1
|
Xie Q, Li K, Chen Y, Li Y, Jiang W, Cao W, Yu H, Fan D, Deng B. Gene therapy breakthroughs in ALS: a beacon of hope for 20% of ALS patients. Transl Neurodegener 2025; 14:19. [PMID: 40234983 PMCID: PMC12001736 DOI: 10.1186/s40035-025-00477-6] [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: 08/01/2024] [Accepted: 03/05/2025] [Indexed: 04/17/2025] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal motor neuron disease that remains incurable. Although the etiologies of ALS are diverse and the precise pathogenic mechanisms are not fully understood, approximately 20% of ALS cases are caused by genetic factors. Therefore, advancing targeted gene therapies holds significant promise, at least for the 20% of ALS patients with genetic etiologies. In this review, we summarize the main strategies and techniques of current ALS gene therapies based on ALS risk genes, and review recent findings from animal studies and clinical trials. Additionally, we highlight ALS-related genes with well-understood pathogenic mechanisms and the potential of numerous emerging gene-targeted therapeutic approaches for ALS.
Collapse
Affiliation(s)
- Qingjian Xie
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 32500, China
- First School of Clinical Medicine, Wenzhou Medical University, Wenzhou, China
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Kezheng Li
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 32500, China
- First School of Clinical Medicine, Wenzhou Medical University, Wenzhou, China
| | - Yinuo Chen
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 32500, China
- First School of Clinical Medicine, Wenzhou Medical University, Wenzhou, China
| | - Yaojia Li
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 32500, China
| | - Wenhua Jiang
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 32500, China
| | - Wen Cao
- Department of Neurology, Peking University Third Hospital, Beijing, China
| | - Huan Yu
- Department of Pediatrics, Second Affiliated Hospital and Yuying Children'S Hospital of Wenzhou Medical University, Wenzhou, China
| | - Dongsheng Fan
- Department of Neurology, Peking University Third Hospital, Beijing, China
| | - Binbin Deng
- Department of Rehabilitation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 32500, China.
| |
Collapse
|
2
|
Singh K, Sethi P, Datta S, Chaudhary JS, Kumar S, Jain D, Gupta JK, Kumar S, Guru A, Panda SP. Advances in gene therapy approaches targeting neuro-inflammation in neurodegenerative diseases. Ageing Res Rev 2024; 98:102321. [PMID: 38723752 DOI: 10.1016/j.arr.2024.102321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 05/03/2024] [Accepted: 05/03/2024] [Indexed: 05/20/2024]
Abstract
Over the last three decades, neurodegenerative diseases (NDs) have increased in frequency. About 15% of the world's population suffers from NDs in some capacity, which causes cognitive and physical impairment. Neurodegenerative diseases, including Amyotrophic Lateral Sclerosis, Parkinson's disease, Alzheimer's disease, and others represent a significant and growing global health challenge. Neuroinflammation is recognized to be related to all NDs, even though NDs are caused by a complex mix of genetic, environmental, and lifestyle factors. Numerous genes and pathways such as NFκB, p38 MAPK, Akt/mTOR, caspase, nitric oxide, and COX are involved in triggering brain immune cells like astrocytes and microglia to secrete inflammatory cytokines such as tumor necrosis factor-α, interleukin (IL)-1β, and IL-6. In AD, the binding of Aβ with CD36, TLR4, and TLR6 receptors results in activation of microglia which start to produce proinflammatory cytokines and chemokines. Consequently, the pro-inflammatory cytokines worsen and spread neuroinflammation, causing the deterioration of healthy neurons and the impairment of brain functions. Gene therapy has emerged as a promising therapeutic approach to modulate the inflammatory response in NDs, offering potential neuroprotective effects and disease-modifying benefits. This review article focuses on recent advances in gene therapy strategies targeting neuroinflammation pathways in NDs. We discussed the molecular pathways involved in neuroinflammation, highlighted key genes and proteins implicated in these processes, and reviewed the latest preclinical and clinical studies utilizing gene therapy to modulate neuroinflammatory responses. Additionally, this review addressed the prospects and challenges in translating gene therapy approaches into effective treatments for NDs.
Collapse
Affiliation(s)
- Kuldeep Singh
- Department of Pharmacology, Institue of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Pranshul Sethi
- Department of Pharmacology, College of Pharmacy, Shri Venkateshwara University, Gajraula, Uttar Pradesh, India
| | - Samaresh Datta
- Department of Pharmaceutical Chemistry, Birbhum Pharmacy School, Sadaipur, Dist-Birbhum, West Bengal, India
| | | | - Sunil Kumar
- Faculty of Pharmacy, P. K. University, Village, Thanra, District, Karera, Shivpuri, Madhya Pradesh, India
| | - Divya Jain
- Department of Microbiology, School of Applied and Life Sciences, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Jeetendra Kumar Gupta
- Department of Pharmacology, Institue of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Shivendra Kumar
- Department of Pharmacology, Rajiv Academy for Pharmacy, Mathura, Uttar Pradesh, India
| | - Ajay Guru
- Department of Cariology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Siva Prasad Panda
- Department of Pharmacology, Institue of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India.
| |
Collapse
|
3
|
García-Bañuelos J, Oceguera-Contreras E, Sandoval-Rodríguez A, Bastidas-Ramírez BE, Lucano-Landeros S, Gordillo-Bastidas D, Gómez-Meda BC, Santos A, Cerda-Reyes E, Armendariz-Borunda J. AdhMMP8 Vector Administration in Muscle: An Alternate Strategy to Regress Hepatic Fibrosis. Cells 2023; 12:2127. [PMID: 37681859 PMCID: PMC10486800 DOI: 10.3390/cells12172127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/10/2023] [Accepted: 08/18/2023] [Indexed: 09/09/2023] Open
Abstract
The development of several vaccines against the SARS-CoV2 virus and their application in millions of people have shown efficacy and safety in the transfer of genes to muscle turning this tissue into a protein-producing factory. Established advanced liver fibrosis, is characterized by replacement of hepatic parenchyma by tissue scar, mostly collagen type I, with increased profibrogenic and proinflammatory molecules gene expression. Matrix metalloproteinase 8 (MMP-8) is an interstitial collagen-degrading proenzyme acting preferentially on collagen type I when activated. This study was carried out to elucidate the effect of an intramuscularly delivered adenoviral vector containing proMMP-8 gene cDNA (AdhMMP8) in male Wistar rats with experimental advanced liver fibrosis induced by thioacetamide. Therapeutic effects were monitored after 1, 2, or 3 weeks of a single dose (3 × 1011 vp/kg) of AdhMMP8. Circulating and liver concentration of MMP-8 protein remained constant; hepatic fibrosis decreased up to 48%; proinflammatory and profibrogenic genes expression diminished: TNF-α 2.28-fold, IL-1 1.95-fold, Col 1A1 4-fold, TGF-β1 3-fold and CTGF 2-fold; and antifibrogenic genes expression raised, MMP-9 2.8-fold and MMP-1 10-fold. Our data proposes that the administration of AdhMMP8 in muscle is safe and effective in achieving liver fibrosis regression at a comparable extent as when the adenoviral vector is delivered systemically to reach the liver, using a minimally invasive procedure.
Collapse
Affiliation(s)
- Jesús García-Bañuelos
- Institute for Molecular Biology in Medicine and Gene Therapy, Department of Molecular Biology and Genomics, Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Edén Oceguera-Contreras
- Laboratorio de Sistemas Biológicos, Centro Universitario de los Valles, Universidad de Guadalajara, Carretera Guadalajara-Ameca km. 45.5, Ameca 46600, Jalisco, Mexico
| | - Ana Sandoval-Rodríguez
- Institute for Molecular Biology in Medicine and Gene Therapy, Department of Molecular Biology and Genomics, Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Blanca Estela Bastidas-Ramírez
- Instituto de Investigación en Enfermedades Crónico Degenerativas, Department of Molecular Biology and Genomics, Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Silvia Lucano-Landeros
- Institute for Molecular Biology in Medicine and Gene Therapy, Department of Molecular Biology and Genomics, Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Daniela Gordillo-Bastidas
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey 64849, Nuevo Leon, Mexico
| | - Belinda C. Gómez-Meda
- Instituto de Genética Humana “Dr. Enrique Corona Rivera”, Department of Molecular Biology and Genomics, Health Sciences University Center, Guadalajara 44340, Jalisco, Mexico
| | - Arturo Santos
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey 64849, Nuevo Leon, Mexico
| | | | - Juan Armendariz-Borunda
- Institute for Molecular Biology in Medicine and Gene Therapy, Department of Molecular Biology and Genomics, Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey 64849, Nuevo Leon, Mexico
| |
Collapse
|
4
|
Benítez-Temiño B, Hernández RG, de la Cruz RR, Pastor AM. BDNF Influence on Adult Terminal Axon Sprouting after Partial Deafferentation. Int J Mol Sci 2023; 24:10660. [PMID: 37445838 DOI: 10.3390/ijms241310660] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/20/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
BDNF is a neurotrophin family member implicated in many different neuronal functions, from neuronal survival during development to synaptic plasticity associated with processes of learning and memory. Its presence in the oculomotor system has previously been demonstrated, as it regulates afferent composition of extraocular motoneurons and their firing pattern. Moreover, BDNF expression increases after extraocular motoneuron partial deafferentation, in parallel with terminal axon sprouting from the remaining axons. To elucidate whether BDNF could play an active role in this process, we performed partial deafferentation of the medial rectus motoneurons through transection of one of the two main afferents, that is, the ascending tract of Deiters, and injected BDNF into the motoneuron target muscle, the medial rectus. Furthermore, to check whether BDNF could stimulate axon sprouting without lesions, we performed the same experiment without any lesions. Axon terminal sprouting was assessed by calretinin immunostaining, which specifically labels the remaining afferent system on medial rectus motoneurons, the abducens internuclear neurons. The results presented herein show that exogenous BDNF stimulated terminal axon growth, allowing the total recovery of synaptic coverage around the motoneuron somata. Moreover, calretinin staining in the neuropil exceeded that present in the control situation. Thus, BDNF could also stimulate axonal sprouting in the neuropil of intact animals. These results point to an active role of BDNF in plastic adaptations that take place after partial deafferentation.
Collapse
Affiliation(s)
- Beatriz Benítez-Temiño
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, 41012 Seville, Spain
| | - Rosendo G Hernández
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, 41012 Seville, Spain
| | - Rosa R de la Cruz
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, 41012 Seville, Spain
| | - Angel M Pastor
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, 41012 Seville, Spain
| |
Collapse
|
5
|
Klann PJ, Wang X, Elfert A, Zhang W, Köhler C, Güttsches AK, Jacobsen F, Weyen U, Roos A, Ehrke-Schulz E, Ehrhardt A, Vorgerd M, Bayer W. Seroprevalence of Binding and Neutralizing Antibodies against 39 Human Adenovirus Types in Patients with Neuromuscular Disorders. Viruses 2022; 15:79. [PMID: 36680119 PMCID: PMC9866721 DOI: 10.3390/v15010079] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/22/2022] [Accepted: 12/24/2022] [Indexed: 12/29/2022] Open
Abstract
High pre-existing antibodies against viral vectors reduce their functionality and may lead to adverse complications. To circumvent this problem in future gene therapy approaches, we tested the seroprevalence of a large range of human adenovirus types in patients with neuromuscular disorders (NMDs) to find appropriate viral vector candidates for gene replacement therapy for NMDs. Binding and neutralizing antibodies against 39 human adenovirus types were tested in the sera of 133 patients with NMDs and 76 healthy controls aged 17-92 years. The influence of age, sex, and NMDs on antibody levels was analyzed. The seroprevalence of different adenoviruses in the cohort varied widely. The highest levels of binding antibodies were detected against HAdV-D27, -C1, -D24, -D70, -B14, -C6, -D13, -B34, and -E4, whereas the lowest reactivity was detected against HAdV-F41, -A31, -B11, -D75, -D8, -D65, -D26, -D80, and -D17. The highest neutralizing reactivity was observed against HAdV-B3, -C2, -E4, -C1, -G52, -C5, and -F41, whereas the lowest neutralizing reactivity was observed against HAdV-D74, -B34, -D73, -B37, -D48, -D13, -D75, -D8, -B35, and -B16. We detected no influence of sex and only minor differences between different age groups. Importantly, there were no significant differences between healthy controls and patients with NMDs. Our data show that patients with NMDs have very similar levels of binding and neutralizing antibodies against HAdV compared to healthy individuals, and we identified HAdV-A31, -B16, -B34, -B35, -D8, -D37, -D48, -D73, -D74, -D75, and -D80 as promising candidates for future vector development due to their low binding and neutralizing antibody prevalence.
Collapse
Affiliation(s)
- Patrick Julian Klann
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, 45122 Essen, Germany
- Heimer Institute for Muscle Research, Department of Neurology, University Hospital Bergmannsheil, Ruhr-University Bochum, 44789 Bochum, Germany
| | - Xiaoyan Wang
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, 45122 Essen, Germany
| | - Anna Elfert
- Heimer Institute for Muscle Research, Department of Neurology, University Hospital Bergmannsheil, Ruhr-University Bochum, 44789 Bochum, Germany
| | - Wenli Zhang
- Virology and Microbiology, Center for Medical Education and Research, Department of Human Medicine, Faculty of Health, Witten/Herdecke University, 58453 Witten, Germany
| | - Cornelia Köhler
- Clinics for Pediatrics and Adolescent Medicine, University Hospital Sankt Josef, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Anne-Katrin Güttsches
- Heimer Institute for Muscle Research, Department of Neurology, University Hospital Bergmannsheil, Ruhr-University Bochum, 44789 Bochum, Germany
| | - Frank Jacobsen
- Heimer Institute for Muscle Research, Department of Neurology, University Hospital Bergmannsheil, Ruhr-University Bochum, 44789 Bochum, Germany
| | - Ute Weyen
- Heimer Institute for Muscle Research, Department of Neurology, University Hospital Bergmannsheil, Ruhr-University Bochum, 44789 Bochum, Germany
| | - Andreas Roos
- Heimer Institute for Muscle Research, Department of Neurology, University Hospital Bergmannsheil, Ruhr-University Bochum, 44789 Bochum, Germany
| | - Eric Ehrke-Schulz
- Virology and Microbiology, Center for Medical Education and Research, Department of Human Medicine, Faculty of Health, Witten/Herdecke University, 58453 Witten, Germany
| | - Anja Ehrhardt
- Virology and Microbiology, Center for Medical Education and Research, Department of Human Medicine, Faculty of Health, Witten/Herdecke University, 58453 Witten, Germany
| | - Matthias Vorgerd
- Heimer Institute for Muscle Research, Department of Neurology, University Hospital Bergmannsheil, Ruhr-University Bochum, 44789 Bochum, Germany
| | - Wibke Bayer
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, 45122 Essen, Germany
| |
Collapse
|
6
|
Miciński B, Jana B, Całka J. Uterine Inflammation Changes the Expression of Cholinergic Neurotransmitters and Decreases the Population of AChE-Positive, Uterus-Innervating Neurons in the Paracervical Ganglion of Sexually Mature Gilts. Animals (Basel) 2022; 12:ani12131676. [PMID: 35804576 PMCID: PMC9264917 DOI: 10.3390/ani12131676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/23/2022] [Accepted: 06/28/2022] [Indexed: 11/27/2022] Open
Abstract
Simple Summary Endometritis, both with non-infectious and infectious backgrounds, is one of the most prevalent pathological states among domestic animals. In animals, it generates severe economic problems, including lowered reproductive indices and rising medical treatment costs, and in women, it might lead to severe fertility impairment. In order to determine how the autonomic nervous system responds to such a pathological state, an experimental group of pigs were treated with Escherichia coli injection into the uterine horns, and several ganglions responsible for innervation of this organ were examined, including the paracervical ganglion located on both sides of the broad ligament of the uterus. The results clearly showed a strong impact of the inflammation on the chemical coding of neurons, some even synthesizing neurotransmitters de novo such as the GAL-expressing perikarya. Additionally, applied injections decreased the number of parasympathetic, acetylcholinesterase-expressing neurons implying the importance of the cholinergic population to keep the inflammation under control. The obtained data serve as a basis for the future implementation of modern treatment and enhancements in animal breeding. Abstract The focus of this study was based on examining the impact of endometritis on the chemical coding of the paracervical ganglion (PCG) perikaryal populations supplying pig uterus. Four weeks after the injection of Fast Blue retrograde tracer into uterine horns, either the Escherichia coli (E. coli) suspension or saline solution was applied to both horns. Laparotomy treatment was performed for the control group. Uterine cervices containing PCG were extracted on the eighth day after previous treatments. Subsequent macroscopic and histopathologic examinations acknowledged the severe form of acute endometritis in the E. coli-treated gilts, whereas double-labeling immunofluorescence procedures allowed changes to be analyzed in the PCG perikaryal populations coded with vesicular acetylcholine transporter (VAChT) and/or somatostatin (SOM), vasoactive intestinal polypeptide (VIP), a neuronal isoform of nitric oxide synthase (nNOS), galanin (GAL). The acetylcholinesterase (AChE) detection method was used to check for the presence and changes in the expression of this enzyme and further confirm the presence of cholinergic perikarya in PCG. Treatment with E. coli resulted in an increase in VAChT+/VIP+, VAChT+/VIP−, VAChT+/SOM+, VAChT+/SOM−, VAChT+/GAL− and VAChT+/nNOS− PCG uterine perikarya. An additional increase was noted in the non-cholinergic VIP-, SOM- and nNOS-immunopositive populations, as well as a decrease in the number of cholinergic nNOS-positive perikarya. Moreover, the population of cholinergic GAL-expressing perikarya that appeared in the E. coli-injected gilts and E. coli injections lowered the number of AChE-positive perikarya. The neurochemical characteristics of the cholinergic uterine perikarya of the PCG were altered and influenced by the pathological state (inflammation of the uterus). These results may indicate the additional influence of such a state on the functioning of this organ.
Collapse
Affiliation(s)
- Bartosz Miciński
- Department of Clinical Physiology, Faculty of Veterinary Medicine, University of Warmia and Mazury, Oczapowskiego 14, 11-041 Olsztyn, Poland;
- Correspondence: (B.M.); (B.J.); Tel.: +48-89-523-44-61 (B.M.); +48-89-539-31-37 (B.J.)
| | - Barbara Jana
- Division of Reproductive Biology, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Tuwima 10, 10-748 Olsztyn, Poland
- Correspondence: (B.M.); (B.J.); Tel.: +48-89-523-44-61 (B.M.); +48-89-539-31-37 (B.J.)
| | - Jarosław Całka
- Department of Clinical Physiology, Faculty of Veterinary Medicine, University of Warmia and Mazury, Oczapowskiego 14, 11-041 Olsztyn, Poland;
| |
Collapse
|
7
|
Parambi DGT, Alharbi KS, Kumar R, Harilal S, Batiha GES, Cruz-Martins N, Magdy O, Musa A, Panda DS, Mathew B. Gene Therapy Approach with an Emphasis on Growth Factors: Theoretical and Clinical Outcomes in Neurodegenerative Diseases. Mol Neurobiol 2022; 59:191-233. [PMID: 34655056 PMCID: PMC8518903 DOI: 10.1007/s12035-021-02555-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 09/05/2021] [Indexed: 12/11/2022]
Abstract
The etiology of many neurological diseases affecting the central nervous system (CNS) is unknown and still needs more effective and specific therapeutic approaches. Gene therapy has a promising future in treating neurodegenerative disorders by correcting the genetic defects or by therapeutic protein delivery and is now an attraction for neurologists to treat brain disorders, like Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, spinal muscular atrophy, spinocerebellar ataxia, epilepsy, Huntington's disease, stroke, and spinal cord injury. Gene therapy allows the transgene induction, with a unique expression in cells' substrate. This article mainly focuses on the delivering modes of genetic materials in the CNS, which includes viral and non-viral vectors and their application in gene therapy. Despite the many clinical trials conducted so far, data have shown disappointing outcomes. The efforts done to improve outcomes, efficacy, and safety in the identification of targets in various neurological disorders are also discussed here. Adapting gene therapy as a new therapeutic approach for treating neurological disorders seems to be promising, with early detection and delivery of therapy before the neuron is lost, helping a lot the development of new therapeutic options to translate to the clinic.
Collapse
Affiliation(s)
- Della Grace Thomas Parambi
- College of Pharmacy, Department of Pharmaceutical Chemistry, Jouf University, Al Jouf-2014, Sakaka, Saudi Arabia
| | - Khalid Saad Alharbi
- College of Pharmacy, Department of Pharmaceutical Chemistry, Jouf University, Al Jouf-2014, Sakaka, Saudi Arabia
| | - Rajesh Kumar
- Kerala University of Health Sciences, Thrissur, Kerala 680596 India
| | - Seetha Harilal
- Kerala University of Health Sciences, Thrissur, Kerala 680596 India
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511 Al Beheira Egypt
| | - Natália Cruz-Martins
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- Institute of Research and Advanced Training in Health Sciences and Technologies (CESPU), Rua Central de Gandra, 1317, 4585-116 Gandra PRD, Portugal
| | - Omnia Magdy
- Department of Clinical Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al Jouf-2014 Kingdom of Saudi Arabia
- Pharmacognosy Department, College of Pharmacy, Jouf University, Sakaka, Aljouf 72341 Kingdom of Saudi Arabia
| | - Arafa Musa
- Pharmacognosy Department, College of Pharmacy, Jouf University, Sakaka, Aljouf 72341 Kingdom of Saudi Arabia
- Pharmacognosy Department, Faculty of Pharmacy, Al-Azhar University, Cairo, 11371 Egypt
| | - Dibya Sundar Panda
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Al Jouf, Sakaka, 72341 Kingdom of Saudi Arabia
| | - Bijo Mathew
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, 682 041 India
| |
Collapse
|
8
|
In Search of a Cure: The Development of Therapeutics to Alter the Progression of Spinal Muscular Atrophy. Brain Sci 2021; 11:brainsci11020194. [PMID: 33562482 PMCID: PMC7915832 DOI: 10.3390/brainsci11020194] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/29/2021] [Accepted: 02/01/2021] [Indexed: 12/19/2022] Open
Abstract
Until the recent development of disease-modifying therapeutics, spinal muscular atrophy (SMA) was considered a devastating neuromuscular disease with a poor prognosis for most affected individuals. Symptoms generally present during early childhood and manifest as muscle weakness and progressive paralysis, severely compromising the affected individual’s quality of life, independence, and lifespan. SMA is most commonly caused by the inheritance of homozygously deleted SMN1 alleles with retention of one or more copies of a paralog gene, SMN2, which inversely correlates with disease severity. The recent advent and use of genetically targeted therapies have transformed SMA into a prototype for monogenic disease treatment in the era of genetic medicine. Many SMA-affected individuals receiving these therapies achieve traditionally unobtainable motor milestones and survival rates as medicines drastically alter the natural progression of this disease. This review discusses historical SMA progression and underlying disease mechanisms, highlights advances made in therapeutic research, clinical trials, and FDA-approved medicines, and discusses possible second-generation and complementary medicines as well as optimal temporal intervention windows in order to optimize motor function and improve quality of life for all SMA-affected individuals.
Collapse
|
9
|
Local and Systemic Humoral Response to Autologous Lineage-Negative Cells Intrathecal Administration in ALS Patients. Int J Mol Sci 2020; 21:ijms21031070. [PMID: 32041109 PMCID: PMC7037134 DOI: 10.3390/ijms21031070] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/31/2020] [Accepted: 02/04/2020] [Indexed: 01/04/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) remains a fatal disease with limited therapeutic options. Signaling via neurotrophins (NTs), neuroinflammation, and certain micro-RNAs are believed to play essential role in ALS pathogenesis. Lineage-negative stem/progenitor cells (Lin−) were obtained from bone marrow of 18 ALS patients and administered intrathecally. Clinical assessment was performed using ALS Functional Rating Scale (FRSr) and Norris scale. Protein concentrations were measured in plasma and cerebrospinal fluid (CSF) by multiplex fluorescent bead-based immunoassay. Gene expression in nucleated blood cells was assessed using gene microarray technique. Finally, miRNA expression was analyzed using qPCR in CSF and plasma samples. We observed a significant decrease of C-reactive protein (CRP) concentration in plasma on the seventh day from the application of cells. Gene array results revealed decreased expression of gene sets responsible for neutrophil activation. Further analysis revealed moderate negative correlation between CRP level in CSF and clinical outcome. Brain-derived neurotrophic factor (BDNF) concentrations in both plasma and CSF significantly correlated with the favorable clinical outcome. On a micro-RNA level, we observed significant increase of miR-16-5p expression one week after transplantation in both body fluids and significant increase of miR-206 expression in plasma. Administration of Lin− cells may decrease inflammatory response and prevent neurodegeneration. However, these issues require further investigations.
Collapse
|
10
|
Raghuram A, Singh A, Chang DK, Nunez M, Reece EM, Schultz BE. The Evolving Landscape of Gene Therapy in Plastic Surgery. Semin Plast Surg 2019; 33:167-172. [PMID: 31384232 DOI: 10.1055/s-0039-1693131] [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: 10/26/2022]
Abstract
With the rapid rise of personalized genomic sequencing and clustered regularly interspaced short palindromic repeat (CRISPR) technology, previous gaps in gene therapy are beginning to be bridged, paving the way for increasing clinical applicability. This article aims to provide an overview of the fundamentals of gene therapy and discuss future potential interventions relevant to plastic surgeons. These interventions include enhancing tissue regeneration and healing, as well as modifying disease processes in congenital anomalies. Though clinical applications are still on the horizon, a deeper understanding of these new advances will help plastic surgeons understand the current landscape of gene therapy and stay abreast of future opportunities.
Collapse
Affiliation(s)
| | - Aspinder Singh
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
| | - Daniel K Chang
- Division of Plastic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
| | - Mervin Nunez
- Division of Plastic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
| | - Edward M Reece
- Division of Plastic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
| | | |
Collapse
|
11
|
Just-Borràs L, Hurtado E, Cilleros-Mañé V, Biondi O, Charbonnier F, Tomàs M, Garcia N, Lanuza MA, Tomàs J. Overview of Impaired BDNF Signaling, Their Coupled Downstream Serine-Threonine Kinases and SNARE/SM Complex in the Neuromuscular Junction of the Amyotrophic Lateral Sclerosis Model SOD1-G93A Mice. Mol Neurobiol 2019; 56:6856-6872. [PMID: 30929165 DOI: 10.1007/s12035-019-1550-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 03/13/2019] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a chronic neurodegenerative disease characterized by progressive motor weakness. It is accepted that it is caused by motoneuron degeneration leading to a decrease in muscle stimulation. However, ALS is being redefined as a distal axonopathy, in that neuromuscular junction dysfunction precedes and may even influence motoneuron loss. In this synapse, several metabotropic receptor-mediated signaling pathways converge on effector kinases that phosphorylate targets that are crucial for synaptic stability and neurotransmission quality. We have previously shown that, in physiological conditions, nerve-induced muscle contraction regulates the brain-derived neurotrophic factor/tropomyosin-related kinase B (BDNF/TrkB) signaling to retrogradely modulate presynaptic protein kinases PKC and PKA, which are directly involved in the modulation of acetylcholine release. In ALS patients, the alteration of this signaling may significantly contribute to a motor impairment. Here, we investigate whether BDNF/TrkB signaling, the downstream PKC (cPKCβI, cPKCα, and nPKCε isoforms), and PKA (regulatory and catalytic subunits) and some SNARE/SM exocytotic machinery proteins (Munc18-1 and SNAP-25) are altered in the skeletal muscle of pre- and symptomatic SOD1-G93A mice. We found that this pathway is strongly affected in symptomatic ALS mice muscles including an unbalance between (I) BDNF and TrkB isoforms, (II) PKC isoforms and PKA subunits, and (III) Munc18-1 and SNAP-25 phosphorylation ratios. Changes in TrkB.T1 and cPKCβI are precociously observed in presymptomatic mice. Altogether, several of these molecular alterations can be partly associated with the known fast-to-slow motor unit transition during the disease process but others can be related with the initial disease pathogenesis.
Collapse
Affiliation(s)
- Laia Just-Borràs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Carrer St Llorenç num 21, 43201, Reus, Spain
| | - Erica Hurtado
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Carrer St Llorenç num 21, 43201, Reus, Spain
| | - Víctor Cilleros-Mañé
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Carrer St Llorenç num 21, 43201, Reus, Spain
| | - Olivier Biondi
- INSERM UMRS 1124 and Université Paris Descartes, 45 rue des Saints-Pères, 75270, Paris Cedex 06, France
| | - Frédéric Charbonnier
- INSERM UMRS 1124 and Université Paris Descartes, 45 rue des Saints-Pères, 75270, Paris Cedex 06, France
| | - Marta Tomàs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Carrer St Llorenç num 21, 43201, Reus, Spain
| | - Neus Garcia
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Carrer St Llorenç num 21, 43201, Reus, Spain
| | - Maria A Lanuza
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Carrer St Llorenç num 21, 43201, Reus, Spain.
| | - Josep Tomàs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Carrer St Llorenç num 21, 43201, Reus, Spain.
| |
Collapse
|
12
|
Tosolini AP, Sleigh JN. Motor Neuron Gene Therapy: Lessons from Spinal Muscular Atrophy for Amyotrophic Lateral Sclerosis. Front Mol Neurosci 2017; 10:405. [PMID: 29270111 PMCID: PMC5725447 DOI: 10.3389/fnmol.2017.00405] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 11/21/2017] [Indexed: 12/11/2022] Open
Abstract
Spinal muscular atrophy (SMA) and amyotrophic lateral sclerosis (ALS) are severe nervous system diseases characterized by the degeneration of lower motor neurons. They share a number of additional pathological, cellular, and genetic parallels suggesting that mechanistic and clinical insights into one disorder may have value for the other. While there are currently no clinical ALS gene therapies, the splice-switching antisense oligonucleotide, nusinersen, was recently approved for SMA. This milestone was achieved through extensive pre-clinical research and patient trials, which together have spawned fundamental insights into motor neuron gene therapy. We have thus tried to distil key information garnered from SMA research, in the hope that it may stimulate a more directed approach to ALS gene therapy. Not only must the type of therapeutic (e.g., antisense oligonucleotide vs. viral vector) be sensibly selected, but considerable thought must be applied to the where, which, what, and when in order to enhance treatment benefit: to where (cell types and tissues) must the drug be delivered and how can this be best achieved? Which perturbed pathways must be corrected and can they be concurrently targeted? What dosing regime and concentration should be used? When should medication be administered? These questions are intuitive, but central to identifying and optimizing a successful gene therapy. Providing definitive solutions to these quandaries will be difficult, but clear thinking about therapeutic testing is necessary if we are to have the best chance of developing viable ALS gene therapies and improving upon early generation SMA treatments.
Collapse
Affiliation(s)
- Andrew P Tosolini
- Sobell Department of Motor Neuroscience and Movement Disorders, Institute of Neurology, University College London, London, United Kingdom
| | - James N Sleigh
- Sobell Department of Motor Neuroscience and Movement Disorders, Institute of Neurology, University College London, London, United Kingdom
| |
Collapse
|
13
|
Alory A, Jacquier A, Gentien D, de la Grange P, Haase G. [Neurotrophic factors for motor neurons: an à la carte menu]. Med Sci (Paris) 2017; 33:835-839. [PMID: 28994373 DOI: 10.1051/medsci/20173310008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Alysson Alory
- Institut de neurosciences de la Timone, UMR 7289 CNRS et Aix-Marseille Université, 27, boulevard Jean Moulin, 13005 Marseille, France
| | - Arnaud Jacquier
- Institut de neurosciences de la Timone, UMR 7289 CNRS et Aix-Marseille Université, 27, boulevard Jean Moulin, 13005 Marseille, France
| | - David Gentien
- Institut Curie, Université PSL, Département de recherche translationnelle, Plateforme de génomique, 26, rue d'Ulm, 75248 Paris, France
| | - Pierre de la Grange
- GenoSplice, iPEPS - ICM, Hôpital Pitié Salpêtrière, 47, boulevard de l'Hôpital, 75013 Paris, France
| | - Georg Haase
- Institut de neurosciences de la Timone, UMR 7289 CNRS et Aix-Marseille Université, 27, boulevard Jean Moulin, 13005 Marseille, France
| |
Collapse
|
14
|
Hernández RG, Silva-Hucha S, Morcuende S, de la Cruz RR, Pastor AM, Benítez-Temiño B. Extraocular Motor System Exhibits a Higher Expression of Neurotrophins When Compared with Other Brainstem Motor Systems. Front Neurosci 2017; 11:399. [PMID: 28744196 PMCID: PMC5504184 DOI: 10.3389/fnins.2017.00399] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 06/26/2017] [Indexed: 01/01/2023] Open
Abstract
Extraocular motoneurons resist degeneration in diseases such as amyotrophic lateral sclerosis. The main objective of the present work was to characterize the presence of neurotrophins in extraocular motoneurons and muscles of the adult rat. We also compared these results with those obtained from other cranial motor systems, such as facial and hypoglossal, which indeed suffer neurodegeneration. Immunocytochemical analysis was used to describe the expression of nerve growth factor, brain-derived neurotrophic factor and neurotrophin-3 in oculomotor, trochlear, abducens, facial, and hypoglossal nuclei of adult rats, and Western blots were used to describe the presence of neurotrophins in extraocular, facial (buccinator), and tongue muscles, which are innervated by the above-mentioned motoneurons. In brainstem samples, brain-derived neurotrophic factor was present both in extraocular and facial motoneuron somata, and to a lesser degree, in hypoglossal motoneurons. Neurotrophin-3 was present in extraocular motor nuclei, while facial and hypoglossal motoneurons were almost devoid of this protein. Finally, nerve growth factor was not present in the soma of any group of motoneurons, although it was present in dendrites of motoneurons located in the neuropil. Neuropil optical density levels were higher in extraocular motoneuron nuclei when compared with facial and hypoglossal nuclei. Neurotrophins could be originated in target muscles, since Western blot analyses revealed the presence of the three molecules in all sampled muscles, to a larger extent in extraocular muscles when compared with facial and tongue muscles. We suggest that the different neurotrophin availability could be related to the particular resistance of extraocular motoneurons to neurodegeneration.
Collapse
|
15
|
Schaller S, Buttigieg D, Alory A, Jacquier A, Barad M, Merchant M, Gentien D, de la Grange P, Haase G. Novel combinatorial screening identifies neurotrophic factors for selective classes of motor neurons. Proc Natl Acad Sci U S A 2017; 114:E2486-E2493. [PMID: 28270618 PMCID: PMC5373341 DOI: 10.1073/pnas.1615372114] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Numerous neurotrophic factors promote the survival of developing motor neurons but their combinatorial actions remain poorly understood; to address this, we here screened 66 combinations of 12 neurotrophic factors on pure, highly viable, and standardized embryonic mouse motor neurons isolated by a unique FACS technique. We demonstrate potent, strictly additive, survival effects of hepatocyte growth factor (HGF), ciliary neurotrophic factor (CNTF), and Artemin through specific activation of their receptor complexes in distinct subsets of lumbar motor neurons: HGF supports hindlimb motor neurons through c-Met; CNTF supports subsets of axial motor neurons through CNTFRα; and Artemin acts as the first survival factor for parasympathetic preganglionic motor neurons through GFRα3/Syndecan-3 activation. These data show that neurotrophic factors can selectively promote the survival of distinct classes of embryonic motor neurons. Similar studies on postnatal motor neurons may provide a conceptual framework for the combined therapeutic use of neurotrophic factors in degenerative motor neuron diseases such as amyotrophic lateral sclerosis, spinal muscular atrophy, and spinobulbar muscular atrophy.
Collapse
Affiliation(s)
- Sébastien Schaller
- Institut de Neurosciences de la Timone, UMR 7289 CNRS, Aix-Marseille University, 13005 Marseille, France
| | - Dorothée Buttigieg
- Institut de Neurosciences de la Timone, UMR 7289 CNRS, Aix-Marseille University, 13005 Marseille, France
| | - Alysson Alory
- Institut de Neurosciences de la Timone, UMR 7289 CNRS, Aix-Marseille University, 13005 Marseille, France
| | - Arnaud Jacquier
- Institut de Neurosciences de la Timone, UMR 7289 CNRS, Aix-Marseille University, 13005 Marseille, France
| | - Marc Barad
- Centre d'Immunologie de Marseille-Luminy (CIML), CNRS, INSERM, Aix-Marseille University, 13288 Marseille, France
| | | | - David Gentien
- Institut Curie, Translational Research Department, Genomic Platform, PSL Research University, 75248 Paris, France
| | - Pierre de la Grange
- GenoSplice Technology, Institut du Cerveau et de la Moëlle (ICM), Hôpital Pitié Salpêtrière, 75013 Paris, France
| | - Georg Haase
- Institut de Neurosciences de la Timone, UMR 7289 CNRS, Aix-Marseille University, 13005 Marseille, France;
| |
Collapse
|
16
|
Janssens J, Lu D, Ni B, Chadwick W, Siddiqui S, Azmi A, Etienne H, Jushaj A, van Gastel J, Martin B, Maudsley S. Development of Precision Small-Molecule Proneurotrophic Therapies for Neurodegenerative Diseases. VITAMINS AND HORMONES 2016; 104:263-311. [PMID: 28215298 DOI: 10.1016/bs.vh.2016.10.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Age-related neurodegenerative diseases, such as Alzheimer's disease, will represent one of the largest future burdens on worldwide healthcare systems due to the increasing proportion of elderly in our society. As deficiencies in neurotrophins are implicated in the pathogenesis of many age-related neurodegenerative disorders, it is reasonable to consider that global neurotrophin resistance may also become a major healthcare threat. Central nervous system networks are effectively maintained through aging by neuroprotective and neuroplasticity signaling mechanisms which are predominantly controlled by neurotrophin receptor signaling. Neurotrophin receptors are single pass receptor tyrosine kinases that form dimeric structures upon ligand binding to initiate cellular signaling events that control many protective and plasticity-related pathways. Declining functionality of the neurotrophin ligand-receptor system is considered one of the hallmarks of neuropathological aging. Therefore, it is imperative to develop effective therapeutic strategies to contend with this significant issue. While the therapeutic applications of cognate ligands for neurotrophin receptors are limited, the development of nonpeptidergic, small-molecule ligands can overcome these limitations, and productively regulate this important receptor system with beneficial effects. Using our advanced knowledge of the high-dimensionality complexity of receptor systems, the future generation of precision medicines targeting these systems will be an attainable goal.
Collapse
Affiliation(s)
- J Janssens
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium
| | - D Lu
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States
| | - B Ni
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States
| | - W Chadwick
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States
| | - S Siddiqui
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States
| | - A Azmi
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium
| | - H Etienne
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium
| | - A Jushaj
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium
| | - J van Gastel
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium
| | - B Martin
- Metabolism Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States
| | - S Maudsley
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium; Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States.
| |
Collapse
|
17
|
Targeting Motor End Plates for Delivery of Adenoviruses: An Approach to Maximize Uptake and Transduction of Spinal Cord Motor Neurons. Sci Rep 2016; 6:33058. [PMID: 27619631 PMCID: PMC5020496 DOI: 10.1038/srep33058] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 08/08/2016] [Indexed: 02/07/2023] Open
Abstract
Gene therapy can take advantage of the skeletal muscles/motor neurons anatomical relationship to restrict gene expression to the spinal cord ventral horn. Furthermore, recombinant adenoviruses are attractive viral-vectors as they permit spatial and temporal modulation of transgene expression. In the literature, however, several inconsistencies exist with regard to the intramuscular delivery parameters of adenoviruses. The present study is an evaluation of the optimal injection sites on skeletal muscle, time course of expression and mice’s age for maximum transgene expression in motor neurons. Targeting motor end plates yielded a 2.5-fold increase in the number of transduced motor neurons compared to injections performed away from this region. Peak adenoviral transgene expression in motor neurons was detected after seven days. Further, greater numbers of transduced motor neurons were found in juvenile (3–7 week old) mice as compared with adults (8+ weeks old). Adenoviral injections produced robust transgene expression in motor neurons and skeletal myofibres. In addition, dendrites of transduced motor neurons were shown to extend well into the white matter where the descending motor pathways are located. These results also provide evidence that intramuscular delivery of adenovirus can be a suitable gene therapy approach to treat spinal cord injury.
Collapse
|
18
|
Gopinath C, Nathar TJ, Ghosh A, Hickstein DD, Nelson EJR. Contemporary Animal Models For Human Gene Therapy Applications. Curr Gene Ther 2016; 15:531-40. [PMID: 26415576 DOI: 10.2174/1566523215666150929110424] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/02/2015] [Accepted: 09/08/2015] [Indexed: 01/18/2023]
Abstract
Over the past three decades, gene therapy has been making considerable progress as an alternative strategy in the treatment of many diseases. Since 2009, several studies have been reported in humans on the successful treatment of various diseases. Animal models mimicking human disease conditions are very essential at the preclinical stage before embarking on a clinical trial. In gene therapy, for instance, they are useful in the assessment of variables related to the use of viral vectors such as safety, efficacy, dosage and localization of transgene expression. However, choosing a suitable disease-specific model is of paramount importance for successful clinical translation. This review focuses on the animal models that are most commonly used in gene therapy studies, such as murine, canine, non-human primates, rabbits, porcine, and a more recently developed humanized mice. Though small and large animals both have their own pros and cons as disease-specific models, the choice is made largely based on the type and length of study performed. While small animals with a shorter life span could be well-suited for degenerative/aging studies, large animals with longer life span could suit longitudinal studies and also help with dosage adjustments to maximize therapeutic benefit. Recently, humanized mice or mouse-human chimaeras have gained interest in the study of human tissues or cells, thereby providing a more reliable understanding of therapeutic interventions. Thus, animal models are of great importance with regard to testing new vector technologies in vivo for assessing safety and efficacy prior to a gene therapy clinical trial.
Collapse
|
19
|
Boido M, Vercelli A. Neuromuscular Junctions as Key Contributors and Therapeutic Targets in Spinal Muscular Atrophy. Front Neuroanat 2016; 10:6. [PMID: 26869891 PMCID: PMC4737916 DOI: 10.3389/fnana.2016.00006] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 01/13/2016] [Indexed: 12/13/2022] Open
Abstract
Spinal muscular atrophy (SMA) is a recessive autosomal neuromuscular disease, representing the most common fatal pediatric pathology. Even though, classically and in a simplistic way, it is categorized as a motor neuron (MN) disease, there is an increasing general consensus that its pathogenesis is more complex than expected. In particular, neuromuscular junctions (NMJs) are affected by dramatic alterations, including immaturity, denervation and neurofilament accumulation, associated to impaired synaptic functions: these abnormalities may in turn have a detrimental effect on MN survival. Here, we provide a description of NMJ development/maintenance/maturation in physiological conditions and in SMA, focusing on pivotal molecules and on the time-course of pathological events. Moreover, since NMJs could represent an important target to be exploited for counteracting the pathology progression, we also describe several therapeutic strategies that, directly or indirectly, aim at NMJs.
Collapse
Affiliation(s)
- Marina Boido
- Department of Neuroscience "Rita Levi Montalcini", Neuroscience Institute Cavalieri Ottolenghi, University of Torino Torino, Italy
| | - Alessandro Vercelli
- Department of Neuroscience "Rita Levi Montalcini", Neuroscience Institute Cavalieri Ottolenghi, University of Torino Torino, Italy
| |
Collapse
|
20
|
Gouarné C, Giraudon-Paoli M, Seimandi M, Biscarrat C, Tardif G, Pruss RM, Bordet T. Olesoxime protects embryonic cortical neurons from camptothecin intoxication by a mechanism distinct from BDNF. Br J Pharmacol 2015; 168:1975-88. [PMID: 23278424 DOI: 10.1111/bph.12094] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Revised: 11/28/2012] [Accepted: 12/10/2012] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND PURPOSE Olesoxime is a small cholesterol-oxime promoting rat embryonic motor neurons survival in the absence of trophic factors. Because olesoxime can substitute for neurotrophic factors in many situations, and to gain further understanding of its mechanism of action, we wondered if it could prevent neuronal death induced by camptothecin (CPT) and compared its effects with those of brain-derived neurotrophic factor (BDNF). EXPERIMENTAL APPROACH E17 rat embryonic cortical neurons were treated with olesoxime, BDNF or vehicle and intoxicated with CPT. Caspase-dependent and caspase-independent death pathways along with pro-survival pathways activation were explored. KEY RESULTS As previously reported for BDNF, olesoxime dose-dependently delayed CPT-induced cell death. Both compounds acted downstream of p53 activation preventing cytochrome c release and caspases activation. When caspase activation was blocked, both olesoxime and BDNF provided additional neuroprotective effect, potentially through the prevention of apoptosis-inducing factor release from mitochondria. While BDNF activates both the PI3K/Akt and the ERK pathway, olesoxime induced only a late activation of the ERK pathways, which did not seem to play a major role in its neuroprotection against CPT. Rather, our results favour preserved mitochondrial membrane integrity by olesoxime. CONCLUSIONS AND IMPLICATIONS Albeit different, olesoxime and BDNF mechanisms for neuroprotection converge to preserve mitochondrial function. These findings emphasize the importance of targeting the mitochondria in the process of neurodegeneration. Importantly olesoxime, by mimicking neurotrophin pro-survival activities without impacting PI3K/Akt and ERK signalling, may have greater therapeutic potential in many diseases where neurotrophins were considered as a therapeutic solution.
Collapse
|
21
|
Bellouze S, Schäfer MK, Buttigieg D, Baillat G, Rabouille C, Haase G. Golgi fragmentation in pmn mice is due to a defective ARF1/TBCE cross-talk that coordinates COPI vesicle formation and tubulin polymerization. Hum Mol Genet 2014; 23:5961-75. [DOI: 10.1093/hmg/ddu320] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
22
|
Ruiz M, Martínez-Vidal AF, Morales JM, Monleón D, Giménez Y Ribotta M. Neurodegenerative changes are prevented by Erythropoietin in the pmn model of motoneuron degeneration. Neuropharmacology 2014; 83:137-53. [PMID: 24769002 DOI: 10.1016/j.neuropharm.2014.04.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 03/02/2014] [Accepted: 04/10/2014] [Indexed: 11/29/2022]
Abstract
Motoneuron diseases are fatal neurodegenerative disorders characterized by a progressive loss of motoneurons, muscle weakness and premature death. The progressive motor neuronopathy (pmn) mutant mouse has been considered a good model for the autosomal recessive childhood form of spinal muscular atrophy (SMA). Here, we investigated the therapeutic potential of Erythropoietin (Epo) on this mutant mouse. Symptomatic or pre-symptomatic treatment with Epo significantly prolongs lifespan by 84.6% or 87.2% respectively. Epo preserves muscle strength and significantly attenuates behavioural motor deficits of mutant pmn mice. Histological and metabolic changes in the spinal cord evaluated by immunohistochemistry, western blot, and high-resolution (1)H-NMR spectroscopy were also greatly prevented by Epo-treatment. Our results illustrate the efficacy of Epo in improving quality of life of mutant pmn mice and open novel therapeutic pathways for motoneuron diseases.
Collapse
Affiliation(s)
- Marta Ruiz
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas (CSIC) - Universidad Miguel Hernández (UMH), Av. Ramón y Cajal s/n, 03550 San Juan de Alicante, Alicante, Spain
| | - Ana Fe Martínez-Vidal
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas (CSIC) - Universidad Miguel Hernández (UMH), Av. Ramón y Cajal s/n, 03550 San Juan de Alicante, Alicante, Spain
| | - José Manuel Morales
- Unidad Central de Investigación en Medicina, Universidad de Valencia, Valencia, Spain
| | - Daniel Monleón
- Fundación de Investigación del Hospital Clínico Universitario de Valencia (FIHCUV), Valencia, Spain
| | - Minerva Giménez Y Ribotta
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas (CSIC) - Universidad Miguel Hernández (UMH), Av. Ramón y Cajal s/n, 03550 San Juan de Alicante, Alicante, Spain.
| |
Collapse
|
23
|
O'Leary VB, Ovsepian SV, Bodeker M, Dolly JO. Improved lentiviral transduction of ALS motoneurons in vivo via dual targeting. Mol Pharm 2013; 10:4195-206. [PMID: 24066863 DOI: 10.1021/mp400247t] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Treatment of amyotrophic lateral sclerosis (ALS), a progressive neurodegenerative disease, is hampered by its complex etiology and lack of efficient means for targeted transfer of therapeutics into motoneurons. The objective of this research was engineering of a versatile motoneuron targeting adapter--a full-length atoxic tetanus toxin fused to core-streptavidin (CS-TeTIM)--for retro-axonal transduction of viral vectors; validation of the targeting efficiency of CS-TeTIM in vivo, by expression of green fluorescence protein (GFP) reporter in motoneurons of presymptomatic and symptomatic ALS-like SOD1(G93A) mice, and comparison with age-matched controls; and appraisal of lentiviral transduction with CS-TeTIM relative to (1) a HC binding fragment of tetanus toxin CS-TeTx(HC), (2) rabies glycoprotein (RG), and (3) a CS-TeTIM-RG dual targeting approach. CS-TeTIM and CS-TeTx(HC) were engineered using recombinant technology and site-directed mutagenesis. Biotinylated vectors, pseudotyped with vesicular stomatitis virus glycoprotein (VSV-G) or RG, were linked to these adaptors and injected intraperitoneally (ip) into presymptomatic (12 weeks old), symptomatic SOD1(G93A) (22 weeks old) or wild type control mice, followed by monitoring of GFP expression in the spinal cord and supraspinal motor structures with quantitative PCR and immuno-histochemistry. Transcripts were detected in the spinal cord and supraspinal motor structures of all mice 2 weeks after receiving a single ip injection, although in symptomatic SOD1(G93A) animals reporter RNA levels were lower compared to presymptomatic and wild-type controls irrespective of the targeting approach. GFP transduction with CS-TeTIM proved more efficient than CS-TeTx(HC) across all groups while CS-TeTIM-RG dual-targeted vectors yielded the highest transcript numbers. Importantly, in both wild-type and presymptomatic SOD1(G93A) mice strong colabeling of choline-acetyltransferase (ChAT) and GFP was visualized in neurons of the brain stem and spinal cord. CS-TeTIM, a versatile adaptor protein for targeted lentiviral transduction of motoneurons, has been engineered and its competence assessed relative to CS-TeTx(HC) and RG. Evidence has been provided that highlights the potential usefulness of this novel recombinant tool for basic research with implications for improved transfer of therapeutic candidates into motoneurons for the amelioration of ALS and related diseases.
Collapse
Affiliation(s)
- Valerie B O'Leary
- International Centre for Neurotherapeutics, Dublin City University , Dublin 9, Ireland
| | | | | | | |
Collapse
|
24
|
Abstract
This overview describes the considerations involved in the preparation and use of a herpes simplex virus type 1 (HSV-1) amplicon as a vector for gene transfer into neurons. Strategies for gene delivery into neurons, either to study the molecular biology of brain function or for gene therapy, must utilize vectors that persist stably in postmitotic cells and that can be targeted both spatially and temporally in the nervous system in vivo. This unit describes the biology of HSV-1 along with a discussion covering development of amplicon and genomic HSV-1 vectors. Advantages and disadvantages of current HSV-1 vectors are presented, and HSV-1 vectors are compared with other vectors for gene transfer into neurons.
Collapse
Affiliation(s)
- Rachael L Neve
- Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
25
|
Pastor D, Viso-León MC, Botella-López A, Jaramillo-Merchan J, Moraleda JM, Jones J, Martínez S. Bone marrow transplantation in hindlimb muscles of motoneuron degenerative mice reduces neuronal death and improves motor function. Stem Cells Dev 2013; 22:1633-44. [PMID: 23282201 DOI: 10.1089/scd.2012.0487] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Bone marrow has proved to be an adequate source of stem cells for the treatment of numerous disorders, including neurodegenerative diseases. Bone marrow can be easily and relatively painlessly extracted from a patient or allogenic donor and then transplanted into the degenerative area. Here, the grafted cells will activate a number of mechanisms in order to protect, repair, and/or regenerate the damaged tissue. These properties make the bone marrow a feasible source for cell therapy. In this work, we transplanted bone marrow cells into a mouse model of motoneuron degeneration, with the particularity of placing the cells in the hindlimb muscles rather than in the spinal cord where neuronal degeneration occurs. To this end, we analyze the possibility for the transplanted cells to increase the survival rate of the spinal cord motoneurons by axonal-guided retrograde neurotrophism. As a result, the mice significantly improved their motor functions. This coincided with an increased number of motoneurons innervating the treated muscle compared with the neurons innervating the non-treated contralateral symmetric muscle. In addition, we detected an increase in glial-derived neurotrophic factor in the spinal cord, a neurotrophic factor known to be involved in the rescue of degenerating motoneurons, exerting a neuroprotective effect. Thus, we have proved that bone marrow injected into the muscles is capable of rescuing these motoneurons from death, which may be a possible therapeutic approach for spinal cord motoneuron degenerative diseases, such as amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Diego Pastor
- Sports Science Research Center, Miguel Hernández University, Elche, Alicante 03202, Spain.
| | | | | | | | | | | | | |
Collapse
|
26
|
Amyotrophic lateral sclerosis. Transl Neurosci 2012. [DOI: 10.1017/cbo9780511980053.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
27
|
Sambataro F, Pennuto M. Cell-autonomous and non-cell-autonomous toxicity in polyglutamine diseases. Prog Neurobiol 2012; 97:152-72. [DOI: 10.1016/j.pneurobio.2011.10.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Revised: 10/21/2011] [Accepted: 10/26/2011] [Indexed: 12/21/2022]
|
28
|
Wadman RI, Bosboom WMJ, van der Pol WL, van den Berg LH, Wokke JHJ, Iannaccone ST, Vrancken AFJE. Drug treatment for spinal muscular atrophy types II and III. Cochrane Database Syst Rev 2012:CD006282. [PMID: 22513940 DOI: 10.1002/14651858.cd006282.pub4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Spinal muscular atrophy (SMA) is caused by degeneration of anterior horn cells, which leads to progressive muscle weakness. Children with SMA type II do not develop the ability to walk without support and have a shortened life expectancy, whereas children with SMA type III develop the ability to walk and have a normal life expectancy. There are no known efficacious drug treatments that influence the disease course of SMA. This is an update of a review first published in 2009. OBJECTIVES To evaluate whether drug treatment is able to slow or arrest the disease progression of SMA types II and III and to assess if such therapy can be given safely. Drug treatment for SMA type I is the topic of a separate updated Cochrane review. SEARCH METHODS We searched the Cochrane Neuromuscular Disease Group Specialized Register (8 March 2011), Cochrane Central Register of Controlled Trials (CENTRAL) (The Cochrane Library 2011, Issue 1), MEDLINE (January 1991 to February 2011), EMBASE (January 1991 to February 2011) and ISI Web of Knowledge (January 1991 to March 8 2011). We also searched clinicaltrials.gov to identify as yet unpublished trials (8 March 2011). SELECTION CRITERIA We sought all randomised or quasi-randomised trials that examined the efficacy of drug treatment for SMA types II and III. Participants had to fulfil the clinical criteria and have a deletion or mutation of the survival motor neuron 1 (SMN1) gene (5q11.2-13.2) that was confirmed by genetic analysis.The primary outcome measure was to be change in disability score within one year after the onset of treatment. Secondary outcome measures within one year after the onset of treatment were to be change in muscle strength, ability to stand or walk, change in quality of life, time from the start of treatment until death or full time ventilation and adverse events attributable to treatment during the trial period. DATA COLLECTION AND ANALYSIS Two authors independently reviewed and extracted data from all potentially relevant trials. Pooled relative risks and pooled standardised mean differences were to be calculated to assess treatment efficacy. Risk of bias was systematically analysed. MAIN RESULTS Six randomised placebo-controlled trials on treatment for SMA types II and III were found and included in the review: the four in the original review and two trials added in this update. The treatments were creatine (55 participants), phenylbutyrate (107 participants), gabapentin (84 participants), thyrotropin releasing hormone (9 participants), hydroxyurea (57 participants), and combination therapy with valproate and acetyl-L-carnitine (61 participants). None of these studies were completely free of bias. All studies had adequate blinding, sequence generation and reports of primary outcomes.None of the included trials showed any statistically significant effects on the outcome measures in participants with SMA types II and III. One participant died due to suffocation in the hydroxyurea trial and one participant died in the creatine trial. No participants in any of the other four trials died or reached the state of full time ventilation. Serious side effects were infrequent. AUTHORS' CONCLUSIONS There is no proven efficacious drug treatment for SMA types II and III.
Collapse
Affiliation(s)
- Renske I Wadman
- Department of Neurology, University Medical Center Utrecht, Utrecht, Netherlands.
| | | | | | | | | | | | | |
Collapse
|
29
|
The combination of a synthetic promoter and a CMV promoter improves foreign gene expression efficiency in myocytes. J Biotechnol 2011; 158:91-6. [PMID: 22206980 DOI: 10.1016/j.jbiotec.2011.11.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Revised: 11/20/2011] [Accepted: 11/22/2011] [Indexed: 12/31/2022]
Abstract
Skeletal muscle is becoming an attractive target tissue for gene therapy. Nevertheless, the low level of gene therapeutic expression in this tissue is the major limitation to it becoming an ideal target for gene transfer. The promoter is important element for gene transcription; however, the gene expression efficiencies and specificities of viral promoters and skeletal muscle-specific promotors are in themselves limiting factors. In this study, we established a dual-promoters system in skeletal muscle using a cytomegalovirus (CMV) promoter and a skeletal muscle-specific synthetic promoter. Mouse myoblast cell line C2C12 cells were transfected with the system. We demonstrated that the dual-promoters system could significantly improve exogenous gene expression rate in vitro when compared with a single CMV promoter system and a skeletal muscle-specific synthetic promoter system in C2C12 cell line, by 69.48% and 41.93%, respectively. Next, we evaluated the system efficiency in vivo, the results showed that the dual-promoters system increased gene expression in mice 1.23-fold and 1.60-fold, respectively compared with expression controlled by the two single promoter vectors. Finally, we tested the dual-promoters system in growth hormone-releasing hormone (GHRH) gene therapy, and revealed that when these two promoters co-drove the GHRH gene expression in vivo animal growth was enhanced significantly. All these results indicate that use of the dual-promoter vector was more efficient for gene expression in skeletal muscle tissue than use of the single promoter vectors. These finding could, hopefully, lead to the development of a high efficiency expression system in myocytes and form an ideal approach for gene therapy.
Collapse
|
30
|
Wadman RI, Bosboom WM, van den Berg LH, Wokke JH, Iannaccone ST, Vrancken AF. Drug treatment for spinal muscular atrophy types II and III. Cochrane Database Syst Rev 2011:CD006282. [PMID: 22161400 DOI: 10.1002/14651858.cd006282.pub3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Spinal muscular atrophy (SMA) is caused by degeneration of anterior horn cells, which leads to progressive muscle weakness. Children with SMA type II do not develop the ability to walk without support and have a shortened life expectancy, whereas children with SMA type III develop the ability to walk and have a normal life expectancy. There are no known efficacious drug treatments that influence the disease course of SMA. This is an update of a review first published in 2009. OBJECTIVES To evaluate whether drug treatment is able to slow or arrest the disease progression of SMA types II and III and to assess if such therapy can be given safely. Drug treatment for SMA type I is the topic of a separate updated Cochrane review. SEARCH METHODS We searched the Cochrane Neuromuscular Disease Group Specialized Register (8 March 2011), Cochrane Central Register of Controlled Trials (CENTRAL) (The Cochrane Library 2011, Issue 1), MEDLINE (January 1991 to February 2011), EMBASE (January 1991 to February 2011) and ISI Web of Knowledge (January 1991 to March 8 2011). We also searched clinicaltrials.gov to identify as yet unpublished trials (8 March 2011). SELECTION CRITERIA We sought all randomised or quasi-randomised trials that examined the efficacy of drug treatment for SMA types II and III. Participants had to fulfil the clinical criteria and have a deletion or mutation of the survival motor neuron 1 (SMN1) gene (5q11.2-13.2) that was confirmed by genetic analysis.The primary outcome measure was to be change in disability score within one year after the onset of treatment. Secondary outcome measures within one year after the onset of treatment were to be change in muscle strength, ability to stand or walk, change in quality of life, time from the start of treatment until death or full time ventilation and adverse events attributable to treatment during the trial period. DATA COLLECTION AND ANALYSIS Two authors independently reviewed and extracted data from all potentially relevant trials. Pooled relative risks and pooled standardised mean differences were to be calculated to assess treatment efficacy. Risk of bias was systematically analysed. MAIN RESULTS Six randomised placebo-controlled trials on treatment for SMA types II and III were found and included in the review: the four in the original review and two trials added in this update. The treatments were creatine (55 participants), phenylbutyrate (107 participants), gabapentin (84 participants), thyrotropin releasing hormone (9 participants), hydroxyurea (57 participants), and combination therapy with valproate and acetyl-L-carnitine (61 participants). None of these studies were completely free of bias. All studies had adequate blinding, sequence generation and reports of primary outcomes.None of the included trials showed any statistically significant effects on the outcome measures in participants with SMA types II and III. One participant died due to suffocation in the hydroxyurea trial and one participant died in the creatine trial. No participants in any of the other four trials died or reached the state of full time ventilation. Serious side effects were infrequent. AUTHORS' CONCLUSIONS There is no proven efficacious drug treatment for SMA types II and III.
Collapse
Affiliation(s)
- Renske I Wadman
- Department of Neurology, University Medical Center Utrecht, Rudolf Magnus Institute for Neuroscience, Universiteitsweg 100, Utrecht, Netherlands, 3584 CG
| | | | | | | | | | | |
Collapse
|
31
|
Gould TW, Oppenheim RW. Motor neuron trophic factors: therapeutic use in ALS? BRAIN RESEARCH REVIEWS 2011; 67:1-39. [PMID: 20971133 PMCID: PMC3109102 DOI: 10.1016/j.brainresrev.2010.10.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2010] [Revised: 10/12/2010] [Accepted: 10/18/2010] [Indexed: 12/12/2022]
Abstract
The modest effects of neurotrophic factor (NTF) treatment on lifespan in both animal models and clinical studies of Amyotropic Lateral Sclerosis (ALS) may result from any one or combination of the four following explanations: 1.) NTFs block cell death in some physiological contexts but not in ALS; 2.) NTFs do not rescue motoneurons (MNs) from death in any physiological context; 3.) NTFs block cell death in ALS but to no avail; and 4.) NTFs are physiologically effective but limited by pharmacokinetic constraints. The object of this review is to critically evaluate the role of both NTFs and the intracellular cell death pathway itself in regulating the survival of spinal and cranial (lower) MNs during development, after injury and in response to disease. Because the role of molecules mediating MN survival has been most clearly resolved by the in vivo analysis of genetically engineered mice, this review will focus on studies of such mice expressing reporter, null or other mutant alleles of NTFs, NTF receptors, cell death or ALS-associated genes.
Collapse
Affiliation(s)
- Thomas W Gould
- Department of Neurobiology and Anatomy, Wake Forest University School of Medicine, Winston-Salem, NC 27157-1010, USA.
| | | |
Collapse
|
32
|
Wang X, Smith GM, Xu XM. Preferential and bidirectional labeling of the rubrospinal tract with adenovirus-GFP for monitoring normal and injured axons. J Neurotrauma 2011; 28:635-47. [PMID: 21299337 DOI: 10.1089/neu.2010.1566] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The rodent rubrospinal tract (RST) has been studied extensively to investigate regeneration and remodeling of central nervous system (CNS) axons. Currently no retrograde tracers can specifically label rubrospinal axons and neurons (RSNs). The RST can be anterogradely labeled by injecting tracers into the red nucleus (RN), but accurately locating the RN is a technical challenge. Here we developed a recombinant adenovirus carrying a green fluorescent protein reporter gene (Adv-GFP) which can preferentially, intensely, and bi-directionally label the RST. When Adv-GFP was injected into the second lumbar spinal cord, the GFP was specifically transported throughout the entire RST, with peak labeling seen at 2 weeks post-injection. When Adv-GFP was injected directly into the RN, GFP was anterogradely transported throughout the RST. Following spinal cord injury (SCI), injection of Adv-GFP resulted in visualization of GFP in transected, spared, or sprouted RST axons bi-directionally. Thus Adv-GFP could be used as a novel tool for monitoring and evaluating strategies designed to maximize RST axonal regeneration and remodeling following SCI.
Collapse
Affiliation(s)
- Xiaofei Wang
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, and Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | | | | |
Collapse
|
33
|
Franz S, Weidner N, Blesch A. Gene therapy approaches to enhancing plasticity and regeneration after spinal cord injury. Exp Neurol 2011; 235:62-9. [PMID: 21281633 DOI: 10.1016/j.expneurol.2011.01.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2010] [Revised: 01/17/2011] [Accepted: 01/24/2011] [Indexed: 01/09/2023]
Abstract
During the past decades, new insights into mechanisms that limit plasticity and functional recovery after spinal cord injury have spurred the development of novel approaches to enhance axonal regeneration and rearrangement of spared circuitry. Gene therapy may provide one means to address mechanisms that underlie the insufficient regenerative response of injured neurons and can also be used to identify factors important for axonal growth. Several genetic approaches aimed to modulate the environment of injured axons, for example by localized expression of growth factors, to enhance axonal sprouting and regeneration and to guide regenerating axons towards their target have been described. In addition, genetic modification of injured neurons via intraparenchymal injection, or via retrograde transport of viral vectors has been used to manipulate the intrinsic growth capacity of injured neurons. In this review we will summarize some of the progress and limitations of cell transplantation and gene therapy to enhance axonal bridging and regeneration across a lesion site, and to maximize the function, collateral sprouting and connectivity of spared axonal systems.
Collapse
Affiliation(s)
- Steffen Franz
- Spinal Cord Injury Center, Heidelberg University Hospital, Germany
| | | | | |
Collapse
|
34
|
Zhang Y, Zheng Y, Zhang YP, Shields LBE, Hu X, Yu P, Burke DA, Wang H, Jun C, Byers J, Whittemore SR, Shields CB. Enhanced adenoviral gene delivery to motor and dorsal root ganglion neurons following injection into demyelinated peripheral nerves. J Neurosci Res 2010; 88:2374-84. [PMID: 20623527 DOI: 10.1002/jnr.22394] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Injection of viral vectors into peripheral nerves may transfer specific genes into their dorsal root ganglion (DRG) neurons and motoneurons. However, myelin sheaths of peripheral axons block the entry of viral particles into nerves. We studied whether mild, transient peripheral nerve demyelination prior to intraneural viral vector injection would enhance gene transfer to target DRG neurons and motoneurons. The right sciatic nerve of C57BL/6 mice was focally demyelinated with 1% lysolecithin, and the left sciatic nerve was similarly injected with saline (control). Five days after demyelination, 0.5 microl of Ad5-GFP was injected into both sciatic nerves at the site of previous injection. The effectiveness of gene transfer was evaluated by counting GFP(+) neurons in the DRGs and ventral horns. After peripheral nerve demyelination, there was a fivefold increase in the number of infected DRG neurons and almost a 15-fold increase in the number of infected motoneurons compared with the control, nondemyelinated side. Focal demyelination reduced the myelin sheath barrier, allowing greater virus-axon contact. Increased CXADR expression on the demyelinated axons facilitated axoplasmic viral entry. No animals sustained any prolonged neurological deficits. Increased gene delivery into DRG neurons and motoneurons may provide effective treatment for amyotrophic lateral sclerosis, pain, and spinal cord injury.
Collapse
Affiliation(s)
- Yongjie Zhang
- Kentucky Spinal Cord Injury Research Center, Louisville, Kentucky, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Brodski C, Vogt Weisenhorn DM, Dechant G. Therapy of neurodegenerative diseases using neurotrophic factors: cell biological perspective. Expert Rev Neurother 2010; 2:417-26. [PMID: 19810873 DOI: 10.1586/14737175.2.3.417] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neurotrophic growth factors are a structurally diverse group of endogenous proteins, which profoundly regulate survival and multiple other cellular functions of healthy and diseased neurons. Their physiological targets include all neuronal cell types affected by neurodegenerative diseases. Initial clinical trials, which were based on strong and convincing neuroprotective effects in animal models of neurodegenerative diseases, have failed so far. In this review, we adopt a cell biological perspective to outline both the potential and problems of the therapeutic use of trophic factors in the nervous system. Emphasis is given to the functional interplay of neurotrophic factors in the regulation of cell death, as well as to the effects these proteins exert on neuronal morphology and synaptic transmission.
Collapse
Affiliation(s)
- Claude Brodski
- GSF Research Center, Clinical Neurogenetics, Ingolstädter, Landstrasse 1, Neuherberg, Germany.
| | | | | |
Collapse
|
36
|
Lim ST, Airavaara M, Harvey BK. Viral vectors for neurotrophic factor delivery: a gene therapy approach for neurodegenerative diseases of the CNS. Pharmacol Res 2009; 61:14-26. [PMID: 19840853 DOI: 10.1016/j.phrs.2009.10.002] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2009] [Revised: 10/11/2009] [Accepted: 10/11/2009] [Indexed: 01/11/2023]
Abstract
The clinical manifestation of most diseases of the central nervous system results from neuronal dysfunction or loss. Diseases such as stroke, epilepsy and neurodegeneration (e.g. Alzheimer's disease and Parkinson's disease) share common cellular and molecular mechanisms (e.g. oxidative stress, endoplasmic reticulum stress, mitochondrial dysfunction) that contribute to the loss of neuronal function. Neurotrophic factors (NTFs) are secreted proteins that regulate multiple aspects of neuronal development including neuronal maintenance, survival, axonal growth and synaptic plasticity. These properties of NTFs make them likely candidates for preventing neurodegeneration and promoting neuroregeneration. One approach to delivering NTFs to diseased cells is through viral vector-mediated gene delivery. Viral vectors are now routinely used as tools for studying gene function as well as developing gene-based therapies for a variety of diseases. Currently, many clinical trials using viral vectors in the nervous system are underway or completed, and seven of these trials involve NTFs for neurodegeneration. In this review, we discuss viral vector-mediated gene transfer of NTFs to treat neurodegenerative diseases of the central nervous system.
Collapse
Affiliation(s)
- Seung T Lim
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC 20057, United States
| | | | | |
Collapse
|
37
|
Tovar-y-Romo LB, Santa-Cruz LD, Tapia R. Experimental models for the study of neurodegeneration in amyotrophic lateral sclerosis. Mol Neurodegener 2009; 4:31. [PMID: 19619317 PMCID: PMC2720968 DOI: 10.1186/1750-1326-4-31] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2009] [Accepted: 07/20/2009] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease of unknown cause, characterized by the selective and progressive death of both upper and lower motoneurons, leading to a progressive paralysis. Experimental animal models of the disease may provide knowledge of the pathophysiological mechanisms and allow the design and testing of therapeutic strategies, provided that they mimic as close as possible the symptoms and temporal progression of the human disease. The principal hypotheses proposed to explain the mechanisms of motoneuron degeneration have been studied mostly in models in vitro, such as primary cultures of fetal motoneurons, organotypic cultures of spinal cord sections from postnatal rodents and the motoneuron-like hybridoma cell line NSC-34. However, these models are flawed in the sense that they do not allow a direct correlation between motoneuron death and its physical consequences like paralysis. In vivo, the most widely used model is the transgenic mouse that bears a human mutant superoxide dismutase 1, the only known cause of ALS. The major disadvantage of this model is that it represents about 2%-3% of human ALS. In addition, there is a growing concern on the accuracy of these transgenic models and the extrapolations of the findings made in these animals to the clinics. Models of spontaneous motoneuron disease, like the wobbler and pmn mice, have been used aiming to understand the basic cellular mechanisms of motoneuron diseases, but these abnormalities are probably different from those occurring in ALS. Therefore, the design and testing of in vivo models of sporadic ALS, which accounts for >90% of the disease, is necessary. The main models of this type are based on the excitotoxic death of spinal motoneurons and might be useful even when there is no definitive demonstration that excitotoxicity is a cause of human ALS. Despite their difficulties, these models offer the best possibility to establish valid correlations between cellular alterations and motor behavior, although improvements are still necessary in order to produce a reliable and integrative model that accurately reproduces the cellular mechanisms of motoneuron degeneration in ALS.
Collapse
Affiliation(s)
- Luis B Tovar-y-Romo
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, AP 70-253, 04510-México, D.F., México
| | - Luz Diana Santa-Cruz
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, AP 70-253, 04510-México, D.F., México
| | - Ricardo Tapia
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, AP 70-253, 04510-México, D.F., México
| |
Collapse
|
38
|
Kosuge Y, Sekikawa-Nishida K, Negi H, Ishige K, Ito Y. Characterization of chronic glutamate-mediated motor neuron toxicity in organotypic spinal cord culture prepared from ALS model mice. Neurosci Lett 2009; 454:165-9. [PMID: 19429077 DOI: 10.1016/j.neulet.2009.03.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2009] [Revised: 03/05/2009] [Accepted: 03/05/2009] [Indexed: 10/21/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease characterized by selective loss of motor neurons. Although organotypic spinal slice cultures (OSCs) exposed to inhibitors of glutamate uptake have been used as a model of ALS for screening of potentially therapeutic drugs, little development of such drugs has been achieved. In the present study we attempted to establish OSCs from G93A SOD1 transgenic mice (G93A) and to characterize the specific cell death pathway in motoneurons using glial cell line-derived neurotrophic factor (GDNF) in these mice. In the presence of GDNF, the number of surviving neurons in the OSCs was dramatically increased in both G93A and control mice. Exposure to threo-hydroxyaspartate (THA), a glutamate transport inhibitor, for 14 days induced loss of motoneurons in OSCs in G93A and control mice. In OSCs cultured with GDNF, THA-induced motoneuronal death was significantly inhibited in G93A mice, whereas that in control mice was not significantly affected. Moreover, the cleaved form of caspase-12 was increased after THA in the OSCs in G93A but not in control mice, and the activation of caspase-12 was attenuated by OSCs cultured with GDNF. These results suggest that the pathway responsible for motoneuronal death induced by THA in OSCs in G93A mice involves not only in excitotoxicity but also other mechanisms, and that the caspase-12-dependent ER stress pathway plays a role in spinal neuronal death in G93A mice. Moreover, OSCs prepared from the G93A mouse model of ALS may provide a suitable in vitro drug screening model for ALS.
Collapse
Affiliation(s)
- Yasuhiro Kosuge
- Research Unit of Pharmacology, Department of Clinical Pharmacy, College of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi-shi, Chiba 274-8555, Japan
| | | | | | | | | |
Collapse
|
39
|
Bosboom WMJ, Vrancken AFJE, van den Berg LH, Wokke JHJ, Iannaccone ST. Drug treatment for spinal muscular atrophy types II and III. Cochrane Database Syst Rev 2009:CD006282. [PMID: 19160275 DOI: 10.1002/14651858.cd006282.pub2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Spinal muscular atrophy (SMA) is caused by degeneration of anterior horn cells, which leads to progressive muscle weakness. Children with SMA type II do not develop the ability to walk without support and have a shortened life expectancy, whereas children with SMA type III develop the ability to walk and have a normal life expectancy. There are no known efficacious drug treatments that influence the disease course of SMA. OBJECTIVES To evaluate if drug treatment is able to slow or arrest the disease progression of SMA type II and III, and to assess if such therapy can be given safely. Drug treatment for SMA type I will be the topic of a separate Cochrane review. SEARCH STRATEGY We searched the Cochrane Neuromuscular Disease Group Trials Register (September 30 2008), The Cochrane Library (Issue 3, 2008), MEDLINE (January 1966 to June 2008), EMBASE (January 1980 to June 2008), ISI (January 1988 to June 2008), and ACP Journal Club (January 1991 to June 2008). SELECTION CRITERIA We sought all randomized or quasi-randomized trials that examined the efficacy of drug treatment for SMA type II and III. Participants had to fulfil the clinical criteria and, in studies including genetic analysis to confirm the diagnosis, have a deletion or mutation of the SMN1 gene (5q11.2-13.2)The primary outcome measure was to be change in disability score within one year after the onset of treatment. Secondary outcome measures within one year after the onset of treatment were to be change in muscle strength, ability to stand or walk, change in quality of life, time from the start of treatment until death or full time ventilation, and adverse events attributable to treatment during the trial period. DATA COLLECTION AND ANALYSIS Two authors independently reviewed and extracted data from all potentially relevant trials. Pooled relative risks and pooled weighted standardized mean differences were to be calculated to assess treatment efficacy MAIN RESULTS Four randomized placebo-controlled trials on treatment for SMA type II and III were found and included in the review. The treatments were creatine, phenylbutyrate, gabapentin and thyrotropin releasing hormone. None of these trials showed any effect on the outcome measures in patients with SMA type II and III. None of the patients in any of the four trials died or reached the state of full time ventilation and serious side effects were infrequent. AUTHORS' CONCLUSIONS There is no proven efficacious drug treatment for SMA type II and III.
Collapse
Affiliation(s)
- Wendy M J Bosboom
- Department of Neurology, Sint Lucas Andreas Hospital, Jan Tooropstraat 164, Amsterdam, Netherlands, 1061 AE.
| | | | | | | | | |
Collapse
|
40
|
Neve RL, Lim F. Overview of gene delivery into cells using HSV-1-based vectors. ACTA ACUST UNITED AC 2008; Chapter 4:Unit 4.12. [PMID: 18428476 DOI: 10.1002/0471142301.ns0100s06] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
This overview describes the considerations involved in the preparation and use of herpes simplex virus type 1 (HSV-1) as a vector for gene transfer into neurons. Strategies for gene delivery into neurons, either to study the molecular biology of brain function or for gene therapy, must utilize vectors that persist stably in postmitotic cells and that can be targeted both spatially and temporally in the nervous system in vivo. This unit describes the biology of HSV-1 along with a discussion covering development of amplicon and genomic HSV-1 vectors. Advantages and disadvantages of current HSV-1 vectors are presented, and HSV-1 vectors are compared with other vectors for gene transfer into neurons.
Collapse
Affiliation(s)
- R L Neve
- Harvard Medical School & McLean Hospital, Belmont, Massachusetts, USA
| | | |
Collapse
|
41
|
Antiapoptotic activity maintenance of Brain Derived Neurotrophic Factor and the C fragment of the tetanus toxin genetic fusion protein. Open Life Sci 2008. [DOI: 10.2478/s11535-008-0011-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractNeurotrophic factors have been widely suggested as a treatment for multiple diseases including motorneuron pathologies, like Amyotrophic Lateral Sclerosis. However, clinical trials in which growth factors have been systematically administered to Amyotrophic Lateral Sclerosis patients have not been effective, owing in part to the short half-life of these factors and their low concentrations at target sites. A possible strategy is the use of the atoxic C fragment of the tetanus toxin as a neurotrophic factor carrier to the motorneurons. The activity of trophic factors should be tested because their genetic fusion to proteins could alter their folding and conformation, thus undermining their neuroprotective properties. For this purpose, in this paper we explored the Brain Derived Neurotrophic Factor (BDNF) activity maintenance after genetic fusion with the C fragment of the tetanus toxin. We demonstrated that BDNF fused with the C fragment of the tetanus toxin induces the neuronal survival Akt kinase pathway in mouse cortical culture neurons and maintains its antiapoptotic neuronal activity in Neuro2A cells.
Collapse
|
42
|
Suzuki M, Svendsen CN. Combining growth factor and stem cell therapy for amyotrophic lateral sclerosis. Trends Neurosci 2008; 31:192-8. [PMID: 18329734 DOI: 10.1016/j.tins.2008.01.006] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2007] [Revised: 01/18/2008] [Accepted: 01/21/2008] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease where motor neurons within the brain and spinal cord are lost, leading to paralysis and death. Certain growth factors should, in principle, be able to protect dying motor neurons. However, targeted delivery to the spinal cord or brain has been a constant problem. There is also accumulating evidence that glial cells might play a crucial role in maintaining motor neuron function and survival in ALS. Stem cells isolated and expanded in culture can be modified to release growth factors and generate glial cells following transplantation into the spinal cord or brain. As such, they might be able to both detoxify the local environment around dying motor neurons and deliver trophic factors. Here we examine the feasibility of translating these findings into new treatments for ALS patients.
Collapse
Affiliation(s)
- Masatoshi Suzuki
- The Waisman Center and Departments of Anatomy and Neurology, University of Wisconsin-Madison, Madison, WI 53707-2280, USA
| | | |
Collapse
|
43
|
Hedlund E, Hefferan MP, Marsala M, Isacson O. REVIEW ARTILCE: Cell therapy and stem cells in animal models of motor neuron disorders. Eur J Neurosci 2007; 26:1721-37. [PMID: 17897390 DOI: 10.1111/j.1460-9568.2007.05780.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS), spinal bulbar muscular atrophy (or Kennedy's disease), spinal muscular atrophy and spinal muscular atrophy with respiratory distress 1 are neurodegenerative disorders mainly affecting motor neurons and which currently lack effective therapies. Recent studies in animal models as well as primary and embryonic stem cell models of ALS, utilizing over-expression of mutated forms of Cu/Zn superoxide dismutase 1, have shown that motor neuron degeneration in these models is in part a non cell-autonomous event and that by providing genetically non-compromised supporting cells such as microglia or growth factor-excreting cells, onset can be delayed and survival increased. Using models of acute motor neuron injury it has been shown that embryonic stem cell-derived motor neurons implanted into the spinal cord can innervate muscle targets and improve functional recovery. Thus, a rationale exists for the development of cell therapies in motor neuron diseases aimed at either protecting and/or replacing lost motor neurons, interneurons as well as non-neuronal cells. This review evaluates approaches used in animal models of motor neuron disorders and their therapeutic relevance.
Collapse
Affiliation(s)
- Eva Hedlund
- Neuroregeneration Laboratory, Center for Neuroregeneration Research, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA.
| | | | | | | |
Collapse
|
44
|
Intraparenchymal spinal cord delivery of adeno-associated virus IGF-1 is protective in the SOD1G93A model of ALS. Brain Res 2007; 1185:256-65. [PMID: 17963733 DOI: 10.1016/j.brainres.2007.09.034] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2007] [Revised: 09/04/2007] [Accepted: 09/05/2007] [Indexed: 12/13/2022]
Abstract
The potent neuroprotective activities of neurotrophic factors, including insulin-like growth factor 1 (IGF-1), make them promising candidates for treatment of amyotrophic lateral sclerosis (ALS). In an effort to maximize rate of motor neuron transduction, achieve high levels of spinal IGF-1 and thus enhance therapeutic benefit, we injected an adeno-associated virus 2 (AAV2)-based vector encoding human IGF-1 (CERE-130) into lumbar spinal cord parenchyma of SOD1(G93A) mice. We observed robust and long-term intraspinal IGF-1 expression and partial rescue of lumbar spinal cord motor neurons, as well as sex-specific delayed disease onset, weight loss, decline in hindlimb grip strength and increased animal survival.
Collapse
|
45
|
Schaefer MKE, Schmalbruch H, Buhler E, Lopez C, Martin N, Guénet JL, Haase G. Progressive motor neuronopathy: a critical role of the tubulin chaperone TBCE in axonal tubulin routing from the Golgi apparatus. J Neurosci 2007; 27:8779-89. [PMID: 17699660 PMCID: PMC6672183 DOI: 10.1523/jneurosci.1599-07.2007] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Axonal degeneration represents one of the earliest pathological features in motor neuron diseases. We here studied the underlying molecular mechanisms in progressive motor neuronopathy (pmn) mice mutated in the tubulin-specific chaperone TBCE. We demonstrate that TBCE is a peripheral membrane-associated protein that accumulates at the Golgi apparatus. In pmn mice, TBCE is destabilized and disappears from the Golgi apparatus of motor neurons, and microtubules are lost in distal axons. The axonal microtubule loss proceeds retrogradely in parallel with the axonal dying back process. These degenerative changes are inhibited in a dose-dependent manner by transgenic TBCE complementation that restores TBCE expression at the Golgi apparatus. In cultured motor neurons, the pmn mutation, interference RNA-mediated TBCE depletion, and brefeldin A-mediated Golgi disruption all compromise axonal tubulin routing. We conclude that motor axons critically depend on axonal tubulin routing from the Golgi apparatus, a process that involves TBCE and possibly other tubulin chaperones.
Collapse
Affiliation(s)
- Michael K. E. Schaefer
- Inserm, Unité 29, Equipe Avenir, 13273 Marseille, France
- Aix Marseille Université, Institut de Neurobiologie de la Méditerranée, 13284 Marseille, France
| | | | - Emmanuelle Buhler
- Inserm, Unité 29, Equipe Avenir, 13273 Marseille, France
- Aix Marseille Université, Institut de Neurobiologie de la Méditerranée, 13284 Marseille, France
| | - Catherine Lopez
- Inserm, Unité 29, Equipe Avenir, 13273 Marseille, France
- Aix Marseille Université, Institut de Neurobiologie de la Méditerranée, 13284 Marseille, France
| | | | | | - Georg Haase
- Inserm, Unité 29, Equipe Avenir, 13273 Marseille, France
- Aix Marseille Université, Institut de Neurobiologie de la Méditerranée, 13284 Marseille, France
| |
Collapse
|
46
|
Nayak MS, Kim YS, Goldman M, Keirstead HS, Kerr DA. Cellular therapies in motor neuron diseases. Biochim Biophys Acta Mol Basis Dis 2006; 1762:1128-38. [PMID: 16872810 DOI: 10.1016/j.bbadis.2006.06.004] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2006] [Revised: 05/28/2006] [Accepted: 06/08/2006] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) and spinal muscular atrophy (SMA) are prototypical motor neuron diseases that result in progressive weakness as a result of motor neuron dysfunction and death. Though much work has been done in both diseases to identify the cellular mechanisms of motor neuron dysfunction, once motor neurons have died, one of potential therapies to restore function would be through the use of cellular transplantation. In this review, we discuss potential strategies whereby cellular therapies, including the use of stem cells, neural progenitors and cells engineered to secrete trophic factors, may be used in motor neuron diseases. We review pre-clinical data in rodents with each of these approaches and discuss advances and regulatory issues regarding the use of cellular therapies in human motor neuron diseases.
Collapse
Affiliation(s)
- Mamatha S Nayak
- Department of Neurology, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Baltimore, MD 21287, USA
| | | | | | | | | |
Collapse
|
47
|
DAS M, Bhargava N, Gregory C, Riedel L, Molnar P, Hickman JJ. Adult rat spinal cord culture on an organosilane surface in a novel serum-free medium. In Vitro Cell Dev Biol Anim 2006; 41:343-8. [PMID: 16448224 DOI: 10.1007/s11626-005-0006-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
In this study, we have documented by morphological analysis, immunocytochemistry, and electrophysiology, the development of a culture system that promotes the growth and long-term survival of dissociated adult rat spinal cord neurons. This system comprises a patternable, nonbiological, cell growth-promoting organosilane substrate coated on a glass surface and an empirically derived novel serum-free medium, supplemented with specific growth factors (acidic fibroblast growth factor, heparin sulfate, neurotrophin-3, brain-derived neurotrophic factor, glial-derived neurotrophic factor, cardiotrophin-1, and vitronectin). Neurons were characterized by immunoreactivity for neurofilament 150, neuron-specific enolase, Islet-1 antibodies, electrophysiology, and the cultures were maintained for 4-6 wk. This culture system could be a useful tool for the study of adult mammalian spinal neurons in a functional in vitro system.
Collapse
Affiliation(s)
- Mainak DAS
- Nanoscience Technology Center, University of Central Florida, Orlando, Florida 32826, USA
| | | | | | | | | | | |
Collapse
|
48
|
Ekestern E. Neurotrophic factors and amyotrophic lateral sclerosis. NEURODEGENER DIS 2006; 1:88-100. [PMID: 16908980 DOI: 10.1159/000080049] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2003] [Accepted: 03/22/2004] [Indexed: 12/15/2022] Open
Abstract
The cause of motor neuron death in amyotrophic lateral sclerosis (ALS) remains a mystery. Initial implications of neurotrophic factor impairment involved in disease progression causing selective motor neuron death were brought forward in the late 1980s. These implications were based on several in vitro studies of motor neuron cultures in which a near to complete rescue of axotomized neonatal motor neurons in the presence of supplementary neurotrophic factors were revealed. These findings pawed the way for extensive investigations in experimental animal models of ALS. Neurotrophic factor administration in rodent ALS models demonstrated a remarkable effect on survival of degenerating motor neurons and rescue of axotomized motor neurons, both in vivo and in vitro. In the absence of efficient therapy for ALS, some of these promising neurotrophic factors have been administered to groups of ALS patients, as they appeared available for clinical trials. Up to date, none of tested factors has lived up to expectations, altering the outcome of the disease. This review summarizes current findings on neurotrophic factor expression in ALS tissue and these factors' potential/debatable clinical relevance to ALS and the treatment of ALS. It also discusses possible interventions improving clinical trial design to obtain efficacy of neurotrophic factor treatment in patients suffering from ALS.
Collapse
Affiliation(s)
- Eva Ekestern
- Department of Neuroimmunology, Brain Research Institute, University of Vienna, Austria.
| |
Collapse
|
49
|
Fabre EE, Bigey P, Orsini C, Scherman D. Comparison of promoter region constructs for in vivo intramuscular expression. J Gene Med 2006; 8:636-45. [PMID: 16477670 DOI: 10.1002/jgm.878] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND High transgene expression is generally expected after gene transfer. However, different level, kinetics and localization of expression might be needed for relevant therapeutic applications. Former studies have compared various promoter regions driving gene expression leading to conflicting results. In the present work, two promoter families have been compared using the efficient in vivo intramuscular electrotransfer technique. METHODS Three promoter regions were constructed by associating the strong ubiquitous cytomegalovirus (CMV) enhancer-promoter to its homologous intron A or to a heterologous intron, or to a hybrid intron. Promoter regions derived from the muscle creatine kinase (MCK) promoter were also studied. The expression of the same transgene (SeAP or neurotrophin-3) under control of these different promoters was compared after plasmid electrotransfer in mouse tibialis-cranialis skeletal muscle. RESULTS Heterologous intron association to the CMV promoter did not modify gene expression kinetics nor increase gene expression level. Usefulness of intron A or hybrid intron association to the CMV promoter depended on the gene. The various MCK promoters drove efficient gene expression but lower than that obtained with the CMV promoter. Furthermore, peak value was reached earlier with MCK promoter regions (14 days). CONCLUSION For applications of gene transfer restricted to skeletal muscle, the MCK promoter or a MCK promoter variant would be a promising alternative to the CMV promoter. Indeed, it has been demonstrated that the use of MCK promoter limits humoral and cell-mediated immune responses. Furthermore, the MCK promoter decreases the initial expression peak that may be detrimental, drives a sustained gene expression, and improves gene transfer safety.
Collapse
|
50
|
Maier A, Zhou Z, Bornemann A. The expression profile of myogenic transcription factors in satellite cells from denervated rat muscle. Brain Pathol 2006; 12:170-7. [PMID: 11958370 PMCID: PMC8095746 DOI: 10.1111/j.1750-3639.2002.tb00431.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
The muscle-specific transcription factors of the MyoD family are altered after denervation. In order to determine whether this shift takes place in satellite cells (SC), we investigated the expression pattern of MyoD, myf5, myogenin, and MRF4 in SC. Hindlimb muscles of rats were denervated for 2 days to 4 weeks. SC were isolated, pooled and the transcription of all 4 factors was assessed by RT-PCR. Protein expression was assessed in histological sections of soleus and anterior tibial (TA) muscles; SC were identified by M-cadherin. Pooled SC from innervated muscles expressed myf5 mRNA, and very weakly MyoD and myogenin mRNA. MyoD and myogenin protein was found in only few SC. After denervation, pooled SC expressed myf5 mRNA, and very weakly myogenin and MRF4 mRNA. Myogenin protein was found in less than about 10% of the cells, whereas MRF4 protein was absent from SC. We conclude that the presence of myf5 and the absence of MyoD and MRF4 protein in SC after denervation indicated the quiescent state of the cell cycle. A subset of SC has additionally acquired myogenin. SC after denervation might be less easily recruited into the mitotic cycle than SC from normal muscle, rendering regeneration of denervated muscle less efficient than normal muscle.
Collapse
Affiliation(s)
- Annette Maier
- Institute of Brain Research, University of Tübingen, Germany
| | - Zhe Zhou
- Institute of Brain Research, University of Tübingen, Germany
| | - Antje Bornemann
- Institute of Brain Research, University of Tübingen, Germany
| |
Collapse
|