51
|
Contingent intramuscular boosting of P2XR7 axis improves motor function in transgenic ALS mice. Cell Mol Life Sci 2021; 79:7. [PMID: 34936028 PMCID: PMC8695421 DOI: 10.1007/s00018-021-04070-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 11/06/2022]
Abstract
Amyotrophic lateral sclerosis is a fatal neurodegenerative disorder that leads to progressive degeneration of motor neurons and severe muscle atrophy without effective treatment. Most research on the disease has been focused on studying motor neurons and supporting cells of the central nervous system. Strikingly, the recent observations have suggested that morpho-functional alterations in skeletal muscle precede motor neuron degeneration, bolstering the interest in studying muscle tissue as a potential target for the delivery of therapies. We previously showed that the systemic administration of the P2XR7 agonist, 2′(3′)-O‐(4-benzoylbenzoyl) adenosine 5-triphosphate (BzATP), enhanced the metabolism and promoted the myogenesis of new fibres in the skeletal muscles of SOD1G93A mice. Here we further corroborated this evidence showing that intramuscular administration of BzATP improved the motor performance of ALS mice by enhancing satellite cells and the muscle pro-regenerative activity of infiltrating macrophages. The preservation of the skeletal muscle retrogradely propagated along with the motor unit, suggesting that backward signalling from the muscle could impinge on motor neuron death. In addition to providing the basis for a suitable adjunct multisystem therapeutic approach in ALS, these data point out that the muscle should be at the centre of ALS research as a target tissue to address novel therapies in combination with those oriented to the CNS.
Collapse
|
52
|
Cudkowicz ME, Lindborg SR, Goyal NA, Miller RG, Burford MJ, Berry JD, Nicholson KA, Mozaffar T, Katz JS, Jenkins LJ, Baloh RH, Lewis RA, Staff N, Owegi MA, Berry DA, Gothelf Y, Levy YS, Aricha R, Kern RZ, Windebank AJ, Brown RH. A Randomized Placebo-Controlled Phase 3 Study of Mesenchymal stem cells induced to secrete high levels of neurotrophic factors in Amyotrophic Lateral Sclerosis. Muscle Nerve 2021; 65:291-302. [PMID: 34890069 PMCID: PMC9305113 DOI: 10.1002/mus.27472] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/04/2021] [Accepted: 12/07/2021] [Indexed: 11/17/2022]
Abstract
Introduction/Aims Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative illness with great unmet patient need. We aimed to evaluate whether mesenchymal stem cells induced to secrete high levels of neurotrophic factors (MSC‐NTF), a novel autologous cell‐therapy capable of targeting multiple pathways, could safely slow ALS disease progression. Methods This randomized, double‐blind, placebo‐controlled study enrolled ALS participants meeting revised El Escorial criteria, revised ALS Functional Rating Scale (ALSFRS‐R) ≥25 (screening) and ≥3 ALSFRS‐R points decline prior to randomization. Participants received three treatments of MSC‐NTF or placebo intrathecally. The primary endpoint evaluated efficacy of MSC‐NTF through a responder analysis and safety. A change in disease progression post‐treatment of ≥1.25 points/mo defines a clinical response. A pre‐specified analysis leveraged baseline ALSFRS‐R of 35 as a subgroup threshold. Results Overall, MSC‐NTF treatment was well tolerated; there were no safety concerns. Thirty‐three percent of MSC‐NTF and 28% of placebo participants met clinical response criteria at 28 wk (odds ratio [OR] = 1.33, P = .45); thus, the primary endpoint was not met. A pre‐specified analysis of participants with baseline ALSFRS‐R ≥ 35 (n = 58) showed a clinical response rate at 28 wk of 35% MSC‐NTF and 16% placebo (OR = 2.6, P = .29). Significant improvements in cerebrospinal biomarkers of neuroinflammation, neurodegeneration, and neurotrophic factor support were observed with MSC‐NTF, with placebo unchanged. Discussion The study did not reach statistical significance on the primary endpoint. However, a pre‐specified subgroup suggests that MSC‐NTF participants with less severe disease may have retained more function compared to placebo. Given the unmet patient need, the results of this trial warrant further investigation.
Collapse
Affiliation(s)
- Merit E Cudkowicz
- Healey Center, Mass General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Stacy R Lindborg
- Research and Development, Brainstorm Cell Therapeutics, New York, New York
| | - Namita A Goyal
- UCI Health ALS & Neuromuscular Center, University of California, Irvine, Irvine, California
| | - Robert G Miller
- Sutter Pacific Medical Foundation, California Pacific Medical Center, San Francisco, California
| | - Matthew J Burford
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, California
| | - James D Berry
- Healey Center, Mass General Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Tahseen Mozaffar
- UCI Health ALS & Neuromuscular Center, University of California, Irvine, Irvine, California
| | - Jonathan S Katz
- Sutter Pacific Medical Foundation, California Pacific Medical Center, San Francisco, California
| | - Liberty J Jenkins
- Sutter Pacific Medical Foundation, California Pacific Medical Center, San Francisco, California
| | - Robert H Baloh
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Richard A Lewis
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, California
| | - NathanP Staff
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Margaret A Owegi
- Neurology Department, University of Massachusetts Medical School, Boston, Massachusetts
| | | | - Yael Gothelf
- Research and Development, Brainstorm Cell Therapeutics, Israel
| | | | - Revital Aricha
- Research and Development, Brainstorm Cell Therapeutics, New York, New York
| | - Ralph Z Kern
- Research and Development, Brainstorm Cell Therapeutics, New York, New York
| | - Anthony J Windebank
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Robert H Brown
- Neurology Department, University of Massachusetts Medical School, Boston, Massachusetts
| |
Collapse
|
53
|
Cocozza G, Garofalo S, Capitani R, D’Alessandro G, Limatola C. Microglial Potassium Channels: From Homeostasis to Neurodegeneration. Biomolecules 2021; 11:1774. [PMID: 34944418 PMCID: PMC8698630 DOI: 10.3390/biom11121774] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/22/2021] [Accepted: 11/23/2021] [Indexed: 12/21/2022] Open
Abstract
The growing interest in the role of microglia in the progression of many neurodegenerative diseases is developing in an ever-expedited manner, in part thanks to emergent new tools for studying the morphological and functional features of the CNS. The discovery of specific biomarkers of the microglia phenotype could find application in a wide range of human diseases, and creates opportunities for the discovery and development of tailored therapeutic interventions. Among these, recent studies highlight the pivotal role of the potassium channels in regulating microglial functions in physiological and pathological conditions such as Alzheimer's Disease, Parkinson's Disease, and Amyotrophic Lateral Sclerosis. In this review, we summarize the current knowledge of the involvement of the microglial potassium channels in several neurodegenerative diseases and their role as modulators of microglial homeostasis and dysfunction in CNS disorders.
Collapse
Affiliation(s)
- Germana Cocozza
- Instituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, 86077 Pozzilli, Italy; (G.C.); (G.D.)
| | - Stefano Garofalo
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy; (S.G.); (R.C.)
| | - Riccardo Capitani
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy; (S.G.); (R.C.)
| | - Giuseppina D’Alessandro
- Instituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, 86077 Pozzilli, Italy; (G.C.); (G.D.)
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy; (S.G.); (R.C.)
| | - Cristina Limatola
- Instituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, 86077 Pozzilli, Italy; (G.C.); (G.D.)
- Department of Physiology and Pharmacology, Laboratory Affiliated to Istituto Pasteur Italia, Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
54
|
Izrael M, Molakandov K, Revel A, Slutsky SG, Sonnenfeld T, Weiss JM, Revel M. Astrocytes Downregulate Inflammation in Lipopolysaccharide-Induced Acute Respiratory Distress Syndrome: Applicability to COVID-19. Front Med (Lausanne) 2021; 8:740071. [PMID: 34778302 PMCID: PMC8585990 DOI: 10.3389/fmed.2021.740071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/27/2021] [Indexed: 11/13/2022] Open
Abstract
Background: An acute respiratory distress syndrome (ARDS) is caused by the increased amounts of pro-inflammatory cytokines and neutrophil-mediated tissue injury. To date, there is no effective treatment for the ARDS available, while the need for one is growing due to the most severe complications of the current coronavirus disease-2019 (COVID-19) pandemic. The human astrocytes (AstroRx) have shown immunomodulatory properties in the central nervous system (CNS). This study aimed to evaluate the capacity of astrocytes to decrease lung inflammation and to be applied as a treatment therapy in ARDS. Methods: First, we assessed the ability of clinical-grade AstroRx to suppress T-cell proliferation in a mixed lymphocyte reaction test. Next, we tested the therapeutical potential of AstroRx cells in a lipopolysaccharide (LPS)-based ARDS mouse model by injecting AstroRx intravenously (i.v). We determined the degree of lung injury by using a severity scoring scale of 0–2, based on the American Thoracic Society. The scoring measured the presence of neutrophils, fibrin deposits, and the thickening of alveolar walls. The state of inflammation was further assessed by quantifying the immune-cell infiltration to the bronchoalveolar lavage fluid (BALF) and by the presence of proinflammatory cytokines and chemokines in the BALF and serum. Results: We detected that AstroRx cells were capable to suppress T-cell proliferation in vitro after exposure to the mitogen concanavalin A (ConA). In vivo, AstroRx cells were able to lower the degree of lung injury in LPS-treated animals compared with the sham injected animals (P = 0.039). In this study, 30% of AstroRx treated mice showed no lung lesions (responder mice), these mice presented a steady number of eosinophils, T cells, and neutrophils comparable with the level of naïve control mice. The inflammatory cytokines and chemokines, such as TNFα, IL1b, IL-6, and CXCL1, were also kept in check in responder AstroRx-treated mice and were not upregulated as in the sham-injected mice (P < 0.05). As a result, the LPS-treated ARDS mice had a higher survival rate when they were treated with AstroRx. Conclusions: Our results demonstrate that the immunosuppressive activity of AstroRx cells support the application of AstroRx cells as a cell therapy treatment for ARDS. The immunoregulatory activity may also be a part of the mechanism of action of AstroRx reported in the amyotrophic lateral sclerosis (ALS) neurodegenerative disease.
Collapse
Affiliation(s)
- Michal Izrael
- Research and Development Department at Kadimastem Ltd, Nes-Ziona, Israel
| | - Kfir Molakandov
- Research and Development Department at Kadimastem Ltd, Nes-Ziona, Israel
| | - Ariel Revel
- Research and Development Department at Kadimastem Ltd, Nes-Ziona, Israel
| | - Shalom Guy Slutsky
- Research and Development Department at Kadimastem Ltd, Nes-Ziona, Israel
| | - Tehila Sonnenfeld
- Research and Development Department at Kadimastem Ltd, Nes-Ziona, Israel
| | - Julia Miriam Weiss
- Research and Development Department at Kadimastem Ltd, Nes-Ziona, Israel
| | - Michel Revel
- Research and Development Department at Kadimastem Ltd, Nes-Ziona, Israel.,Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
55
|
Novel P2X7 Antagonist Ameliorates the Early Phase of ALS Disease and Decreases Inflammation and Autophagy in SOD1-G93A Mouse Model. Int J Mol Sci 2021; 22:ijms221910649. [PMID: 34638992 PMCID: PMC8508678 DOI: 10.3390/ijms221910649] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/21/2021] [Accepted: 09/28/2021] [Indexed: 12/18/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a disease with a resilient neuroinflammatory component caused by activated microglia and infiltrated immune cells. How to successfully balance neuroprotective versus neurotoxic actions through the use of anti-inflammatory agents is still under debate. There has been a boost of awareness regarding the role of extracellular ATP and purinergic receptors in modulating the physiological and pathological mechanisms in the nervous system. Particularly in ALS, it is known that the purinergic ionotropic P2X7 receptor plays a dual role in disease progression by acting at different cellular and molecular levels. In this context, we previously demonstrated that the P2X7 receptor antagonist, brilliant blue G, reduces neuroinflammation and ameliorates some of the pathological features of ALS in the SOD1-G93A mouse model. Here, we test the novel, noncommercially available, and centrally permeant Axxam proprietary P2X7 antagonist, AXX71, in SOD1-G93A mice, by assessing some behavioral and molecular parameters, among which are disease progression, survival, gliosis, and motor neuron wealth. We demonstrate that AXX71 affects the early symptomatic phase of the disease by reducing microglia-related proinflammatory markers and autophagy without affecting the anti-inflammatory markers or motor neuron survival. Our results suggest that P2X7 modulation can be further investigated as a therapeutic strategy in preclinical studies, and exploited in ALS clinical trials.
Collapse
|
56
|
Vucic S, Henderson RD, Mathers S, Needham M, Schultz D, Kiernan MC. Safety and efficacy of dimethyl fumarate in ALS: randomised controlled study. Ann Clin Transl Neurol 2021; 8:1991-1999. [PMID: 34477330 PMCID: PMC8528453 DOI: 10.1002/acn3.51446] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/02/2021] [Accepted: 08/03/2021] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVE Neuroinflammation is an important pathogenic mechanism in amyotrophic lateral sclerosis (ALS), with regulatory T cells (Tregs) mediating a slower rate of disease progression. Dimethyl fumarate enhances Treg levels and suppresses pro-inflammatory T cells. The present study assessed the safety and efficacy of dimethyl fumarate in ALS. METHODS Phase-2, double-blind, placebo-controlled randomised clinical trial recruited participants from May 1, 2018 to September 25, 2019, across six Australian sites. Participants were randomised (2:1 ratio) to dimethyl fumarate (480 mg/day) or matching placebo, completing visits at screening, baseline, weeks 12, 24 and 36. The primary efficacy endpoint was a change in Amyotrophic Lateral Sclerosis Functional Rating Scale-Revised (ALSFRS-R) at week 36. Secondary outcome measures included survival, neurophysiological index (NI), respiratory function, urinary neurotrophin-receptor p75 and quality of life. RESULTS A total of 107 participants were randomised to dimethyl fumarate (n = 72) or placebo (n = 35). ALSFRS-R score was not significantly different at week 36 (-1.12 [-3.75 to 1.52, p = 0.41]). Dimethyl fumarate was associated with a reduced NI decline week 36 (differences in the least-squares mean: (0.84 [-0.51 to 2.22, p = 0.22]). There were no significant differences in other secondary outcome measures. Safety profiles were comparable between groups. INTERPRETATION Dimethyl fumarate, in combination with riluzole, was safe and well-tolerated in ALS. There was no significant improvement in the primary endpoint. The trial provides class I evidence for safety and lack of efficacy of dimethyl fumarate in ALS.
Collapse
Affiliation(s)
- Steve Vucic
- Brain and Nerve Research Centre, Concord Clinical School, Concord Hospital, University of Sydney, Sydney, Australia
| | - Robert D Henderson
- Department of Neurology, Royal Brisbane & Women's Hospital, Brisbane, Australia
| | - Susan Mathers
- Department of Neurology, Calvary Health Care Bethlehem, Caulfield South, Australia
| | - Merrilee Needham
- Department of Neurology, Fiona Stanley Hospital, Murdoch University, Murdoch, Australia.,Perron Institute for Neurological and Translational Science, Nedlands, Australia.,University of Notre Dame, Fremantle, Australia
| | | | | | | |
Collapse
|
57
|
Wei QQ, Hou YB, Zhang LY, Ou RW, Cao B, Chen YP, Shang HF. Neutrophil-to-lymphocyte ratio in sporadic amyotrophic lateral sclerosis. Neural Regen Res 2021; 17:875-880. [PMID: 34472488 PMCID: PMC8530123 DOI: 10.4103/1673-5374.322476] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The neutrophil-to-lymphocyte ratio (NLR) is considered a robust prognostic biomarker for predicting patient survival outcomes in many diseases. However, it remains unclear whether it can be used as a biomarker for amyotrophic lateral sclerosis (ALS). To correlate NLR with disease progression and survival in sporadic ALS, 1030 patients with ALS between January 2012 and December 2018 were included in this study. These patients were assigned into three groups according to their NLR values: Group 1 (NLR < 2, n = 544 [52.8%]), Group 2 (NLR = 2-3, n = 314 [30.5%]), and Group 3 (NLR > 3, n = 172 [16.7%]). All patients were followed up until April 2020. Patients in Group 3 had a significantly older onset age, a lower score on the Revised ALS Functional Rating Scale, and rapidly progressing disease conditions. Furthermore, faster disease progression rates were associated with higher NLR values (odds ratio = 1.211, 95% confidence interval [CI]: 1.090-1.346, P < 0.001) after adjusting for other risk factors. Compared with Groups 1 and 2, the survival time in Group 3 was significantly shorter (log-rank P = 0.002). The NLR value was considered an independent parameter for the prediction of survival in ALS patients after normalizing for all other potential parameters (hazard ratio [HR] = 1.079, 95% CI: 1.016-1.146, P = 0.014). The effects on ALS survival remained significant when adjusted for treatment (HR = 1.074, 95% CI: 1.012-1.141, Ptrend = 0.019) or when considering the stratified NLR value (HR = 1.115, 95% CI: 1.009-1.232, Ptrend = 0.033). Thus, the NLR may help to predict the rate of disease progression and survival in patients with sporadic ALS. The study was approved by the Institutional Ethics Committee of West China Hospital of Sichuan University, China (approval No. 2015 (236)) on December 23, 2015.
Collapse
Affiliation(s)
- Qian-Qian Wei
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yan-Bing Hou
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ling-Yu Zhang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ru-Wei Ou
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Bei Cao
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yong-Ping Chen
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Hui-Fang Shang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
58
|
Higgins NR, Greenslade JE, Wu JJ, Miranda E, Galliciotti G, Monteiro MJ. Serpin neuropathology in the P497S UBQLN2 mouse model of ALS/FTD. Brain Pathol 2021; 31:e12948. [PMID: 33780087 PMCID: PMC8387369 DOI: 10.1111/bpa.12948] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 02/17/2021] [Accepted: 03/08/2021] [Indexed: 01/12/2023] Open
Abstract
Accumulating evidence suggests X-linked dominant mutations in UBQLN2 cause amyotrophic lateral sclerosis (ALS) with frontotemporal dementia (FTD) through both loss- and gain-of-function mechanisms. However, the mechanisms by which the mutations cause disease are still unclear. The goal of the study was to uncover the possible pathomechanism(s) by which UBQLN2 mutations cause ALS/FTD. An analysis of proteomic changes in neuronal tissue was used to identify proteins with altered accumulation in the P497S UBQLN2 transgenic mouse model of ALS/FTD. We then used immunocytochemistry and biochemical techniques to confirm protein changes in the mutant P497S mice. Additionally, we used cell lines inactivated of UBQLN2 expression to determine whether its loss underlies the alteration in the proteins seen in P497S mice. The proteome screen identified a dramatic alteration of serine protease inhibitor (serpin) proteins in the mutant P497S animals. Double immunofluorescent staining of brain and spinal cord tissues of the mutant and control mice revealed an age-dependent change in accumulation of Serpin A1, C1, and I1 in puncta whose staining colocalized with UBQLN2 puncta in the mutant P497S mice. Serpin A1 aggregation in P497S animals was confirmed by biochemical extraction and filter retardation assays. A similar phenomenon of serpin protein aggregation was found in HeLa and NSC34 motor neuron cells with inactivated UBQLN2 expression. We found aberrant aggregation of serpin proteins, particularly Serpin A1, in the brain and spinal cord of the P497S UBQLN2 mouse model of ALS/FTD. Similar aggregation of serpin proteins was found in UBQLN2 knockout cells suggesting that serpin aggregation in the mutant P497S animals may stem from loss of UBQLN2 function. Because serpin aggregation is known to cause disease through both loss- and gain-of-function mechanisms, we speculate that their accumulation in the P497S mouse model of ALS/FTD may contribute to disease pathogenesis through similar mechanism(s).
Collapse
Affiliation(s)
- Nicole R. Higgins
- Program in Molecular MedicineUniversity of Maryland School of MedicineBaltimoreMDUSA
- Center for Biomedical Engineering and TechnologyDepartment of Anatomy and NeurobiologyUniversity of Maryland School of MedicineBaltimoreMDUSA
| | - Jessie E. Greenslade
- Center for Biomedical Engineering and TechnologyDepartment of Anatomy and NeurobiologyUniversity of Maryland School of MedicineBaltimoreMDUSA
| | - Josephine J. Wu
- Center for Biomedical Engineering and TechnologyDepartment of Anatomy and NeurobiologyUniversity of Maryland School of MedicineBaltimoreMDUSA
| | - Elena Miranda
- Department of Biology and Biotechnologies ‘Charles Darwin’Pasteur Institute – Cenci Bolognetti FoundationSapienza University of RomeRomeItaly
| | - Giovanna Galliciotti
- Institute of NeuropathologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Mervyn J. Monteiro
- Program in Molecular MedicineUniversity of Maryland School of MedicineBaltimoreMDUSA
- Center for Biomedical Engineering and TechnologyDepartment of Anatomy and NeurobiologyUniversity of Maryland School of MedicineBaltimoreMDUSA
| |
Collapse
|
59
|
Xu X, Shen D, Gao Y, Zhou Q, Ni Y, Meng H, Shi H, Le W, Chen S, Chen S. A perspective on therapies for amyotrophic lateral sclerosis: can disease progression be curbed? Transl Neurodegener 2021; 10:29. [PMID: 34372914 PMCID: PMC8353789 DOI: 10.1186/s40035-021-00250-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 07/09/2021] [Indexed: 01/17/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease involving both upper and lower motor neurons, leading to paralysis and eventually death. Symptomatic treatments such as inhibition of salivation, alleviation of muscle cramps, and relief of spasticity and pain still play an important role in enhancing the quality of life. To date, riluzole and edaravone are the only two drugs approved by the Food and Drug Administration for the treatment of ALS in a few countries. While there is adequate consensus on the modest efficacy of riluzole, there are still open questions concerning the efficacy of edaravone in slowing the disease progression. Therefore, identification of novel therapeutic strategies is urgently needed. Impaired autophagic process plays a critical role in ALS pathogenesis. In this review, we focus on therapies modulating autophagy in the context of ALS. Furthermore, stem cell therapies, gene therapies, and newly-developed biomaterials have great potentials in alleviating neurodegeneration, which might halt the disease progression. In this review, we will summarize the current and prospective therapies for ALS.
Collapse
Affiliation(s)
- Xiaojiao Xu
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China.,Institute of Neurology, Sichuan Academy of Medical Sciences-Sichuan Provincial Hospital, Chengdu, 610031, China
| | - Dingding Shen
- Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200020, China
| | - Yining Gao
- Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200020, China
| | - Qinming Zhou
- Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200020, China
| | - You Ni
- Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200020, China
| | - Huanyu Meng
- Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200020, China
| | - Hongqin Shi
- Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200020, China.,Department of Neurology, Xinrui Hospital, Wuxi, 214028, China
| | - Weidong Le
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China. .,Institute of Neurology, Sichuan Academy of Medical Sciences-Sichuan Provincial Hospital, Chengdu, 610031, China. .,Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China.
| | - Shengdi Chen
- Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200020, China.
| | - Sheng Chen
- Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200020, China.
| |
Collapse
|
60
|
De Marchi F, Munitic I, Amedei A, Berry JD, Feldman EL, Aronica E, Nardo G, Van Weehaeghe D, Niccolai E, Prtenjaca N, Sakowski SA, Bendotti C, Mazzini L. Interplay between immunity and amyotrophic lateral sclerosis: Clinical impact. Neurosci Biobehav Rev 2021; 127:958-978. [PMID: 34153344 PMCID: PMC8428677 DOI: 10.1016/j.neubiorev.2021.06.027] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 05/07/2021] [Accepted: 06/17/2021] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a debilitating and rapidly fatal neurodegenerative disease. Despite decades of research and many new insights into disease biology over the 150 years since the disease was first described, causative pathogenic mechanisms in ALS remain poorly understood, especially in sporadic cases. Our understanding of the role of the immune system in ALS pathophysiology, however, is rapidly expanding. The aim of this manuscript is to summarize the recent advances regarding the immune system involvement in ALS, with particular attention to clinical translation. We focus on the potential pathophysiologic mechanism of the immune system in ALS, discussing local and systemic factors (blood, cerebrospinal fluid, and microbiota) that influence ALS onset and progression in animal models and people. We also explore the potential of Positron Emission Tomography to detect neuroinflammation in vivo, and discuss ongoing clinical trials of therapies targeting the immune system. With validation in human patients, new evidence in this emerging field will serve to identify novel therapeutic targets and provide realistic hope for personalized treatment strategies.
Collapse
Affiliation(s)
- Fabiola De Marchi
- Department of Neurology and ALS Centre, University of Piemonte Orientale, Maggiore Della Carità Hospital, Corso Mazzini 18, Novara, 28100, Italy
| | - Ivana Munitic
- Laboratory for Molecular Immunology, Department of Biotechnology, University of Rijeka, R. Matejcic 2, 51000, Rijeka, Croatia
| | - Amedeo Amedei
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - James D Berry
- Sean M. Healey & AMG Center for ALS, Department of Neurology, Massachusetts General Hospital, 165 Cambridge Street, Suite 600, Boston, MA, 02114, USA
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Eleonora Aronica
- Amsterdam UMC, University of Amsterdam, Department of (Neuro) Pathology, Amsterdam Neuroscience, Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - Giovanni Nardo
- Laboratory of Molecular Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milanm, 20156, Italy
| | - Donatienne Van Weehaeghe
- Division of Nuclear Medicine, Department of Imaging and Pathology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Elena Niccolai
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Nikolina Prtenjaca
- Laboratory for Molecular Immunology, Department of Biotechnology, University of Rijeka, R. Matejcic 2, 51000, Rijeka, Croatia
| | - Stacey A Sakowski
- Department of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Caterina Bendotti
- Laboratory of Molecular Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milanm, 20156, Italy
| | - Letizia Mazzini
- Department of Neurology and ALS Centre, University of Piemonte Orientale, Maggiore Della Carità Hospital, Corso Mazzini 18, Novara, 28100, Italy.
| |
Collapse
|
61
|
Passaro AP, Lebos AL, Yao Y, Stice SL. Immune Response in Neurological Pathology: Emerging Role of Central and Peripheral Immune Crosstalk. Front Immunol 2021; 12:676621. [PMID: 34177918 PMCID: PMC8222736 DOI: 10.3389/fimmu.2021.676621] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/17/2021] [Indexed: 12/13/2022] Open
Abstract
Neuroinflammation is a key component of neurological disorders and is an important therapeutic target; however, immunotherapies have been largely unsuccessful. In cases where these therapies have succeeded, particularly multiple sclerosis, they have primarily focused on one aspect of the disease and leave room for improvement. More recently, the impact of the peripheral immune system is being recognized, since it has become evident that the central nervous system is not immune-privileged, as once thought. In this review, we highlight key interactions between central and peripheral immune cells in neurological disorders. While traditional approaches have examined these systems separately, the immune responses and processes in neurological disorders consist of substantial crosstalk between cells of the central and peripheral immune systems. Here, we provide an overview of major immune effector cells and the role of the blood-brain barrier in regard to neurological disorders and provide examples of this crosstalk in various disorders, including stroke and traumatic brain injury, multiple sclerosis, neurodegenerative diseases, and brain cancer. Finally, we propose targeting central-peripheral immune interactions as a potential improved therapeutic strategy to overcome failures in clinical translation.
Collapse
Affiliation(s)
- Austin P. Passaro
- Regenerative Bioscience Center, University of Georgia, Athens, GA, United States
- Division of Neuroscience, Biomedical Health and Sciences Institute, University of Georgia, Athens, GA, United States
| | - Abraham L. Lebos
- Regenerative Bioscience Center, University of Georgia, Athens, GA, United States
- Department of Biochemistry and Microbiology, University of Georgia, Athens, GA, United States
| | - Yao Yao
- Regenerative Bioscience Center, University of Georgia, Athens, GA, United States
- Department of Animal and Dairy Science, University of Georgia, Athens, GA, United States
| | - Steven L. Stice
- Regenerative Bioscience Center, University of Georgia, Athens, GA, United States
- Division of Neuroscience, Biomedical Health and Sciences Institute, University of Georgia, Athens, GA, United States
- Department of Animal and Dairy Science, University of Georgia, Athens, GA, United States
| |
Collapse
|
62
|
Some CSF Kynurenine Pathway Intermediates Associated with Disease Evolution in Amyotrophic Lateral Sclerosis. Biomolecules 2021; 11:biom11050691. [PMID: 34063031 PMCID: PMC8147980 DOI: 10.3390/biom11050691] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 04/30/2021] [Accepted: 05/02/2021] [Indexed: 12/15/2022] Open
Abstract
The aim of this study was to evaluate the kynurenine pathway (KP) and amino acids profile, using mass spectrometry, in the cerebrospinal fluid (CSF) of 42 amyotrophic lateral sclerosis (ALS) patients at the diagnosis and 40 controls to detect early disorders of these pathways. Diagnostic and predictive ability (based on weight loss, forced vital capacity, ALS Functional Rating Scale-Revised evolution over 12 months, and survival time) of these metabolites were evaluated using univariate followed by supervised multivariate analysis. The multivariate model between ALS and controls was not significant but highlighted some KP metabolites (kynurenine (KYN), kynurenic acid (KYNA), 3-Hydroxynurenine (3-HK)/KYNA ratio), and amino acids (Lysine, asparagine) as involved in the discrimination between groups (accuracy 62%). It revealed a probable KP impairment toward neurotoxicity in ALS patients and in bulbar forms. Regarding the prognostic effect of metabolites, 12 were commonly discriminant for at least 3 of 4 disease evolution criteria. This investigation was crucial as it did not show significant changes in CSF concentrations of amino acids and KP intermediates in early ALS evolution. However, trends of KP modifications suggest further exploration. The unclear kinetics of neuroinflammation linked to KP support the interest in exploring these pathways during disease evolution through a longitudinal strategy.
Collapse
|
63
|
A State of the Art of Antioxidant Properties of Curcuminoids in Neurodegenerative Diseases. Int J Mol Sci 2021; 22:ijms22063168. [PMID: 33804658 PMCID: PMC8003642 DOI: 10.3390/ijms22063168] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 01/07/2023] Open
Abstract
Neurodegenerative diseases represent a set of pathologies characterized by an irreversible and progressive, and a loss of neuronal cells in specific areas of the brain. Oxidative phosphorylation is a source of energy production by which many cells, such as the neuronal cells, meet their energy needs. Dysregulations of oxidative phosphorylation induce oxidative stress, which plays a key role in the onset of neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). To date, for most neurodegenerative diseases, there are no resolute treatments, but only interventions capable of alleviating the symptoms or slowing the course of the disease. Therefore, effective neuroprotection strategies are needed. In recent years, natural products, such as curcuminoids, have been intensively explored and studied for their therapeutic potentials in several neurodegenerative diseases. Curcuminoids are, nutraceutical compouns, that owen several therapeutic properties such as anti-oxidant, anti-inflammatory and neuroprotective effects. In this context, the aim of this review was to provide an overview of preclinical and clinical evidence aimed to illustrate the antioxidant effects of curcuminoids in neurodegenerative diseases. Promising results from preclinical studies encourage the use of curcuminoids for neurodegeneration prevention and treatment.
Collapse
|
64
|
Clarke BE, Patani R. The microglial component of amyotrophic lateral sclerosis. Brain 2021; 143:3526-3539. [PMID: 33427296 PMCID: PMC7805793 DOI: 10.1093/brain/awaa309] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/06/2020] [Accepted: 07/12/2020] [Indexed: 12/11/2022] Open
Abstract
Microglia are the primary immune cells of the CNS, carrying out key homeostatic roles and undergoing context-dependent and temporally regulated changes in response to injury and neurodegenerative diseases. Microglia have been implicated in playing a role in amyotrophic lateral sclerosis (ALS), a neurodegenerative disease characterized by extensive motor neuron loss leading to paralysis and premature death. However, as the pathomechansims of ALS are increasingly recognized to involve a multitude of different cell types, it has been difficult to delineate the specific contribution of microglia to disease. Here, we review the literature of microglial involvement in ALS and discuss the evidence for the neurotoxic and neuroprotective pathways that have been attributed to microglia in this disease. We also discuss accumulating evidence for spatiotemporal regulation of microglial activation in this context. A deeper understanding of the role of microglia in the ‘cellular phase’ of ALS is crucial in the development of mechanistically rationalized therapies.
Collapse
Affiliation(s)
- Benjamin E Clarke
- Department of Neuromuscular disease, Institute of Neurology, University College London, Queen Square, London, UK.,The Francis Crick Institute, 1 Midland Road, London, UK
| | - Rickie Patani
- Department of Neuromuscular disease, Institute of Neurology, University College London, Queen Square, London, UK.,The Francis Crick Institute, 1 Midland Road, London, UK
| |
Collapse
|
65
|
Li CY, Yang TM, Ou RW, Wei QQ, Shang HF. Genome-wide genetic links between amyotrophic lateral sclerosis and autoimmune diseases. BMC Med 2021; 19:27. [PMID: 33541344 PMCID: PMC7863260 DOI: 10.1186/s12916-021-01903-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 01/05/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Epidemiological and clinical studies have suggested comorbidity between amyotrophic lateral sclerosis (ALS) and autoimmune disorders. However, little is known about their shared genetic architecture. METHODS To examine the relation between ALS and 10 autoimmune diseases, including asthma, celiac disease (CeD), Crohn's disease (CD), inflammatory bowel disease (IBD), multiple sclerosis (MS), psoriasis, rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), type 1 diabetes (T1D), and ulcerative colitis (UC), and identify shared risk loci, we first estimated the genetic correlation using summary statistics from genome-wide association studies, and then analyzed the genetic enrichment leveraging the conditional false discovery rate statistical method. RESULTS We identified a significant positive genetic correlation between ALS and CeD, MS, RA, and SLE, as well as a significant negative genetic correlation between ALS and IBD, UC, and CD. Robust genetic enrichment was observed between ALS and CeD and MS, and moderate enrichment was found between ALS and UC and T1D. Thirteen shared genetic loci were identified, among which five were suggestively significant in another ALS GWAS, namely rs3828599 (GPX3), rs3849943 (C9orf72), rs7154847 (G2E3), rs6571361 (SCFD1), and rs9903355 (GGNBP2). By integrating cis-expression quantitative trait loci analyses in Braineac and GTEx, we further identified GGNBP2, ATXN3, and SLC9A8 as novel ALS risk genes. Functional enrichment analysis indicated that the shared risk genes were involved in four pathways including membrane trafficking, vesicle-mediated transport, ER to Golgi anterograde transport, and transport to the Golgi and subsequent modification. CONCLUSIONS Our findings demonstrate a specific genetic correlation between ALS and autoimmune diseases and identify shared risk loci, including three novel ALS risk genes. These results provide a better understanding for the pleiotropy of ALS and have implications for future therapeutic trials.
Collapse
Affiliation(s)
- Chun Yu Li
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatric, West China Hospital, Sichuan University, Chengdu, China
| | - Tian Mi Yang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatric, West China Hospital, Sichuan University, Chengdu, China
| | - Ru Wei Ou
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatric, West China Hospital, Sichuan University, Chengdu, China
| | - Qian Qian Wei
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatric, West China Hospital, Sichuan University, Chengdu, China
| | - Hui Fang Shang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatric, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
66
|
Abstract
This is an historical account of Canadian pioneers working in amyotrophic lateral sclerosis (ALS) in the 1970s and 1980s. Key contributions included the development of specialized clinics, the ALS Society of Canada, human motor unit estimates in vivo, use of transcranial magnetic stimulation (TMS), the dementias of ALS, the importance of neurofilaments and axonal flow, neuroinflammation and immunity related to ALS, use of tissue culture to study pathogenesis, and the story of ALS in Guam. Their work set the stage for future generations of ALS physicians and scientists to bring about meaningful therapies and hopefully a cure for ALS.
Collapse
|
67
|
Trolese MC, Mariani A, Terao M, de Paola M, Fabbrizio P, Sironi F, Kurosaki M, Bonanno S, Marcuzzo S, Bernasconi P, Trojsi F, Aronica E, Bendotti C, Nardo G. CXCL13/CXCR5 signalling is pivotal to preserve motor neurons in amyotrophic lateral sclerosis. EBioMedicine 2020; 62:103097. [PMID: 33161233 PMCID: PMC7670099 DOI: 10.1016/j.ebiom.2020.103097] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 10/13/2020] [Accepted: 10/13/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND CXCL13 is a B and T lymphocyte chemokine that mediates neuroinflammation through its receptor CXCR5. This chemokine is highly expressed by motoneurons (MNs) in Amyotrophic Lateral Sclerosis (ALS) SOD1G93A (mSOD1) mice during the disease, particularly in fast-progressing mice. Accordingly, in this study, we investigated the role of this chemokine in ALS. METHODS We used in vitro and in vivo experimental paradigms derived from ALS mice and patients to investigate the expression level and distribution of CXCL13/CXCR5 axis and its role in MN death and disease progression. Moreover, we compared the levels of CXCL13 in the CSF and serum of ALS patients and controls. FINDINGS CXCL13 and CXCR5 are overexpressed in the spinal MNs and peripheral axons in mSOD1 mice. CXCL13 inhibition in the CNS of ALS mice resulted in the exacerbation of motor impairment (n = 4/group;Mean_Diff.=27.81) and decrease survival (n = 14_Treated:19.2 ± 1.05wks, n = 17_Controls:20.2 ± 0.6wks; 95% CI: 0.4687-1.929). This was corroborated by evidence from primary spinal cultures where the inhibition or activation of CXCL13 exacerbated or prevented the MN loss. Besides, we found that CXCL13/CXCR5 axis is overexpressed in the spinal cord MNs of ALS patients, and CXCL13 levels in the CSF discriminate ALS (n = 30) from Multiple Sclerosis (n = 16) patients with a sensitivity of 97.56%. INTERPRETATION We hypothesise that MNs activate CXCL13 signalling to attenuate CNS inflammation and prevent the neuromuscular denervation. The low levels of CXCL13 in the CSF of ALS patients might reflect the MN dysfunction, suggesting this chemokine as a potential clinical adjunct to discriminate ALS from other neurological diseases. FUNDING Vaccinex, Inc.; Regione Lombardia (TRANS-ALS).
Collapse
Affiliation(s)
- Maria Chiara Trolese
- Laboratory of Molecular Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milan 20156, Italy
| | - Alessandro Mariani
- Laboratory of Biology of Neurodegenerative Disorders, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milan 20156, Italy
| | - Mineko Terao
- Laboratory of Molecular Biology, Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche IRCCS, Via Mario Negri 2, Milan 20156, Italy
| | - Massimiliano de Paola
- Laboratory of Biology of Neurodegenerative Disorders, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milan 20156, Italy
| | - Paola Fabbrizio
- Laboratory of Molecular Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milan 20156, Italy
| | - Francesca Sironi
- Laboratory of Molecular Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milan 20156, Italy
| | - Mami Kurosaki
- Laboratory of Molecular Biology, Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche IRCCS, Via Mario Negri 2, Milan 20156, Italy
| | - Silvia Bonanno
- Neurology IV-Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan 20133, Italy
| | - Stefania Marcuzzo
- Neurology IV-Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan 20133, Italy
| | - Pia Bernasconi
- Neurology IV-Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan 20133, Italy
| | - Francesca Trojsi
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", P.zza Miraglia 2, Naples 80138, Italy
| | - Eleonora Aronica
- Department of Pathology, Academic Medic\\\al Centre, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, Netherlands
| | - Caterina Bendotti
- Laboratory of Molecular Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milan 20156, Italy.
| | - Giovanni Nardo
- Laboratory of Molecular Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milan 20156, Italy.
| |
Collapse
|
68
|
Air Pollution-Related Brain Metal Dyshomeostasis as a Potential Risk Factor for Neurodevelopmental Disorders and Neurodegenerative Diseases. ATMOSPHERE 2020. [DOI: 10.3390/atmos11101098] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Increasing evidence links air pollution (AP) exposure to effects on the central nervous system structure and function. Particulate matter AP, especially the ultrafine (nanoparticle) components, can carry numerous metal and trace element contaminants that can reach the brain in utero and after birth. Excess brain exposure to either essential or non-essential elements can result in brain dyshomeostasis, which has been implicated in both neurodevelopmental disorders (NDDs; autism spectrum disorder, schizophrenia, and attention deficit hyperactivity disorder) and neurodegenerative diseases (NDGDs; Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, and amyotrophic lateral sclerosis). This review summarizes the current understanding of the extent to which the inhalational or intranasal instillation of metals reproduces in vivo the shared features of NDDs and NDGDs, including enlarged lateral ventricles, alterations in myelination, glutamatergic dysfunction, neuronal cell death, inflammation, microglial activation, oxidative stress, mitochondrial dysfunction, altered social behaviors, cognitive dysfunction, and impulsivity. Although evidence is limited to date, neuronal cell death, oxidative stress, and mitochondrial dysfunction are reproduced by numerous metals. Understanding the specific contribution of metals/trace elements to this neurotoxicity can guide the development of more realistic animal exposure models of human AP exposure and consequently lead to a more meaningful approach to mechanistic studies, potential intervention strategies, and regulatory requirements.
Collapse
|
69
|
Chen S, Liao Q, Lu K, Zhou J, Huang C, Bi F. Riluzole Exhibits No Therapeutic Efficacy on a Transgenic Rat model of Amyotrophic Lateral Sclerosis. Curr Neurovasc Res 2020; 17:275-285. [PMID: 32271694 DOI: 10.2174/1567202617666200409125227] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/10/2020] [Accepted: 03/12/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a neurological disorder clinically characterized by motor system dysfunction, with intraneuronal accumulation of the TAR DNAbinding protein 43 (TDP-43) being a pathological hallmark. Riluzole is a primarily prescribed medicine for ALS patients, while its therapeutical efficacy appears limited. TDP-43 transgenic mice are existing animal models for mechanistic/translational research into ALS. METHODS We developed a transgenic rat model of ALS expressing a mutant human TDP-43 transgene (TDP-43M337V) and evaluated the therapeutic effect of Riluzole on this model. Relative to control, rats with TDP-43M337V expression promoted by the neurofilament heavy subunit (NEF) gene or specifically in motor neurons promoted by the choline acetyltransferase (ChAT) gene showed progressive worsening of mobility and grip strength, along with loss of motor neurons, microglial activation, and intraneuronal accumulation of TDP-43 and ubiquitin aggregations in the spinal cord. RESULTS Compared to vehicle control, intragastric administration of Riluzole (30 mg/kg/d) did not mitigate the behavioral deficits nor alter the neuropathologies in the transgenics. CONCLUSION These findings indicate that transgenic rats recapitulate the basic neurological and neuropathological characteristics of human ALS, while Riluzole treatment can not halt the development of the behavioral and histopathological phenotypes in this new transgenic rodent model of ALS.
Collapse
Affiliation(s)
- Si Chen
- Department of Neurology, Central South University, Xiangya Hospital, Changsha, Hunan, China
| | - Qiao Liao
- Department of Neurology, Central South University, Xiangya Hospital, Changsha, Hunan, China
| | - Ke Lu
- Department of Neurology, Central South University, Xiangya Hospital, Changsha, Hunan, China
| | - Jinxia Zhou
- Department of Neurology, Central South University, Xiangya Hospital, Changsha, Hunan, China
| | - Cao Huang
- Department of Pathology Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Fangfang Bi
- Department of Neurology, Central South University, Xiangya Hospital, Changsha, Hunan, China
| |
Collapse
|
70
|
Finkbeiner S. Functional genomics, genetic risk profiling and cell phenotypes in neurodegenerative disease. Neurobiol Dis 2020; 146:105088. [PMID: 32977020 PMCID: PMC7686089 DOI: 10.1016/j.nbd.2020.105088] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 09/11/2020] [Accepted: 09/18/2020] [Indexed: 12/03/2022] Open
Abstract
Human genetics provides unbiased insights into the causes of human disease, which can be used to create a foundation for effective ways to more accurately diagnose patients, stratify patients for more successful clinical trials, discover and develop new therapies, and ultimately help patients choose the safest and most promising therapeutic option based on their risk profile. But the process for translating basic observations from human genetics studies into pathogenic disease mechanisms and treatments is laborious and complex, and this challenge has particularly slowed the development of interventions for neurodegenerative disease. In this review, we discuss the many steps in the process, the important considerations at each stage, and some of the latest tools and technologies that are available to help investigators translate insights from human genetics into diagnostic and therapeutic strategies that will lead to the sort of advances in clinical care that make a difference for patients.
Collapse
Affiliation(s)
- Steven Finkbeiner
- Center for Systems and Therapeutics, USA; Taube/Koret Center for Neurodegenerative Disease Research, Gladstone Institutes, San Francisco, CA 94158, USA; Departments of Neurology and Physiology, University of Califorina, San Francisco, CA 94158, USA.
| |
Collapse
|
71
|
Mannie MD, DeOca KB, Bastian AG, Moorman CD. Tolerogenic vaccines: Targeting the antigenic and cytokine niches of FOXP3 + regulatory T cells. Cell Immunol 2020; 355:104173. [PMID: 32712270 PMCID: PMC7444458 DOI: 10.1016/j.cellimm.2020.104173] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/06/2020] [Accepted: 07/10/2020] [Indexed: 02/06/2023]
Abstract
FOXP3+ regulatory T cells (Tregs) constitute a critical barrier that enforces tolerance to both the self-peptidome and the extended-self peptidome to ensure tissue-specific resistance to autoimmune, allergic, and other inflammatory disorders. Here, we review intuitive models regarding how T cell antigen receptor (TCR) specificity and antigen recognition efficiency shape the Treg and conventional T cell (Tcon) repertoires to adaptively regulate T cell maintenance, tissue-residency, phenotypic stability, and immune function in peripheral tissues. Three zones of TCR recognition efficiency are considered, including Tcon recognition of specific low-efficiency self MHC-ligands, Treg recognition of intermediate-efficiency agonistic self MHC-ligands, and Tcon recognition of cross-reactive high-efficiency agonistic foreign MHC-ligands. These respective zones of TCR recognition efficiency are key to understanding how tissue-resident immune networks integrate the antigenic complexity of local environments to provide adaptive decisions setting the balance of suppressive and immunogenic responses. Importantly, deficiencies in the Treg repertoire appear to be an important cause of chronic inflammatory disease. Deficiencies may include global deficiencies in Treg numbers or function, subtle 'holes in the Treg repertoire' in tissue-resident Treg populations, or simply Treg insufficiencies that are unable to counter an overwhelming molecular mimicry stimulus. Tolerogenic vaccination and Treg-based immunotherapy are two therapeutic modalities meant to restore dominance of Treg networks to reverse chronic inflammatory disease. Studies of these therapeutic modalities in a preclinical setting have provided insight into the Treg niche, including the concept that intermediate-efficiency TCR signaling, high IFN-β concentrations, and low IL-2 concentrations favor Treg responses and active dominant mechanisms of immune tolerance. Overall, the purpose here is to assimilate new and established concepts regarding how cognate TCR specificity of the Treg repertoire and the contingent cytokine networks provide a foundation for understanding Treg suppressive strategy.
Collapse
Affiliation(s)
- Mark D Mannie
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States.
| | - Kayla B DeOca
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States
| | - Alexander G Bastian
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States
| | - Cody D Moorman
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States
| |
Collapse
|
72
|
Harrison JM, Rafuse VF. Muscle fiber-type specific terminal Schwann cell pathology leads to sprouting deficits following partial denervation in SOD1 G93A mice. Neurobiol Dis 2020; 145:105052. [PMID: 32827689 DOI: 10.1016/j.nbd.2020.105052] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 07/20/2020] [Accepted: 08/13/2020] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an adult-onset disease characterized by the progressive death of motoneurons and denervation of muscle fibers. To restore motor function, surviving motoneurons in partially denervated muscles typically sprout axons to reinnervate denervated endplates. However, studies on the SOD1G93A rodent models of ALS indicate that sprouting is significantly limited in fast, but not slow, twitch muscles after disease onset. This limitation hastens the rate of muscle weakness and loss of motor function. The causes of this limitation are currently unknown. Sprouting could be limited because the SOD1G93A mutation weakens motoneurons making them incapable of expanding their field of innervation. Alternatively, motoneurons may be capable of sprouting, but unable to do so due to the loss of a permissive sprouting environment. To distinguish between the two possibilities, we compared the sprouting capacity of motoneuron subtypes by partially denervating the fast twitch plantaris (composed of type IIa/IIb muscle fibers) and slow twitch soleus muscles (type I/IIa fibers) prior to disease onset and weakening in SOD1G93A and WT mice. We found that only motoneurons innervating the SOD1G93A plantaris had a limited sprouting capacity. This was correlated with the selective loss of terminal Schwann cells (TSCs) at IIb fibers and an increase in macrophage infiltration. Treating SOD1G93A mice with the tyrosine kinase inhibitor, masitinib, significantly reduced infiltration, prevented TSC loss, and increased the sprouting capacity to near normal. These results suggest that TSCs at denervated type IIb muscle fibers are aberrantly targeted by infiltrating macrophages in SOD1G93A mice, and their loss accounts, at least in part, for the compromised sprouting capacity of the largest motoneurons during early stages of ALS.
Collapse
Affiliation(s)
- Julia M Harrison
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia B3H 1X5, Canada; Brain Repair Centre, Life Sciences Research Institute, Halifax, Nova Scotia B3H 4R2, Canada
| | - Victor F Rafuse
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia B3H 1X5, Canada; Brain Repair Centre, Life Sciences Research Institute, Halifax, Nova Scotia B3H 4R2, Canada.
| |
Collapse
|
73
|
Repeated 5-day cycles of low dose aldesleukin in amyotrophic lateral sclerosis (IMODALS): A phase 2a randomised, double-blind, placebo-controlled trial. EBioMedicine 2020; 59:102844. [PMID: 32651161 PMCID: PMC7502670 DOI: 10.1016/j.ebiom.2020.102844] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/29/2020] [Accepted: 06/03/2020] [Indexed: 12/17/2022] Open
Abstract
Background Low-dose interleukin-2 (ld-IL-2) enhances regulatory T-cell (Treg) function in auto-inflammatory conditions. Neuroinflammation being a pathogenic feature of amyotrophic lateral sclerosis (ALS), we evaluated the pharmacodynamics and safety of ld-IL-2 in ALS subjects. Methods We performed a single centre, parallel three-arm, randomised, double-blind, placebo-controlled study. Eligibility criteria included age < 75 years, disease duration < 5 years, riluzole treatment > 3 months, and a slow vital capacity ≥ 70% of normal. Patients were randomised (1:1:1) to aldesleukin 2 MIU, 1 MIU, or placebo once daily for 5 days every 4 weeks for 3 cycles. Primary outcome was change from baseline in Treg percentage of CD4+ T cells (%Tregs) following a first cycle. Secondary laboratory outcomes included: %Treg and Treg number following repeated cycles, and plasma CCL2 and neurofilament light chain protein (NFL) concentrations as surrogate markers of efficacy. Safety outcomes included motor-function (ALSFRS-R), slow vital capacity (SVC), and adverse event reports. This trial is registered with ClinicalTrials.gov, NCT02059759. Findings All randomised patients (12 per group), recruited from October 2015 to December 2015, were alive at the end of follow-up and included in the intent-to-treat (ITT) analysis. No drug-related serious adverse event was observed. Non-serious adverse events occurred more frequently with the 1 and 2 MIU IL-2 doses compared to placebo, including injection site reactions and flu-like symptoms. Primary outcome analysis showed a significant increase (p < 0·0001) in %Tregs in the 2 MIU and 1 MIU arms (mean [SD]: 2 MIU: +6·2% [2·2]; 1 MIU: +3·9% [1·2]) as compared to placebo (mean [SD]: -0·49% [1·3]). Effect sizes (ES) were large in treated groups: 2 MIU ES=3·7 (IC95%: 2·3–4·9) and 1 MIU ES=3·5 (IC95%: 2·1–4·6). Secondary outcomes showed a significant increase in %Tregs following repeated cycles (p < 0·0001) as compared to placebo, and a dose-dependent decrease in plasma CCL2 (p = 0·0049). There were no significant differences amongst the three groups on plasma NFL levels. Interpretation Ld-IL-2 is well tolerated and immunologically effective in subjects with ALS. These results warrant further investigation into their eventual therapeutic impact on slowing ALS disease progression. Funding : The French Health Ministry (PHRC-I-14-056), EU H2020 (grant #633413), and the Association pour la Recherche sur la SLA (ARSLA).
Collapse
|
74
|
Abstract
Organs-on-chips are broadly defined as microfabricated surfaces or devices designed to engineer cells into microscale tissues with native-like features and then extract physiologically relevant readouts at scale. Because they are generally compatible with patient-derived cells, these technologies can address many of the human relevance limitations of animal models. As a result, organs-on-chips have emerged as a promising new paradigm for patient-specific disease modeling and drug development. Because neuromuscular diseases span a broad range of rare conditions with diverse etiology and complex pathophysiology, they have been especially challenging to model in animals and thus are well suited for organ-on-chip approaches. In this Review, we first briefly summarize the challenges in neuromuscular disease modeling with animal models. Next, we describe a variety of existing organ-on-chip approaches for neuromuscular tissues, including a survey of cell sources for both muscle and nerve, and two- and three-dimensional neuromuscular tissue-engineering techniques. Although researchers have made tremendous advances in modeling neuromuscular diseases on a chip, the remaining challenges in cell sourcing, cell maturity, tissue assembly and readout capabilities limit their integration into the drug development pipeline today. However, as the field advances, models of healthy and diseased neuromuscular tissues on a chip, coupled with animal models, have vast potential as complementary tools for modeling multiple aspects of neuromuscular diseases and identifying new therapeutic strategies. Summary: Modeling neuromuscular diseases is challenging due to their complex etiology and pathophysiology. Here, we review the cell sources and tissue-engineering procedures that are being integrated as emerging neuromuscular disease models.
Collapse
Affiliation(s)
- Jeffrey W Santoso
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Megan L McCain
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA .,Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
75
|
|
76
|
Petrozziello T, Mills AN, Farhan SM, Mueller KA, Granucci EJ, Glajch KE, Chan J, Chew S, Berry JD, Sadri‐Vakili G. Lipocalin‐2 is increased in amyotrophic lateral sclerosis. Muscle Nerve 2020; 62:272-283. [DOI: 10.1002/mus.26911] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 04/24/2020] [Accepted: 04/29/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Tiziana Petrozziello
- Sean M. Healey & AMG Center for ALS at Mass GeneralMassachusetts General Hospital Boston Massachusetts
| | - Alexandra N. Mills
- Sean M. Healey & AMG Center for ALS at Mass GeneralMassachusetts General Hospital Boston Massachusetts
| | - Sali M.K. Farhan
- Analytic and Translational Genetics Unit, Department of MedicineMassachusetts General Hospital and Harvard Medical School Boston Massachusetts
- Program in Medical and Population GeneticsBroad Institute of MIT and Harvard Cambridge Massachusetts
| | - Kaly A. Mueller
- Sean M. Healey & AMG Center for ALS at Mass GeneralMassachusetts General Hospital Boston Massachusetts
| | - Eric J. Granucci
- Sean M. Healey & AMG Center for ALS at Mass GeneralMassachusetts General Hospital Boston Massachusetts
| | - Kelly E. Glajch
- Sean M. Healey & AMG Center for ALS at Mass GeneralMassachusetts General Hospital Boston Massachusetts
| | - James Chan
- Biostatistics Center, Department of MedicineMassachusetts General Hospital Boston Massachusetts
| | - Sheena Chew
- Sean M. Healey & AMG Center for ALS at Mass GeneralMassachusetts General Hospital Boston Massachusetts
| | - James D. Berry
- Sean M. Healey & AMG Center for ALS at Mass GeneralMassachusetts General Hospital Boston Massachusetts
| | - Ghazaleh Sadri‐Vakili
- Sean M. Healey & AMG Center for ALS at Mass GeneralMassachusetts General Hospital Boston Massachusetts
| |
Collapse
|
77
|
Moreno-Martinez L, de la Torre M, Muñoz MJ, Zaragoza P, Aguilera J, Calvo AC, Osta R. Neuroprotective Fragment C of Tetanus Toxin Modulates IL-6 in an ALS Mouse Model. Toxins (Basel) 2020; 12:toxins12050330. [PMID: 32429516 PMCID: PMC7290364 DOI: 10.3390/toxins12050330] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/15/2020] [Accepted: 05/13/2020] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation plays a significant role in amyotrophic lateral sclerosis (ALS) pathology, leading to the development of therapies targeting inflammation in recent years. Our group has studied the tetanus toxin C-terminal fragment (TTC) as a therapeutic molecule, showing neuroprotective properties in the SOD1G93A mouse model. However, it is unknown whether TTC could have some effect on inflammation. The objective of this study was to assess the effect of TTC on the regulation of inflammatory mediators to elucidate its potential role in modulating inflammation occurring in ALS. After TTC treatment in SOD1G93A mice, levels of eotaxin-1, interleukin (IL)-2, IL-6 and macrophage inflammatory protein (MIP)-1 alpha (α) and galectin-1 were analyzed by immunoassays in plasma samples, whilst protein expression of caspase-1, IL-1β, IL-6 and NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) was measured in the spinal cord, extensor digitorum longus (EDL) muscle and soleus (SOL) muscle. The results showed reduced levels of IL-6 in spinal cord, EDL and SOL in treated SOD1G93A mice. In addition, TTC showed a different role in the modulation of NLRP3 and caspase-1 depending on the tissue analyzed. In conclusion, our results suggest that TTC could have a potential anti-inflammatory effect by reducing IL-6 levels in tissues drastically affected by the disease. However, further research is needed to study more in depth the anti-inflammatory effect of TTC in ALS.
Collapse
Affiliation(s)
- Laura Moreno-Martinez
- Laboratory of Genetics and Biochemistry (LAGENBIO), Faculty of Veterinary, Institute for Health Research Aragon (IIS Aragón), AgriFood Institute of Aragon (IA2), Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), University of Zaragoza, Miguel Servet 177, 50013 Zaragoza, Spain; (L.M.-M.); (M.d.l.T.); (M.J.M.); (P.Z.)
| | - Miriam de la Torre
- Laboratory of Genetics and Biochemistry (LAGENBIO), Faculty of Veterinary, Institute for Health Research Aragon (IIS Aragón), AgriFood Institute of Aragon (IA2), Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), University of Zaragoza, Miguel Servet 177, 50013 Zaragoza, Spain; (L.M.-M.); (M.d.l.T.); (M.J.M.); (P.Z.)
| | - María J. Muñoz
- Laboratory of Genetics and Biochemistry (LAGENBIO), Faculty of Veterinary, Institute for Health Research Aragon (IIS Aragón), AgriFood Institute of Aragon (IA2), Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), University of Zaragoza, Miguel Servet 177, 50013 Zaragoza, Spain; (L.M.-M.); (M.d.l.T.); (M.J.M.); (P.Z.)
| | - Pilar Zaragoza
- Laboratory of Genetics and Biochemistry (LAGENBIO), Faculty of Veterinary, Institute for Health Research Aragon (IIS Aragón), AgriFood Institute of Aragon (IA2), Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), University of Zaragoza, Miguel Servet 177, 50013 Zaragoza, Spain; (L.M.-M.); (M.d.l.T.); (M.J.M.); (P.Z.)
| | - José Aguilera
- Institut de Neurociències and Departament de Bioquímica i de Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona (UAB) and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 08193 Barcelona, Spain;
| | - Ana C. Calvo
- Laboratory of Genetics and Biochemistry (LAGENBIO), Faculty of Veterinary, Institute for Health Research Aragon (IIS Aragón), AgriFood Institute of Aragon (IA2), Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), University of Zaragoza, Miguel Servet 177, 50013 Zaragoza, Spain; (L.M.-M.); (M.d.l.T.); (M.J.M.); (P.Z.)
- Correspondence: (A.C.C.); (R.O.); Tel.: +34-976761622 (A.C.C.); +34-976761621 (R.O.)
| | - Rosario Osta
- Laboratory of Genetics and Biochemistry (LAGENBIO), Faculty of Veterinary, Institute for Health Research Aragon (IIS Aragón), AgriFood Institute of Aragon (IA2), Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), University of Zaragoza, Miguel Servet 177, 50013 Zaragoza, Spain; (L.M.-M.); (M.d.l.T.); (M.J.M.); (P.Z.)
- Correspondence: (A.C.C.); (R.O.); Tel.: +34-976761622 (A.C.C.); +34-976761621 (R.O.)
| |
Collapse
|
78
|
Valsecchi V, Boido M, Montarolo F, Guglielmotto M, Perga S, Martire S, Cutrupi S, Iannello A, Gionchiglia N, Signorino E, Calvo A, Fuda G, Chiò A, Bertolotto A, Vercelli A. The transcription factor Nurr1 is upregulated in amyotrophic lateral sclerosis patients and SOD1-G93A mice. Dis Model Mech 2020; 13:dmm043513. [PMID: 32188741 PMCID: PMC7240304 DOI: 10.1242/dmm.043513] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 03/13/2020] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease that affects both lower and upper motor neurons (MNs) in the central nervous system. ALS etiology is highly multifactorial and multifarious, and an effective treatment is still lacking. Neuroinflammation is a hallmark of ALS and could be targeted to develop new therapeutic approaches. Interestingly, the transcription factor Nurr1 has been demonstrated to have an important role in the inflammatory process in several neurological disorders, such as Parkinson's disease and multiple sclerosis. In the present paper, we demonstrate for the first time that Nurr1 expression levels are upregulated in the peripheral blood of ALS patients. Moreover, we investigated Nurr1 function in the SOD1-G93A mouse model of ALS. Nurr1 was strongly upregulated in the spinal cord during the asymptomatic and early symptomatic phases of the disease, where it promoted the expression of brain-derived neurotrophic factor mRNA and the repression of NFκB pro-inflammatory targets, such as inducible nitric oxide synthase. Therefore, we hypothesize that Nurr1 is activated in an early phase of the disease as a protective endogenous anti-inflammatory mechanism, although not sufficient to reverse disease progression. On the basis of these observations, Nurr1 could represent a potential biomarker for ALS and a promising target for future therapies.
Collapse
MESH Headings
- Amyotrophic Lateral Sclerosis/blood
- Amyotrophic Lateral Sclerosis/genetics
- Animals
- Astrocytes/metabolism
- Astrocytes/pathology
- Brain-Derived Neurotrophic Factor/metabolism
- Female
- Gene Expression Regulation
- Humans
- Male
- Mice
- Mice, Transgenic
- Middle Aged
- Motor Neurons/metabolism
- Motor Neurons/pathology
- NF-kappa B/genetics
- NF-kappa B/metabolism
- Nitric Oxide Synthase Type II/genetics
- Nitric Oxide Synthase Type II/metabolism
- Nuclear Receptor Subfamily 4, Group A, Member 2/blood
- Nuclear Receptor Subfamily 4, Group A, Member 2/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism
- Promoter Regions, Genetic/genetics
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Spinal Cord/metabolism
- Spinal Cord/pathology
- Superoxide Dismutase-1/genetics
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transcriptional Activation/genetics
- Up-Regulation/genetics
Collapse
Affiliation(s)
- Valeria Valsecchi
- Department of Neuroscience Rita Levi Montalcini, University of Turin, via Cherasco 15, 10126 Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
- Department of Neuroscience, Reproductive and Dentistry Sciences, University of Naples "Federico II", via S. Pansini 5, 80131, Naples, Italy
| | - Marina Boido
- Department of Neuroscience Rita Levi Montalcini, University of Turin, via Cherasco 15, 10126 Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Francesca Montarolo
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
- Neurobiology Unit, Neurology - CReSM (Regional Referring Center of Multiple Sclerosis), AOU San Luigi Gonzaga, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Michela Guglielmotto
- Department of Neuroscience Rita Levi Montalcini, University of Turin, via Cherasco 15, 10126 Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Simona Perga
- Department of Neuroscience Rita Levi Montalcini, University of Turin, via Cherasco 15, 10126 Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
- Neurobiology Unit, Neurology - CReSM (Regional Referring Center of Multiple Sclerosis), AOU San Luigi Gonzaga, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Serena Martire
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
- Neurobiology Unit, Neurology - CReSM (Regional Referring Center of Multiple Sclerosis), AOU San Luigi Gonzaga, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Santina Cutrupi
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Andrea Iannello
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Nadia Gionchiglia
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Elena Signorino
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Andrea Calvo
- Department of Neuroscience Rita Levi Montalcini, Amyotrophic Lateral Sclerosis Expert Center (CRESLA), University of Turin, via Cherasco 15, 10126 Turin, Italy
- University Hospital Città della Scienza e della Salute, corso Bramante 88, 10126 Turin, Italy
| | - Giuseppe Fuda
- Department of Neuroscience Rita Levi Montalcini, Amyotrophic Lateral Sclerosis Expert Center (CRESLA), University of Turin, via Cherasco 15, 10126 Turin, Italy
- University Hospital Città della Scienza e della Salute, corso Bramante 88, 10126 Turin, Italy
| | - Adriano Chiò
- Department of Neuroscience Rita Levi Montalcini, Amyotrophic Lateral Sclerosis Expert Center (CRESLA), University of Turin, via Cherasco 15, 10126 Turin, Italy
- University Hospital Città della Scienza e della Salute, corso Bramante 88, 10126 Turin, Italy
| | - Antonio Bertolotto
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
- Neurobiology Unit, Neurology - CReSM (Regional Referring Center of Multiple Sclerosis), AOU San Luigi Gonzaga, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Alessandro Vercelli
- Department of Neuroscience Rita Levi Montalcini, University of Turin, via Cherasco 15, 10126 Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| |
Collapse
|
79
|
|
80
|
McCombe PA, Lee JD, Woodruff TM, Henderson RD. The Peripheral Immune System and Amyotrophic Lateral Sclerosis. Front Neurol 2020; 11:279. [PMID: 32373052 PMCID: PMC7186478 DOI: 10.3389/fneur.2020.00279] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 03/25/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a severe neurodegenerative disease that is defined by loss of upper and lower motor neurons, associated with accumulation of protein aggregates in cells. There is also pathology in extra-motor areas of the brain, Possible causes of cell death include failure to deal with the aggregated proteins, glutamate toxicity and mitochondrial failure. ALS also involves abnormalities of metabolism and the immune system, including neuroinflammation in the brain and spinal cord. Strikingly, there are also abnormalities of the peripheral immune system, with alterations of T lymphocytes, monocytes, complement and cytokines in the peripheral blood of patients with ALS. The precise contribution of the peripheral immune system in ALS pathogenesis is an active area of research. Although some trials of immunomodulatory agents have been negative, there is strong preclinical evidence of benefit from immune modulation and further trials are currently underway. Here, we review the emerging evidence implicating peripheral immune alterations contributing to ALS, and their potential as future therapeutic targets for clinical intervention.
Collapse
Affiliation(s)
- Pamela A. McCombe
- Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia
- Wesley Medical Research, The Wesley Hospital, Brisbane, QLD, Australia
| | - John D. Lee
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Trent M. Woodruff
- Wesley Medical Research, The Wesley Hospital, Brisbane, QLD, Australia
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | | |
Collapse
|
81
|
Natural killer cells modulate motor neuron-immune cell cross talk in models of Amyotrophic Lateral Sclerosis. Nat Commun 2020; 11:1773. [PMID: 32286313 PMCID: PMC7156729 DOI: 10.1038/s41467-020-15644-8] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 03/18/2020] [Indexed: 12/13/2022] Open
Abstract
In amyotrophic lateral sclerosis (ALS), immune cells and glia contribute to motor neuron (MN) degeneration. We report the presence of NK cells in post-mortem ALS motor cortex and spinal cord tissues, and the expression of NKG2D ligands on MNs. Using a mouse model of familial-ALS, hSOD1G93A, we demonstrate NK cell accumulation in the motor cortex and spinal cord, with an early CCL2-dependent peak. NK cell depletion reduces the pace of MN degeneration, delays motor impairment and increases survival. This is confirmed in another ALS mouse model, TDP43A315T. NK cells are neurotoxic to hSOD1G93A MNs which express NKG2D ligands, while IFNγ produced by NK cells instructs microglia toward an inflammatory phenotype, and impairs FOXP3+/Treg cell infiltration in the spinal cord of hSOD1G93A mice. Together, these data suggest a role of NK cells in determining the onset and progression of MN degeneration in ALS, and in modulating Treg recruitment and microglia phenotype. Neuroimmune interactions are important in amyotrophic lateral sclerosis (ALS). Here the authors characterize the role of NK cells in mouse models of ALS, and in patient tissue.
Collapse
|
82
|
Jin M, Günther R, Akgün K, Hermann A, Ziemssen T. Peripheral proinflammatory Th1/Th17 immune cell shift is linked to disease severity in amyotrophic lateral sclerosis. Sci Rep 2020; 10:5941. [PMID: 32246039 PMCID: PMC7125229 DOI: 10.1038/s41598-020-62756-8] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 03/18/2020] [Indexed: 12/11/2022] Open
Abstract
Neuroinflammation is involved in the pathogenesis of amyotrophic lateral sclerosis (ALS), but only limited data are available on systematic peripheral and central immune cell profiles in ALS. We studied detailed immune profiles of 73 ALS patients and 48 healthy controls (controls) in peripheral blood by fluorescence-activated cell sorting as well as cytokine expression profiles in serum. In a subgroup of 16 ALS patients and 10 controls we additionally studied cerebrospinal fluid (CSF) samples. In peripheral blood, T cell subtypes presented a shift towards pro-inflammatory Th 1 and Th 17 cells whereas anti-inflammatory Th2 and T regulatory cells were decreased. Important players in innate immunity including distinct monocyte (Mo) and natural killer (NK) cell subtypes were changed in ALS patients compared to controls. Pro-inflammatory serum cytokines such as interleukin (IL)-1 beta, IL-6 and interferon-gamma (IFN-gamma) were increased and the anti-inflammatory cytokine IL-10 was decreased. Correlation analysis revealed moderate negative correlations between Th1 and Th17 to the ALS functional rating scale revised (ALSFRS-R) and to forced vital capacity. In CSF samples, no relevant alteration of the immune profile was found. In conclusion, the immune profile in ALS was shifted towards a Th1/Th17 cell-mediated pro-inflammatory immune response and correlated to disease severity and progression. Large prospective studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Mengmeng Jin
- Department of Neurology, Technische Universität Dresden, Dresden, Germany.,Center for Clinical Neuroscience, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Rene Günther
- Department of Neurology, Technische Universität Dresden, Dresden, Germany. .,German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany.
| | - Katja Akgün
- Department of Neurology, Technische Universität Dresden, Dresden, Germany.,Center for Clinical Neuroscience, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Andreas Hermann
- Department of Neurology, Technische Universität Dresden, Dresden, Germany.,German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany.,Translational Neurodegeneration Section "Albrecht-Kossel", Department of Neurology, University Medical Center Rostock, University of Rostock, Rostock, Germany.,German Center for Neurodegenerative Diseases (DZNE) Rostock/Greifswald, Rostock, Germany
| | - Tjalf Ziemssen
- Department of Neurology, Technische Universität Dresden, Dresden, Germany.,Center for Clinical Neuroscience, University Hospital Carl Gustav Carus, Dresden, Germany
| |
Collapse
|
83
|
Banfi G, Diani M, Pigatto PD, Reali E. T Cell Subpopulations in the Physiopathology of Fibromyalgia: Evidence and Perspectives. Int J Mol Sci 2020; 21:ijms21041186. [PMID: 32054062 PMCID: PMC7072736 DOI: 10.3390/ijms21041186] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/06/2020] [Accepted: 02/10/2020] [Indexed: 12/11/2022] Open
Abstract
Fibromyalgia is one of the most important “rheumatic” disorders, after osteoarthritis. The etiology of the disease is still not clear. At the moment, the most defined pathological mechanism is the alteration of central pain pathways, and emotional conditions can trigger or worsen symptoms. Increasing evidence supports the role of mast cells in maintaining pain conditions such as musculoskeletal pain and central sensitization. Importantly, mast cells can mediate microglia activation through the production of proinflammatory cytokines such as IL-1β, IL-6, and TNFα. In addition, levels of chemokines and proinflammatory cytokines are enhanced in serum and could contribute to inflammation at systemic level. Despite the well-characterized relationship between the nervous system and inflammation, the mechanism that links the different pathological features of fibromyalgia, including stress-related manifestations, central sensitization, and dysregulation of the innate and adaptive immune responses is largely unknown. This review aims to provide an overview of the current understanding of the role of adaptive immune cells, in particular T cells, in the physiopathology of fibromyalgia. It also aims at linking the latest advances emerging from basic science to envisage new perspectives to explain the role of T cells in interconnecting the psychological, neurological, and inflammatory symptoms of fibromyalgia.
Collapse
Affiliation(s)
- Giuseppe Banfi
- IRCCS Istituto Ortopedico Galeazzi, 20161Milan, Italy; (G.B.); (M.D.); (P.D.P.)
- School of Medicine, Università Vita-Salute San Raffaele, 20132 Milan, Italy
| | - Marco Diani
- IRCCS Istituto Ortopedico Galeazzi, 20161Milan, Italy; (G.B.); (M.D.); (P.D.P.)
| | - Paolo D. Pigatto
- IRCCS Istituto Ortopedico Galeazzi, 20161Milan, Italy; (G.B.); (M.D.); (P.D.P.)
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy
| | - Eva Reali
- IRCCS Istituto Ortopedico Galeazzi, 20161Milan, Italy; (G.B.); (M.D.); (P.D.P.)
- Correspondence:
| |
Collapse
|
84
|
Motataianu A, Barcutean L, Balasa R. Neuroimmunity in amyotrophic lateral sclerosis: focus on microglia. Amyotroph Lateral Scler Frontotemporal Degener 2020; 21:159-166. [PMID: 31903792 DOI: 10.1080/21678421.2019.1708949] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS), an incurable, devastating condition of the central nervous system, is characterized by selective destruction of motoneurons with an important contribution of innate and adaptative immunity. Microglia and immune cells are key players in neuroinflammation and active participants in ALS pathogenesis. Recent experiments in animal models have shown that microglia display both neuroprotective and neurotoxic properties, depending on the stage of disease progression and cytokine secretion. A better knowledge of the interactions between T cells and microglia in the immunopathogenesis of ALS is desirable for the development of ALS therapeutic strategies.
Collapse
Affiliation(s)
- Anca Motataianu
- Neurology Department, University of Medicine, Pharmacy, Science and Technology "George Emil Palade", Targu Mures, Romania
| | - Laura Barcutean
- Neurology Department, University of Medicine, Pharmacy, Science and Technology "George Emil Palade", Targu Mures, Romania
| | - Rodica Balasa
- Neurology Department, University of Medicine, Pharmacy, Science and Technology "George Emil Palade", Targu Mures, Romania
| |
Collapse
|
85
|
Figueroa-Romero C, Guo K, Murdock BJ, Paez-Colasante X, Bassis CM, Mikhail KA, Pawlowski KD, Evans MC, Taubman GF, McDermott AJ, O'Brien PD, Savelieff MG, Hur J, Feldman EL. Temporal evolution of the microbiome, immune system and epigenome with disease progression in ALS mice. Dis Model Mech 2019; 13:dmm041947. [PMID: 31597644 PMCID: PMC6906635 DOI: 10.1242/dmm.041947] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 10/05/2019] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a terminal neurodegenerative disease. Genetic predisposition, epigenetic changes, aging and accumulated life-long environmental exposures are known ALS risk factors. The complex and dynamic interplay between these pathological influences plays a role in disease onset and progression. Recently, the gut microbiome has also been implicated in ALS development. In addition, immune cell populations are differentially expanded and activated in ALS compared to healthy individuals. However, the temporal evolution of both the intestinal flora and the immune system relative to symptom onset in ALS is presently not fully understood. To better elucidate the timeline of the various potential pathological factors, we performed a longitudinal study to simultaneously assess the gut microbiome, immunophenotype and changes in ileum and brain epigenetic marks relative to motor behavior and muscle atrophy in the mutant superoxide dismutase 1 (SOD1G93A) familial ALS mouse model. We identified alterations in the gut microbial environment early in the life of SOD1G93A animals followed by motor dysfunction and muscle atrophy, and immune cell expansion and activation, particularly in the spinal cord. Global brain cytosine hydroxymethylation was also altered in SOD1G93A animals at disease end-stage compared to control mice. Correlation analysis confirmed interrelationships with the microbiome and immune system. This study serves as a starting point to more deeply comprehend the influence of gut microorganisms and the immune system on ALS onset and progression. Greater insight may help pinpoint novel biomarkers and therapeutic interventions to improve diagnosis and treatment for ALS patients.This article has an associated First Person interview with the joint first authors of the paper.
Collapse
Affiliation(s)
| | - Kai Guo
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, USA
| | - Benjamin J Murdock
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Christine M Bassis
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kristen A Mikhail
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Matthew C Evans
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | | | - Andrew J McDermott
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Phillipe D O'Brien
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Masha G Savelieff
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Junguk Hur
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, USA
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
86
|
Healy LM, Yaqubi M, Ludwin S, Antel JP. Species differences in immune-mediated CNS tissue injury and repair: A (neuro)inflammatory topic. Glia 2019; 68:811-829. [PMID: 31724770 DOI: 10.1002/glia.23746] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 10/04/2019] [Accepted: 10/11/2019] [Indexed: 12/13/2022]
Abstract
Cells of the adaptive and innate immune systems in the brain parenchyma and in the meningeal spaces contribute to physiologic functions and disease states in the central nervous system (CNS). Animal studies have demonstrated the involvement of immune constituents, along with major histocompatibility complex (MHC) molecules, in neural development and rare genetic disorders (e.g., colony stimulating factor 1 receptor [CSF1R] deficiency). Genome wide association studies suggest a comparable role of the immune system in humans. Although the CNS can be the target of primary autoimmune disorders, no current experimental model captures all of the features of the most common human disorder placed in this category, multiple sclerosis (MS). Such features include spontaneous onset, environmental contributions, and a recurrent/progressive disease course in a genetically predisposed host. Numerous therapeutic interventions related to antigen and cytokine specific therapies have demonstrated effectiveness in experimental autoimmune encephalomyelitis (EAE), the animal model used to define principles underlying immune-mediated mechanisms in MS. Despite the similarities in the two diseases, most treatments used to ameliorate EAE have failed to translate to the human disease. As directly demonstrated in animal models and implicated by correlative studies in humans, adaptive and innate immune constituents within the systemic compartment and resident in the CNS contribute to the disease course of neurodegenerative and neurobehavioral disorders. The expanding knowledge of the molecular properties of glial cells provides increasing insights into species related variables. These variables affect glial bidirectional interactions with the immune system as well as their own production of "immune molecules" that mediate tissue injury and repair.
Collapse
Affiliation(s)
- Luke M Healy
- Neuroimmunology Unit, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montréal, Quebec, Canada
| | - Moein Yaqubi
- Neuroimmunology Unit, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montréal, Quebec, Canada
| | - Samuel Ludwin
- Neuroimmunology Unit, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montréal, Quebec, Canada.,Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Jack P Antel
- Neuroimmunology Unit, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montréal, Quebec, Canada
| |
Collapse
|
87
|
Batra G, Jain M, Singh RS, Sharma AR, Singh A, Prakash A, Medhi B. Novel therapeutic targets for amyotrophic lateral sclerosis. Indian J Pharmacol 2019; 51:418-425. [PMID: 32029967 PMCID: PMC6984016 DOI: 10.4103/ijp.ijp_823_19] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 01/01/2020] [Accepted: 01/04/2020] [Indexed: 11/04/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an untreatable and fatal neurodegenerative disease that is identified by the loss of motor neurons in the spinal cord, brain stem, and motor cortex which theatrically reduces life expectancy. Although the primary cause of ALS remains unclear, its heterogeneity put forward for consideration of association with various factors, including endogenous and/or environmental ones, which may be involved in progressive motor neuron stress that causes activation of different cell death pathways. It is hypothesized that this disease is triggered by factors related to genetic, environmental, and age-dependent risk. In spite of large neurobiological, molecular and genetic research, at the beginning of the 21st century, ALS still remains one of the most devastating neurodegenerative diseases because of the lack of effective therapeutic targets. It is a challenge for the clinical and scientific community. A better understanding of the etiology of ALS is necessary to develop specific targets of this progressive neurodegenerative disease. This review states about the current knowledge of targets in ALS research. This review provides an overview of the contribution of different targets like mitochondrial dysfunction, glutamate transport and excitotoxicity, protein accumulation, Oxidative stress, neuromuscular junction, microglia, and other molecular targets in the pathogenesis of ALS.
Collapse
Affiliation(s)
- Gitika Batra
- Department of Neurology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Manav Jain
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Rahul Soloman Singh
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Amit Raj Sharma
- Department of Neurology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Ashutosh Singh
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Ajay Prakash
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Bikash Medhi
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
88
|
Serrano A, Apolloni S, Rossi S, Lattante S, Sabatelli M, Peric M, Andjus P, Michetti F, Carrì MT, Cozzolino M, D'Ambrosi N. The S100A4 Transcriptional Inhibitor Niclosamide Reduces Pro-Inflammatory and Migratory Phenotypes of Microglia: Implications for Amyotrophic Lateral Sclerosis. Cells 2019; 8:cells8101261. [PMID: 31623154 PMCID: PMC6829868 DOI: 10.3390/cells8101261] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/11/2019] [Accepted: 10/14/2019] [Indexed: 02/07/2023] Open
Abstract
S100A4, belonging to a large multifunctional S100 protein family, is a Ca2+-binding protein with a significant role in stimulating the motility of cancer and immune cells, as well as in promoting pro-inflammatory properties in different cell types. In the CNS, there is limited information concerning S100A4 presence and function. In this study, we analyzed the expression of S100A4 and the effect of the S100A4 transcriptional inhibitor niclosamide in murine activated primary microglia. We found that S100A4 was strongly up-regulated in reactive microglia and that niclosamide prevented NADPH oxidase 2, mTOR (mammalian target of rapamycin), and NF-κB (nuclear factor-kappa B) increase, cytoskeletal rearrangements, migration, and phagocytosis. Furthermore, we found that S100A4 was significantly up-regulated in astrocytes and microglia in the spinal cord of a transgenic rat SOD1-G93A model of amyotrophic lateral sclerosis. Finally, we demonstrated the increased expression of S100A4 also in fibroblasts derived from amyotrophic lateral sclerosis (ALS) patients carrying SOD1 pathogenic variants. These results ascribe S100A4 as a marker of microglial reactivity, suggesting the contribution of S100A4-regulated pathways to neuroinflammation, and identify niclosamide as a possible drug in the control and attenuation of reactive phenotypes of microglia, thus opening the way to further investigation for a new application in neurodegenerative conditions.
Collapse
Affiliation(s)
- Alessia Serrano
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Savina Apolloni
- Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy.
| | - Simona Rossi
- Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy.
- Institute of Translational Pharmacology, CNR, 00133 Rome, Italy.
| | - Serena Lattante
- Unità Operativa Complessa di Genetica Medica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy.
- Istituto di Medicina Genomica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Mario Sabatelli
- Unità Operativa Complessa di Neurologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy.
- Centro Clinico NEMO, 00168 Rome, Italy.
- Istituto di Neurologia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Mina Peric
- Institute of Physiology and Biochemistry "Ivan Djaja", Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia.
| | - Pavle Andjus
- Institute of Physiology and Biochemistry "Ivan Djaja", Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia.
| | - Fabrizio Michetti
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Maria Teresa Carrì
- Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy.
| | - Mauro Cozzolino
- Institute of Translational Pharmacology, CNR, 00133 Rome, Italy.
| | - Nadia D'Ambrosi
- Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy.
| |
Collapse
|
89
|
Abrahao A, Meng Y, Llinas M, Huang Y, Hamani C, Mainprize T, Aubert I, Heyn C, Black SE, Hynynen K, Lipsman N, Zinman L. First-in-human trial of blood-brain barrier opening in amyotrophic lateral sclerosis using MR-guided focused ultrasound. Nat Commun 2019; 10:4373. [PMID: 31558719 PMCID: PMC6763482 DOI: 10.1038/s41467-019-12426-9] [Citation(s) in RCA: 316] [Impact Index Per Article: 52.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 09/06/2019] [Indexed: 12/17/2022] Open
Abstract
MR-guided focused ultrasound (MRgFUS) is an emerging technology that can accurately and transiently permeabilize the blood-brain barrier (BBB) for targeted drug delivery to the central nervous system. We conducted a single-arm, first-in-human trial to investigate the safety and feasibility of MRgFUS-induced BBB opening in eloquent primary motor cortex in four volunteers with amyotrophic lateral sclerosis (ALS). Here, we show successful BBB opening using MRgFUS as demonstrated by gadolinium leakage at the target site immediately after sonication in all subjects, which normalized 24 hours later. The procedure was well-tolerated with no serious clinical, radiologic or electroencephalographic adverse events. This study demonstrates that non-invasive BBB permeabilization over the motor cortex using MRgFUS is safe, feasible, and reversible in ALS subjects. In future, MRgFUS can be coupled with promising therapeutics providing a targeted delivery platform in ALS.
Collapse
Affiliation(s)
- Agessandro Abrahao
- Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, M4N 3M5, Canada. .,Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, M4N 3M5, Canada. .,Harquail Centre for Neuromodulation, Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada.
| | - Ying Meng
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, M4N 3M5, Canada.,Harquail Centre for Neuromodulation, Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada.,Division of Neurosurgery, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, M4N 3M5, Canada
| | - Maheleth Llinas
- Harquail Centre for Neuromodulation, Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada.,Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, M4N 3M5, Canada
| | - Yuexi Huang
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, M4N 3M5, Canada
| | - Clement Hamani
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, M4N 3M5, Canada.,Harquail Centre for Neuromodulation, Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada.,Division of Neurosurgery, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, M4N 3M5, Canada
| | - Todd Mainprize
- Division of Neurosurgery, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, M4N 3M5, Canada
| | - Isabelle Aubert
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, M4N 3M5, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 3H7, Canada
| | - Chinthaka Heyn
- Department of Medical Imaging, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, M4N 3M5, Canada.,Odette Cancer Research, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, M4N 3M5, Canada
| | - Sandra E Black
- Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, M4N 3M5, Canada.,Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, M4N 3M5, Canada.,Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, M4N 3M5, Canada.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3H7, Canada
| | - Kullervo Hynynen
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, M4N 3M5, Canada.,Odette Cancer Research, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, M4N 3M5, Canada.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3H7, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON, M5S 3H7, Canada
| | - Nir Lipsman
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, M4N 3M5, Canada.,Harquail Centre for Neuromodulation, Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada.,Division of Neurosurgery, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, M4N 3M5, Canada
| | - Lorne Zinman
- Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, M4N 3M5, Canada.,Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, M4N 3M5, Canada
| |
Collapse
|
90
|
Theoharides TC, Tsilioni I, Bawazeer M. Mast Cells, Neuroinflammation and Pain in Fibromyalgia Syndrome. Front Cell Neurosci 2019; 13:353. [PMID: 31427928 PMCID: PMC6687840 DOI: 10.3389/fncel.2019.00353] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 07/16/2019] [Indexed: 02/04/2023] Open
Abstract
Fibromyalgia Syndrome (FMS) is a disorder of chronic, generalized muscular pain, accompanied by sleep disturbances, fatigue and cognitive dysfunction. There is no definitive pathogenesis except for altered central pain pathways. We previously reported increased serum levels of the neuropeptides substance P (SP) and its structural analogue hemokinin-1 (HK-1) together with the pro-inflammatory cytokines IL-6 and TNF in FMS patients as compared to sedentary controls. We hypothesize that thalamic mast cells contribute to inflammation and pain, by releasing neuro-sensitizing molecules that include histamine, IL-1β, IL-6 and TNF, as well as calcitonin-gene related peptide (CGRP), HK-1 and SP. These molecules could either stimulate thalamic nociceptive neurons directly, or via stimulation of microglia in the diencephalon. As a result, inhibiting mast cell stimulation could be used as a novel approach for reducing pain and the symptoms of FMS.
Collapse
Affiliation(s)
- Theoharis C Theoharides
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, Tufts University School of Medicine, Boston, MA, United States.,Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, United States.,Department of Internal Medicine, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, United States.,Department of Psychiatry, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, United States
| | - Irene Tsilioni
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, Tufts University School of Medicine, Boston, MA, United States
| | - Mona Bawazeer
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, Tufts University School of Medicine, Boston, MA, United States.,Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, United States.,Department of Basic Medical Sciences, College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| |
Collapse
|
91
|
Volonté C, Apolloni S, Sabatelli M. Histamine beyond its effects on allergy: Potential therapeutic benefits for the treatment of Amyotrophic Lateral Sclerosis (ALS). Pharmacol Ther 2019; 202:120-131. [PMID: 31233766 DOI: 10.1016/j.pharmthera.2019.06.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 06/03/2019] [Indexed: 02/06/2023]
Abstract
ALS currently remains a challenge despite many efforts in performing successful clinical trials and formulating therapeutic solutions. By learning from current failures and striving for success, scientists and clinicians are checking every possibility to search for missing hints and efficacious treatments. Because the disease is very complex and heterogeneous and, moreover, targeting not only motor neurons but also several different cell types including muscle, glial, and immune cells, the right answer to ALS is conceivably a multidrug strategy or the use of broad-spectrum molecules. The aim of the present work is to gather evidence about novel perspectives on ALS pathogenesis and to present recent and innovative paradigms for therapy. In particular, we describe how an old molecule possessing immunomodulatory and neuroprotective functions beyond its recognized effects on allergy, histamine, might have a renewed and far-reaching momentum in ALS.
Collapse
Affiliation(s)
- Cinzia Volonté
- CNR-Institute of Cell Biology and Neurobiology/UCSC, Largo Francesco Vito 1, 00168 Rome, Italy; Fondazione Santa Lucia IRCCS, Preclinical Neuroscience, Via Del Fosso di Fiorano 65, 00143 Rome, Italy.
| | - Savina Apolloni
- Fondazione Santa Lucia IRCCS, Preclinical Neuroscience, Via Del Fosso di Fiorano 65, 00143 Rome, Italy
| | - Mario Sabatelli
- Institute of Neurology-Catholic University of Sacro Cuore, Clinic Center NEMO- Fondazione Pol. A. Gemelli IRCCS, Largo Agostino Gemelli, 8, 00168 Rome, Italy
| |
Collapse
|
92
|
McCombe PA, Henderson RD, Lee A, Lee JD, Woodruff TM, Restuadi R, McRae A, Wray NR, Ngo S, Steyn FJ. Gut microbiota in ALS: possible role in pathogenesis? Expert Rev Neurother 2019; 19:785-805. [PMID: 31122082 DOI: 10.1080/14737175.2019.1623026] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction: The gut microbiota has important roles in maintaining human health. The microbiota and its metabolic byproducts could play a role in the pathogenesis of neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). Areas covered: The authors evaluate the methods of assessing the gut microbiota, and also review how the gut microbiota affects the various physiological functions of the gut. The authors then consider how gut dysbiosis could theoretically affect the pathogenesis of ALS. They present the current evidence regarding the composition of the gut microbiota in ALS and in rodent models of ALS. Finally, the authors review therapies that could improve gut dysbiosis in the context of ALS. Expert opinion: Currently reported studies suggest some instances of gut dysbiosis in ALS patients and mouse models; however, these studies are limited, and more information with well-controlled larger datasets is required to make a definitive judgment about the role of the gut microbiota in ALS pathogenesis. Overall this is an emerging field that is worthy of further investigation. The authors advocate for larger studies using modern metagenomic techniques to address the current knowledge gaps.
Collapse
Affiliation(s)
- Pamela A McCombe
- Centre for Clinical Research, The University of Queensland , Brisbane , Australia.,Wesley Medical Research, Level 8 East Wing, The Wesley Hospital , Brisbane , Australia.,Department of Neurology, Royal Brisbane & Women's Hospital , Brisbane , Australia.,School of Medicine, The University of Queensland , Brisbane , Australia
| | - Robert D Henderson
- Wesley Medical Research, Level 8 East Wing, The Wesley Hospital , Brisbane , Australia.,Department of Neurology, Royal Brisbane & Women's Hospital , Brisbane , Australia.,School of Medicine, The University of Queensland , Brisbane , Australia.,Queensland Brain Institute, The University of Queensland , Brisbane , Australia
| | - Aven Lee
- Centre for Clinical Research, The University of Queensland , Brisbane , Australia
| | - John D Lee
- School of Biomedical Sciences, The University of Queensland , Brisbane , Australia
| | - Trent M Woodruff
- School of Biomedical Sciences, The University of Queensland , Brisbane , Australia
| | - Restuadi Restuadi
- Institute for Molecular Bioscience, The University of Queensland , Brisbane , Australia
| | - Allan McRae
- Institute for Molecular Bioscience, The University of Queensland , Brisbane , Australia
| | - Naomi R Wray
- Queensland Brain Institute, The University of Queensland , Brisbane , Australia.,Institute for Molecular Bioscience, The University of Queensland , Brisbane , Australia
| | - Shyuan Ngo
- Centre for Clinical Research, The University of Queensland , Brisbane , Australia.,Wesley Medical Research, Level 8 East Wing, The Wesley Hospital , Brisbane , Australia.,Department of Neurology, Royal Brisbane & Women's Hospital , Brisbane , Australia.,Queensland Brain Institute, The University of Queensland , Brisbane , Australia.,Australian Institute for Bioengineering and Nanotechnology, The University of Queensland , Brisbane , Australia
| | - Frederik J Steyn
- Centre for Clinical Research, The University of Queensland , Brisbane , Australia.,Wesley Medical Research, Level 8 East Wing, The Wesley Hospital , Brisbane , Australia.,Department of Neurology, Royal Brisbane & Women's Hospital , Brisbane , Australia.,Australian Institute for Bioengineering and Nanotechnology, The University of Queensland , Brisbane , Australia
| |
Collapse
|
93
|
Human Bone Marrow Endothelial Progenitor Cell Transplantation into Symptomatic ALS Mice Delays Disease Progression and Increases Motor Neuron Survival by Repairing Blood-Spinal Cord Barrier. Sci Rep 2019; 9:5280. [PMID: 30918315 PMCID: PMC6437219 DOI: 10.1038/s41598-019-41747-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 03/14/2019] [Indexed: 12/11/2022] Open
Abstract
Convincing evidence demonstrated impairment of the blood-spinal cord barrier (BSCB) in Amyotrophic Lateral Sclerosis (ALS), mainly by endothelial cell (EC) alterations. Replacing damaged ECs by cell transplantation is a potential barrier repair strategy. Recently, we showed that intravenous (iv) administration of human bone marrow CD34+ (hBM34+) cells into symptomatic ALS mice benefits BSCB restoration and postpones disease progression. However, delayed effect on motor function and some severely damaged capillaries were noted. We hypothesized that hematopoietic cells from a restricted lineage would be more effective. This study aimed to establish the effects of human bone marrow-derived endothelial progenitor cells (hBMEPCs) systemically transplanted into G93A mice at symptomatic disease stage. Results showed that transplanted hBMEPCs significantly improved behavioral disease outcomes, engrafted widely into capillaries of the gray/white matter spinal cord and brain motor cortex/brainstem, substantially restored capillary ultrastructure, significantly decreased EB extravasation into spinal cord parenchyma, meaningfully re-established perivascular astrocyte end-feet, and enhanced spinal cord motor neuron survival. These results provide novel evidence that transplantation of hBMEPCs effectively repairs the BSCB, potentially preventing entry of detrimental peripheral factors, including immune/inflammatory cells, which contribute to motor neuron dysfunction. Transplanting EC progenitor cells may be a promising strategy for barrier repair therapy in this disease.
Collapse
|
94
|
Abati E, Bresolin N, Comi G, Corti S. Advances, Challenges, and Perspectives in Translational Stem Cell Therapy for Amyotrophic Lateral Sclerosis. Mol Neurobiol 2019; 56:6703-6715. [DOI: 10.1007/s12035-019-1554-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 03/13/2019] [Indexed: 12/13/2022]
|
95
|
Abstract
INTRODUCTION Amyotrophic lateral sclerosis (ALS) is a progressive fatal disorder that affects all skeletal muscles, leading to death, mostly within 2-4 years from onset. To date, the anti-glutamatergic drug riluzole is the only drug that has been approved for the treatment of this disease; however, its efficacy is modest. Oxidative stress is considered to be involved in the pathology of ALS, and in this regard, the free radical scavenger edaravone, which was originally developed for the treatment of acute ischemic stroke, has also been developed for the treatment of ALS. Areas covered: This review describes the pharmacological properties of edaravone and the progress of clinical trials conducted to evaluate the efficacy of this drug in the treatment of ALS. Expert commentary: Edaravone is the first drug to show effective inhibition of the motor function deterioration experienced by ALS patients with early-stage probable and definite types. In order to effectively prolong the quality of motor function, edaravone treatment should be initiated as soon as the diagnosis has been confirmed; however, the respiratory function should be carefully monitored when a deterioration in breathing capacity is detected.
Collapse
Affiliation(s)
- Hiide Yoshino
- a Department of Neurology , Yoshino Neurology Clinic , Ichikawa-city , Japan
| |
Collapse
|