301
|
Mariosa D, Kamel F, Bellocco R, Ye W, Fang F. Association between diabetes and amyotrophic lateral sclerosis in Sweden. Eur J Neurol 2015; 22:1436-42. [PMID: 25600257 DOI: 10.1111/ene.12632] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Accepted: 10/28/2014] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND PURPOSE Energy metabolism is altered in patients with amyotrophic lateral sclerosis (ALS) but the role of diabetes is largely unknown. METHODS A population-based case-control study was conducted of 5108 ALS cases and 25,540 individually matched population controls during 1991-2010. Information on ALS and pre-existing diabetes was retrieved from the Swedish Patient Register to explore the association of ALS with diabetes overall and with insulin-dependent or non-insulin-dependent diabetes specifically. Variation of the association by diabetes duration and age was also studied. RESULTS In total, 224 ALS cases (4.39%) and 1437 controls (5.63%) had diabetes before the index date, leading to an overall inverse association between diabetes and ALS risk [odds ratio (OR) 0.79, 95% confidence interval (CI) 0.68-0.91]. The association was strong for non-insulin-dependent diabetes (OR 0.66, 95% CI 0.53-0.81) but not for insulin-dependent diabetes (OR 0.83, 95% CI 0.60-1.15) and varied as a function of diabetes duration, with the strongest association observed around 6 years after first ascertainment of diabetes. The association was age-specific; the inverse association was noted only amongst individuals aged 70 or older. In contrast, for younger individuals (<50 years), pre-existing insulin-dependent diabetes was associated with a higher ALS risk (OR 5.38, 95% CI 1.87-15.51). CONCLUSIONS Our study suggests that there is an association between diabetes and ALS, and highlights the importance of taking into account age, insulin dependence and diabetes duration. Future studies should explore whether the association is independent of body mass index.
Collapse
Affiliation(s)
- D Mariosa
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - F Kamel
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - R Bellocco
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.,Department of Statistics and Quantitative Methods, University of Milano-Bicocca, Milan, Italy
| | - W Ye
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - F Fang
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
302
|
Ahuja A, Dev K, Tanwar RS, Selwal KK, Tyagi PK. Copper mediated neurological disorder: visions into amyotrophic lateral sclerosis, Alzheimer and Menkes disease. J Trace Elem Med Biol 2015; 29:11-23. [PMID: 24975171 DOI: 10.1016/j.jtemb.2014.05.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Revised: 04/16/2014] [Accepted: 05/08/2014] [Indexed: 12/31/2022]
Abstract
Copper (Cu) is a vital redox dynamic metal that is possibly poisonous in superfluous. Metals can traditionally or intricately cause propagation in reactive oxygen species (ROS) accretion in cells and this may effect in programmed cell death. Accumulation of Cu causes necrosis that looks to be facilitated by DNA damage, followed by activation of P53. Cu dyshomeostasis has also been concerned in neurodegenerative disorders such as Alzheimer, Amyotrophic lateral sclerosis (ALS) or Menkes disease and is directly related to neurodegenerative syndrome that usually produces senile dementia. These mortal syndromes are closely related with an immense damage of neurons and synaptic failure in the brain. This review focuses on copper mediated neurological disorders with insights into amyotrophic lateral sclerosis, Alzheimer and Menkes disease.
Collapse
Affiliation(s)
- Anami Ahuja
- Department of Biotechnology, NIMS University, Jaipur, India.
| | - Kapil Dev
- Faculty of Medicine in Hradec Kralove, University of Charles, Prague, Czech Republic
| | - Ranjeet S Tanwar
- Department of Biotechnology, N.C. College of Engineering, Israna, India
| | - Krishan K Selwal
- Department of Biotechnology, Deenbandhu Chotu Ram University of Science and Technology, Murthal, India
| | - Pankaj K Tyagi
- Department of Biotechnology, Meerut Institute of Engineering and Technology, Meerut, India
| |
Collapse
|
303
|
Devenney E, Foxe D, Dobson-Stone C, Kwok JB, Kiernan MC, Hodges JR. Clinical heterogeneity of the C9orf72 genetic mutation in frontotemporal dementia. Neurocase 2015; 21:535-41. [PMID: 25138488 DOI: 10.1080/13554794.2014.951058] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The C9orf72 genetic mutation represents the most common cause of familial frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS). Studies over the last 2 years have revealed a number of key features of this mutation in the fields of clinical neurology, imaging, pathology, and genetics. Despite these efforts, the clinical phenotype appears to extend beyond FTD and ALS into the realm of psychiatric disease, and while highly variable survival rates have been reported, the clinical course of carriers remains relatively unexplored. This report describes two contrasting C9orf72 cases, one with a protracted indolent course dominated by neuropsychiatric features and the other with a rapidly progressive dementia. In both cases, initial structural brain imaging was relatively normal.
Collapse
Affiliation(s)
- E Devenney
- a Neuroscience Research Australia , Sydney , NSW , Australia
| | | | | | | | | | | |
Collapse
|
304
|
Paganoni S, Karam C, Joyce N, Bedlack R, Carter GT. Comprehensive rehabilitative care across the spectrum of amyotrophic lateral sclerosis. NeuroRehabilitation 2015; 37:53-68. [PMID: 26409693 PMCID: PMC5223769 DOI: 10.3233/nre-151240] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS or Lou Gehrig's disease) is a neurodegenerative disease that results in progressive muscle weakness and wasting. There is no known cure and the disease is uniformly fatal. PURPOSE This review discusses current concepts in ALS care, from breaking the diagnosis to end-of-life care. People with ALS have several multidisciplinary needs due to a complex and dynamic disease process. They benefit from rehabilitation interventions that are individualized and have the goal of optimizing independence, function, and safety. These strategies also help minimize symptomatic burden and maximize quality of life. CONCLUSION Patient-centered, multidisciplinary care has a significant impact on the life of people with ALS and is the current standard of care for this patient population.
Collapse
Affiliation(s)
- Sabrina Paganoni
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Spaulding Rehabilitation Hospital, Massachusetts General Hospital and Boston VA Healthcare System, Boston, MA, USA
| | - Chafic Karam
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Nanette Joyce
- Department of Physical Medicine and Rehabilitation, Neuromuscular Section, University of California, Davis School of Medicine, Sacramento, CA, USA
| | - Richard Bedlack
- Duke University School of Medicine and Durham Veterans Affairs Medical Center, Durham, NC, USA
| | - Gregory T Carter
- Department of Physical Medicine and Rehabilitation, St Luke's Rehabilitation Institute, Spokane, WA, USA
| |
Collapse
|
305
|
Turner MR, Benatar M. Ensuring continued progress in biomarkers for amyotrophic lateral sclerosis. Muscle Nerve 2015; 51:14-8. [PMID: 25288265 PMCID: PMC4270289 DOI: 10.1002/mus.24470] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Revised: 09/19/2014] [Accepted: 09/29/2014] [Indexed: 12/31/2022]
Abstract
Multiple candidate biomarkers for amyotrophic lateral sclerosis (ALS) have emerged across a range of platforms. Replication of results, however, has been absent in all but a few cases, and the range of control samples has been limited. If progress toward clinical translation is to continue, the specific biomarker needs of ALS, which differ from those of other neurodegenerative disorders, as well as the challenges inherent to longitudinal ALS biomarker cohorts, must be understood. Appropriate application of multimodal approaches, international collaboration, presymptomatic studies, and biomarker integration into future therapeutic trials are among the essential priorities going forward.
Collapse
Affiliation(s)
- Martin R Turner
- Oxford University, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, West Wing Level 3, Oxford, OX3 9DU, UK
| | | |
Collapse
|
306
|
Joyce PI, Mcgoldrick P, Saccon RA, Weber W, Fratta P, West SJ, Zhu N, Carter S, Phatak V, Stewart M, Simon M, Kumar S, Heise I, Bros-Facer V, Dick J, Corrochano S, Stanford MJ, Luong TV, Nolan PM, Meyer T, Brandner S, Bennett DLH, Ozdinler PH, Greensmith L, Fisher EMC, Acevedo-Arozena A. A novel SOD1-ALS mutation separates central and peripheral effects of mutant SOD1 toxicity. Hum Mol Genet 2014; 24:1883-97. [PMID: 25468678 PMCID: PMC4355022 DOI: 10.1093/hmg/ddu605] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Transgenic mouse models expressing mutant superoxide dismutase 1 (SOD1) have been critical in furthering our understanding of amyotrophic lateral sclerosis (ALS). However, such models generally overexpress the mutant protein, which may give rise to phenotypes not directly relevant to the disorder. Here, we have analysed a novel mouse model that has a point mutation in the endogenous mouse Sod1 gene; this mutation is identical to a pathological change in human familial ALS (fALS) which results in a D83G change in SOD1 protein. Homozgous Sod1D83G/D83G mice develop progressive degeneration of lower (LMN) and upper motor neurons, likely due to the same unknown toxic gain of function as occurs in human fALS cases, but intriguingly LMN cell death appears to stop in early adulthood and the mice do not become paralyzed. The D83 residue coordinates zinc binding, and the D83G mutation results in loss of dismutase activity and SOD1 protein instability. As a result, Sod1D83G/D83G mice also phenocopy the distal axonopathy and hepatocellular carcinoma found in Sod1 null mice (Sod1−/−). These unique mice allow us to further our understanding of ALS by separating the central motor neuron body degeneration and the peripheral effects from a fALS mutation expressed at endogenous levels.
Collapse
Affiliation(s)
- Peter I Joyce
- MRC Mammalian Genetics Unit, Harwell, Oxfordshire OX11 0RD, UK
| | - Philip Mcgoldrick
- MRC Centre for Neuromuscular Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Rachele A Saccon
- MRC Centre for Neuromuscular Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - William Weber
- Department of Neurology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Pietro Fratta
- MRC Centre for Neuromuscular Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Steven J West
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Ning Zhu
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Sarah Carter
- MRC Mammalian Genetics Unit, Harwell, Oxfordshire OX11 0RD, UK
| | - Vinaya Phatak
- MRC Mammalian Genetics Unit, Harwell, Oxfordshire OX11 0RD, UK
| | | | - Michelle Simon
- MRC Mammalian Genetics Unit, Harwell, Oxfordshire OX11 0RD, UK
| | - Saumya Kumar
- MRC Mammalian Genetics Unit, Harwell, Oxfordshire OX11 0RD, UK
| | - Ines Heise
- MRC Mammalian Genetics Unit, Harwell, Oxfordshire OX11 0RD, UK
| | - Virginie Bros-Facer
- MRC Centre for Neuromuscular Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - James Dick
- MRC Centre for Neuromuscular Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | | | - Macdonnell J Stanford
- Department of Neurology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Tu Vinh Luong
- Department of Cellular Pathology, Royal Free London NHS Foundation Trust, Pond Street, London NW3 2QG, UK
| | - Patrick M Nolan
- MRC Mammalian Genetics Unit, Harwell, Oxfordshire OX11 0RD, UK
| | - Timothy Meyer
- UCL Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, London WC1E 6BT, UK
| | - Sebastian Brandner
- MRC Centre for Neuromuscular Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - David L H Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - P Hande Ozdinler
- Department of Neurology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Linda Greensmith
- MRC Centre for Neuromuscular Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK,
| | - Elizabeth M C Fisher
- MRC Centre for Neuromuscular Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK,
| | | |
Collapse
|
307
|
Fitzgerald KC, O'Reilly ÉJ, Falcone GJ, McCullough ML, Park Y, Kolonel LN, Ascherio A. Dietary ω-3 polyunsaturated fatty acid intake and risk for amyotrophic lateral sclerosis. JAMA Neurol 2014; 71:1102-10. [PMID: 25023276 DOI: 10.1001/jamaneurol.2014.1214] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
IMPORTANCE Amyotrophic lateral sclerosis (ALS) is a severe progressive disease that cannot be prevented or cured. Diet-derived long-chain polyunsaturated fatty acids (PUFAs) are incorporated in brain lipids and modulate oxidative and inflammatory processes and could thus affect ALS risk and progression. OBJECTIVE To examine the association between ω-6 and ω-3 PUFA consumption and ALS risk. DESIGN, SETTING, AND PARTICIPANTS Longitudinal analyses based on 1,002,082 participants (479,114 women and 522,968 men) in 5 prospective cohorts: the National Institutes of Health-AARP Diet and Health Study, the Cancer Prevention Study II Nutrition Cohort, the Health Professionals Follow-up Study, the Multiethnic Cohort Study, and the Nurses' Health Study. Diet was assessed via food frequency questionnaire developed or modified for each cohort. Participants were categorized into cohort-specific quintiles of intake of energy-adjusted dietary variables. MAIN OUTCOMES AND MEASURES Cohort-specific multivariable-adjusted risk ratios (RRs) of ALS incidence or death estimated by Cox proportional hazards regression and pooled using random-effects methods. RESULTS A total of 995 ALS cases were documented during the follow-up. A greater ω-3 PUFA intake was associated with a reduced risk for ALS. The pooled, multivariable-adjusted RR for the highest to the lowest quintile was 0.66 (95% CI, 0.53-0.81; P < .001 for trend). Consumption of both α-linolenic acid (RR, 0.73; 95% CI, 0.59-0.89; P = .003 for trend) and marine ω-3 PUFAs (RR, 0.84; 95% CI, 0.65-1.08; P = .03 for trend) contributed to this inverse association. Intakes of ω-6 PUFA were not associated with ALS risk. CONCLUSIONS AND RELEVANCE Consumption of foods high in ω-3 PUFAs may help prevent or delay the onset of ALS.
Collapse
Affiliation(s)
| | - Éilis J O'Reilly
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts7Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Guido J Falcone
- Department of Neurology, Massachusetts General Hospital, Boston6Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts
| | | | - Yikyung Park
- Nutritional Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | | | - Alberto Ascherio
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts6Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts7Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical Scho
| |
Collapse
|
308
|
Prognostic factors for the course of functional status of patients with ALS: a systematic review. J Neurol 2014; 262:1407-23. [PMID: 25385051 DOI: 10.1007/s00415-014-7564-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Revised: 10/24/2014] [Accepted: 10/25/2014] [Indexed: 01/31/2023]
Abstract
The progressive course of amyotrophic lateral sclerosis (ALS) results in an ever-changing spectrum of the care needs of patients with ALS. Knowledge of prognostic factors for the functional course of ALS may enhance clinical prediction and improve the timing of appropriate interventions. Our objective was to systematically review the evidence regarding prognostic factors for the rate of functional decline of patients with ALS, assessed with versions of the ALS Functional Rating Scale (ALSFRS). Two reviewers independently assessed the methodological quality of the thirteen included studies using the Quality in Prognosis Studies (QUIPS) tool. The overall quality of evidence for each prognostic factor was assessed using the Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach, considering risk of bias, imprecision, inconsistency, indirectness, and publication bias. The quality of evidence for the prognostic value of age at onset, site of onset, time from symptom onset to diagnosis, and ALSFRS-Revised baseline score was low, mainly due to the limited data and inconsistency of results in the small number of studies included. The prognostic value of initial rate of disease progression, age at diagnosis, forced vital capacity, frontotemporal dementia, body mass index, and comorbidity remains unclear. We conclude that the current evidence on prognostic factors for functional decline in ALS is insufficient to allow the development of a prediction tool that can support clinical decisions. Given the limited data, future prognostic studies may need to focus on factors that have a predictive value for a decline in ALSFRS(-R) domain scores, preferably based on internationally collected and shared data.
Collapse
|
309
|
Chiò A, Pagani M, Agosta F, Calvo A, Cistaro A, Filippi M. Neuroimaging in amyotrophic lateral sclerosis: insights into structural and functional changes. Lancet Neurol 2014; 13:1228-40. [PMID: 25453462 DOI: 10.1016/s1474-4422(14)70167-x] [Citation(s) in RCA: 166] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
In the past two decades, structural and functional neuroimaging findings have greatly modified longstanding notions regarding the pathophysiology of amyotrophic lateral sclerosis (ALS). Neuroimaging studies have shown that anatomical and functional lesions spread beyond precentral cortices and corticospinal tracts, to include the corpus callosum; frontal, sensory, and premotor cortices; thalamus; and midbrain. Both MRI and PET studies have shown early and diffuse loss of inhibitory cortical interneurons in the motor cortex (increased levels of functional connectivity and loss of GABAergic neurons, respectively) and diffuse gliosis in white-matter tracts. In ALS endophenotypes, neuroimaging has also shown a diverse spreading of lesions and a dissimilar impairment of functional and structural connections. A possible role of PET in the diagnosis of ALS has recently been proposed. However, most neuroimaging studies have pitfalls, such as a small number and poor clinical characterisation of patients, absence of adequate controls, and scarcity of longitudinal assessments. Studies involving international collaborations, standardised assessments, and large patient cohorts will overcome these shortcomings and provide further insight into the pathogenesis of ALS.
Collapse
Affiliation(s)
- Adriano Chiò
- ALS Center, "Rita Levi Montalcini" Department of Neuroscience, University of Turin, Turin, Italy.
| | - Marco Pagani
- Institute of Cognitive Sciences and Technologies, Consiglio Nazionale delle Ricerche (CNR), Rome, Italy; Department of Nuclear Medicine, Karolinska Hospital, Stockholm, Sweden
| | - Federica Agosta
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Andrea Calvo
- ALS Center, "Rita Levi Montalcini" Department of Neuroscience, University of Turin, Turin, Italy
| | - Angelina Cistaro
- Institute of Cognitive Sciences and Technologies, Consiglio Nazionale delle Ricerche (CNR), Rome, Italy; Positron Emission Tomography Center IRMET S.p.A, Euromedic Inc, Torino, Italy
| | - Massimo Filippi
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
310
|
Eisen A, Kiernan M, Mitsumoto H, Swash M. Amyotrophic lateral sclerosis: a long preclinical period? J Neurol Neurosurg Psychiatry 2014; 85:1232-8. [PMID: 24648037 DOI: 10.1136/jnnp-2013-307135] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The onset of amyotrophic lateral sclerosis (ALS) is conventionally considered as commencing with the recognition of clinical symptoms. We propose that, in common with other neurodegenerations, the pathogenic mechanisms culminating in ALS phenotypes begin much earlier in life. Animal models of genetically determined ALS exhibit pathological abnormalities long predating clinical deficits. The overt clinical ALS phenotype may develop when safety margins are exceeded subsequent to years of mitochondrial dysfunction, neuroinflammation or an imbalanced environment of excitation and inhibition in the neuropil. Somatic mutations, the epigenome and external environmental influences may interact to trigger a metabolic cascade that in the adult eventually exceeds functional threshold. A long preclinical and subsequent presymptomatic period pose a challenge for recognition, since it offers an opportunity for protective and perhaps even preventive therapeutic intervention to rescue dysfunctional neurons. We suggest, by analogy with other neurodegenerations and from SOD1 ALS mouse studies, that vulnerability might be induced in the perinatal period.
Collapse
Affiliation(s)
- Andrew Eisen
- Division of Neurology, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Hiroshi Mitsumoto
- Wesley J. Howe Professor of Neurology at CUMC, Eleanor and Lou Gehrig MDA/ALS Research Center, The Neurological Institute of New York, Columbia University Medical Center, New York, USA
| | - Michael Swash
- Queen Mary University of London, UK Institute of Neuroscience, University of Lisbon, Portugal
| |
Collapse
|
311
|
Mitsumoto H, Brooks BR, Silani V. Clinical trials in amyotrophic lateral sclerosis: why so many negative trials and how can trials be improved? Lancet Neurol 2014; 13:1127-1138. [DOI: 10.1016/s1474-4422(14)70129-2] [Citation(s) in RCA: 200] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
312
|
Heiman A, Pallottie A, Heary RF, Elkabes S. Toll-like receptors in central nervous system injury and disease: a focus on the spinal cord. Brain Behav Immun 2014; 42:232-45. [PMID: 25063708 DOI: 10.1016/j.bbi.2014.06.203] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 06/17/2014] [Accepted: 06/28/2014] [Indexed: 12/12/2022] Open
Abstract
Toll-like receptors (TLRs) are best known for recognizing pathogens and initiating an innate immune response to protect the host. However, they also detect tissue damage and induce sterile inflammation upon the binding of endogenous ligands released by stressed or injured cells. In addition to immune system-related cells, TLRs have been identified in central nervous system (CNS) neurons and glial subtypes including microglia, astrocytes and oligodendrocytes. Direct and indirect effects of TLR ligands on neurons and glial subtypes have been documented in vitro. Likewise, the effects of TLR ligands have been demonstrated in vivo using animal models of CNS trauma and disease including spinal cord injury (SCI), amyotrophic lateral sclerosis (ALS) and neuropathic pain. The indirect effects are most likely mediated via microglia or immune system cells that infiltrate the diseased or injured CNS. Despite considerable progress over the past decade, the role of TLRs in the physiological and pathological function of the spinal cord remains inadequately defined. Published reports collectively highlight TLRs as promising targets for therapeutic interventions in spinal cord pathology. The findings also underscore the complexity of TLR-mediated mechanisms and the necessity for further research in this field. The goals of the current review are to recapitulate the studies that investigated the role of TLRs in the spinal cord, to discuss potential future research directions, and to examine some of the challenges associated with pre-clinical studies pertinent to TLRs in the injured or diseased spinal cord.
Collapse
Affiliation(s)
- Adee Heiman
- Reynolds Family Spine Laboratory, Department of Neurological Surgery, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Alexandra Pallottie
- Reynolds Family Spine Laboratory, Department of Neurological Surgery, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States; Graduate School of Biomedical Sciences, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Robert F Heary
- Reynolds Family Spine Laboratory, Department of Neurological Surgery, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Stella Elkabes
- Reynolds Family Spine Laboratory, Department of Neurological Surgery, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States.
| |
Collapse
|
313
|
Simon NG, Lomen-Hoerth C, Kiernan MC. Patterns of clinical and electrodiagnostic abnormalities in early amyotrophic lateral sclerosis. Muscle Nerve 2014; 50:894-9. [DOI: 10.1002/mus.24244] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2014] [Indexed: 12/12/2022]
Affiliation(s)
- Neil G. Simon
- Department of Neurology; University of California at San Francisco; San Francisco California USA
- Prince of Wales Clinical School and Neuroscience Research Australia, University of New South Wales; Sydney New South Wales Australia
| | - Catherine Lomen-Hoerth
- Department of Neurology; University of California at San Francisco; San Francisco California USA
| | - Matthew C. Kiernan
- Brain and Mind Research Institute, University of Sydney; 100 Mallett Street Camperdown New South Wales 2050 Australia
| |
Collapse
|
314
|
Neurodegenerative diseases in a dish: the promise of iPSC technology in disease modeling and therapeutic discovery. Neurol Sci 2014; 36:21-7. [PMID: 25354658 PMCID: PMC4282683 DOI: 10.1007/s10072-014-1989-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 10/22/2014] [Indexed: 02/07/2023]
Abstract
The study of stem-cell biology has been a flourishing research area because of its multi-differentiation potential. The emergence of induced pluripotent stem cells (iPSCs) open up the possibility of addressing obstructs, such as the limited cell source, inherent complexity of the human brain, and ethical constrains. Though still at its infancy phase, reprogramming of somatic cells has been demonstrating the ability to enhance in vitro study of neurodegenerative diseases and potential treatment. However, iPSCs would not thoroughly translate to the clinic before limitations are addressed. In this review, by summarizing the recent development of iPSC-based models, we will discuss the feasibility of iPSC technology on relevant diseases depth and illustrate how this new tool applies to drug screening and celluar therapy.
Collapse
|
315
|
Rademakers R, van Blitterswijk M. Excess of rare damaging TUBA4A variants suggests cytoskeletal defects in ALS. Neuron 2014; 84:241-3. [PMID: 25374348 DOI: 10.1016/j.neuron.2014.10.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Identifying disease genes implicated in late-onset neurodegenerative disorders can be challenging due to the lack of DNA samples from multiple affected family members. To overcome this limitation, Smith et al. (2014) report in this issue of Neuron the first exome-wide rare variant analysis in unrelated familial amyotrophic lateral sclerosis (ALS) patients associating TUBA4A with ALS.
Collapse
Affiliation(s)
- Rosa Rademakers
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA.
| | | |
Collapse
|
316
|
McCluskey L, Vandriel S, Elman L, Van Deerlin VM, Powers J, Boller A, Wood EM, Woo J, McMillan CT, Rascovsky K, Grossman M. ALS-Plus syndrome: non-pyramidal features in a large ALS cohort. J Neurol Sci 2014; 345:118-24. [PMID: 25086858 PMCID: PMC4177937 DOI: 10.1016/j.jns.2014.07.022] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Revised: 05/03/2014] [Accepted: 07/10/2014] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Autopsy studies show widespread pathology in amyotrophic lateral sclerosis (ALS), but clinical surveys of multisystem disease in ALS are rare. We investigated ALS-Plus syndrome, an understudied group of patients with clinical features extending beyond pyramidal and neuromuscular systems with or without cognitive/behavioral deficits. METHODS In a large, consecutively-ascertained cohort of 550 patients with ALS, we documented atypical clinical manifestations. Genetic screening for C9orf72 hexanucleotide expansions was performed in 343 patients, and SOD1, TARDBP, and VCP were tested in the subgroup of patients with a family history of ALS. Gray matter and white matter imaging was available in a subgroup of 30 patients. RESULTS Seventy-five (13.6%) patients were identified with ALS-Plus syndrome. We found disorders of ocular motility, cerebellar, extrapyramidal and autonomic functioning. Relative to those without ALS-Plus, cognitive impairment (8.0% vs 2.9%, p=0.029), bulbar-onset (49.3% vs 23.2%, p<0.001), and pathogenic mutations (20.0% vs 8.4%, p=0.015) were more than twice as common in ALS-Plus. Survival was significantly shorter in ALS-Plus (29.66 months vs 42.50 months, p=0.02), regardless of bulbar-onset or mutation status. Imaging revealed significantly greater cerebellar and cerebral disease in ALS-Plus compared to those without ALS-Plus. CONCLUSIONS ALS-Plus syndrome is not uncommon, and the presence of these atypical features is consistent with neuropathological observations that ALS is a multisystem disorder. ALS-Plus syndrome is associated with increased risk for poor survival and the presence of a pathogenic mutation.
Collapse
Affiliation(s)
- Leo McCluskey
- Department of Neurology, University of Pennsylvania, United States
| | - Shannon Vandriel
- Department of Neurology, University of Pennsylvania, United States
| | - Lauren Elman
- Department of Neurology, University of Pennsylvania, United States
| | - Vivianna M Van Deerlin
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, United States
| | - John Powers
- Department of Neurology, University of Pennsylvania, United States
| | - Ashley Boller
- Department of Neurology, University of Pennsylvania, United States
| | - Elisabeth McCarty Wood
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, United States
| | - John Woo
- Department of Radiology, University of Pennsylvania, United States
| | - Corey T McMillan
- Department of Neurology, University of Pennsylvania, United States
| | - Katya Rascovsky
- Department of Neurology, University of Pennsylvania, United States
| | - Murray Grossman
- Department of Neurology, University of Pennsylvania, United States.
| |
Collapse
|
317
|
Rogers ML, Smith KS, Matusica D, Fenech M, Hoffman L, Rush RA, Voelcker NH. Non-viral gene therapy that targets motor neurons in vivo. Front Mol Neurosci 2014; 7:80. [PMID: 25352776 PMCID: PMC4196515 DOI: 10.3389/fnmol.2014.00080] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 09/18/2014] [Indexed: 12/11/2022] Open
Abstract
A major challenge in neurological gene therapy is safe delivery of transgenes to sufficient cell numbers from the circulation or periphery. This is particularly difficult for diseases involving spinal cord motor neurons such as amyotrophic lateral sclerosis (ALS). We have examined the feasibility of non-viral gene delivery to spinal motor neurons from intraperitoneal injections of plasmids carried by “immunogene” nanoparticles targeted for axonal retrograde transport using antibodies. PEGylated polyethylenimine (PEI-PEG12) as DNA carrier was conjugated to an antibody (MLR2) to the neurotrophin receptor p75 (p75NTR). We used a plasmid (pVIVO2) designed for in vivo gene delivery that produces minimal immune responses, has improved nuclear entry into post mitotic cells and also expresses green fluorescent protein (GFP). MLR2-PEI-PEG12 carried pVIVO2 and was specific for mouse motor neurons in mixed cultures containing astrocytes. While only 8% of motor neurons expressed GFP 72 h post transfection in vitro, when the immunogene was given intraperitonealy to neonatal C57BL/6J mice, GFP specific motor neuron expression was observed in 25.4% of lumbar, 18.3% of thoracic and 17.0% of cervical motor neurons, 72 h post transfection. PEI-PEG12 carrying pVIVO2 by itself did not transfect motor neurons in vivo, demonstrating the need for specificity via the p75NTR antibody MLR2. This is the first time that specific transfection of spinal motor neurons has been achieved from peripheral delivery of plasmid DNA as part of a non-viral gene delivery agent. These results stress the specificity and feasibility of immunogene delivery targeted for p75NTR expressing motor neurons, but suggests that further improvements are required to increase the transfection efficiency of motor neurons in vivo.
Collapse
Affiliation(s)
- Mary-Louise Rogers
- Department of Human Physiology, Centre for Neuroscience, Flinders University Adelaide, SA, Australia
| | - Kevin S Smith
- Department of Human Physiology, Centre for Neuroscience, Flinders University Adelaide, SA, Australia
| | - Dusan Matusica
- Department of Anatomy and Histology, Centre for Neuroscience, Flinders University Adelaide, SA, Australia
| | - Matthew Fenech
- Department of Human Physiology, Centre for Neuroscience, Flinders University Adelaide, SA, Australia
| | - Lee Hoffman
- Department of Chemistry and Biochemistry, South Dakota State University Brookings, SD, USA
| | - Robert A Rush
- Department of Human Physiology, Centre for Neuroscience, Flinders University Adelaide, SA, Australia
| | - Nicolas H Voelcker
- Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Mawson Institute, University of South Australia Adelaide, SA, Australia
| |
Collapse
|
318
|
Günther R, Saal KA, Suhr M, Scheer D, Koch JC, Bähr M, Lingor P, Tönges L. The rho kinase inhibitor Y-27632 improves motor performance in male SOD1(G93A) mice. Front Neurosci 2014; 8:304. [PMID: 25339858 PMCID: PMC4187656 DOI: 10.3389/fnins.2014.00304] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 09/09/2014] [Indexed: 12/11/2022] Open
Abstract
Disease progression in amyotrophic lateral sclerosis (ALS) is characterized by degeneration of motoneurons and their axons which results in a progressive muscle weakness and ultimately death from respiratory failure. The only approved drug, riluzole, lacks clinical efficacy so that more potent treatment options are needed. We have identified rho kinase (ROCK) as a target, which can be manipulated to beneficially influence disease progression in models of ALS. Here, we examined the therapeutic potential of the ROCK inhibitor Y-27632 in both an in vitro and in an in vivo paradigm of motoneuron disease. Application of Y-27632 to primary motoneurons in vitro increased survival and promoted neurite outgrowth. In vivo, SOD1G93A mice were orally treated with 2 or 30 mg/kg body weight of Y-27632. The 2 mg/kg group did not benefit from Y-27632 treatment, whereas treatment with 30 mg/kg resulted in improved motor function in male mice. Female mice showed only limited improvement and overall survival was not modified in both 2 and 30 mg/kg Y-27632 groups. In conclusion, we provide evidence that inhibition of ROCK by Y-27632 is neuroprotective in vitro but has limited beneficial effects in vivo being restricted to male mice. Therefore, the evaluation of ROCK inhibitors in preclinical models of ALS should always take gender differences into account.
Collapse
Affiliation(s)
- René Günther
- Department of Neurology, University Medicine Göttingen Göttingen, Germany
| | - Kim-Ann Saal
- Department of Neurology, University Medicine Göttingen Göttingen, Germany
| | - Martin Suhr
- Department of Neurology, University Medicine Göttingen Göttingen, Germany
| | - David Scheer
- Department of Neurology, University Medicine Göttingen Göttingen, Germany
| | - Jan Christoph Koch
- Department of Neurology, University Medicine Göttingen Göttingen, Germany
| | - Mathias Bähr
- Department of Neurology, University Medicine Göttingen Göttingen, Germany ; Nanoscale Microscopy and Molecular Physiology of the Brain, Cluster of Excellence 171-DFG Research Center 103 (CNMPB) Göttingen, Germany
| | - Paul Lingor
- Department of Neurology, University Medicine Göttingen Göttingen, Germany ; Nanoscale Microscopy and Molecular Physiology of the Brain, Cluster of Excellence 171-DFG Research Center 103 (CNMPB) Göttingen, Germany
| | - Lars Tönges
- Department of Neurology, University Medicine Göttingen Göttingen, Germany ; Nanoscale Microscopy and Molecular Physiology of the Brain, Cluster of Excellence 171-DFG Research Center 103 (CNMPB) Göttingen, Germany
| |
Collapse
|
319
|
Vitamin D deficiency in patients with primary immune-mediated peripheral neuropathies. J Neurol Sci 2014; 345:184-8. [DOI: 10.1016/j.jns.2014.07.040] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 07/15/2014] [Accepted: 07/17/2014] [Indexed: 12/14/2022]
|
320
|
Gonçalves M, Tillack L, de Carvalho M, Pinto S, Conradt HS, Costa J. Phosphoneurofilament heavy chain and N-glycomics from the cerebrospinal fluid in amyotrophic lateral sclerosis. Clin Chim Acta 2014; 438:342-9. [PMID: 25261856 DOI: 10.1016/j.cca.2014.09.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 08/26/2014] [Accepted: 09/10/2014] [Indexed: 01/04/2023]
Abstract
BACKGROUND Amyotrophic Lateral Sclerosis (ALS) is a fatal neurodegenerative disease of the motor neuron for which no clinically validated biomarkers have been identified. METHODS We have quantified by ELISA the biomarker phosphoneurofilament heavy chain (pNFH) in the cerebrospinal fluid (CSF) of ALS patients (n=29) and age-matched control patients with other diseases (n=19) by ELISA. Furthermore, we compared protein N-glycosylation of the CSF in ALS patients and controls, by applying a glycomics approach based on liquid chromatography and mass spectrometry. RESULTS pNFH levels were significantly higher in ALS patients in comparison with controls (P<0.0001) in particular in fast progressors. The N-glycans found in the CSF were predominantly complex diantennary with sialic acid in α2,3- and α2,6-linkage, and bisecting N-acetylglucosamine-containing structures as well as peripherally fucosylated structures were found. As compared with controls the ALS group had a significant increase of a peak composed of the monosialylated diantennary glycans A2G2S(6)1 and FA2G2S(3)1 (P=0.0348). CONCLUSIONS Our results underscore the value of pNFH as a biomarker in ALS. In addition, we identified a variation of the N-glycosylation pattern in ALS, suggesting that this change should be explored in future studies as potential biomarker.
Collapse
Affiliation(s)
- Margarida Gonçalves
- Laboratory of Glycobiology, Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal
| | - Linda Tillack
- GlycoThera GmbH, Feodor-Lynen Strasse 35, 30625 Hannover, Germany
| | - Mamede de Carvalho
- Department Neurosciences, Hospital de Santa Maria, Lisbon, Portugal; Translational Clinical Physiology Unit, Instituto de Medicina Molecular, Institute of Physiology, Faculty of Medicine - University of Lisbon, Portugal
| | - Susana Pinto
- Department Neurosciences, Hospital de Santa Maria, Lisbon, Portugal; Translational Clinical Physiology Unit, Instituto de Medicina Molecular, Institute of Physiology, Faculty of Medicine - University of Lisbon, Portugal
| | - Harald S Conradt
- GlycoThera GmbH, Feodor-Lynen Strasse 35, 30625 Hannover, Germany
| | - Júlia Costa
- Laboratory of Glycobiology, Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal.
| |
Collapse
|
321
|
Freischmidt A, Müller K, Zondler L, Weydt P, Volk AE, Božič AL, Walter M, Bonin M, Mayer B, von Arnim CAF, Otto M, Dieterich C, Holzmann K, Andersen PM, Ludolph AC, Danzer KM, Weishaupt JH. Serum microRNAs in patients with genetic amyotrophic lateral sclerosis and pre-manifest mutation carriers. ACTA ACUST UNITED AC 2014; 137:2938-50. [PMID: 25193138 DOI: 10.1093/brain/awu249] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Knowledge about the nature of pathomolecular alterations preceding onset of symptoms in amyotrophic lateral sclerosis is largely lacking. It could not only pave the way for the discovery of valuable therapeutic targets but might also govern future concepts of pre-manifest disease modifying treatments. MicroRNAs are central regulators of transcriptome plasticity and participate in pathogenic cascades and/or mirror cellular adaptation to insults. We obtained comprehensive expression profiles of microRNAs in the serum of patients with familial amyotrophic lateral sclerosis, asymptomatic mutation carriers and healthy control subjects. We observed a strikingly homogenous microRNA profile in patients with familial amyotrophic lateral sclerosis that was largely independent from the underlying disease gene. Moreover, we identified 24 significantly downregulated microRNAs in pre-manifest amyotrophic lateral sclerosis mutation carriers up to two decades or more before the estimated time window of disease onset; 91.7% of the downregulated microRNAs in mutation carriers overlapped with the patients with familial amyotrophic lateral sclerosis. Bioinformatic analysis revealed a consensus sequence motif present in the vast majority of downregulated microRNAs identified in this study. Our data thus suggest specific common denominators regarding molecular pathogenesis of different amyotrophic lateral sclerosis genes. We describe the earliest pathomolecular alterations in amyotrophic lateral sclerosis mutation carriers known to date, which provide a basis for the discovery of novel therapeutic targets and strongly argue for studies evaluating presymptomatic disease-modifying treatment in amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
| | | | - Lisa Zondler
- 1 Department of Neurology, Ulm University, Ulm, Germany
| | - Patrick Weydt
- 1 Department of Neurology, Ulm University, Ulm, Germany
| | | | | | - Michael Walter
- 4 Department of Medical Genetics, University of Tübingen, Tübingen, Germany
| | - Michael Bonin
- 4 Department of Medical Genetics, University of Tübingen, Tübingen, Germany
| | - Benjamin Mayer
- 5 Institute for Epidemiology and Medical Biometry, Ulm University, Ulm, Germany
| | | | - Markus Otto
- 1 Department of Neurology, Ulm University, Ulm, Germany
| | | | - Karlheinz Holzmann
- 6 Genomics-Core Facility, University Hospital Ulm, Centre for Biomedical Research, Ulm, Germany
| | - Peter M Andersen
- 1 Department of Neurology, Ulm University, Ulm, Germany 7 The Institute of Pharmacology and Clinical Neuroscience, Umeå University, Umeå, Sweden 8 Virtual Helmholtz Institute RNA dysmetabolism in Amyotrophic Lateral Sclerosis and Fronto-temporal Dementia, Germany
| | - Albert C Ludolph
- 1 Department of Neurology, Ulm University, Ulm, Germany 8 Virtual Helmholtz Institute RNA dysmetabolism in Amyotrophic Lateral Sclerosis and Fronto-temporal Dementia, Germany
| | | | | |
Collapse
|
322
|
Marrali G, Casale F, Salamone P, Fuda G, Caorsi C, Amoroso A, Brunetti M, Restagno G, Barberis M, Bertuzzo D, Canosa A, Moglia C, Calvo A, Chiò A. NADPH oxidase (NOX2) activity is a modifier of survival in ALS. J Neurol 2014; 261:2178-83. [PMID: 25178511 DOI: 10.1007/s00415-014-7470-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 08/08/2014] [Accepted: 08/10/2014] [Indexed: 12/11/2022]
Abstract
NADPH-oxidases (NOX) catalyze the formation of reactive oxygen species (ROS), which play a role in the development of neurological diseases, particularly those generated by the phagocytic isoform NOX2. Increased ROS has been observed in the amyotrophic lateral sclerosis (ALS) SOD1 transgenic mouse, and in this preclinical model the inactivation of NOX2 decreases ROS production and extends survival. Our aim was to evaluate NOX2 activity measuring neutrophil oxidative burst in a cohort of 83 ALS patients, and age- and gender-matched healthy controls. Oxidative burst was measured directly in fresh blood using Phagoburst™ assay by flow cytometry. Mean fluorescence intensity (MFI), emitted in response to different stimuli, leads to produce ROS and corresponds to the percentage of oxidizing cells and their enzymatic activity (GeoMean). No difference was found between the MFI values in cases and controls. NOX2 activity was independent from gender and age, and in patients was not related to disease duration, site of onset (bulbar vs. spinal), or ALSFRS-R score. However, patients with a NOX2 activity lower than the median value showed a 1-year increase of survival from onset (p = 0.011). The effect of NOX2 was independent from other known prognostic factors. These findings are in keeping with the observations in the mouse model of ALS, and demonstrate the strong role of NOX2 in modifying progression in ALS patients. A proper modulation of NOX2 activity might hold therapeutic potential for ALS.
Collapse
Affiliation(s)
- Giuseppe Marrali
- "Rita Levi Montalcini" Department of Neuroscience, ALS Centre, University of Torino, Via Cherasco 15, 10126, Turin, Italy,
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
323
|
Cerebellar integrity in the amyotrophic lateral sclerosis-frontotemporal dementia continuum. PLoS One 2014; 9:e105632. [PMID: 25144223 PMCID: PMC4140802 DOI: 10.1371/journal.pone.0105632] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 07/22/2014] [Indexed: 01/31/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and behavioural variant frontotemporal dementia (bvFTD) are multisystem neurodegenerative disorders that manifest overlapping cognitive, neuropsychiatric and motor features. The cerebellum has long been known to be crucial for intact motor function although emerging evidence over the past decade has attributed cognitive and neuropsychiatric processes to this structure. The current study set out i) to establish the integrity of cerebellar subregions in the amyotrophic lateral sclerosis-behavioural variant frontotemporal dementia spectrum (ALS-bvFTD) and ii) determine whether specific cerebellar atrophy regions are associated with cognitive, neuropsychiatric and motor symptoms in the patients. Seventy-eight patients diagnosed with ALS, ALS-bvFTD, behavioural variant frontotemporal dementia (bvFTD), most without C9ORF72 gene abnormalities, and healthy controls were investigated. Participants underwent cognitive, neuropsychiatric and functional evaluation as well as structural imaging using voxel-based morphometry (VBM) to examine the grey matter subregions of the cerebellar lobules, vermis and crus. VBM analyses revealed: i) significant grey matter atrophy in the cerebellum across the whole ALS-bvFTD continuum; ii) atrophy predominantly of the superior cerebellum and crus in bvFTD patients, atrophy of the inferior cerebellum and vermis in ALS patients, while ALS-bvFTD patients had both patterns of atrophy. Post-hoc covariance analyses revealed that cognitive and neuropsychiatric symptoms were particularly associated with atrophy of the crus and superior lobule, while motor symptoms were more associated with atrophy of the inferior lobules. Taken together, these findings indicate an important role of the cerebellum in the ALS-bvFTD disease spectrum, with all three clinical phenotypes demonstrating specific patterns of subregional atrophy that associated with different symptomology.
Collapse
|
324
|
A longer diagnostic interval is a risk for depression in amyotrophic lateral sclerosis. Palliat Support Care 2014; 13:1019-24. [DOI: 10.1017/s1478951514000881] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
AbstractObjective:Recognizing depressive symptoms in patients with amyotrophic lateral sclerosis (ALS) remains problematic given the potential overlap with the normal psychological responses to a terminal illness. Understanding mental health and disease-related risk factors for depression is key to identifying psychological morbidity. The present study aimed to determine the prevalence of depressive symptoms in ALS and to explore mental health and disease-related risk factors for depression.Method:Structured medical and psychiatric history questionnaires and a validated depression scale (Depression, Anxiety, Stress Scale–21) were completed by 27 ALS patients (60% female; 59% limb onset; age 65.11 ± SE 2.21) prior to their initial review at a multidisciplinary clinic. Physical function was assessed with the Amyotrophic Lateral Sclerosis Functional Rating Scale (ALSFRS–R).Results:At the time of initial assessment, 44% of patients had a previous psychiatric history, although the majority (62%) reported no symptoms of depression. The mean ALSFRS–R score was 37.78 ± SE 1.22, with an average diagnostic interval of 16.04 ± SE 2.39 months. Logistic regression analysis revealed that the length of the diagnostic interval alone predicted depressive symptoms (χ2(3, n = 26) = 9.21, Odds Ratio (OR) = 1.12, p < 0.05.Significance of Results:The illness experiences of ALS patients rather than established mental health risk factors influence the manifestation of depressive symptoms in the early stages of the disease, with clinical implications for the assessment and treatment of psychological morbidity. Patients with lengthy diagnostic intervals may be prime targets for psychological assessment and intervention, especially in the absence of ALS-specific tests and biomarkers.
Collapse
|
325
|
Bozik ME, Mitsumoto H, Brooks BR, Rudnicki SA, Moore DH, Zhang B, Ludolph A, Cudkowicz ME, van den Berg LH, Mather J, Petzinger T, Archibald D. A post hoc analysis of subgroup outcomes and creatinine in the phase III clinical trial (EMPOWER) of dexpramipexole in ALS. Amyotroph Lateral Scler Frontotemporal Degener 2014; 15:406-13. [PMID: 25125035 DOI: 10.3109/21678421.2014.943672] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Our objective was to compare the phase II and phase III (EMPOWER) studies of dexpramipexole in ALS and evaluate potential EMPOWER responder subgroups and biomarkers based on significant inter-study population differences. In a post hoc analysis, we compared the baseline population characteristics of both dexpramipexole studies and analyzed EMPOWER efficacy outcomes and laboratory measures in subgroups defined by significant inter-study differences. Results showed that, compared with phase II, the proportion of El Escorial criteria (EEC) definite participants decreased (p = 0.005), riluzole use increased (p = 0.002), and mean symptom duration increased (p = 0.037) significantly in EMPOWER. Baseline creatinine (p < 0.001) and on-study creatinine change (p < 0.001) correlated significantly with ALSFRS-R in EMPOWER. In the EMPOWER subgroup defined by EEC-definite ALS, riluzole use, and < median symptom duration (15.3 months), dexpramipexole-treated participants had reduced ALSFRS-R slope decline (p = 0.015), decreased mortality (p = 0.011), and reduced creatinine loss (p = 0.003). In conclusion, significant differences existed between the phase II and EMPOWER study populations in ALS clinical trials of dexpramipexole. In a post hoc analysis of EMPOWER subgroups defined by these differences, potential clinical benefits of dexpramipexole were identified in the subgroup of riluzole-treated, short-symptom duration, EEC-definite ALS participants. Creatinine loss correlated with disease progression and was reduced in dexpramipexole-treated participants, suggesting it as a candidate biomarker.
Collapse
|
326
|
Nascimento F, Pousinha PA, Correia AM, Gomes R, Sebastião AM, Ribeiro JA. Adenosine A2A receptors activation facilitates neuromuscular transmission in the pre-symptomatic phase of the SOD1(G93A) ALS mice, but not in the symptomatic phase. PLoS One 2014; 9:e104081. [PMID: 25093813 PMCID: PMC4122437 DOI: 10.1371/journal.pone.0104081] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 07/05/2014] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a neurodegenerative disease leading to motor neuron dysfunction resulting in impairment of neuromuscular transmission. A2A adenosine receptors have already been considered as a potential therapeutical target for ALS but their neuromodulatory role at the neuromuscular junction in ALS remains to be clarified. In the present work, we evaluated the effects of A2A receptors on neuromuscular transmission of an animal model of ALS: SOD1(G93A) mice either in the pre-symptomatic (4-6 weeks old) or in the symptomatic (12-14 weeks old) stage. Electrophysiological experiments were performed obtaining intracellular recordings in Mg2+ paralyzed phrenic nerve-hemidiaphragm preparations. Endplate potentials (EPPs), quantal content (q. c.) of EPPs, miniature endplate potentials (MEPPs) and giant miniature endplate potential (GMEPPs) were recorded. In the pre-symptomatic phase of the disease (4-6 weeks old mice), the selective A2A receptor agonist, CGS 21680, significantly enhanced (p<0.05 Unpaired t-test) the mean amplitude and q.c. of EPPs, and the frequency of MEPPs and GMEPPs at SOD1(G93A) neuromuscular junctions, the effect being of higher magnitude (p<0.05, Unpaired t-test) than age-matched control littermates. On the contrary, in symptomatic mice (12-14 weeks old), CGS 21680 was devoid of effect on both the amplitude and q.c. of EPPs and the frequency of MEPPs and GMEPPs (p<0.05 Paired t-test). The results herein reported clearly document that at the neuromuscular junction of SOD1(G93A) mice there is an exacerbation of A2A receptor-mediated excitatory effects at the pre-symptomatic phase, whereas in the symptomatic phase A2A receptor activation is absent. The results thus suggest that A2A receptors function changes with ALS progression.
Collapse
Affiliation(s)
- Filipe Nascimento
- Institute of Pharmacology and Neurosciences, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
- Unit of Neurosciences, Instituto de Medicina Molecular, University of Lisbon, Lisbon, Portugal
| | - Paula A. Pousinha
- Institute of Pharmacology and Neurosciences, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
- Unit of Neurosciences, Instituto de Medicina Molecular, University of Lisbon, Lisbon, Portugal
| | - Alexandra M. Correia
- Unit of Neurosciences, Instituto de Medicina Molecular, University of Lisbon, Lisbon, Portugal
- National Museum of Natural History and Science, University of Lisbon, Lisbon, Portugal
| | - Rui Gomes
- Unit of Neurosciences, Instituto de Medicina Molecular, University of Lisbon, Lisbon, Portugal
- Faculty of Sciences, University of Lisbon, Lisbon, Portugal
| | - Ana M. Sebastião
- Institute of Pharmacology and Neurosciences, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
- Unit of Neurosciences, Instituto de Medicina Molecular, University of Lisbon, Lisbon, Portugal
| | - Joaquim A. Ribeiro
- Institute of Pharmacology and Neurosciences, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
- Unit of Neurosciences, Instituto de Medicina Molecular, University of Lisbon, Lisbon, Portugal
| |
Collapse
|
327
|
Tan W, Pasinelli P, Trotti D. Role of mitochondria in mutant SOD1 linked amyotrophic lateral sclerosis. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1842:1295-301. [PMID: 24568860 PMCID: PMC4074562 DOI: 10.1016/j.bbadis.2014.02.009] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 02/13/2014] [Accepted: 02/15/2014] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease with an adult onset characterized by loss of both upper and lower motor neurons. In ~10% of cases, patients developed ALS with an apparent genetic linkage (familial ALS or fALS). Approximately 20% of fALS displays mutations in the SOD1 gene encoding superoxide dismutase 1. There are many proposed cellular and molecular mechanisms among which, mitochondrial dysfunctions occur early, prior to symptoms occurrence. In this review, we modeled the effect of mutant SOD1 protein via the formation of a toxic complex with Bcl2 on mitochondrial bioenergetics. Furthermore, we discuss that the shutdown of ATP permeation through mitochondrial outer membrane could lead to both respiration inhibition and temporary mitochondrial hyperpolarization. Moreover, we reviewed mitochondrial calcium signaling, oxidative stress, fission and fusion, autophagy and apoptosis in mutant SOD1-linked ALS. Functional defects in mitochondria appear early before symptoms are manifested in ALS. Therefore, mitochondrial dysfunction is a promising therapeutic target in ALS.
Collapse
Affiliation(s)
- Wenzhi Tan
- Frances and Joseph Weinberg Unit for ALS Research, Farber Institute for the Neurosciences, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Piera Pasinelli
- Frances and Joseph Weinberg Unit for ALS Research, Farber Institute for the Neurosciences, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Davide Trotti
- Frances and Joseph Weinberg Unit for ALS Research, Farber Institute for the Neurosciences, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
328
|
Increased IL-17, a Pathogenic Link between Hepatosplenic Schistosomiasis and Amyotrophic Lateral Sclerosis: A Hypothesis. Case Reports Immunol 2014; 2014:804761. [PMID: 25379310 PMCID: PMC4207377 DOI: 10.1155/2014/804761] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Accepted: 07/15/2014] [Indexed: 01/05/2023] Open
Abstract
The immune system protects the organism from foreign invaders and foreign substances and is involved in physiological functions that range from tissue repair to neurocognition. However, an excessive or dysregulated immune response can cause immunopathology and disease. A 39-year-old man was affected by severe hepatosplenic schistosomiasis mansoni and by amyotrophic lateral sclerosis. One question that arose was, whether there was a relation between the parasitic and the neurodegenerative disease. IL-17, a proinflammatory cytokine, is produced mainly by T helper-17 CD4 cells, a recently discovered new lineage of effector CD4 T cells. Experimental mouse models of schistosomiasis have shown that IL-17 is a key player in the immunopathology of schistosomiasis. There are also reports that suggest that IL-17 might have an important role in the pathogenesis of amyotrophic lateral sclerosis. It is hypothesized that the factors that might have led to increased IL-17 in the hepatosplenic schistosomiasis mansoni might also have contributed to the development of amyotrophic lateral sclerosis in the described patient. A multitude of environmental factors, including infections, xenobiotic substances, intestinal microbiota, and vitamin D deficiency, that are able to induce a proinflammatory immune response polarization, might favor the development of amyotrophic lateral sclerosis in predisposed individuals.
Collapse
|
329
|
Wolf J, Safer A, Wöhrle JC, Palm F, Nix WA, Maschke M, Grau AJ. Variability and prognostic relevance of different phenotypes in amyotrophic lateral sclerosis - data from a population-based registry. J Neurol Sci 2014; 345:164-7. [PMID: 25086855 DOI: 10.1016/j.jns.2014.07.033] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 07/14/2014] [Indexed: 12/11/2022]
Abstract
OBJECTIVES The clinical spectrum of amyotrophic lateral sclerosis (ALS) is characterized by a considerable variation. Different phenotypes have been described by previous studies. We assessed clinical variability and prognostic relevance of these phenotypes in a prospective, population-based cohort of ALS patients in Rhineland-Palatinate, Germany. METHODS Incident ALS cases, diagnosed between October 2009 and September 2012, were prospectively enrolled and classified according to established ALS phenotype classification (bulbar, classic, flail arm, flail leg, pyramidal, respiratory). Survival probability was described using Kaplan-Meier method. Moreover, the influence of an additional frontotemporal dementia (FTD) was analysed. RESULTS Phenotypes of all 200 patients were determined. Bulbar and classic phenotypes accounted for 75% of all cases. Deterioration of functional impairment during disease progression was lowest in flail leg and pyramidal variants, and most pronounced in bulbar and classic phenotypes. A poor survival prognosis was observed for bulbar, classic or respiratory phenotypes. Patients with an additional FTD showed an even worse outcome. CONCLUSIONS Results suggest that ALS is a heterogeneous disease, as ALS phenotypes differ in disease progression and survival time. Patients classified as suffering from bulbar, classic and respiratory ALS, as well as those with an additional FTD, show a marked reduction of survival time.
Collapse
Affiliation(s)
- Joachim Wolf
- Department of Neurology, Klinikum der Stadt Ludwigshafen, Germany.
| | - Anton Safer
- Institute of Public Health, Medical Faculty, Ruprecht-Karls-University, Heidelberg, Germany
| | - Johannes C Wöhrle
- Department of Neurology, Katholisches Klinikum, Brüderhaus, Koblenz, Germany
| | - Frederick Palm
- Department of Neurology, Klinikum der Stadt Ludwigshafen, Germany
| | - Wilfred A Nix
- Department of Neurology, Universitätsmedizin, Mainz, Germany
| | - Matthias Maschke
- Department of Neurology, Krankenhaus der Barmherzigen Brüder, Trier, Germany
| | - Armin J Grau
- Department of Neurology, Klinikum der Stadt Ludwigshafen, Germany
| |
Collapse
|
330
|
Milan L, Barrière G, De Deurwaerdère P, Cazalets JR, Bertrand SS. Monoaminergic control of spinal locomotor networks in SOD1G93A newborn mice. Front Neural Circuits 2014; 8:77. [PMID: 25071458 PMCID: PMC4081764 DOI: 10.3389/fncir.2014.00077] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 06/18/2014] [Indexed: 12/11/2022] Open
Abstract
Mutations in the gene that encodes Cu/Zn-superoxide dismutase (SOD1) are the cause of approximately 20% of familial forms of amyotrophic lateral sclerosis (ALS), a fatal neurodegenerative disease characterized by the progressive loss of motor neurons. While ALS symptoms appear in adulthood, spinal motoneurons exhibit functional alterations as early as the embryonic and postnatal stages in the murine model of ALS, the SOD1 mice. Monoaminergic - i.e., dopaminergic (DA), serotoninergic (5-HT), and noradrenergic (NA) - pathways powerfully control spinal networks and contribute significantly to their embryonic and postnatal maturation. Alterations in monoaminergic neuromodulation during development could therefore lead to impairments in the motoneuronal physiology. In this study, we sought to determine whether the monoaminergic spinal systems are modified in the early stages of development in SOD1 mice. Using a post-mortem analysis by high performance liquid chromatography (HPLC), monoaminergic neuromodulators and their metabolites were quantified in the lumbar spinal cord of SOD1 and wild-type (WT) mice aged one postnatal day (P1) and P10. This analysis underscores an increased content of DA in the SOD1 lumbar spinal cord compared to that of WT mice but failed to reveal any modification of the other monoaminergic contents. In a next step, we compared the efficiency of the monoaminergic compounds in triggering and modulating fictive locomotion in WT and SOD1 mice. This study was performed in P1-P3 SOD1 mice and age-matched control littermates using extracellular recordings from the lumbar ventral roots in the in vitro isolated spinal cord preparation. This analysis revealed that the spinal networks of SOD1(G93A) mice could generate normal locomotor activity in the presence of NMA-5-HT. Interestingly, we also observed that SOD1 spinal networks have an increased sensitivity to NA compared to WT spinal circuits but exhibited similar DA responses.
Collapse
Affiliation(s)
- Léa Milan
- CNRS, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, UMR5287, Université de Bordeaux Bordeaux, France
| | - Grégory Barrière
- CNRS, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, UMR5287, Université de Bordeaux Bordeaux, France
| | | | - Jean-René Cazalets
- CNRS, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, UMR5287, Université de Bordeaux Bordeaux, France
| | - Sandrine S Bertrand
- CNRS, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, UMR5287, Université de Bordeaux Bordeaux, France
| |
Collapse
|
331
|
SQSTM1 mutations – Bridging Paget disease of bone and ALS/FTLD. Exp Cell Res 2014; 325:27-37. [DOI: 10.1016/j.yexcr.2014.01.020] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 01/17/2014] [Accepted: 01/21/2014] [Indexed: 12/14/2022]
|
332
|
Side of limb-onset predicts laterality of gray matter loss in amyotrophic lateral sclerosis. BIOMED RESEARCH INTERNATIONAL 2014; 2014:473250. [PMID: 25093168 PMCID: PMC4100370 DOI: 10.1155/2014/473250] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 06/03/2014] [Indexed: 11/17/2022]
Abstract
Conflicting findings have been reported regarding the lateralized brain abnormality in patients with amyotrophic lateral sclerosis (ALS). In this study, we aimed to investigate the probable lateralization of gray matter (GM) atrophy in ALS patients. We focused on the relationship between the asymmetry in decreased GM volume and the side of disease onset in patients with limb-onset. Structural imaging evaluation of normalized atrophy (SIENAX) and voxel-based morphometry (VBM) were used to assess differences in global and local brain regions in patients with heterogeneous body onset and subgroups with different side of limb-onset. We found global brain atrophy and GM losses in the frontal and parietal areas in each patient group as well as left predominant GM losses in the total cohort. The intriguing findings in subgroup analyses demonstrated that the motor cortex in the contralateral hemisphere of the initially involved limb was most affected. We also found that regional brain atrophy was related to disease progression rate. Our observations suggested that side of limb-onset can predict laterality of GM loss in ALS patients and disease progression correlates with the extent of cortical abnormality.
Collapse
|
333
|
Body mass index delineates ALS from FTD: implications for metabolic health. J Neurol 2014; 261:1774-80. [DOI: 10.1007/s00415-014-7416-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 06/12/2014] [Accepted: 06/13/2014] [Indexed: 12/12/2022]
|
334
|
Menke RAL, Körner S, Filippini N, Douaud G, Knight S, Talbot K, Turner MR. Widespread grey matter pathology dominates the longitudinal cerebral MRI and clinical landscape of amyotrophic lateral sclerosis. ACTA ACUST UNITED AC 2014; 137:2546-55. [PMID: 24951638 PMCID: PMC4132644 DOI: 10.1093/brain/awu162] [Citation(s) in RCA: 141] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Menke/Koerner et al. use structural MRI to explore the extent of longitudinal changes in cerebral pathology in amyotrophic lateral sclerosis, and their relationship to clinical features. A characteristic white matter tract pathological signature is seen cross-sectionally, while cortical involvement dominates longitudinally. This has implications for the development of biomarkers for diagnosis versus therapeutic monitoring. Diagnosis, stratification and monitoring of disease progression in amyotrophic lateral sclerosis currently rely on clinical history and examination. The phenotypic heterogeneity of amyotrophic lateral sclerosis, including extramotor cognitive impairments is now well recognized. Candidate biomarkers have shown variable sensitivity and specificity, and studies have been mainly undertaken only cross-sectionally. Sixty patients with sporadic amyotrophic lateral sclerosis (without a family history of amyotrophic lateral sclerosis or dementia) underwent baseline multimodal magnetic resonance imaging at 3 T. Grey matter pathology was identified through analysis of T1-weighted images using voxel-based morphometry. White matter pathology was assessed using tract-based spatial statistics analysis of indices derived from diffusion tensor imaging. Cross-sectional analyses included group comparison with a group of healthy controls (n = 36) and correlations with clinical features, including regional disability, clinical upper motor neuron signs and cognitive impairment. Patients were offered 6-monthly follow-up MRI, and the last available scan was used for a separate longitudinal analysis (n = 27). In cross-sectional study, the core signature of white matter pathology was confirmed within the corticospinal tract and callosal body, and linked strongly to clinical upper motor neuron burden, but also to limb disability subscore and progression rate. Localized grey matter abnormalities were detected in a topographically appropriate region of the left motor cortex in relation to bulbar disability, and in Broca’s area and its homologue in relation to verbal fluency. Longitudinal analysis revealed progressive and widespread changes in the grey matter, notably including the basal ganglia. In contrast there was limited white matter pathology progression, in keeping with a previously unrecognized limited change in individual clinical upper motor neuron scores, despite advancing disability. Although a consistent core white matter pathology was found cross-sectionally, grey matter pathology was dominant longitudinally, and included progression in clinically silent areas such as the basal ganglia, believed to reflect their wider cortical connectivity. Such changes were significant across a range of apparently sporadic patients rather than being a genotype-specific effect. It is also suggested that the upper motor neuron lesion in amyotrophic lateral sclerosis may be relatively constant during the established symptomatic period. These findings have implications for the development of effective diagnostic versus therapeutic monitoring magnetic resonance imaging biomarkers. Amyotrophic lateral sclerosis may be characterized initially by a predominantly white matter tract pathological signature, evolving as a widespread cortical network degeneration.
Collapse
Affiliation(s)
- Ricarda A L Menke
- 1 Oxford Centre for Functional Magnetic Resonance of the Brain, University of Oxford, UK2 Nuffield Department of Clinical Neurosciences, University of Oxford, UK
| | - Sonja Körner
- 3 Department of Neurology, Hannover Medical School, Germany
| | - Nicola Filippini
- 1 Oxford Centre for Functional Magnetic Resonance of the Brain, University of Oxford, UK4 Department of Psychiatry, University of Oxford, UK
| | - Gwenaëlle Douaud
- 1 Oxford Centre for Functional Magnetic Resonance of the Brain, University of Oxford, UK2 Nuffield Department of Clinical Neurosciences, University of Oxford, UK
| | - Steven Knight
- 5 Oxford Centre for Magnetic Resonance Imaging Research, University of Oxford, UK
| | - Kevin Talbot
- 2 Nuffield Department of Clinical Neurosciences, University of Oxford, UK
| | - Martin R Turner
- 1 Oxford Centre for Functional Magnetic Resonance of the Brain, University of Oxford, UK2 Nuffield Department of Clinical Neurosciences, University of Oxford, UK5 Oxford Centre for Magnetic Resonance Imaging Research, University of Oxford, UK
| |
Collapse
|
335
|
Grossman M, Elman L, McCluskey L, McMillan CT, Boller A, Powers J, Rascovsky K, Hu W, Shaw L, Irwin DJ, Lee VMY, Trojanowski JQ. Phosphorylated tau as a candidate biomarker for amyotrophic lateral sclerosis. JAMA Neurol 2014; 71:442-8. [PMID: 24492862 DOI: 10.1001/jamaneurol.2013.6064] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
IMPORTANCE An increasingly varied clinical spectrum of cases with amyotrophic lateral sclerosis (ALS) has been identified, and objective criteria for clinical trial eligibility are necessary. OBJECTIVE To develop a cerebrospinal fluid (CSF) biomarker sensitive and specific for the diagnosis of ALS. DESIGN, SETTING, AND PARTICIPANTS A case-control study including 51 individuals with ALS and 23 individuals with a disorder associated with a 4-repeat tauopathy was conducted at an academic medical center. MAIN OUTCOMES AND MEASURES The CSF level of tau phosphorylated at threonine 181 (ptau) and ratio of ptau to total tau (ttau). RESULTS Using a cross-validation prediction procedure, we found significantly reduced CSF levels of ptau and the ptau:ttau ratio in ALS relative to 4-repeat tauopathy and to controls. In the validation cohort, the receiver operating characteristic area under the curve for the ptau:ttau ratio was 0.916, and the comparison of ALS with 4-repeat tauopathy showed 92.0% sensitivity and 91.7% specificity. Correct classification based on a low CSF ptau:ttau ratio was confirmed in 18 of 21 cases (86%) with autopsy-proved or genetically determined disease. In patients with available measures, ptau:ttau in ALS correlated with clinical measures of disease severity, such as the Mini-Mental State Examination (n = 51) and ALS Functional Rating Scale-Revised (n = 42), and regression analyses related the ptau:ttau ratio to magnetic resonance imaging (n = 10) evidence of disease in the corticospinal tract and white matter projections involving the prefrontal cortex. CONCLUSIONS AND RELEVANCE The CSF ptau:ttau ratio may be a candidate biomarker to provide objective support for the diagnosis of ALS.
Collapse
Affiliation(s)
- Murray Grossman
- Penn Frontotemporal Degeneration Center, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Lauren Elman
- Penn Frontotemporal Degeneration Center, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Leo McCluskey
- Penn Frontotemporal Degeneration Center, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Corey T McMillan
- Penn Frontotemporal Degeneration Center, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Ashley Boller
- Penn Frontotemporal Degeneration Center, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - John Powers
- Penn Frontotemporal Degeneration Center, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Katya Rascovsky
- Penn Frontotemporal Degeneration Center, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - William Hu
- Department of Neurology, Emory University, Atlanta, Georgia
| | - Les Shaw
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - David J Irwin
- Penn Frontotemporal Degeneration Center, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia3Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medi
| | - Virginia M-Y Lee
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - John Q Trojanowski
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
336
|
McLean JR, Smith GA, Rocha EM, Osborn TM, Dib S, Hayes MA, Beagan JA, Brown TB, Lawson TFS, Hallett PJ, Robertson J, Isacson O. ALS-associated peripherin spliced transcripts form distinct protein inclusions that are neuroprotective against oxidative stress. Exp Neurol 2014; 261:217-29. [PMID: 24907400 DOI: 10.1016/j.expneurol.2014.05.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 05/16/2014] [Accepted: 05/26/2014] [Indexed: 01/27/2023]
Abstract
Intracellular proteinaceous inclusions are well-documented hallmarks of the fatal motor neuron disorder amyotrophic lateral sclerosis (ALS). The pathological significance of these inclusions remains unknown. Peripherin, a type III intermediate filament protein, is upregulated in ALS and identified as a component within different types of ALS inclusions. The formation of these inclusions may be associated with abnormal peripherin splicing, whereby an increase in mRNA retaining introns 3 and 4 (Per-3,4) leads to the generation of an aggregation-prone isoform, Per-28. During the course of evaluating peripherin filament assembly in SW-13 cells, we identified that expression of both Per-3,4 and Per-28 transcripts formed inclusions with categorically distinct morphology: Per-3,4 was associated with cytoplasmic condensed/bundled filaments, small inclusions (<10μM), or large inclusions (≥10μM); while Per-28 was associated with punctate inclusions in the nucleus and/or cytoplasm. We found temporal and spatial changes in inclusion morphology between 12 and 48h post-transfected cells, which were accompanied by unique immunofluorescent and biochemical changes of other ALS-relevant proteins, including TDP-43 and ubiquitin. Despite mild cytotoxicity associated with peripherin transfection, Per-3,4 and Per-28 expression increased cell viability during H2O2-mediated oxidative stress in BE(2)-M17 neuroblastoma cells. Taken together, this study shows that ALS-associated peripherin isoforms form dynamic cytoplasmic and intranuclear inclusions, effect changes in local endogenous protein expression, and afford cytoprotection against oxidative stress. These findings may have important relevance to understanding the pathophysiological role of inclusions in ALS.
Collapse
Affiliation(s)
- Jesse R McLean
- Neuroregeneration Research Institute, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - Gaynor A Smith
- Neuroregeneration Research Institute, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - Emily M Rocha
- Neuroregeneration Research Institute, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - Teresia M Osborn
- Neuroregeneration Research Institute, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - Samar Dib
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Melissa A Hayes
- Neuroregeneration Research Institute, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - Jonathan A Beagan
- Neuroregeneration Research Institute, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - Tana B Brown
- Neuroregeneration Research Institute, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - Tristan F S Lawson
- Neuroregeneration Research Institute, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - Penelope J Hallett
- Neuroregeneration Research Institute, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - Janice Robertson
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Ole Isacson
- Neuroregeneration Research Institute, McLean Hospital, Harvard Medical School, Belmont, MA, USA.
| |
Collapse
|
337
|
Pinto S, Swash M, de Carvalho M. Does surgery accelerate progression of amyotrophic lateral sclerosis? J Neurol Neurosurg Psychiatry 2014; 85:643-6. [PMID: 23922387 DOI: 10.1136/jnnp-2013-305770] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Surgery is not a recognised potential amyotrophic lateral sclerosis (ALS) risk factor that might modify the onset or course of ALS. METHODS We studied our database of ALS patients, which includes questions concerning surgical procedures. We defined surgery as an operative procedure requiring general or regional anaesthesia, but not local anaesthesia. Patients were classified as G1-no surgery; G2-surgery performed ≥3 months before disease onset; G3-surgery <3 months before disease onset; and G4-surgery after disease onset. The ALS-FRS score was evaluated every 3 months from presentation. The maximal ALS-FRS score was ascribed to disease onset, itself defined as symptom onset. RESULTS 657 patients with ALS were studied. In G3 there was a positive correlation between onset-region and surgery-region (p=0.032). In G4, 35 (57.6%) patients had surgery, probably due to initial misdiagnosis. The rate of functional change (%) in G4 was significantly greater in the 3-month period immediately after surgery as compared with the 3-month period before (1.46%±1.35 vs. 6.30%±8.10, p=0.005) and the following 3 months (3.30%±3.10, p=0.006). CONCLUSIONS The site of surgery before ALS onset correlates with the region of onset of ALS. Patients with slower disease progression are at an increased risk of undergoing surgery, probably as part of initial difficulty in diagnosis. We noted accelerated disease progression during the 3-month period after surgery. Definite diagnosis is important to avoid unnecessary surgical trauma and subsequent more rapid deterioration.
Collapse
Affiliation(s)
- Susana Pinto
- Translational Clinical Physiology Unit, Faculty of Medicine, Instituto de Medicina Molecular, Institute of Physiology, University of Lisbon, , Lisbon, Portugal
| | | | | |
Collapse
|
338
|
Ismailov SM, Barykova YA, Shmarov MM, Tarantul VZ, Barskov IV, Kucheryanu VG, Brylev LV, Logunov DY, Tutykhina IL, Bocharov EV, Zakharova MN, Narodizky BS, Illarioshkin SN. Experimental approach to the gene therapy of motor neuron disease with the use of genes hypoxia-inducible factors. RUSS J GENET+ 2014. [DOI: 10.1134/s1022795414050056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
339
|
Gratten J, Wray NR, Keller MC, Visscher PM. Large-scale genomics unveils the genetic architecture of psychiatric disorders. Nat Neurosci 2014; 17:782-90. [PMID: 24866044 DOI: 10.1038/nn.3708] [Citation(s) in RCA: 286] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 03/27/2014] [Indexed: 12/11/2022]
Abstract
Family study results are consistent with genetic effects making substantial contributions to risk of psychiatric disorders such as schizophrenia, yet robust identification of specific genetic variants that explain variation in population risk had been disappointing until the advent of technologies that assay the entire genome in large samples. We highlight recent progress that has led to a better understanding of the number of risk variants in the population and the interaction of allele frequency and effect size. The emerging genetic architecture implies a large number of contributing loci (that is, a high genome-wide mutational target) and suggests that genetic risk of psychiatric disorders involves the combined effects of many common variants of small effect, as well as rare and de novo variants of large effect. The capture of a substantial proportion of genetic risk facilitates new study designs to investigate the combined effects of genes and the environment.
Collapse
Affiliation(s)
- Jacob Gratten
- 1] Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia. [2] These authors contributed equally to this work
| | - Naomi R Wray
- 1] Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia. [2] These authors contributed equally to this work
| | - Matthew C Keller
- 1] Department of Psychology and Neuroscience, University of Colorado, Boulder, Colorado, USA. [2] These authors contributed equally to this work
| | - Peter M Visscher
- 1] Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia. [2] The Translational Research Institute, University of Queensland Diamantina Institute, Brisbane, Queensland, Australia. [3] These authors contributed equally to this work
| |
Collapse
|
340
|
Mioshi E, Hsieh S, Caga J, Ramsey E, Chen K, Lillo P, Simon N, Vucic S, Hornberger M, Hodges JR, Kiernan MC. A novel tool to detect behavioural symptoms in ALS. Amyotroph Lateral Scler Frontotemporal Degener 2014; 15:298-304. [DOI: 10.3109/21678421.2014.896927] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
341
|
Brites D, Vaz AR. Microglia centered pathogenesis in ALS: insights in cell interconnectivity. Front Cell Neurosci 2014; 8:117. [PMID: 24904276 PMCID: PMC4033073 DOI: 10.3389/fncel.2014.00117] [Citation(s) in RCA: 160] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 04/10/2014] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is the most common and most aggressive form of adult motor neuron (MN) degeneration. The cause of the disease is still unknown, but some protein mutations have been linked to the pathological process. Loss of upper and lower MNs results in progressive muscle paralysis and ultimately death due to respiratory failure. Although initially thought to derive from the selective loss of MNs, the pathogenic concept of non-cell-autonomous disease has come to the forefront for the contribution of glial cells in ALS, in particular microglia. Recent studies suggest that microglia may have a protective effect on MN in an early stage. Conversely, activated microglia contribute and enhance MN death by secreting neurotoxic factors, and impaired microglial function at the end-stage may instead accelerate disease progression. However, the nature of microglial–neuronal interactions that lead to MN degeneration remains elusive. We review the contribution of the neurodegenerative network in ALS pathology, with a special focus on each glial cell type from data obtained in the transgenic SOD1G93A rodents, the most widely used model. We further discuss the diverse roles of neuroinflammation and microglia phenotypes in the modulation of ALS pathology. We provide information on the processes associated with dysfunctional cell–cell communication and summarize findings on pathological cross-talk between neurons and astroglia, and neurons and microglia, as well as on the spread of pathogenic factors. We also highlight the relevance of neurovascular disruption and exosome trafficking to ALS pathology. The harmful and beneficial influences of NG2 cells, oligodendrocytes and Schwann cells will be discussed as well. Insights into the complex intercellular perturbations underlying ALS, including target identification, will enhance our efforts to develop effective therapeutic approaches for preventing or reversing symptomatic progression of this devastating disease.
Collapse
Affiliation(s)
- Dora Brites
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa Lisbon, Portugal ; Department of Biochemistry and Human Biology, Faculdade de Farmácia, Universidade de Lisboa Lisbon, Portugal
| | - Ana R Vaz
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa Lisbon, Portugal ; Department of Biochemistry and Human Biology, Faculdade de Farmácia, Universidade de Lisboa Lisbon, Portugal
| |
Collapse
|
342
|
Sako W, Ishimoto S. Can cystatin C in cerebrospinal fluid be a biomarker for amyotrophic lateral sclerosis? A lesson from previous studies. ACTA ACUST UNITED AC 2014. [DOI: 10.1111/ncn3.82] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Wataru Sako
- Center for Neurosciences; The Feinstein Institute for Medical Research; Manhasset NY USA
| | - Shinji Ishimoto
- Department of Neurology; Wakayama Rosai Hospital; Wakayama Japan
| |
Collapse
|
343
|
Meininger V, Pradat PF, Corse A, Al-Sarraj S, Rix Brooks B, Caress JB, Cudkowicz M, Kolb SJ, Lange D, Leigh PN, Meyer T, Milleri S, Morrison KE, Orrell RW, Peters G, Rothstein JD, Shefner J, Lavrov A, Williams N, Overend P, Price J, Bates S, Bullman J, Krull D, Berges A, Abila B, Meno-Tetang G, Wurthner J. Safety, pharmacokinetic, and functional effects of the nogo-a monoclonal antibody in amyotrophic lateral sclerosis: a randomized, first-in-human clinical trial. PLoS One 2014; 9:e97803. [PMID: 24841795 PMCID: PMC4026380 DOI: 10.1371/journal.pone.0097803] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 04/22/2014] [Indexed: 01/11/2023] Open
Abstract
UNLABELLED The neurite outgrowth inhibitor, Nogo-A, has been shown to be overexpressed in skeletal muscle in amyotrophic lateral sclerosis (ALS); it is both a potential biomarker and therapeutic target. We performed a double-blind, two-part, dose-escalation study, in subjects with ALS, assessing safety, pharmacokinetics (PK) and functional effects of ozanezumab, a humanized monoclonal antibody against Nogo-A. In Part 1, 40 subjects were randomized (3∶1) to receive single dose intravenous ozanezumab (0.01, 0.1, 1, 5, or 15 mg/kg) or placebo. In Part 2, 36 subjects were randomized (3∶1) to receive two repeat doses of intravenous ozanezumab (0.5, 2.5, or 15 mg/kg) or placebo, approximately 4 weeks apart. The primary endpoints were safety and tolerability (adverse events [AEs], vital signs, electrocardiogram (ECG), and clinical laboratory tests). Secondary endpoints included PK, immunogenicity, functional endpoints (clinical and electrophysiological), and biomarker parameters. Overall, ozanezumab treatment (0.01-15 mg/kg) was well tolerated. The overall incidence of AEs in the repeat dose 2.5 mg/kg and 15 mg/kg ozanezumab groups was higher than in the repeat dose placebo group and repeat dose 0.5 mg/kg ozanezumab group. The majority were considered not related to study drug by the investigators. Six serious AEs were reported in three subjects receiving ozanezumab; none were considered related to study drug. No study drug-related patterns were identified for ECG, laboratory, or vital signs parameters. One subject (repeat dose 15 mg/kg ozanezumab) showed a weak, positive anti-ozanezumab-antibody result. PK results were generally consistent with monoclonal antibody treatments. No apparent treatment effects were observed for functional endpoints or muscle biomarkers. Immunohistochemical staining showed dose-dependent co-localization of ozanezumab with Nogo-A in skeletal muscle. In conclusion, single and repeat dose ozanezumab treatment was well tolerated and demonstrated co-localization at the site of action. These findings support future studies with ozanezumab in ALS. TRIAL REGISTRATION ClinicalTrials.gov NCT00875446 GSK-ClinicalStudyRegister.com GSK ID 111330.
Collapse
MESH Headings
- Administration, Intravenous
- Amyotrophic Lateral Sclerosis/drug therapy
- Amyotrophic Lateral Sclerosis/metabolism
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/adverse effects
- Antibodies, Monoclonal/pharmacokinetics
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antibodies, Monoclonal, Humanized/pharmacology
- Biomarkers/metabolism
- Dose-Response Relationship, Drug
- Female
- Humans
- Immunohistochemistry
- Male
- Middle Aged
- Myelin Proteins/metabolism
- Nogo Proteins
Collapse
Affiliation(s)
- Vincent Meininger
- Département des Maladies du Système Nerveux, Assistance Publique – Hôpitaux de Paris, Centre de Référence Maladies Rares SLA, Groupe Hospitalier Pitié-Salpêtrière, Université Pierre-et-Marie-Curie, Paris, France
| | - Pierre-François Pradat
- Département des Maladies du Système Nerveux, Assistance Publique – Hôpitaux de Paris, Centre de Référence Maladies Rares SLA, Groupe Hospitalier Pitié-Salpêtrière, Université Pierre-et-Marie-Curie, Paris, France
- Unité Mixte de Recherche-678, Institut National de la Santé et de la Recherche Médicale - Université Pierre-et-Marie-Curie, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - Andrea Corse
- Neuromuscular Pathology Lab, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Safa Al-Sarraj
- Department of Clinical Neuropathology, Kings College Hospital/Kings College London, London, United Kingdom
| | - Benjamin Rix Brooks
- Carolinas Neuromuscular/Amyotrophic Lateral Sclerosis-Muscular Dystrophy Association Center, Department of Neurology, Carolinas Medical Center and University of North Carolina School of Medicine-Charlotte Campus, Charlotte, North Carolina, United States of America
| | - James B. Caress
- Wake Forest School of Medicine, M Reynolds Tower, Medical Center Boulevard, Winston-Salem, North Carolina, United States of America
| | - Merit Cudkowicz
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Stephen J. Kolb
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Dale Lange
- Department of Neurology, Weill Cornell School of Medicine, New York, New York, United States of America
| | - P. Nigel Leigh
- Trafford Centre for Biomedical Research, Brighton and Sussex Medical School, Falmer, Sussex, United Kingdom
| | - Thomas Meyer
- Department of Neurology, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Stefano Milleri
- Centro Ricerche Cliniche, University Hospital G.B. Rossi, Verona, Italy
| | - Karen E. Morrison
- School of Clinical and Experimental Medicine, University of Birmingham and Neurosciences Department, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - Richard W. Orrell
- Department of Clinical Neuroscience, Institute of Neurology, University College London, London, United Kingdom
- Department of Neurology, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Gary Peters
- GlaxoSmithKline Clinical Unit Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Jeffrey D. Rothstein
- Brain Science Institute, Johns Hopkins University, Department of Neurology, Baltimore, Maryland, United States of America
| | - Jeremy Shefner
- Department of Neurology, SUNY Upstate Medical University, Syracuse, New York, United States of America
| | - Arseniy Lavrov
- Neurosciences Therapy Area Unit, Medicines Discovery and Development, GlaxoSmithKline, Stockley Park, Uxbridge, Middlesex, United Kingdom
| | - Nicola Williams
- Clinical Statistics, GlaxoSmithKline, Stevenage, Hertfordshire, United Kingdom
| | - Phil Overend
- Clinical Statistics, GlaxoSmithKline, Stevenage, Hertfordshire, United Kingdom
| | - Jeffrey Price
- Clinical Pharmacology, Science and Study Operations, BioPharm and Infectious Diseases, GlaxoSmithKline, Stevenage, Hertfordshire, United Kingdom
| | - Stewart Bates
- BioPharm Translational Medicine, GlaxoSmithKline, Stevenage, Hertfordshire, United Kingdom
| | - Jonathan Bullman
- Clinical Pharmacology Modelling & Simulation (Neurosciences), GlaxoSmithKline, Stevenage, Hertfordshire, United Kingdom
| | - David Krull
- Safety Assessment, GlaxoSmithKline, Research Triangle Park, North Carolina, United States of America
| | - Alienor Berges
- Clinical Pharmacology Modelling & Simulation, GlaxoSmithKline, Stockley Park, Uxbridge, Middlesex, United Kingdom
| | - Bams Abila
- BioPharm Translational Medicine, GlaxoSmithKline, Stevenage, Hertfordshire, United Kingdom
| | - Guy Meno-Tetang
- Clinical Pharmacology Modelling & Simulation, GlaxoSmithKline, Stockley Park, Uxbridge, Middlesex, United Kingdom
| | - Jens Wurthner
- Oncology Translational Medicine, Novartis Basel, Switzerland
| |
Collapse
|
344
|
Devine MS, Kiernan MC, Heggie S, McCombe PA, Henderson RD. Study of motor asymmetry in ALS indicates an effect of limb dominance on onset and spread of weakness, and an important role for upper motor neurons. Amyotroph Lateral Scler Frontotemporal Degener 2014; 15:481-7. [DOI: 10.3109/21678421.2014.906617] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Matthew S. Devine
- Department of Neurology, Royal Brisbane and Women’s Hospital,
Herston, QLD, Australia
- The University of Queensland,
St Lucia, QLD, Australia
| | - Matthew C. Kiernan
- Neuroscience Research Australia,
Randwick, NSW, Australia
- Prince of Wales Clinical School, University of New South Wales,
NSW, Australia
| | - Susan Heggie
- Department of Neurology, Royal Brisbane and Women’s Hospital,
Herston, QLD, Australia
| | - Pamela A. McCombe
- Department of Neurology, Royal Brisbane and Women’s Hospital,
Herston, QLD, Australia
- The University of Queensland,
St Lucia, QLD, Australia
| | - Robert D. Henderson
- Department of Neurology, Royal Brisbane and Women’s Hospital,
Herston, QLD, Australia
| |
Collapse
|
345
|
|
346
|
Miquel E, Cassina A, Martínez-Palma L, Souza JM, Bolatto C, Rodríguez-Bottero S, Logan A, Smith RAJ, Murphy MP, Barbeito L, Radi R, Cassina P. Neuroprotective effects of the mitochondria-targeted antioxidant MitoQ in a model of inherited amyotrophic lateral sclerosis. Free Radic Biol Med 2014; 70:204-13. [PMID: 24582549 DOI: 10.1016/j.freeradbiomed.2014.02.019] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 02/10/2014] [Accepted: 02/17/2014] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder characterized by motor neuron degeneration that ultimately results in progressive paralysis and death. Growing evidence indicates that mitochondrial dysfunction and oxidative stress contribute to motor neuron degeneration in ALS. To further explore the hypothesis that mitochondrial dysfunction and nitroxidative stress contribute to disease pathogenesis at the in vivo level, we assessed whether the mitochondria-targeted antioxidant [10-(4,5-dimethoxy-2-methyl-3,6-dioxo-1,4-cyclohexadien-1-yl)decyl]triphenylphosphonium methane sulfonate (MitoQ) can modify disease progression in the SOD1(G93A) mouse model of ALS. To do this, we administered MitoQ (500 µM) in the drinking water of SOD1(G93A) mice from a time when early symptoms of neurodegeneration become evident at 90 days of age until death. This regime is a clinically plausible scenario and could be more easily translated to patients as this corresponds to initiating treatment of patients after they are first diagnosed with ALS. MitoQ was detected in all tested tissues by liquid chromatography/mass spectrometry after 20 days of administration. MitoQ treatment slowed the decline of mitochondrial function, in both the spinal cord and the quadriceps muscle, as measured by high-resolution respirometry. Importantly, nitroxidative markers and pathological signs in the spinal cord of MitoQ-treated animals were markedly reduced and neuromuscular junctions were recovered associated with a significant increase in hindlimb strength. Finally, MitoQ treatment significantly prolonged the life span of SOD1(G93A) mice. Our results support a role for mitochondrial nitroxidative damage and dysfunction in the pathogenesis of ALS and suggest that mitochondria-targeted antioxidants may be of pharmacological use for ALS treatment.
Collapse
Affiliation(s)
- Ernesto Miquel
- Departamento de Histología y Embriología, Facultad de Medicina, Universidad de la República, 11800 Montevideo, Uruguay
| | - Adriana Cassina
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, 11800 Montevideo, Uruguay; Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, 11800 Montevideo, Uruguay
| | - Laura Martínez-Palma
- Departamento de Histología y Embriología, Facultad de Medicina, Universidad de la República, 11800 Montevideo, Uruguay
| | - José M Souza
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, 11800 Montevideo, Uruguay; Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, 11800 Montevideo, Uruguay
| | - Carmen Bolatto
- Departamento de Histología y Embriología, Facultad de Medicina, Universidad de la República, 11800 Montevideo, Uruguay
| | - Sebastián Rodríguez-Bottero
- Departamento de Histología y Embriología, Facultad de Medicina, Universidad de la República, 11800 Montevideo, Uruguay
| | - Angela Logan
- Mitochondrial Biology Unit, Medical Research Council, Cambridge CB2 0XY, UK
| | - Robin A J Smith
- Department of Chemistry, University of Otago, Dunedin 9054, New Zealand
| | - Michael P Murphy
- Mitochondrial Biology Unit, Medical Research Council, Cambridge CB2 0XY, UK
| | - Luis Barbeito
- Institut Pasteur de Montevideo, 11400 Montevideo, Uruguay
| | - Rafael Radi
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, 11800 Montevideo, Uruguay; Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, 11800 Montevideo, Uruguay
| | - Patricia Cassina
- Departamento de Histología y Embriología, Facultad de Medicina, Universidad de la República, 11800 Montevideo, Uruguay; Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, 11800 Montevideo, Uruguay.
| |
Collapse
|
347
|
|
348
|
Chen X, Guo X, Huang R, Zheng Z, Chen Y, Shang HF. An exploratory study of serum creatinine levels in patients with amyotrophic lateral sclerosis. Neurol Sci 2014; 35:1591-7. [PMID: 24782098 DOI: 10.1007/s10072-014-1807-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 04/16/2014] [Indexed: 02/05/2023]
Abstract
The etiology of amyotrophic lateral sclerosis (ALS) remains unknown, but existing data argue for a role of creatinine in ALS pathophysiology. Our aim is to clarify the correlation between serum creatinine and ALS in Chinese population. A total of 512 sporadic ALS (SALS) patients and 501 age- and gender-matched healthy controls were included. Revised ALS Functional Rating Scale (ALS-FRS-R) was used to assess the motor functional status of SALS patients. Survival analysis was performed using Kaplan-Meier method. Serum creatinine levels were significantly lower in SALS patients than in controls (p < 0.001). Patients with the second, the third and highest quartiles of creatinine levels had a significantly lower presence of ALS compared to those with the lowest quartile (p for trend <0.001). However, decreased presence of ALS was not found in the highest quartiles compared with the lowest quartiles in females. Sporadic ALS patients with different site of onset have similar serum creatinine levels, but underweight patients presented lower levels of serum creatinine. Patients with low serum creatinine levels are more likely to have severe motor impairment and low body mass index (BMI) values. This study demonstrates that SALS patients have lower serum creatinine levels than well-matched controls. Higher levels of serum creatinine are less likely to be associated with the presence of ALS in Chinese populations. Low serum creatinine levels may be related to severe motor impairment in SALS patients, after adjusting the confounding factor-BMI. However, serum creatinine has no deleterious impact on survival in ALS.
Collapse
Affiliation(s)
- Xueping Chen
- Department of Neurology, West China Hospital Sichuan University, No. 37 Guoxue Road, Chengdu, 610041, Sichuan, China
| | | | | | | | | | | |
Collapse
|
349
|
Pronto-Laborinho AC, Pinto S, de Carvalho M. Roles of vascular endothelial growth factor in amyotrophic lateral sclerosis. BIOMED RESEARCH INTERNATIONAL 2014; 2014:947513. [PMID: 24987705 PMCID: PMC4022172 DOI: 10.1155/2014/947513] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 03/24/2014] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal devastating neurodegenerative disorder, involving progressive degeneration of motor neurons in spinal cord, brainstem, and motor cortex. Riluzole is the only drug approved in ALS but it only confers a modest improvement in survival. In spite of a high number of clinical trials no other drug has proved effectiveness. Recent studies support that vascular endothelial growth factor (VEGF), originally described as a key angiogenic factor, also plays a key role in the nervous system, including neurogenesis, neuronal survival, neuronal migration, and axon guidance. VEGF has been used in exploratory clinical studies with promising results in ALS and other neurological disorders. Although VEGF is a very promising compound, translating the basic science breakthroughs into clinical practice is the major challenge ahead. VEGF-B, presenting a single safety profile, protects motor neurons from degeneration in ALS animal models and, therefore, it will be particularly interesting to test its effects in ALS patients. In the present paper the authors make a brief description of the molecular properties of VEGF and its receptors and review its different features and therapeutic potential in the nervous system/neurodegenerative disease, particularly in ALS.
Collapse
Affiliation(s)
- Ana Catarina Pronto-Laborinho
- Institute of Physiology, Faculty of Medicine, University of Lisbon, Avenida Professor Egas Moniz, 1649-028 Lisbon, Portugal
- Instituto de Medicina Molecular (IMM), Translational Clinical Physiology Unit, Avenida Professor Egas Moniz, 1649-028 Lisbon, Portugal
| | - Susana Pinto
- Institute of Physiology, Faculty of Medicine, University of Lisbon, Avenida Professor Egas Moniz, 1649-028 Lisbon, Portugal
- Instituto de Medicina Molecular (IMM), Translational Clinical Physiology Unit, Avenida Professor Egas Moniz, 1649-028 Lisbon, Portugal
| | - Mamede de Carvalho
- Institute of Physiology, Faculty of Medicine, University of Lisbon, Avenida Professor Egas Moniz, 1649-028 Lisbon, Portugal
- Instituto de Medicina Molecular (IMM), Translational Clinical Physiology Unit, Avenida Professor Egas Moniz, 1649-028 Lisbon, Portugal
- Department of Neurosciences, Hospital Santa Maria, Centro Hospitalar Lisboa Norte, Avenida Professor Egas Moniz, 1649-028 Lisbon, Portugal
| |
Collapse
|
350
|
Therrien M, Parker JA. Worming forward: amyotrophic lateral sclerosis toxicity mechanisms and genetic interactions in Caenorhabditis elegans. Front Genet 2014; 5:85. [PMID: 24860590 PMCID: PMC4029022 DOI: 10.3389/fgene.2014.00085] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 03/30/2014] [Indexed: 12/11/2022] Open
Abstract
Neurodegenerative diseases share pathogenic mechanisms at the cellular level including protein misfolding, excitotoxicity and altered RNA homeostasis among others. Recent advances have shown that the genetic causes underlying these pathologies overlap, hinting at the existence of a genetic network for neurodegeneration. This is perhaps best illustrated by the recent discoveries of causative mutations for amyotrophic lateral sclerosis (ALS) and frontotemporal degeneration (FTD). Once thought to be distinct entities, it is now recognized that these diseases exist along a genetic spectrum. With this wealth of discoveries comes the need to develop new genetic models of ALS and FTD to investigate not only pathogenic mechanisms linked to causative mutations, but to uncover potential genetic interactions that may point to new therapeutic targets. Given the conservation of many disease genes across evolution, Caenorhabditis elegans is an ideal system to investigate genetic interactions amongst these genes. Here we review the use of C. elegans to model ALS and investigate a putative genetic network for ALS/FTD that may extend to other neurological disorders.
Collapse
Affiliation(s)
- Martine Therrien
- Départment de Pathologie et Biologie Cellulaire, CRCHUM-Centre Hospitalier de l'Université de Montréal, Université de Montréal Montréal, QC, Canada
| | - J Alex Parker
- Départment de Pathologie et Biologie Cellulaire, Départment de Neurosciences, CRCHUM-Centre Hospitalier de l'Université de Montréal, Université de Montréal Montréal, QC, Canada
| |
Collapse
|