glioma in Minipigs. Sci Rep 2020; 10:5291. [PMID: 32210315 PMCID: PMC7093438 DOI: 10.1038/s41598-020-62167-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/09/2020] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Prior studies have applied driver mutations targeting the RTK/RAS/PI3K and p53 pathways to induce the formation of high-grade gliomas in rodent models. In the present study, we report the production of a high-grade spinal cord glioma model in pigs using lentiviral gene transfer. METHODS Six Gottingen Minipigs received thoracolumbar (T14-L1) lateral white matter injections of a combination of lentiviral vectors, expressing platelet-derived growth factor beta (PDGF-B), constitutive HRAS, and shRNA-p53 respectively. All animals received injection of control vectors into the contralateral cord. Animals underwent baseline and endpoint magnetic resonance imaging (MRI) and were evaluated daily for clinical deficits. Hematoxylin and eosin (H&E) and immunohistochemical analysis was conducted. Data are presented using descriptive statistics including relative frequencies, mean, standard deviation, and range. RESULTS 100% of animals (n = 6/6) developed clinical motor deficits ipsilateral to the oncogenic lentiviral injections by a three-week endpoint. MRI scans at endpoint demonstrated contrast enhancing mass lesions at the site of oncogenic lentiviral injection and not at the site of control injections. Immunohistochemistry demonstrated positive staining for GFAP, Olig2, and a high Ki-67 proliferative index. Histopathologic features demonstrate consistent and reproducible growth of a high-grade glioma in all animals. CONCLUSIONS Lentiviral gene transfer represents a feasible pathway to glioma modeling in higher order species. The present model is the first lentiviral vector induced pig model of high-grade spinal cord glioma and may potentially be used in preclinical therapeutic development programs.
Collapse
|
71
|
Padmakumar S, Taha MS, Kadakia E, Bleier BS, Amiji MM. Delivery of neurotrophic factors in the treatment of age-related chronic neurodegenerative diseases. Expert Opin Drug Deliv 2020; 17:323-340. [DOI: 10.1080/17425247.2020.1727443] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Smrithi Padmakumar
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA, USA
| | - Maie S. Taha
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA, USA
- Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Ekta Kadakia
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA, USA
- Drug Metabolism and Pharmacokinetics (DMPK), Biogen Inc, Cambridge, MA, USA
| | - Benjamin S. Bleier
- Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| | - Mansoor M. Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA, USA
| |
Collapse
|
72
|
Denninger JK, Chen X, Turkoglu AM, Sarchet P, Volk AR, Rieskamp JD, Yan P, Kirby ED. Defining the adult hippocampal neural stem cell secretome: In vivo versus in vitro transcriptomic differences and their correlation to secreted protein levels. Brain Res 2020; 1735:146717. [PMID: 32035887 DOI: 10.1016/j.brainres.2020.146717] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 02/03/2020] [Accepted: 02/05/2020] [Indexed: 01/08/2023]
Abstract
Adult hippocampal neural stem and progenitor cells (NSPCs) secrete a variety of proteins that affect tissue function. Though several individual NSPC-derived proteins have been shown to impact key cellular processes, a broad characterization is lacking. Secretome profiling of low abundance stem cell populations is typically achieved via proteomic characterization of in vitro, isolated cells. Here, we identified hundreds of secreted proteins in conditioned media from in vitro adult mouse hippocampal NSPCs using an antibody array and mass spectrometry. Comparison of protein abundance between antibody array and mass spectrometry plus quantification of several key secreted proteins by ELISA revealed notable disconnect between methods in what proteins were identified as being high versus low abundance, suggesting that data from antibody arrays in particular should be approached with caution. We next assessed the NSPC secretome on a transcriptional level with single cell and bulk RNA sequencing (RNAseq) of cultured NSPCs. Comparison of RNAseq transcript levels of highly secreted proteins revealed that quantification of gene expression did not necessarily predict relative protein abundance. Interestingly, comparing our in vitro NSPC gene expression data with similar data from freshly isolated, in vivo hippocampal NSPCs revealed strong correlations in global gene expression between in vitro and in vivo NSPCs. Understanding the components and functions of the NSPC secretome is essential to understanding how these cells may modulate the hippocampal neurogenic niche. Cumulatively, our data emphasize the importance of using proteomics in conjunction with transcriptomics and highlights the need for better methods of unbiased secretome profiling.
Collapse
Affiliation(s)
- Jiyeon K Denninger
- Department of Psychology, College of Arts and Sciences, The Ohio State University, United States
| | - Xi Chen
- Comprehensive Cancer Center, The Ohio State University, United States
| | - Altan M Turkoglu
- College of Arts and Sciences, The Ohio State University, United States
| | - Patricia Sarchet
- Comprehensive Cancer Center, The Ohio State University, United States
| | - Abby R Volk
- College of Arts and Sciences, The Ohio State University, United States
| | - Joshua D Rieskamp
- Neuroscience Graduate Program, The Ohio State University, United States
| | - Pearlly Yan
- Comprehensive Cancer Center, The Ohio State University, United States; Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, United States
| | - Elizabeth D Kirby
- Department of Psychology, College of Arts and Sciences, The Ohio State University, United States; Department of Neuroscience, The Ohio State University, United States; Chronic Brain Injury Initiative, The Ohio State University, United States.
| |
Collapse
|
73
|
Willis CM, Nicaise AM, Peruzzotti-Jametti L, Pluchino S. The neural stem cell secretome and its role in brain repair. Brain Res 2020; 1729:146615. [DOI: 10.1016/j.brainres.2019.146615] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 12/05/2019] [Accepted: 12/16/2019] [Indexed: 12/13/2022]
|
74
|
Atala A. Most-read articles 2019: A year of noteworthy research. Stem Cells Transl Med 2020; 9:4-5. [PMID: 31926060 PMCID: PMC6954737 DOI: 10.1002/sctm.19-0437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 12/19/2019] [Indexed: 11/19/2022] Open
Affiliation(s)
- Anthony Atala
- Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
75
|
Atkinson SP. A Preview of Selected Articles. Stem Cells Transl Med 2019. [PMCID: PMC6708068 DOI: 10.1002/sctm.19-0225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|