1
|
Kim S, Sharma C, Hong J, Kim JH, Nam Y, Kim MS, Lee TY, Kim KS, Suk K, Lee HW, Kim SR. Post-symptomatic administration of hMSCs exerts therapeutic effects in SCA2 mice. Stem Cell Res Ther 2024; 15:411. [PMID: 39521966 PMCID: PMC11550562 DOI: 10.1186/s13287-024-04020-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Defects in the ataxin-2 (ATXN-2) protein and CAG trinucleotide repeat expansion in its coding gene, Atxn-2, cause the neurodegenerative disorder spinocerebellar ataxia type 2 (SCA2). While clinical studies suggest potential benefits of human-derived mesenchymal stem cells (hMSCs) for treating various ataxias, the exact mechanisms underlying their therapeutic effects and interaction with host tissue to stimulate neurotrophin expression remain unclear specifically in the context of SCA2. METHODS Human bone marrow-derived MSCs (hMSCs) were injected into the cisterna magna of 26-week-old wild-type and SCA2 mice. Mice were assessed for impaired motor coordination using the accelerating rotarod, open field test, and composite phenotype scoring. At 50 weeks, the cerebellum vermis was harvested for protein assessment and immunohistochemical analysis. RESULTS Significant loss of NeuN and calbindin was observed in 25-week-old SCA2 mice. However, after receiving multiple injections of hMSCs starting at 26 weeks of age, these mice exhibited a significant improvement in abnormal motor performance and a protective effect on Purkinje cells. This beneficial effect persisted until the mice reached 50 weeks of age, at which point they were sacrificed to study further mechanistic events triggered by the administration of hMSCs. Calbindin-positive cells in the Purkinje cell layer expressed bone-derived neurotrophic factor after hMSC administration, contributing to the protection of cerebellar neurons from cell death. CONCLUSION In conclusion, repeated administration of hMSCs shows promise in alleviating SCA2 symptoms by preserving Purkinje cells, improving neurotrophic support, and reducing inflammation, ultimately leading to the preservation of locomotor function in SCA2 mice.
Collapse
Affiliation(s)
- Sehwan Kim
- School of Life Science and Biotechnology, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, Korea
- Brain Science and Engineering Institute, Kyungpook National University, Daegu, 41944, Korea
| | - Chanchal Sharma
- School of Life Science and Biotechnology, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, Korea
- Byrd Alzheimer's Centre and Research Institute, University of South Florida, Tampa, FL, 33620, USA
| | - Jungwan Hong
- Brain Science and Engineering Institute, Kyungpook National University, Daegu, 41944, Korea
| | - Jong-Heon Kim
- Brain Science and Engineering Institute, Kyungpook National University, Daegu, 41944, Korea
| | - Youngpyo Nam
- Brain Science and Engineering Institute, Kyungpook National University, Daegu, 41944, Korea
| | - Min Sung Kim
- Bioengineering Institute, Corestemchemon Inc, Seoul, 13486, Korea
| | - Tae Yong Lee
- Bioengineering Institute, Corestemchemon Inc, Seoul, 13486, Korea
| | - Kyung-Suk Kim
- Bioengineering Institute, Corestemchemon Inc, Seoul, 13486, Korea
| | - Kyoungho Suk
- Brain Science and Engineering Institute, Kyungpook National University, Daegu, 41944, Korea
- Department of Pharmacology and Biomedical Science, School of Medicine, Kyungpook National University, Daegu, 41944, Korea
| | - Ho-Won Lee
- Brain Science and Engineering Institute, Kyungpook National University, Daegu, 41944, Korea
- Department of Neurology, Kyungpook National University Chilgok Hospital, Daegu, 41404, Korea
| | - Sang Ryong Kim
- School of Life Science and Biotechnology, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, Korea.
- Brain Science and Engineering Institute, Kyungpook National University, Daegu, 41944, Korea.
| |
Collapse
|
2
|
Serafini MM, Sepehri S, Midali M, Stinckens M, Biesiekierska M, Wolniakowska A, Gatzios A, Rundén-Pran E, Reszka E, Marinovich M, Vanhaecke T, Roszak J, Viviani B, SenGupta T. Recent advances and current challenges of new approach methodologies in developmental and adult neurotoxicity testing. Arch Toxicol 2024; 98:1271-1295. [PMID: 38480536 PMCID: PMC10965660 DOI: 10.1007/s00204-024-03703-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/06/2024] [Indexed: 03/27/2024]
Abstract
Adult neurotoxicity (ANT) and developmental neurotoxicity (DNT) assessments aim to understand the adverse effects and underlying mechanisms of toxicants on the human nervous system. In recent years, there has been an increasing focus on the so-called new approach methodologies (NAMs). The Organization for Economic Co-operation and Development (OECD), together with European and American regulatory agencies, promote the use of validated alternative test systems, but to date, guidelines for regulatory DNT and ANT assessment rely primarily on classical animal testing. Alternative methods include both non-animal approaches and test systems on non-vertebrates (e.g., nematodes) or non-mammals (e.g., fish). Therefore, this review summarizes the recent advances of NAMs focusing on ANT and DNT and highlights the potential and current critical issues for the full implementation of these methods in the future. The status of the DNT in vitro battery (DNT IVB) is also reviewed as a first step of NAMs for the assessment of neurotoxicity in the regulatory context. Critical issues such as (i) the need for test batteries and method integration (from in silico and in vitro to in vivo alternatives, e.g., zebrafish, C. elegans) requiring interdisciplinarity to manage complexity, (ii) interlaboratory transferability, and (iii) the urgent need for method validation are discussed.
Collapse
Affiliation(s)
- Melania Maria Serafini
- Department of Pharmacological and Biomolecular Sciences, "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy.
| | - Sara Sepehri
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussels, Brussels, Belgium
| | - Miriam Midali
- Department of Pharmacological and Biomolecular Sciences, "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Marth Stinckens
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussels, Brussels, Belgium
| | - Marta Biesiekierska
- Department of Translational Research, Nofer Institute of Occupational Medicine, Lodz, Poland
| | - Anna Wolniakowska
- Department of Translational Research, Nofer Institute of Occupational Medicine, Lodz, Poland
| | - Alexandra Gatzios
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussels, Brussels, Belgium
| | - Elise Rundén-Pran
- The Climate and Environmental Research Institute NILU, Kjeller, Norway
| | - Edyta Reszka
- Department of Translational Research, Nofer Institute of Occupational Medicine, Lodz, Poland
| | - Marina Marinovich
- Department of Pharmacological and Biomolecular Sciences, "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
- Center of Research on New Approach Methodologies (NAMs) in chemical risk assessment (SAFE-MI), Università degli Studi di Milano, Milan, Italy
| | - Tamara Vanhaecke
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussels, Brussels, Belgium
| | - Joanna Roszak
- Department of Translational Research, Nofer Institute of Occupational Medicine, Lodz, Poland
| | - Barbara Viviani
- Department of Pharmacological and Biomolecular Sciences, "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
- Center of Research on New Approach Methodologies (NAMs) in chemical risk assessment (SAFE-MI), Università degli Studi di Milano, Milan, Italy
| | - Tanima SenGupta
- The Climate and Environmental Research Institute NILU, Kjeller, Norway
| |
Collapse
|
3
|
Okarmus J, Agergaard JB, Stummann TC, Haukedal H, Ambjørn M, Freude KK, Fog K, Meyer M. USP30 inhibition induces mitophagy and reduces oxidative stress in parkin-deficient human neurons. Cell Death Dis 2024; 15:52. [PMID: 38225227 PMCID: PMC10789816 DOI: 10.1038/s41419-024-06439-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 12/22/2023] [Accepted: 01/04/2024] [Indexed: 01/17/2024]
Abstract
Ubiquitination of mitochondrial proteins plays an important role in the cellular regulation of mitophagy. The E3 ubiquitin ligase parkin (encoded by PARK2) and the ubiquitin-specific protease 30 (USP30) have both been reported to regulate the ubiquitination of outer mitochondrial proteins and thereby mitophagy. Loss of E3 ligase activity is thought to be pathogenic in both sporadic and inherited Parkinson's disease (PD), with loss-of-function mutations in PARK2 being the most frequent cause of autosomal recessive PD. The aim of the present study was to evaluate whether mitophagy induced by USP30 inhibition provides a functional rescue in isogenic human induced pluripotent stem cell-derived dopaminergic neurons with and without PARK2 knockout (KO). Our data show that healthy neurons responded to CCCP-induced mitochondrial damage by clearing the impaired mitochondria and that this process was accelerated by USP30 inhibition. Parkin-deficient neurons showed an impaired mitophagic response to the CCCP challenge, although mitochondrial ubiquitination was enhanced. USP30 inhibition promoted mitophagy in PARK2 KO neurons, independently of whether left in basal conditions or treated with CCCP. In PARK2 KO, as in control neurons, USP30 inhibition balanced oxidative stress levels by reducing excessive production of reactive oxygen species. Interestingly, non-dopaminergic neurons were the main driver of the beneficial effects of USP30 inhibition. Our findings demonstrate that USP30 inhibition is a promising approach to boost mitophagy and improve cellular health, also in parkin-deficient cells, and support the potential relevance of USP30 inhibitors as a novel therapeutic approach in diseases with a need to combat neuronal stress mediated by impaired mitochondria.
Collapse
Affiliation(s)
- Justyna Okarmus
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, J.B. Winsløws Vej 21, 5000, Odense C, Denmark
| | - Jette Bach Agergaard
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, J.B. Winsløws Vej 21, 5000, Odense C, Denmark
| | - Tina C Stummann
- Neuroscience, H. Lundbeck A/S, Ottiliavej 9, 2500, Valby, Denmark
| | - Henriette Haukedal
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Grønnegaardsvej 7, 1870, Frederiksberg C, Denmark
| | - Malene Ambjørn
- Neuroscience, H. Lundbeck A/S, Ottiliavej 9, 2500, Valby, Denmark
| | - Kristine K Freude
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Grønnegaardsvej 7, 1870, Frederiksberg C, Denmark
| | - Karina Fog
- Neuroscience, H. Lundbeck A/S, Ottiliavej 9, 2500, Valby, Denmark
| | - Morten Meyer
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, J.B. Winsløws Vej 21, 5000, Odense C, Denmark.
- Department of Neurology, Odense University Hospital, J.B. Winsløws Vej 4, 5000, Odense C, Denmark.
- BRIDGE-Brain Research Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, J.B. Winsløws Vej 19, 5000, Odense C, Denmark.
| |
Collapse
|
4
|
Adegunsoye A, Gonzales NM, Gilad Y. Induced Pluripotent Stem Cells in Disease Biology and the Evidence for Their In Vitro Utility. Annu Rev Genet 2023; 57:341-360. [PMID: 37708421 DOI: 10.1146/annurev-genet-022123-090319] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2023]
Abstract
Many human phenotypes are impossible to recapitulate in model organisms or immortalized human cell lines. Induced pluripotent stem cells (iPSCs) offer a way to study disease mechanisms in a variety of differentiated cell types while circumventing ethical and practical issues associated with finite tissue sources and postmortem states. Here, we discuss the broad utility of iPSCs in genetic medicine and describe how they are being used to study musculoskeletal, pulmonary, neurologic, and cardiac phenotypes. We summarize the particular challenges presented by each organ system and describe how iPSC models are being used to address them. Finally, we discuss emerging iPSC-derived organoid models and the potential value that they can bring to studies of human disease.
Collapse
Affiliation(s)
- Ayodeji Adegunsoye
- Genetics, Genomics, and Systems Biology, Section of Pulmonary and Critical Care, and the Department of Medicine, University of Chicago, Chicago, Illinois, USA;
| | - Natalia M Gonzales
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, USA; ,
| | - Yoav Gilad
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, USA; ,
- Department of Human Genetics, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
5
|
Sharlow ER, Llaneza DC, Tewari BP, Mingledorff GA, Mendelson AJ, Sontheimer H, Bloom GS, Lazo JS. Pharmacological profiling identifies divergent chemosensitivities of differentiating and maturing iPSC-derived human cortical neuron populations. FEBS J 2023; 290:4950-4965. [PMID: 37428551 PMCID: PMC10592385 DOI: 10.1111/febs.16901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 06/16/2023] [Accepted: 07/07/2023] [Indexed: 07/11/2023]
Abstract
Neuronal differentiation and maturation are extended developmental processes. To determine whether neurons at different developmental stages have divergent chemosensitivities, we screened differentiating and maturing neuronal populations using a small compound library comprising FDA-approved and investigational drugs. Using a neurotoxicity assay format, both respective neuronal population-based screening campaigns performed robustly (Z-factors = 0.7-0.8), although the hit rate for the differentiating neurons (2.8%) was slightly higher than for maturing neurons (1.9%). While the majority of hits were toxic to both neuronal populations, these hits predominantly represented promiscuous drugs. Other drugs were selectively neurotoxic, with receptor tyrosine kinase inhibitors disproportionally represented after confirmation. Ponatinib and amuvatinib were neuroinhibitory for differentiating and maturing neurons, respectively. Chemoinformatic analyses confirmed differences in potential drug targets that may be differentially expressed during neuronal development. Subsequent studies demonstrated neuronal expression of AXL, an amuvatinib target, in both neuronal populations. However, functional AXL activity was confirmed only in the maturing neuronal population as determined by AXL phosphorylation in response to GAS6, the cognate ligand of AXL, and concurrent STAT3Y705 phosphorylation. Differentiating neurons were unresponsive to the effects of GAS6 suggesting that the AXL-STAT3 signaling axis was nonfunctional. Amuvatinib treatment of maturing neuronal cultures significantly reduced pAXL levels. These studies indicate that neuronal developmental states may exhibit unique chemosensitivities and that drugs may have different neuro-inhibitory effects depending upon the developmental stage of the neuronal population.
Collapse
Affiliation(s)
| | - Danielle C. Llaneza
- Department of Pharmacology, University of Virginia, Charlottesville, VA, 22908
| | - Bhanu P. Tewari
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22908
| | | | - Anna J. Mendelson
- Department of Pharmacology, University of Virginia, Charlottesville, VA, 22908
| | - Harald Sontheimer
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22908
| | - George S. Bloom
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22908
- Department of Biology, University of Virginia, Charlottesville, VA 22904
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908
| | - John S. Lazo
- Department of Pharmacology, University of Virginia, Charlottesville, VA, 22908
| |
Collapse
|
6
|
Radoszkiewicz K, Hribljan V, Isakovic J, Mitrecic D, Sarnowska A. Critical points for optimizing long-term culture and neural differentiation capacity of rodent and human neural stem cells to facilitate translation into clinical settings. Exp Neurol 2023; 363:114353. [PMID: 36841464 DOI: 10.1016/j.expneurol.2023.114353] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 02/03/2023] [Accepted: 02/18/2023] [Indexed: 02/27/2023]
Abstract
Despite several decades of research on the nature and functional properties of neural stem cells, which brought great advances in regenerative medicine, there is still a plethora of ambiguous protocols and interpretations linked to their applications. Here, we present a whole spectrum of protocol elements that should be standardized in order to obtain viable cell cultures and facilitate their translation into clinical settings. Additionally, this review also presents outstanding limitations and possible problems to be encountered when dealing with protocol optimization. Most importantly, we also outline the critical points that should be considered before starting any experiments utilizing neural stem cells or interpreting their results.
Collapse
Affiliation(s)
- Klaudia Radoszkiewicz
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawinskiego 5 Street, 02-106 Warsaw, Poland
| | - Valentina Hribljan
- Laboratory for Stem Cells, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Šalata 12, Zagreb, Croatia
| | - Jasmina Isakovic
- Omnion Research International Ltd, Heinzelova 4, 10000 Zagreb, Croatia
| | - Dinko Mitrecic
- Laboratory for Stem Cells, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Šalata 12, Zagreb, Croatia
| | - Anna Sarnowska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawinskiego 5 Street, 02-106 Warsaw, Poland.
| |
Collapse
|
7
|
Chow SYA, Hu H, Osaki T, Levi T, Ikeuchi Y. Advances in construction and modeling of functional neural circuits in vitro. Neurochem Res 2022; 47:2529-2544. [PMID: 35943626 PMCID: PMC9463289 DOI: 10.1007/s11064-022-03682-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/26/2022] [Accepted: 07/08/2022] [Indexed: 11/26/2022]
Abstract
Over the years, techniques have been developed to culture and assemble neurons, which brought us closer to creating neuronal circuits that functionally and structurally mimic parts of the brain. Starting with primary culture of neurons, preparations of neuronal culture have advanced substantially. Development of stem cell research and brain organoids has opened a new path for generating three-dimensional human neural circuits. Along with the progress in biology, engineering technologies advanced and paved the way for construction of neural circuit structures. In this article, we overview research progress and discuss perspective of in vitro neural circuits and their ability and potential to acquire functions. Construction of in vitro neural circuits with complex higher-order functions would be achieved by converging development in diverse major disciplines including neuroscience, stem cell biology, tissue engineering, electrical engineering and computer science.
Collapse
Affiliation(s)
- Siu Yu A Chow
- Institute of Industrial Science, The University of Tokyo, Tokyo, Japan
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo, Japan
- Institute for AI and Beyond, The University of Tokyo, Tokyo, Japan
| | - Huaruo Hu
- Institute of Industrial Science, The University of Tokyo, Tokyo, Japan
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Tatsuya Osaki
- Institute of Industrial Science, The University of Tokyo, Tokyo, Japan
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo, Japan
- Institute for AI and Beyond, The University of Tokyo, Tokyo, Japan
| | - Timothée Levi
- Institute of Industrial Science, The University of Tokyo, Tokyo, Japan
- Institute for AI and Beyond, The University of Tokyo, Tokyo, Japan
- IMS laboratory, CNRS UMR 5218, University of Bordeaux, Talence, France
| | - Yoshiho Ikeuchi
- Institute of Industrial Science, The University of Tokyo, Tokyo, Japan.
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo, Japan.
- Institute for AI and Beyond, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
8
|
Devina T, Wong YH, Hsiao CW, Li YJ, Lien CC, Cheng IHJ. Endoplasmic reticulum stress induces Alzheimer's disease-like phenotypes in the neuron derived from the induced pluripotent stem cell with D678H mutation on amyloid precursor protein. J Neurochem 2022; 163:26-39. [PMID: 35943292 DOI: 10.1111/jnc.15687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 07/12/2022] [Accepted: 08/06/2022] [Indexed: 11/28/2022]
Abstract
Alzheimer's disease (AD), a progressive neurodegenerative disorder, is mainly caused by the interaction of genetic and environmental factors. The impact of environmental factors on the genetic mutation in the amyloid precursor protein (APP) is not well characterized. We hypothesized that Endoplasmic Reticulum (ER) stress would promote disease for the patient carrying the APP D678H mutation. Therefore, we analyzed the impact of a familial AD mutation on amyloid precursor protein (APP D678H) under ER stress. Induced pluripotent stem cell (iPSC) from APP D678H mutant carrier was differentiated into neurons, which were then analyzed for AD-like changes. Immunocytochemistry and whole-cell patch-clamp recording revealed that the derived neurons on day 28 after differentiation showed neuronal markers and electrophysiological properties similar to those of mature neurons. However, the APP D678H mutant neurons did not have significant alterations in the levels of amyloid-β (Aβ) and phosphorylated tau (pTau) compared to its isogenic wild-type neuron. Only under ER stress, the neurons with the APP D678H mutation had more Aβ and pTau via immune detection assays. The higher level of Aβ in the APP D678H mutant neurons was probably due to the increased level of β-site APP cleaving enzyme (BACE1) and decreased level of Aβ degrading enzymes under ER stress. Increased Aβ and pTau under ER stress reduced the N-methyl-D-aspartate receptor (NMDAR) in Western blot analysis and altered electrophysiological properties in the mutant neurons. Our study provides evidence that the interaction between genetic mutation and ER stress would induce AD-like changes.
Collapse
Affiliation(s)
- Tania Devina
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan.,Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Hui Wong
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Life Science and Institute of Genome Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chiao-Wan Hsiao
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| | - Yu-Jui Li
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Cheng-Chang Lien
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan.,Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan.,Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Irene Han-Juo Cheng
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan.,Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| |
Collapse
|
9
|
Pavlova G, Kolesnikova V, Samoylenkova N, Drozd S, Revishchin A, Shamadykova D, Usachev DY, Kopylov A. A Combined Effect of G-Quadruplex and Neuro-Inducers as an Alternative Approach to Human Glioblastoma Therapy. Front Oncol 2022; 12:880740. [PMID: 35586496 PMCID: PMC9109612 DOI: 10.3389/fonc.2022.880740] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 03/28/2022] [Indexed: 11/22/2022] Open
Abstract
Cancer cell reprogramming based on treatment with G-quadruplex, having antiproliferative power, along with small molecules able to develop iPSCs into neurons, could create a novel approach to diminish the chance of glioblastoma recurrence and circumvent tumor resistance to conventional therapy. In this research, we have tested several combinations of factors to affect both total cell cultures, derived from tumor tissue of patients after surgical resection and two subfractions of this cell culture after dividing them into CD133-enriched and CD133-depleted populations (assuming CD133 to be a marker of glioblastoma stem-like cells). CD133+ and CD133− cells exhibit different responses to the same combinations of factors; CD133+ cells have stem-like properties and are more resistant. Therefore, the ability to affect CD133+ cells provides a possibility to circumvent resistance to conventional therapy and to build a promising strategy for translation to improve the treatment of patients with glioblastoma.
Collapse
Affiliation(s)
- Galina Pavlova
- Laboratory of Neurogenetics and Genetics Development, Institute of Higher Nervous Activity and Neurophysiology of Russian Academy of Sciences (RAS), Moscow, Russia.,Federal State Autonomous Institution «N. N. Burdenko National Medical Research Center of Neurosurgery» of the Ministry of Health of the Russian Federation, Moscow, Russia.,Department of Medical Genetics, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Varvara Kolesnikova
- Laboratory of Neurogenetics and Genetics Development, Institute of Higher Nervous Activity and Neurophysiology of Russian Academy of Sciences (RAS), Moscow, Russia
| | - Nadezhda Samoylenkova
- Federal State Autonomous Institution «N. N. Burdenko National Medical Research Center of Neurosurgery» of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Sergey Drozd
- Federal State Autonomous Institution «N. N. Burdenko National Medical Research Center of Neurosurgery» of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Alexander Revishchin
- Laboratory of Neurogenetics and Genetics Development, Institute of Higher Nervous Activity and Neurophysiology of Russian Academy of Sciences (RAS), Moscow, Russia
| | - Dzhirgala Shamadykova
- Laboratory of Neurogenetics and Genetics Development, Institute of Higher Nervous Activity and Neurophysiology of Russian Academy of Sciences (RAS), Moscow, Russia
| | - Dmitry Y Usachev
- Federal State Autonomous Institution «N. N. Burdenko National Medical Research Center of Neurosurgery» of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Alexey Kopylov
- Chemistry Department, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
10
|
Benchoua A, Lasbareilles M, Tournois J. Contribution of Human Pluripotent Stem Cell-Based Models to Drug Discovery for Neurological Disorders. Cells 2021; 10:cells10123290. [PMID: 34943799 PMCID: PMC8699352 DOI: 10.3390/cells10123290] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/19/2021] [Accepted: 11/23/2021] [Indexed: 02/07/2023] Open
Abstract
One of the major obstacles to the identification of therapeutic interventions for central nervous system disorders has been the difficulty in studying the step-by-step progression of diseases in neuronal networks that are amenable to drug screening. Recent advances in the field of human pluripotent stem cell (PSC) biology offers the capability to create patient-specific human neurons with defined clinical profiles using reprogramming technology, which provides unprecedented opportunities for both the investigation of pathogenic mechanisms of brain disorders and the discovery of novel therapeutic strategies via drug screening. Many examples not only of the creation of human pluripotent stem cells as models of monogenic neurological disorders, but also of more challenging cases of complex multifactorial disorders now exist. Here, we review the state-of-the art brain cell types obtainable from PSCs and amenable to compound-screening formats. We then provide examples illustrating how these models contribute to the definition of new molecular or functional targets for drug discovery and to the design of novel pharmacological approaches for rare genetic disorders, as well as frequent neurodegenerative diseases and psychiatric disorders.
Collapse
Affiliation(s)
- Alexandra Benchoua
- Neuroplasticity and Therapeutics, CECS, I-STEM, AFM, 91100 Corbeil-Essonnes, France;
- High Throughput Screening Platform, CECS, I-STEM, AFM, 91100 Corbeil-Essonnes, France;
- Correspondence:
| | - Marie Lasbareilles
- Neuroplasticity and Therapeutics, CECS, I-STEM, AFM, 91100 Corbeil-Essonnes, France;
- UEVE UMR 861, I-STEM, AFM, 91100 Corbeil-Essonnes, France
| | - Johana Tournois
- High Throughput Screening Platform, CECS, I-STEM, AFM, 91100 Corbeil-Essonnes, France;
| |
Collapse
|
11
|
MacMahon Copas AN, McComish SF, Fletcher JM, Caldwell MA. The Pathogenesis of Parkinson's Disease: A Complex Interplay Between Astrocytes, Microglia, and T Lymphocytes? Front Neurol 2021; 12:666737. [PMID: 34122308 PMCID: PMC8189423 DOI: 10.3389/fneur.2021.666737] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/20/2021] [Indexed: 12/12/2022] Open
Abstract
Parkinson's disease (PD), the second most common neurodegenerative disease, is characterised by the motor symptoms of bradykinesia, rigidity and resting tremor and non-motor symptoms of sleep disturbances, constipation, and depression. Pathological hallmarks include neuroinflammation, degeneration of dopaminergic neurons in the substantia nigra pars compacta, and accumulation of misfolded α-synuclein proteins as intra-cytoplasmic Lewy bodies and neurites. Microglia and astrocytes are essential to maintaining homeostasis within the central nervous system (CNS), including providing protection through the process of gliosis. However, dysregulation of glial cells results in disruption of homeostasis leading to a chronic pro-inflammatory, deleterious environment, implicated in numerous CNS diseases. Recent evidence has demonstrated a role for peripheral immune cells, in particular T lymphocytes in the pathogenesis of PD. These cells infiltrate the CNS, and accumulate in the substantia nigra, where they secrete pro-inflammatory cytokines, stimulate surrounding immune cells, and induce dopaminergic neuronal cell death. Indeed, a greater understanding of the integrated network of communication that exists between glial cells and peripheral immune cells may increase our understanding of disease pathogenesis and hence provide novel therapeutic approaches.
Collapse
Affiliation(s)
- Adina N MacMahon Copas
- Department of Physiology, School of Medicine, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin, Ireland.,Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Sarah F McComish
- Department of Physiology, School of Medicine, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin, Ireland.,Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Jean M Fletcher
- Department of Physiology, School of Medicine, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin, Ireland.,School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Maeve A Caldwell
- Department of Physiology, School of Medicine, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin, Ireland.,Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
12
|
Nisin and non-essential amino acids: new perspective in differentiation of neural progenitors from human-induced pluripotent stem cells in vitro. Hum Cell 2021; 34:1142-1152. [PMID: 33899160 DOI: 10.1007/s13577-021-00537-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 04/16/2021] [Indexed: 12/23/2022]
Abstract
Over the past decades, stem cell therapy has been investigated as a promising approach towards various diseases, including neurodegenerative disorders. Stem cells show the capability to differentiate into neuronal progenitor cells in vitro. In the present study, the differentiation potential of human-induced pluripotent stem cells (hiPSCs) into neural lineages was examined under the efficient induction media containing forskolin and 3-isobutyl-1-methyl-xanthine (IBMX) in the presence of nisin (Ni), non-essential amino acids (NEAA) and combination of those (NEAA-Ni) in vitro. The optimum concentrations of these factors were obtained by MTT assay and acridine orange (AO) staining. The effect of Ni and NEAA on the expression rate of neural-specific markers including NSE, MAP2, and ß-tubulin III was studied via immunocytochemistry (ICC) and real-time RT-PCR analyses. Our results indicated that the induction medium containing Ni or NEAA increased the gene and protein expression of NSE, MAP2, and β-tubulin III on the 14th differentiation day. On the other hand, NEAA-Ni showed a less-differentiated hiPSCs compared to Ni and NEAA alone. In conclusion, the obtained results illustrated that Ni and NEAA could be applied as effective factors for neural differentiation of hiPSCs in the future.
Collapse
|
13
|
Chen Q, Yuan C, Jiang S, Heng BC, Zou T, Shen Z, Wang P, Zhang C. Small molecules efficiently reprogram apical papilla stem cells into neuron-like cells. Exp Ther Med 2021; 21:546. [PMID: 33850518 DOI: 10.3892/etm.2021.9978] [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: 11/05/2019] [Accepted: 09/08/2020] [Indexed: 12/13/2022] Open
Abstract
Stem cell-based therapy may provide a novel approach for neural tissue regeneration. A small molecule cocktail-based culture protocol was previously shown to enhance neurogenic differentiation of stem cells from dental tissues. The present study aimed to investigate the early phase of small molecule-induced neurogenic differentiation of stem cells from the apical papilla (SCAP). SCAP were cultured in neural-induction medium or neural-induction medium with small molecules (NIMS-SCAP) and examined for their cell morphologies. Expression levels of neural progenitor cell-related markers, including Nestin, paired-box gene 6 (Pax6) and Sry-related HMG box 2 (Sox2), were examined using western blotting and immunocytofluorescence. Expression of differentiated neuron-related markers, including neurofilament protein (NFM), neuron-specific nuclear protein (NeuN) and microtubule-associated protein (MAP)-2, were also examined using western blotting, while NFM and MAP2 gene expression and cell proliferation were assessed using reverse transcription-quantitative (RT-q)PCR and Cell Counting Kit (CCK)-8 assays, respectively. SCAP morphology was affected by small molecules after as little as 30 min. Specifically, Nestin, Pax6 and Sox2 expression detected using western blotting was increased by day 3 but then decreased over the course of 7 days with neural induction, while immunocytofluorescence revealed expression of all three markers in NIMS-SCAP. The protein levels of NFM, NeuN and MAP2 on day 7 were significantly upregulated in NIMS-SCAP, as detected using western blotting, while NFM and MAP2 gene expression levels detected using RT-qPCR were significantly increased on days 5 and 7. Proliferation of NIMS-SCAP ceased after 5 days. Electrophysiological analysis showed that only SCAP cultured in NIMS had the functional activity of neuronal cells. Thus, small molecules reprogrammed SCAP into neural progenitor cells within the first 3 days, followed by further differentiation into neuron-like cells.
Collapse
Affiliation(s)
- Qixin Chen
- Restorative Dental Sciences, Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong 999077, SAR, P.R. China.,Department of Implant Dentistry, The Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Changyong Yuan
- Department of Implant Dentistry, The Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Shan Jiang
- Restorative Dental Sciences, Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong 999077, SAR, P.R. China
| | - Boon Chin Heng
- School of Stomatology, Peking University, Beijing 100081, P.R. China
| | - Ting Zou
- Restorative Dental Sciences, Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong 999077, SAR, P.R. China
| | - Zhongshan Shen
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Penglai Wang
- Department of Implant Dentistry, The Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Chengfei Zhang
- Restorative Dental Sciences, Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong 999077, SAR, P.R. China
| |
Collapse
|
14
|
Acioglu C, Li L, Elkabes S. Contribution of astrocytes to neuropathology of neurodegenerative diseases. Brain Res 2021; 1758:147291. [PMID: 33516810 DOI: 10.1016/j.brainres.2021.147291] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 12/10/2020] [Accepted: 01/05/2021] [Indexed: 02/08/2023]
Abstract
Classically, the loss of vulnerable neuronal populations in neurodegenerative diseases was considered to be the consequence of cell autonomous degeneration of neurons. However, progress in the understanding of glial function, the availability of improved animal models recapitulating the features of the human diseases, and the development of new approaches to derive glia and neurons from induced pluripotent stem cells obtained from patients, provided novel information that altered this view. Current evidence strongly supports the notion that non-cell autonomous mechanisms contribute to the demise of neurons in neurodegenerative disorders, and glia causally participate in the pathogenesis and progression of these diseases. In addition to microglia, astrocytes have emerged as key players in neurodegenerative diseases and will be the focus of the present review. Under the influence of pathological stimuli present in the microenvironment of the diseased CNS, astrocytes undergo morphological, transcriptional, and functional changes and become reactive. Reactive astrocytes are heterogeneous and exhibit neurotoxic (A1) or neuroprotective (A2) phenotypes. In recent years, single-cell or single-nucleus transcriptome analyses unraveled new, disease-specific phenotypes beyond A1/A2. These investigations highlighted the complexity of the astrocytic responses to CNS pathology. The present review will discuss the contribution of astrocytes to neurodegenerative diseases with particular emphasis on Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis and frontotemporal dementia. Some of the commonalties and differences in astrocyte-mediated mechanisms that possibly drive the pathogenesis or progression of the diseases will be summarized. The emerging view is that astrocytes are potential new targets for therapeutic interventions. A comprehensive understanding of astrocyte heterogeneity and disease-specific phenotypic complexity could facilitate the design of novel strategies to treat neurodegenerative disorders.
Collapse
Affiliation(s)
- Cigdem Acioglu
- The Reynolds Family Spine Laboratory, Department of Neurological Surgery, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, United States.
| | - Lun Li
- The Reynolds Family Spine Laboratory, Department of Neurological Surgery, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, United States.
| | - Stella Elkabes
- The Reynolds Family Spine Laboratory, Department of Neurological Surgery, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, United States.
| |
Collapse
|
15
|
Cenini G, Hebisch M, Iefremova V, Flitsch LJ, Breitkreuz Y, Tanzi RE, Kim DY, Peitz M, Brüstle O. Dissecting Alzheimer's disease pathogenesis in human 2D and 3D models. Mol Cell Neurosci 2021; 110:103568. [DOI: 10.1016/j.mcn.2020.103568] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/30/2020] [Accepted: 10/12/2020] [Indexed: 02/06/2023] Open
|
16
|
Hauser S, Schuster S, Heuten E, Höflinger P, Admard J, Schelling Y, Velic A, Macek B, Ossowski S, Schöls L. Comparative Transcriptional Profiling of Motor Neuron Disorder-Associated Genes in Various Human Cell Culture Models. Front Cell Dev Biol 2020; 8:544043. [PMID: 33072739 PMCID: PMC7543453 DOI: 10.3389/fcell.2020.544043] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 09/04/2020] [Indexed: 01/06/2023] Open
Abstract
Disease modeling requires appropriate cellular models that best mimic the underlying pathophysiology. Human origin and an adequate expression of the disease protein are pre-requisites that support information from a model to be meaningful. In this study we investigated expression profiles of (i) PBMCs and (ii) fibroblasts as patient derived cells as well as (iii) lymphoblasts and (iv) induced pluripotent stem cells (iPSC) as immortalized sources, and (v) iPSC-derived cortical neurons to assess their aptitude to model motor neuron diseases (MNDs) including hereditary spastic paraplegia (HSP), amyotrophic lateral sclerosis (ALS) and spinal muscular atrophy (SMA). We generated all five different cell types from two healthy donors and performed RNA sequencing to display expression patterns in MND-related genes. For the ten most common HSP genotypes we validated gene expression by qPCR. To verify the results on protein level, proteome analysis of fibroblasts, iPSCs and cortical neurons was performed. Depending on the specific MND gene we found largely different expression patterns. Out of 168 MND-related genes, 50 had their highest expression in iPSC-derived cortical neurons, 41 were most strongly expressed in fibroblasts, 26 in lymphoblasts, 22 in iPSCs, and 14 in PBMCs. Pathophysiologically related MNDs like HSPs associated with axonal transport deficits shared highest expression in cortical neurons. 15 MND-related genes were not detectable in any of the analyzed cell types. This may reflect the critical dependency of motor neurons on support of other cell types like oligodendrocytes which express myelin proteins like L1CAM (SPG1), PLP1 (SPG2) and MAG (SPG75) which are lacking in neurons but cause MNDs if mutated. This study provides comprehensive information on expression of genes associated with a large spectrum of MNDs. Expression profiles can be used to inform on appropriate cell models for genotype specific motor neuron research.
Collapse
Affiliation(s)
- Stefan Hauser
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.,Department of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Stefanie Schuster
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.,Department of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,Graduate School of Cellular and Molecular Neuroscience, University of Tübingen, Tübingen, Germany
| | - Elena Heuten
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Philip Höflinger
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.,Department of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,Graduate School of Cellular and Molecular Neuroscience, University of Tübingen, Tübingen, Germany
| | - Jakob Admard
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Yvonne Schelling
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.,Department of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Ana Velic
- Proteome Center Tübingen, University of Tübingen, Tübingen, Germany
| | - Boris Macek
- Proteome Center Tübingen, University of Tübingen, Tübingen, Germany
| | - Stephan Ossowski
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany.,Center of Rare Diseases, University of Tübingen, Tübingen, Germany
| | - Ludger Schöls
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.,Department of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,Center of Rare Diseases, University of Tübingen, Tübingen, Germany
| |
Collapse
|
17
|
Mechanistic Insights of Astrocyte-Mediated Hyperactive Autophagy and Loss of Motor Neuron Function in SOD1 L39R Linked Amyotrophic Lateral Sclerosis. Mol Neurobiol 2020; 57:4117-4133. [PMID: 32676988 DOI: 10.1007/s12035-020-02006-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 06/26/2020] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder with no cure. The reports showed the role of nearby astrocytes around the motor neurons as one among the causes of the disease. However, the exact mechanistic insights are not explored so far. Thus, in the present investigations, we employed the induced pluripotent stem cells (iPSCs) of Cu/Zn-SOD1L39R linked ALS patient to convert them into the motor neurons (MNs) and astrocytes. We report that the higher expression of stress granule (SG) marker protein G3BP1, and its co-localization with the mutated Cu/Zn-SOD1L39R protein in patient's MNs and astrocytes are linked with AIF1-mediated upregulation of caspase 3/7 and hyper activated autophagy. We also observe the astrocyte-mediated non-cell autonomous neurotoxicity on MNs in ALS. The secretome of the patient's iPSC-derived astrocytes exerts significant oxidative stress in MNs. The findings suggest the hyperactive status of autophagy in MNs, as witnessed by the co-distribution of LAMP1, P62 and LC3 I/II with the autolysosomes. Conversely, the secretome of normal astrocytes has shown neuroprotection in patient's iPSC-derived MNs. The whole-cell patch-clamp assay confirms our findings at a physiological functional level in MNs. Perhaps for the first time, we are reporting that the MN degeneration in ALS triggered by the hyper-activation of autophagy and induced apoptosis in both cell-autonomous and non-cell autonomous conditions.
Collapse
|
18
|
Fischer I, Dulin JN, Lane MA. Transplanting neural progenitor cells to restore connectivity after spinal cord injury. Nat Rev Neurosci 2020; 21:366-383. [PMID: 32518349 PMCID: PMC8384139 DOI: 10.1038/s41583-020-0314-2] [Citation(s) in RCA: 168] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2020] [Indexed: 12/12/2022]
Abstract
Spinal cord injury remains a scientific and therapeutic challenge with great cost to individuals and society. The goal of research in this field is to find a means of restoring lost function. Recently we have seen considerable progress in understanding the injury process and the capacity of CNS neurons to regenerate, as well as innovations in stem cell biology. This presents an opportunity to develop effective transplantation strategies to provide new neural cells to promote the formation of new neuronal networks and functional connectivity. Past and ongoing clinical studies have demonstrated the safety of cell therapy, and preclinical research has used models of spinal cord injury to better elucidate the underlying mechanisms through which donor cells interact with the host and thus increase long-term efficacy. While a variety of cell therapies have been explored, we focus here on the use of neural progenitor cells obtained or derived from different sources to promote connectivity in sensory, motor and autonomic systems.
Collapse
Affiliation(s)
- Itzhak Fischer
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA.
| | - Jennifer N Dulin
- Department of Biology, Texas A&M University, College Station, TX, USA
| | - Michael A Lane
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
19
|
Klimmt J, Dannert A, Paquet D. Neurodegeneration in a dish: advancing human stem-cell-based models of Alzheimer's disease. Curr Opin Neurobiol 2020; 61:96-104. [PMID: 32112992 DOI: 10.1016/j.conb.2020.01.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/03/2020] [Accepted: 01/21/2020] [Indexed: 12/11/2022]
Abstract
Induced pluripotent stem-cell-based models enable investigation of pathomechanisms in disease-relevant human brain cell types and therefore offer great potential for mechanistic and translational studies on neurodegenerative disorders, such as Alzheimer's disease (AD). While current AD models allow analysis of early disease phenotypes including Aβ accumulation and Tau hyperphosphorylation, they still fail to fully recapitulate later hallmarks such as protein aggregation and neurodegeneration. This impedes the identification of pathomechanisms and novel therapeutic targets. We discuss strategies to overcome these drawbacks and optimize physiological properties and translational potential of iPSC-based models by improving culture formats, increasing cellular diversity, applying genome editing, and implementing maturation and ageing paradigms.
Collapse
Affiliation(s)
- Julien Klimmt
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Feodor-Lynen-Str. 17, 81377 Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynen-Str. 17, 81377 Munich, Germany
| | - Angelika Dannert
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Feodor-Lynen-Str. 17, 81377 Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynen-Str. 17, 81377 Munich, Germany
| | - Dominik Paquet
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Feodor-Lynen-Str. 17, 81377 Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynen-Str. 17, 81377 Munich, Germany.
| |
Collapse
|
20
|
Neural In Vitro Models for Studying Substances Acting on the Central Nervous System. Handb Exp Pharmacol 2020; 265:111-141. [PMID: 32594299 DOI: 10.1007/164_2020_367] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Animal models have been greatly contributing to our understanding of physiology, mechanisms of diseases, and toxicity. Yet, their limitations due to, e.g., interspecies variation are reflected in the high number of drug attrition rates, especially in central nervous system (CNS) diseases. Therefore, human-based neural in vitro models for studying safety and efficacy of substances acting on the CNS are needed. Human iPSC-derived cells offer such a platform with the unique advantage of reproducing the "human context" in vitro by preserving the genetic and molecular phenotype of their donors. Guiding the differentiation of hiPSC into cells of the nervous system and combining them in a 2D or 3D format allows to obtain complex models suitable for investigating neurotoxicity or brain-related diseases with patient-derived cells. This chapter will give an overview over stem cell-based human 2D neuronal and mixed neuronal/astrocyte models, in vitro cultures of microglia, as well as CNS disease models and considers new developments in the field, more specifically the use of brain organoids and 3D bioprinted in vitro models for safety and efficacy evaluation.
Collapse
|
21
|
Marcatili M, Sala C, Dakanalis A, Colmegna F, D'Agostino A, Gambini O, Dell'Osso B, Benatti B, Conti L, Clerici M. Human induced pluripotent stem cells technology in treatment resistant depression: novel strategies and opportunities to unravel ketamine's fast-acting antidepressant mechanisms. Ther Adv Psychopharmacol 2020; 10:2045125320968331. [PMID: 33224469 PMCID: PMC7649879 DOI: 10.1177/2045125320968331] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 05/11/2020] [Indexed: 12/12/2022] Open
Abstract
Approximately 30% of Major Depressive Disorder (MDD) patients develop treatment-resistant depression (TRD). Among the different causes that make TRD so challenging in both clinical and research contexts, major roles are played by the inadequate understanding of MDD pathophysiology and the limitations of current pharmacological treatments. Nevertheless, the field of psychiatry is facing exciting times. Combined with recent advances in genome editing techniques, human induced pluripotent stem cell (hiPSC) technology is offering novel and unique opportunities in both disease modelling and drug discovery. This technology has allowed innovative disease-relevant patient-specific in vitro models to be set up for many psychiatric disorders. Such models hold great potential in enhancing our understanding of MDD pathophysiology and overcoming many of the well-known practical limitations inherent to animal and post-mortem models. Moreover, the field is approaching the advent of (es)ketamine, a glutamate N-methyl-d-aspartate (NMDA) receptor antagonist, claimed as one of the first and exemplary agents with rapid (in hours) antidepressant effects, even in TRD patients. Although ketamine seems poised to transform the treatment of depression, its exact mechanisms of action are still unclear but greatly demanded, as the resulting knowledge may provide a model to understand the mechanisms behind rapid-acting antidepressants, which may lead to the discovery of novel compounds for the treatment of depression. After reviewing insights into ketamine's mechanisms of action (derived from preclinical animal studies) and depicting the current state of the art of hiPSC technology below, we will consider the implementation of an hiPSC technology-based TRD model for the study of ketamine's fast acting antidepressant mechanisms of action.
Collapse
Affiliation(s)
- Matteo Marcatili
- Psychiatric Department, San Gerardo Hospital, ASST Monza, Monza, Italy
| | - Carlo Sala
- National Research Council Neuroscience Institute, Milan, Italy
| | - Antonios Dakanalis
- Department of Medicine and Surgery, University of Milano Bicocca, Monza, Italy
| | - Fabrizia Colmegna
- Psychiatric Department, San Gerardo Hospital, ASST Monza, Monza, Italy
| | - Armando D'Agostino
- Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Orsola Gambini
- Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Bernardo Dell'Osso
- Psychiatry Unit, Department of Biomedical and Clinical Sciences "Luigi Sacco", University of Milan, Milan, Italy
| | - Beatrice Benatti
- Psychiatry Unit, Department of Biomedical and Clinical Sciences "Luigi Sacco", University of Milan, Milan, Italy
| | - Luciano Conti
- Laboratory of Stem Cell Biology, Department of Cellular, Computational and Integrative Biology (CIBIO), Università degli Studi di Trento, Trento, Italy
| | - Massimo Clerici
- Psychiatric Department, San Gerardo Hospital, ASST Monza, Monza, Italy
| |
Collapse
|
22
|
Hay DC, O'Farrelly C. Designer human tissue: coming to a lab near you. Philos Trans R Soc Lond B Biol Sci 2019; 373:rstb.2017.0212. [PMID: 29786548 PMCID: PMC5974436 DOI: 10.1098/rstb.2017.0212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2018] [Indexed: 11/12/2022] Open
Abstract
Human pluripotent stem cells (PSCs) offer a scalable alternative to primary and transformed human tissue. PSCs include human embryonic stem cells, derived from the inner cell mass of blastocysts unsuitable for human implantation; and induced PSCs, generated by the reprogramming of somatic cells. Both cell types display the ability to self-renew and retain pluripotency, promising an unlimited supply of human somatic cells for biomedical application. A distinct advantage of using PSCs is the ability to select for genetic background, promising personalized modelling of human biology ‘in a dish’ or immune-matched cell-based therapies for the clinic. This special issue will guide the reader through stem cell self-renewal, pluripotency and differentiation. The first articles focus on improving cell fidelity, understanding the innate immune system and the importance of materials chemistry, biofabrication and bioengineering. These are followed by articles that focus on industrial application, commercialization and label-free assessment of tissue formation. The special issue concludes with an article discussing human liver cell-based therapies past, present and future. This article is part of the theme issue ‘Designer human tissue: coming to a lab near you’.
Collapse
Affiliation(s)
- David C Hay
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, EH16 4UU Edinburgh, UK
| | - Cliona O'Farrelly
- Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse St, Dublin 2, Dublin, Republic of Ireland
| |
Collapse
|
23
|
Legradi JB, Di Paolo C, Kraak MHS, van der Geest HG, Schymanski EL, Williams AJ, Dingemans MML, Massei R, Brack W, Cousin X, Begout ML, van der Oost R, Carion A, Suarez-Ulloa V, Silvestre F, Escher BI, Engwall M, Nilén G, Keiter SH, Pollet D, Waldmann P, Kienle C, Werner I, Haigis AC, Knapen D, Vergauwen L, Spehr M, Schulz W, Busch W, Leuthold D, Scholz S, vom Berg CM, Basu N, Murphy CA, Lampert A, Kuckelkorn J, Grummt T, Hollert H. An ecotoxicological view on neurotoxicity assessment. ENVIRONMENTAL SCIENCES EUROPE 2018; 30:46. [PMID: 30595996 PMCID: PMC6292971 DOI: 10.1186/s12302-018-0173-x] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 10/31/2018] [Indexed: 05/04/2023]
Abstract
The numbers of potential neurotoxicants in the environment are raising and pose a great risk for humans and the environment. Currently neurotoxicity assessment is mostly performed to predict and prevent harm to human populations. Despite all the efforts invested in the last years in developing novel in vitro or in silico test systems, in vivo tests with rodents are still the only accepted test for neurotoxicity risk assessment in Europe. Despite an increasing number of reports of species showing altered behaviour, neurotoxicity assessment for species in the environment is not required and therefore mostly not performed. Considering the increasing numbers of environmental contaminants with potential neurotoxic potential, eco-neurotoxicity should be also considered in risk assessment. In order to do so novel test systems are needed that can cope with species differences within ecosystems. In the field, online-biomonitoring systems using behavioural information could be used to detect neurotoxic effects and effect-directed analyses could be applied to identify the neurotoxicants causing the effect. Additionally, toxic pressure calculations in combination with mixture modelling could use environmental chemical monitoring data to predict adverse effects and prioritize pollutants for laboratory testing. Cheminformatics based on computational toxicological data from in vitro and in vivo studies could help to identify potential neurotoxicants. An array of in vitro assays covering different modes of action could be applied to screen compounds for neurotoxicity. The selection of in vitro assays could be guided by AOPs relevant for eco-neurotoxicity. In order to be able to perform risk assessment for eco-neurotoxicity, methods need to focus on the most sensitive species in an ecosystem. A test battery using species from different trophic levels might be the best approach. To implement eco-neurotoxicity assessment into European risk assessment, cheminformatics and in vitro screening tests could be used as first approach to identify eco-neurotoxic pollutants. In a second step, a small species test battery could be applied to assess the risks of ecosystems.
Collapse
Affiliation(s)
- J. B. Legradi
- Institute for Environmental Research, Department of Ecosystem Analysis, ABBt–Aachen Biology and Biotechnology, RWTH Aachen University, Worringerweg 1, 52074 Aachen, Germany
- Environment and Health, VU University, 1081 HV Amsterdam, The Netherlands
| | - C. Di Paolo
- Institute for Environmental Research, Department of Ecosystem Analysis, ABBt–Aachen Biology and Biotechnology, RWTH Aachen University, Worringerweg 1, 52074 Aachen, Germany
| | - M. H. S. Kraak
- FAME-Freshwater and Marine Ecology, Institute for Biodiversity and Ecosystem Dynamics, University of Amsterdam, P.O. Box 94248, 1090 GE Amsterdam, The Netherlands
| | - H. G. van der Geest
- FAME-Freshwater and Marine Ecology, Institute for Biodiversity and Ecosystem Dynamics, University of Amsterdam, P.O. Box 94248, 1090 GE Amsterdam, The Netherlands
| | - E. L. Schymanski
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 6 Avenue du Swing, 4367 Belvaux, Luxembourg
| | - A. J. Williams
- National Center for Computational Toxicology, Office of Research and Development, U.S. Environmental Protection Agency, 109 T.W. Alexander Dr., Research Triangle Park, NC 27711 USA
| | - M. M. L. Dingemans
- KWR Watercycle Research Institute, Groningenhaven 7, 3433 PE Nieuwegein, The Netherlands
| | - R. Massei
- Department Effect-Directed Analysis, Helmholtz Centre for Environmental Research-UFZ, Permoserstr. 15, Leipzig, Germany
| | - W. Brack
- Department Effect-Directed Analysis, Helmholtz Centre for Environmental Research-UFZ, Permoserstr. 15, Leipzig, Germany
| | - X. Cousin
- Ifremer, UMR MARBEC, Laboratoire Adaptation et Adaptabilités des Animaux et des Systèmes, Route de Maguelone, 34250 Palavas-les-Flots, France
- INRA, UMR GABI, INRA, AgroParisTech, Domaine de Vilvert, Batiment 231, 78350 Jouy-en-Josas, France
| | - M.-L. Begout
- Ifremer, Laboratoire Ressources Halieutiques, Place Gaby Coll, 17137 L’Houmeau, France
| | - R. van der Oost
- Department of Technology, Research and Engineering, Waternet Institute for the Urban Water Cycle, Amsterdam, The Netherlands
| | - A. Carion
- Laboratory of Evolutionary and Adaptive Physiology, Institute of Life, Earth and Environment, University of Namur, 5000 Namur, Belgium
| | - V. Suarez-Ulloa
- Laboratory of Evolutionary and Adaptive Physiology, Institute of Life, Earth and Environment, University of Namur, 5000 Namur, Belgium
| | - F. Silvestre
- Laboratory of Evolutionary and Adaptive Physiology, Institute of Life, Earth and Environment, University of Namur, 5000 Namur, Belgium
| | - B. I. Escher
- Department of Cell Toxicology, Helmholtz Centre for Environmental Research-UFZ, Permoserstr. 15, 04318 Leipzig, Germany
- Eberhard Karls University Tübingen, Environmental Toxicology, Center for Applied Geosciences, 72074 Tübingen, Germany
| | - M. Engwall
- MTM Research Centre, School of Science and Technology, Örebro University, Fakultetsgatan 1, 70182 Örebro, Sweden
| | - G. Nilén
- MTM Research Centre, School of Science and Technology, Örebro University, Fakultetsgatan 1, 70182 Örebro, Sweden
| | - S. H. Keiter
- MTM Research Centre, School of Science and Technology, Örebro University, Fakultetsgatan 1, 70182 Örebro, Sweden
| | - D. Pollet
- Faculty of Chemical Engineering and Biotechnology, University of Applied Sciences Darmstadt, Stephanstrasse 7, 64295 Darmstadt, Germany
| | - P. Waldmann
- Faculty of Chemical Engineering and Biotechnology, University of Applied Sciences Darmstadt, Stephanstrasse 7, 64295 Darmstadt, Germany
| | - C. Kienle
- Swiss Centre for Applied Ecotoxicology Eawag-EPFL, Überlandstrasse 133, 8600 Dübendorf, Switzerland
| | - I. Werner
- Swiss Centre for Applied Ecotoxicology Eawag-EPFL, Überlandstrasse 133, 8600 Dübendorf, Switzerland
| | - A.-C. Haigis
- Institute for Environmental Research, Department of Ecosystem Analysis, ABBt–Aachen Biology and Biotechnology, RWTH Aachen University, Worringerweg 1, 52074 Aachen, Germany
| | - D. Knapen
- Zebrafishlab, Veterinary Physiology and Biochemistry, University of Antwerp, Wilrijk, Belgium
| | - L. Vergauwen
- Zebrafishlab, Veterinary Physiology and Biochemistry, University of Antwerp, Wilrijk, Belgium
| | - M. Spehr
- Institute for Biology II, Department of Chemosensation, RWTH Aachen University, Aachen, Germany
| | - W. Schulz
- Zweckverband Landeswasserversorgung, Langenau, Germany
| | - W. Busch
- Department of Bioanalytical Ecotoxicology, UFZ–Helmholtz Centre for Environmental Research, Leipzig, Germany
| | - D. Leuthold
- Department of Bioanalytical Ecotoxicology, UFZ–Helmholtz Centre for Environmental Research, Leipzig, Germany
| | - S. Scholz
- Department of Bioanalytical Ecotoxicology, UFZ–Helmholtz Centre for Environmental Research, Leipzig, Germany
| | - C. M. vom Berg
- Department of Environmental Toxicology, Swiss Federal Institute of Aquatic Science and Technology, Eawag, Dübendorf, 8600 Switzerland
| | - N. Basu
- Faculty of Agricultural and Environmental Sciences, McGill University, Montreal, Canada
| | - C. A. Murphy
- Department of Fisheries and Wildlife, Michigan State University, East Lansing, USA
| | - A. Lampert
- Institute of Physiology (Neurophysiology), Aachen, Germany
| | - J. Kuckelkorn
- Section Toxicology of Drinking Water and Swimming Pool Water, Federal Environment Agency (UBA), Heinrich-Heine-Str. 12, 08645 Bad Elster, Germany
| | - T. Grummt
- Section Toxicology of Drinking Water and Swimming Pool Water, Federal Environment Agency (UBA), Heinrich-Heine-Str. 12, 08645 Bad Elster, Germany
| | - H. Hollert
- Institute for Environmental Research, Department of Ecosystem Analysis, ABBt–Aachen Biology and Biotechnology, RWTH Aachen University, Worringerweg 1, 52074 Aachen, Germany
| |
Collapse
|