1
|
Fereidan‐Esfahani M, Decker PA, Weigand SD, Lopez Chiriboga AS, Flanagan EP, Tillema J, Lucchinetti CF, Eckel‐Passow JE, Tobin WO. Defining the natural history of tumefactive demyelination: A retrospective cohort of 257 patients. Ann Clin Transl Neurol 2023; 10:1544-1555. [PMID: 37443413 PMCID: PMC10502639 DOI: 10.1002/acn3.51844] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 06/20/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
OBJECTIVE To describe demographic, clinical, and radiographic features of tumefactive demyelination (TD) and identify factors associated with severe attacks and poor outcomes. METHODS Retrospective review of TD cases seen at Mayo Clinic, 1990-2021. RESULTS Of 257 patients with TD, 183/257 (71%) fulfilled the 2017 multiple sclerosis (MS) McDonald criteria at the last follow-up, 12/257 (5%) had myelin oligodendrocyte glycoprotein antibody-associated disease (MOGAD), 0 had aquaporin-4-IgG seropositive neuromyelitis optic spectrum disorders (AQP4+ NMOSD), and 62/257 (24%) were cryptogenic. Onset before age 18 was present in 18/257 (7%). Female to male ratio was 1.3:1. Cerebrospinal fluid oligoclonal (CSF) bands were present in 95/153 (62%). TD was the first demyelinating attack in 176/257 (69%). At presentation, 59/126 (47%) fulfilled Barkhof criteria for dissemination in space, 59/100 (59%) had apparent diffusion coefficient (ADC) restriction, and 57/126 (45%) had mass effect. Despite aggressive clinical presentation at onset, 181/257 (70%) of patients remained fully ambulatory (Expanded Disability Status Scale [EDSS] ≤4) after a 3.0-year median follow-up duration. Severe initial attack-related disability (EDSS ≥4) was more common in patients with motor symptoms (81/143 vs. 35/106, p < 0.0001), encephalopathy (20/143 vs. 2/106, p < 0.0001) and ADC restriction on initial MRI (42/63 vs. 15/33, p = 0.04). Poor long-term outcome (EDSS ≥4) was more common in patients with older onset age (41.9 ± 15 vs. 36.8 ± 15.6, p = 0.02) and motor symptoms at onset (49/76 vs. 66/171, p < 0.0001). INTERPRETATION Most TD patients should be considered part of the MS spectrum after excluding MOGAD and NMOSD. Motor symptoms and older age at presentation portend a poor outcome.
Collapse
Affiliation(s)
- Mahboubeh Fereidan‐Esfahani
- Department of NeurologyMayo ClinicRochesterMinnesotaUSA
- Center for Multiple Sclerosis and Autoimmune NeurologyMayo ClinicRochesterMinnesotaUSA
- Dell Medical SchoolUniversity of TexasAustinTexasUSA
| | - Paul A Decker
- Department of Quantitative Health SciencesMayo ClinicRochesterMinnesotaUSA
| | - Stephen D. Weigand
- Department of Quantitative Health SciencesMayo ClinicRochesterMinnesotaUSA
| | | | - Eoin P Flanagan
- Department of NeurologyMayo ClinicRochesterMinnesotaUSA
- Center for Multiple Sclerosis and Autoimmune NeurologyMayo ClinicRochesterMinnesotaUSA
- Department of Laboratory Medicine and PathologyMinneapolisMinnesotaUSA
| | - Jan‐Mendelt Tillema
- Department of NeurologyMayo ClinicRochesterMinnesotaUSA
- Center for Multiple Sclerosis and Autoimmune NeurologyMayo ClinicRochesterMinnesotaUSA
| | - Claudia F Lucchinetti
- Department of NeurologyMayo ClinicRochesterMinnesotaUSA
- Center for Multiple Sclerosis and Autoimmune NeurologyMayo ClinicRochesterMinnesotaUSA
| | | | - W. Oliver Tobin
- Department of NeurologyMayo ClinicRochesterMinnesotaUSA
- Center for Multiple Sclerosis and Autoimmune NeurologyMayo ClinicRochesterMinnesotaUSA
| |
Collapse
|
2
|
Alsameen MH, Gong Z, Qian W, Kiely M, Triebswetter C, Bergeron CM, Cortina LE, Faulkner ME, Laporte JP, Bouhrara M. C-NODDI: a constrained NODDI model for axonal density and orientation determinations in cerebral white matter. Front Neurol 2023; 14:1205426. [PMID: 37602266 PMCID: PMC10435293 DOI: 10.3389/fneur.2023.1205426] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 07/14/2023] [Indexed: 08/22/2023] Open
Abstract
Purpose Neurite orientation dispersion and density imaging (NODDI) provides measures of neurite density and dispersion through computation of the neurite density index (NDI) and the orientation dispersion index (ODI). However, NODDI overestimates the cerebrospinal fluid water fraction in white matter (WM) and provides physiologically unrealistic high NDI values. Furthermore, derived NDI values are echo-time (TE)-dependent. In this work, we propose a modification of NODDI, named constrained NODDI (C-NODDI), for NDI and ODI mapping in WM. Methods Using NODDI and C-NODDI, we investigated age-related alterations in WM in a cohort of 58 cognitively unimpaired adults. Further, NDI values derived using NODDI or C-NODDI were correlated with the neurofilament light chain (NfL) concentration levels, a plasma biomarker of axonal degeneration. Finally, we investigated the TE dependence of NODDI or C-NODDI derived NDI and ODI. Results ODI derived values using both approaches were virtually identical, exhibiting constant trends with age. Further, our results indicated a quadratic relationship between NDI and age suggesting that axonal maturation continues until middle age followed by a decrease. This quadratic association was notably significant in several WM regions using C-NODDI, while limited to a few regions using NODDI. Further, C-NODDI-NDI values exhibited a stronger correlation with NfL concentration levels as compared to NODDI-NDI, with lower NDI values corresponding to higher levels of NfL. Finally, we confirmed the previous finding that NDI estimation using NODDI was dependent on TE, while NDI derived values using C-NODDI exhibited lower sensitivity to TE in WM. Conclusion C-NODDI provides a complementary method to NODDI for determination of NDI in white matter.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Mustapha Bouhrara
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| |
Collapse
|
3
|
Bhaskaran S, Kumar G, Thadathil N, Piekarz KM, Mohammed S, Lopez SD, Qaisar R, Walton D, Brown JL, Murphy A, Smith N, Saunders D, Beckstead MJ, Plafker S, Lewis TL, Towner R, Deepa SS, Richardson A, Axtell RC, Van Remmen H. Neuronal deletion of MnSOD in mice leads to demyelination, inflammation and progressive paralysis that mimics phenotypes associated with progressive multiple sclerosis. Redox Biol 2023; 59:102550. [PMID: 36470129 PMCID: PMC9720104 DOI: 10.1016/j.redox.2022.102550] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/17/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022] Open
Abstract
Neuronal oxidative stress has been implicated in aging and neurodegenerative disease. Here we investigated the impact of elevated oxidative stress induced in mouse spinal cord by deletion of Mn-Superoxide dismutase (MnSOD) using a neuron specific Cre recombinase in Sod2 floxed mice (i-mn-Sod2 KO). Sod2 deletion in spinal cord neurons was associated with mitochondrial alterations and peroxide generation. Phenotypically, i-mn-Sod2 KO mice experienced hindlimb paralysis and clasping behavior associated with extensive demyelination and reduced nerve conduction velocity, axonal degeneration, enhanced blood brain barrier permeability, elevated inflammatory cytokines, microglia activation, infiltration of neutrophils and necroptosis in spinal cord. In contrast, spinal cord motor neuron number, innervation of neuromuscular junctions, muscle mass, and contractile function were not altered. Overall, our findings show that loss of MnSOD in spinal cord promotes a phenotype of demyelination, inflammation and progressive paralysis that mimics phenotypes associated with progressive multiple sclerosis.
Collapse
Affiliation(s)
- Shylesh Bhaskaran
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, OK, USA
| | - Gaurav Kumar
- Arthritis & Clinical Immunology, Oklahoma Medical Research Foundation, OK, USA
| | - Nidheesh Thadathil
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, OK, USA
| | - Katarzyna M Piekarz
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, OK, USA
| | - Sabira Mohammed
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | | | - Rizwan Qaisar
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, OK, USA
| | - Dorothy Walton
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, OK, USA
| | - Jacob L Brown
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, OK, USA; Oklahoma City VA Medical Center, Oklahoma City, OK, USA
| | - Ashley Murphy
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, OK, USA
| | - Nataliya Smith
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, OK, USA
| | - Debra Saunders
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, OK, USA
| | - Michael J Beckstead
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, OK, USA; Oklahoma City VA Medical Center, Oklahoma City, OK, USA
| | - Scott Plafker
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, OK, USA
| | - Tommy L Lewis
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, OK, USA
| | - Rheal Towner
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, OK, USA
| | - Sathyaseelan S Deepa
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Arlan Richardson
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Oklahoma City VA Medical Center, Oklahoma City, OK, USA
| | - Robert C Axtell
- Arthritis & Clinical Immunology, Oklahoma Medical Research Foundation, OK, USA.
| | - Holly Van Remmen
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, OK, USA; Oklahoma City VA Medical Center, Oklahoma City, OK, USA.
| |
Collapse
|
4
|
Harris G, Rickard JJS, Butt G, Kelleher L, Blanch RJ, Cooper J, Oppenheimer PG. Review: Emerging Eye-Based Diagnostic Technologies for Traumatic Brain Injury. IEEE Rev Biomed Eng 2023; 16:530-559. [PMID: 35320105 PMCID: PMC9888755 DOI: 10.1109/rbme.2022.3161352] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 02/11/2022] [Accepted: 03/15/2022] [Indexed: 11/06/2022]
Abstract
The study of ocular manifestations of neurodegenerative disorders, Oculomics, is a growing field of investigation for early diagnostics, enabling structural and chemical biomarkers to be monitored overtime to predict prognosis. Traumatic brain injury (TBI) triggers a cascade of events harmful to the brain, which can lead to neurodegeneration. TBI, termed the "silent epidemic" is becoming a leading cause of death and disability worldwide. There is currently no effective diagnostic tool for TBI, and yet, early-intervention is known to considerably shorten hospital stays, improve outcomes, fasten neurological recovery and lower mortality rates, highlighting the unmet need for techniques capable of rapid and accurate point-of-care diagnostics, implemented in the earliest stages. This review focuses on the latest advances in the main neuropathophysiological responses and the achievements and shortfalls of TBI diagnostic methods. Validated and emerging TBI-indicative biomarkers are outlined and linked to ocular neuro-disorders. Methods detecting structural and chemical ocular responses to TBI are categorised along with prospective chemical and physical sensing techniques. Particular attention is drawn to the potential of Raman spectroscopy as a non-invasive sensing of neurological molecular signatures in the ocular projections of the brain, laying the platform for the first tangible path towards alternative point-of-care diagnostic technologies for TBI.
Collapse
Affiliation(s)
- Georgia Harris
- School of Chemical Engineering, Advanced Nanomaterials Structures and Applications Laboratories, College of Engineering and Physical SciencesUniversity of BirminghamB15 2TTBirminghamU.K.
| | - Jonathan James Stanley Rickard
- School of Chemical Engineering, Advanced Nanomaterials Structures and Applications Laboratories, College of Engineering and Physical SciencesUniversity of BirminghamB15 2TTBirminghamU.K.
- Department of Physics, Cavendish LaboratoryUniversity of CambridgeCB3 0HECambridgeU.K.
| | - Gibran Butt
- Ophthalmology DepartmentUniversity Hospitals Birmingham NHS Foundation TrustB15 2THBirminghamU.K.
| | - Liam Kelleher
- School of Chemical Engineering, Advanced Nanomaterials Structures and Applications Laboratories, College of Engineering and Physical SciencesUniversity of BirminghamB15 2TTBirminghamU.K.
| | - Richard James Blanch
- Department of Military Surgery and TraumaRoyal Centre for Defence MedicineB15 2THBirminghamU.K.
- Neuroscience and Ophthalmology, Department of Ophthalmology, University Hospitals Birmingham NHS Foundation TrustcBirminghamU.K.
| | - Jonathan Cooper
- School of Biomedical EngineeringUniversity of GlasgowG12 8LTGlasgowU.K.
| | - Pola Goldberg Oppenheimer
- School of Chemical Engineering, Advanced Nanomaterials Structures and Applications Laboratories, College of Engineering and Physical SciencesUniversity of BirminghamB15 2TTBirminghamU.K.
- Healthcare Technologies Institute, Institute of Translational MedicineB15 2THBirminghamU.K.
| |
Collapse
|
5
|
Di Liegro CM, Schiera G, Schirò G, Di Liegro I. RNA-Binding Proteins as Epigenetic Regulators of Brain Functions and Their Involvement in Neurodegeneration. Int J Mol Sci 2022; 23:ijms232314622. [PMID: 36498959 PMCID: PMC9739182 DOI: 10.3390/ijms232314622] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022] Open
Abstract
A central aspect of nervous system development and function is the post-transcriptional regulation of mRNA fate, which implies time- and site-dependent translation, in response to cues originating from cell-to-cell crosstalk. Such events are fundamental for the establishment of brain cell asymmetry, as well as of long-lasting modifications of synapses (long-term potentiation: LTP), responsible for learning, memory, and higher cognitive functions. Post-transcriptional regulation is in turn dependent on RNA-binding proteins that, by recognizing and binding brief RNA sequences, base modifications, or secondary/tertiary structures, are able to control maturation, localization, stability, and translation of the transcripts. Notably, most RBPs contain intrinsically disordered regions (IDRs) that are thought to be involved in the formation of membrane-less structures, probably due to liquid-liquid phase separation (LLPS). Such structures are evidenced as a variety of granules that contain proteins and different classes of RNAs. The other side of the peculiar properties of IDRs is, however, that, under altered cellular conditions, they are also prone to form aggregates, as observed in neurodegeneration. Interestingly, RBPs, as part of both normal and aggregated complexes, are also able to enter extracellular vesicles (EVs), and in doing so, they can also reach cells other than those that produced them.
Collapse
Affiliation(s)
- Carlo Maria Di Liegro
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche) (STEBICEF), University of Palermo, 90128 Palermo, Italy
| | - Gabriella Schiera
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche) (STEBICEF), University of Palermo, 90128 Palermo, Italy
| | - Giuseppe Schirò
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata) (Bi.N.D.), University of Palermo, 90127 Palermo, Italy
| | - Italia Di Liegro
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata) (Bi.N.D.), University of Palermo, 90127 Palermo, Italy
- Correspondence: ; Tel.: +39-091-238-97 (ext. 415/446)
| |
Collapse
|
6
|
Savya SP, Li F, Lam S, Wellman SM, Stieger KC, Chen K, Eles JR, Kozai TDY. In vivo spatiotemporal dynamics of astrocyte reactivity following neural electrode implantation. Biomaterials 2022; 289:121784. [PMID: 36103781 PMCID: PMC10231871 DOI: 10.1016/j.biomaterials.2022.121784] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/24/2022] [Accepted: 08/29/2022] [Indexed: 11/02/2022]
Abstract
Brain computer interfaces (BCIs), including penetrating microelectrode arrays, enable both recording and stimulation of neural cells. However, device implantation inevitably causes injury to brain tissue and induces a foreign body response, leading to reduced recording performance and stimulation efficacy. Astrocytes in the healthy brain play multiple roles including regulating energy metabolism, homeostatic balance, transmission of neural signals, and neurovascular coupling. Following an insult to the brain, they are activated and gather around the site of injury. These reactive astrocytes have been regarded as one of the main contributors to the formation of a glial scar which affects the performance of microelectrode arrays. This study investigates the dynamics of astrocytes within the first 2 weeks after implantation of an intracortical microelectrode into the mouse brain using two-photon microscopy. From our observation astrocytes are highly dynamic during this period, exhibiting patterns of process extension, soma migration, morphological activation, and device encapsulation that are spatiotemporally distinct from other glial cells, such as microglia or oligodendrocyte precursor cells. This detailed characterization of astrocyte reactivity will help to better understand the tissue response to intracortical devices and lead to the development of more effective intervention strategies to improve the functional performance of neural interfacing technology.
Collapse
Affiliation(s)
- Sajishnu P Savya
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Northwestern University, USA
| | - Fan Li
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, USA; Computational Modeling & Simulation PhD Program, University of Pittsburgh, Pittsburgh, PA, USA
| | - Stephanie Lam
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, USA
| | - Steven M Wellman
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kevin C Stieger
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, USA
| | - Keying Chen
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, USA
| | - James R Eles
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, USA
| | - Takashi D Y Kozai
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neuroscience, University of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA; NeuroTech Center, University of Pittsburgh Brain Institute, Pittsburgh, PA, USA.
| |
Collapse
|
7
|
Feng X, Hu W, Hong Y, Ruan L, Hu Y, Liu D. Taurine Ameliorates Iron Overload-Induced Hepatocyte Injury via the Bcl-2/VDAC1-Mediated Mitochondrial Apoptosis Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4135752. [PMID: 35879990 PMCID: PMC9308541 DOI: 10.1155/2022/4135752] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 05/20/2022] [Accepted: 05/31/2022] [Indexed: 12/29/2022]
Abstract
Iron overload can induce reactive oxygen species (ROS) burst and liver damage. Taurine can reduce ROS production and ameliorate liver injury caused by iron overload; however, the underlying molecular mechanism remains elusive. Herein, L02 cells treated with 120 μM iron dextran for 48 h showed marked oxidative stress damage and significantly increased apoptosis. Taurine protected hepatocytes by stabilizing mitochondrial membranes and resisting oxidative stress damage caused by iron overload. However, transfection with siRNA Bcl-2 virus abrogated the observed protective effects. Following treatment with taurine, B cell lymphoma-2 (Bcl-2) could inhibit the opening of the mitochondrial permeability transition pore (mPTP), subsequently stabilizing the mitochondrial membrane potential by interacting with voltage-dependent anion channel 1 (VDAC1) of mPTP. The present study is the first to clarify the mechanism underlying taurine-afforded hepatocyte protection against iron overload-induced oxidative stress via Bcl-2-mediated inhibition of mPTP opening and the antiapoptotic pathway.
Collapse
Affiliation(s)
- Xiaoyu Feng
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Wenfeng Hu
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
- Department of Pharmacy, Jiujiang Traditional Chinese Medicine Hospital, Jiujiang 332900, China
| | - Yujiao Hong
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Linlin Ruan
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Yueben Hu
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Dan Liu
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| |
Collapse
|
8
|
Preventing Axonal Sodium Overload or Mitochondrial Calcium Uptake Protects Axonal Mitochondria from Oxidative Stress-Induced Alterations. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6125711. [PMID: 35663200 PMCID: PMC9157283 DOI: 10.1155/2022/6125711] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 05/02/2022] [Accepted: 05/05/2022] [Indexed: 11/26/2022]
Abstract
In neuroinflammatory and neurodegenerative disorders such as multiple sclerosis, mitochondrial damage caused by oxidative stress is believed to contribute to neuroaxonal damage. Previously, we demonstrated that exposure to hydrogen peroxide (H2O2) alters mitochondrial morphology and motility in myelinated axons and that these changes initiate at the nodes of Ranvier, where numerous sodium channels are located. Therefore, we suggested that mitochondrial damage may lead to ATP deficit, thereby affecting the efficiency of the sodium-potassium ATPase and eventually leading to sodium overload in axons. The increased intra-axonal sodium may revert the axonal sodium-calcium exchangers and thus may lead to a pathological calcium overload in the axoplasm and mitochondria. Here, we used the explanted murine ventral spinal roots to investigate whether modulation of sodium or calcium influx may prevent mitochondrial alterations in myelinated axons during exogenous application of H2O2 inducing oxidative stress. For that, tetrodotoxin, an inhibitor of voltage-gated sodium ion channels, and ruthenium 360, an inhibitor of the mitochondrial calcium uniporter, were applied simultaneously with hydrogen peroxide to axons. Mitochondrial shape and motility were analyzed. We showed that inhibition of axonal sodium influx prevented oxidative stress-induced morphological changes (i.e., increase in circularity and area and decrease in length) and preserved mitochondrial membrane potential, which is crucial for ATP production. Blocking mitochondrial calcium uptake prevented decrease in mitochondrial motility and also preserved membrane potential. Our findings indicate that alterations of both mitochondrial morphology and motility in the contexts of oxidative stress can be counterbalanced by modulating intramitochondrial ion concentrations pharmacologically. Moreover, motile mitochondria show preserved membrane potentials, pointing to a close association between mitochondrial motility and functionality.
Collapse
|
9
|
Wenzel N, Wittayer M, Weber CE, Schirmer L, Platten M, Gass A, Eisele P. MRI predictors for the conversion from contrast-enhancing to iron rim multiple sclerosis lesions. J Neurol 2022; 269:4414-4420. [PMID: 35332392 PMCID: PMC9293822 DOI: 10.1007/s00415-022-11082-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/11/2022] [Accepted: 03/11/2022] [Indexed: 12/22/2022]
Abstract
BACKGROUND In multiple sclerosis (MS), iron rim lesions (IRLs) are characterized by progressive tissue matrix damage. Therefore, early identification could represent an interesting target for therapeutic intervention to minimize evolving tissue damage. The aim of this study was to identify magnetic resonance imaging (MRI) parameters predicting the conversion from contrast-enhancing to IRLs. METHODS We retrospective identified MS patients scanned on the same 3 T MRI system presenting at least one supratentorial contrast-enhancing lesion (CEL) and a second MRI including susceptibility-weighted images after at least 3 months. On baseline MRI, pattern of contrast-enhancement was categorized as "nodular" or "ring-like", apparent diffusion coefficient (ADC) maps were assessed for the presence of a peripheral hypointense rim. Lesion localization, quantitative volumes (ADC, lesion volume) and the presence of a central vein were assessed. RESULTS Eighty-nine acute contrast-enhancing lesions in 54 MS patients were included. On follow-up, 16/89 (18%) initially CELs converted into IRLs. CELs that converted into IRLs were larger and demonstrated significantly more often a ring-like contrast-enhancement pattern and a peripheral hypointense rim on ADC maps. Logistic regression model including the covariables pattern of contrast-enhancement and presence of a hypointense rim on ADC maps showed the best predictive performance (area under the curve = 0.932). DISCUSSION The combination of a ring-like contrast-enhancement pattern and a peripheral hypointense rim on ADC maps has the ability to predict the evolution from acute to IRLs. This could be of prognostic value and become a target for early therapeutic intervention to minimize the associated tissue damage.
Collapse
Affiliation(s)
- Nicolas Wenzel
- Department of Neurology, Medical Faculty Mannheim and Mannheim Center of Translational Neurosciences (MCTN), Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Matthias Wittayer
- Department of Neurology, Medical Faculty Mannheim and Mannheim Center of Translational Neurosciences (MCTN), Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Claudia E Weber
- Department of Neurology, Medical Faculty Mannheim and Mannheim Center of Translational Neurosciences (MCTN), Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Lucas Schirmer
- Department of Neurology, Medical Faculty Mannheim and Mannheim Center of Translational Neurosciences (MCTN), Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.,Institute for Innate Immunoscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Michael Platten
- Department of Neurology, Medical Faculty Mannheim and Mannheim Center of Translational Neurosciences (MCTN), Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.,DKTK CCU Neuroimmunology and Brain Tumor Immunology, DKFZ, Heidelberg, Germany
| | - Achim Gass
- Department of Neurology, Medical Faculty Mannheim and Mannheim Center of Translational Neurosciences (MCTN), Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.
| | - Philipp Eisele
- Department of Neurology, Medical Faculty Mannheim and Mannheim Center of Translational Neurosciences (MCTN), Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| |
Collapse
|
10
|
Shen D, Liu K, Wang H, Wang H. Autophagy modulation in multiple sclerosis and experimental autoimmune encephalomyelitis. Clin Exp Immunol 2022; 209:140-150. [PMID: 35641229 PMCID: PMC9390842 DOI: 10.1093/cei/uxac017] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 02/01/2022] [Accepted: 02/14/2022] [Indexed: 11/14/2022] Open
Abstract
Multiple sclerosis (MS), a white matter demyelinating disease of the central nervous system (CNS), is characterized by neuroinflammatory and neurodegenerative. Experimental autoimmune encephalomyelitis (EAE) is a commonly used animal model for investigating pathogenic mechanisms of MS, representing the destruction of the blood-brain barrier (BBB), the activation of T cells, and the infiltration of myeloid cells. An increasing number of studies have documented that autophagy plays a critical role in the pathogenesis of both MS and EAE. Autophagy maintains CNS homeostasis by degrading the damaged organelles and abnormal proteins. Furthermore, autophagy is involved in inflammatory responses by regulating the activation of immune cells and the secretion of inflammatory factors. However, the specific mechanisms of autophagy involved in MS and EAE are not completely understood. In this review, we will summarize the complex mechanisms of autophagy in MS and EAE, providing potential therapeutic approaches for the management of MS.
Collapse
Affiliation(s)
- Donghui Shen
- Department of Neurology, Qingdao Municipal Hospital, Qingdao 266000, Shan Dong Province, China
| | - Kang Liu
- Department of Stomatology, Qingdao Municipal Hospital, Qingdao 266000, Shan Dong Province, China
| | - Hongyan Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao 266000, Shan Dong Province, China
| | - Haifeng Wang
- Correspondence: Haifeng Wang, Department of Neurology, Qingdao Municipal Hospital, Qingdao, Shan Dong Province, China.
| |
Collapse
|
11
|
Cho J, Nguyen TD, Huang W, Sweeney EM, Luo X, Kovanlikaya I, Zhang S, Gillen KM, Spincemaille P, Gupta A, Gauthier SA, Wang Y. Brain oxygen extraction fraction mapping in patients with multiple sclerosis. J Cereb Blood Flow Metab 2022; 42:338-348. [PMID: 34558996 PMCID: PMC9122515 DOI: 10.1177/0271678x211048031] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
We aimed to demonstrate the feasibility of whole brain oxygen extraction fraction (OEF) mapping for measuring lesion specific and regional OEF abnormalities in multiple sclerosis (MS) patients. In 22 MS patients and 11 healthy controls (HC), OEF and neural tissue susceptibility (χn) maps were computed from MRI multi-echo gradient echo data. In MS patients, 80 chronic active lesions with hyperintense rim on quantitative susceptibility mapping were identified, and the mean OEF and χn within the rim and core were compared using linear mixed-effect model analysis. The rim showed higher OEF and χn than the core: relative to their adjacent normal appearing white matter, OEF contrast = -6.6 ± 7.0% vs. -9.8 ± 7.8% (p < 0.001) and χn contrast = 33.9 ± 20.3 ppb vs. 25.7 ± 20.5 ppb (p = 0.017). Between MS and HC, OEF and χn were compared using a linear regression model in subject-based regions of interest. In the whole brain, compared to HC, MS had lower OEF, 30.4 ± 3.3% vs. 21.4 ± 4.4% (p < 0.001), and higher χn, -23.7 ± 7.0 ppb vs. -11.3 ± 7.7 ppb (p = 0.018). Our feasibility study suggests that OEF may serve as a useful quantitative marker of tissue oxygen utilization in MS.
Collapse
Affiliation(s)
- Junghun Cho
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Thanh D Nguyen
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Weiyuan Huang
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Elizabeth M Sweeney
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Xianfu Luo
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | | | - Shun Zhang
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Kelly M Gillen
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | | | - Ajay Gupta
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Susan A Gauthier
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA.,Department of Neurology, Weill Cornell Medicine, New York, NY, USA
| | - Yi Wang
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA.,Department of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| |
Collapse
|
12
|
Elsadany M, Elghaish RA, Khalil AS, Ahmed AS, Mansour RH, Badr E, Elserafy M. Transcriptional Analysis of Nuclear-Encoded Mitochondrial Genes in Eight Neurodegenerative Disorders: The Analysis of Seven Diseases in Reference to Friedreich’s Ataxia. Front Genet 2021; 12:749792. [PMID: 34987545 PMCID: PMC8721009 DOI: 10.3389/fgene.2021.749792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/20/2021] [Indexed: 11/25/2022] Open
Abstract
Neurodegenerative diseases (NDDs) are challenging to understand, diagnose, and treat. Revealing the genomic and transcriptomic changes in NDDs contributes greatly to the understanding of the diseases, their causes, and development. Moreover, it enables more precise genetic diagnosis and novel drug target identification that could potentially treat the diseases or at least ease the symptoms. In this study, we analyzed the transcriptional changes of nuclear-encoded mitochondrial (NEM) genes in eight NDDs to specifically address the association of these genes with the diseases. Previous studies show strong links between defects in NEM genes and neurodegeneration, yet connecting specific genes with NDDs is not well studied. Friedreich’s ataxia (FRDA) is an NDD that cannot be treated effectively; therefore, we focused first on FRDA and compared the outcome with seven other NDDs, including Alzheimer’s disease, amyotrophic lateral sclerosis, Creutzfeldt–Jakob disease, frontotemporal dementia, Huntington’s disease, multiple sclerosis, and Parkinson’s disease. First, weighted correlation network analysis was performed on an FRDA RNA-Seq data set, focusing only on NEM genes. We then carried out differential gene expression analysis and pathway enrichment analysis to pinpoint differentially expressed genes that are potentially associated with one or more of the analyzed NDDs. Our findings propose a strong link between NEM genes and NDDs and suggest that our identified candidate genes can be potentially used as diagnostic markers and therapeutic targets.
Collapse
Affiliation(s)
- Muhammad Elsadany
- University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
| | - Reem A. Elghaish
- University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
| | - Aya S. Khalil
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
| | - Alaa S. Ahmed
- University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
| | - Rana H. Mansour
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
| | - Eman Badr
- University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
- Faculty of Computers and Artificial Intelligence, Cairo University, Giza, Egypt
- *Correspondence: Eman Badr, ; Menattallah Elserafy,
| | - Menattallah Elserafy
- University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- *Correspondence: Eman Badr, ; Menattallah Elserafy,
| |
Collapse
|
13
|
Bromley LE, Horvath PJ, Bennett SE, Weinstock-Guttman B, Rideout TC, Browne RW, Ray AD. Three-Day Dietary Manipulation in Multiple Sclerosis: Exercise and Fatigue Outcomes. Int J MS Care 2021; 23:199-205. [PMID: 34720759 DOI: 10.7224/1537-2073.2020-036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Background In persons with multiple sclerosis (MS), the effect of nutrition on exercise performance and fatigue remains unknown. The objective was to determine whether a 3-day diet high in triglycerides (FAT) compared with a 3-day diet high in carbohydrates (CARB) would improve fatigue and exercise performance in persons with MS. Methods A randomized controlled crossover design was incorporated to study FAT versus CARB on submaximal cycling endurance (60% of peak oxygen consumption), substrate utilization, and fatigue in 12 persons with mild-to-moderate MS (Expanded Disability Status Scale score, 2.0-5.0) and 12 age- and sex-matched controls. Results There were no differences in cycling time between diets in either group (P = .29). The MS group had no changes in fatigue between diets (P = .64); the control group demonstrated increased total mental fatigue after FAT (P = .05). The control group increased carbohydrate oxidation by 24% at rest and 13% during exercise after CARB. Similarly, the control group significantly increased fat oxidation after FAT by 22% at rest and 68% during exercise (P = .01). These changes were not seen in the MS group. Compared with controls, persons with MS oxidized approximately 50% less fat during exercise after FAT (P = .05). Conclusions Neither CARB nor FAT altered submaximal exercise performance or baseline fatigue in the MS group. The results suggest that persons with MS are unable to adapt to dietary changes and oxidize fatty acids as efficiently as controls.
Collapse
Affiliation(s)
- Lacey E Bromley
- Department of Physical Therapy, D'Youville, Buffalo, NY, USA (LEB)
| | - Peter J Horvath
- Department of Exercise and Nutritional Sciences (PJH, TCR), University at Buffalo, Buffalo, NY
| | - Susan E Bennett
- Department of Rehabilitation Science (SEB), University at Buffalo, Buffalo, NY, USA
| | | | - Todd C Rideout
- Department of Exercise and Nutritional Sciences (PJH, TCR), University at Buffalo, Buffalo, NY
| | - Richard W Browne
- Department of Biotechnical and Clinical Laboratory Sciences (RWB), University at Buffalo, Buffalo, NY, USA
| | - Andrew D Ray
- Department of Cancer Prevention and Control, Roswell Park Cancer Center, Buffalo NY, USA (ADR)
| |
Collapse
|
14
|
Pukoli D, Polyák H, Rajda C, Vécsei L. Kynurenines and Neurofilament Light Chain in Multiple Sclerosis. Front Neurosci 2021; 15:658202. [PMID: 34113231 PMCID: PMC8185147 DOI: 10.3389/fnins.2021.658202] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 04/29/2021] [Indexed: 12/30/2022] Open
Abstract
Multiple sclerosis is an autoimmune, demyelinating, and neurodegenerative disease of the central nervous system. In recent years, it has been proven that the kynurenine system plays a significant role in the development of several nervous system disorders, including multiple sclerosis. Kynurenine pathway metabolites have both neurotoxic and neuroprotective effects. Moreover, the enzymes of the kynurenine pathway play an important role in immunomodulation processes, among others, as well as interacting with neuronal energy balance and various redox reactions. Dysregulation of many of the enzymatic steps in kynurenine pathway and upregulated levels of these metabolites locally in the central nervous system, contribute to the progression of multiple sclerosis pathology. This process can initiate a pathogenic cascade, including microglia activation, glutamate excitotoxicity, chronic oxidative stress or accumulated mitochondrial damage in the axons, that finally disrupt the homeostasis of neurons, leads to destabilization of neuronal cell cytoskeleton, contributes to neuro-axonal damage and neurodegeneration. Neurofilaments are good biomarkers of the neuro-axonal damage and their level reliably indicates the severity of multiple sclerosis and the treatment response. There is increasing evidence that connections exist between the molecules generated in the kynurenine metabolic pathway and the change in neurofilament concentrations. Thus the alterations in the kynurenine pathway may be an important biomarker of the course of multiple sclerosis. In our present review, we report the possible relationship and connection between neurofilaments and the kynurenine system in multiple sclerosis based on the available evidences.
Collapse
Affiliation(s)
- Dániel Pukoli
- Department of Neurology, Faculty of Medicine, University of Szeged, Szeged, Hungary.,Department of Neurology, Vaszary Kolos Hospital, Esztergom, Hungary
| | - Helga Polyák
- Department of Neurology, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Cecilia Rajda
- Department of Neurology, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - László Vécsei
- Department of Neurology, Faculty of Medicine, University of Szeged, Szeged, Hungary.,MTA-SZTE Neuroscience Research Group, Department of Neurology, Faculty of Medicine, Interdisciplinary Excellence Centre, University of Szeged, Szeged, Hungary
| |
Collapse
|
15
|
Plantone D, Pardini M, Rinaldi G. Riboflavin in Neurological Diseases: A Narrative Review. Clin Drug Investig 2021; 41:513-527. [PMID: 33886098 DOI: 10.1007/s40261-021-01038-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2021] [Indexed: 12/11/2022]
Abstract
Riboflavin is classified as one of the water-soluble B vitamins. It is part of the functional group of flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD) cofactors and is required for numerous flavoprotein-catalysed reactions. Riboflavin has important antioxidant properties, essential for correct cell functioning. It is required for the conversion of oxidised glutathione to the reduced form and for the mitochondrial respiratory chain as complexes I and II contain flavoprotein reductases and electron transferring flavoproteins. Riboflavin deficiency has been demonstrated to impair the oxidative state of the body, especially in relation to lipid peroxidation status, in both animal and human studies. In the nervous system, riboflavin is essential for the synthesis of myelin and its deficiency can determine the disruption of myelin lamellae. The inherited condition of restricted riboflavin absorption and utilisation, reported in about 10-15% of world population, warrants further investigation in relation to its association with the main neurodegenerative diseases. Several successful trials testing riboflavin for migraine prevention were performed, and this drug is currently classified as a Level B medication for migraine according to the American Academy of Neurology evidence-based rating, with evidence supporting its efficacy. Brown-Vialetto-Van Laere syndrome and Fazio-Londe diseases are now renamed as "riboflavin transporter deficiency" because these are autosomal recessive diseases caused by mutations of SLC52A2 and SLC52A3 genes that encode riboflavin transporters. High doses of riboflavin represent the mainstay of the therapy of these diseases and high doses of riboflavin should be rapidly started as soon as the diagnosis is suspected and continued lifelong. Remarkably, some mitochondrial diseases respond to supplementation with riboflavin. These include multiple acyl-CoA-dehydrogenase deficiency (which is caused by ETFDH gene mutations in the majority of the cases, or mutations in the ETFA and ETFB genes in a minority), mutations of ACAD9 gene, mutations of AIFM1 gene, mutations of the NDUFV1 and NDUFV2 genes. Therapeutic riboflavin administration has been tried in other neurological diseases, including stroke, multiple sclerosis, Friedreich's ataxia and Parkinson's disease. Unfortunately, the design of these clinical trials was not uniform, not allowing to accurately assess the real effects of this molecule on the disease course. In this review we analyse the properties of riboflavin and its possible effects on the pathogenesis of different neurological diseases, and we will review the current indications of this vitamin as a therapeutic intervention in neurology.
Collapse
Affiliation(s)
- Domenico Plantone
- Neurology Unit, Azienda Sanitaria Locale della Provincia di Bari, Di Venere Teaching Hospital, Via Ospedale Di Venere 1, 70131, Bari, Italy.
| | - Matteo Pardini
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy.,Ospedale Policlinico San Martino, IRCCS, Genoa, Italy
| | - Giuseppe Rinaldi
- Neurology Unit, Azienda Sanitaria Locale della Provincia di Bari, Di Venere Teaching Hospital, Via Ospedale Di Venere 1, 70131, Bari, Italy
| |
Collapse
|
16
|
Bergaglio T, Luchicchi A, Schenk GJ. Engine Failure in Axo-Myelinic Signaling: A Potential Key Player in the Pathogenesis of Multiple Sclerosis. Front Cell Neurosci 2021; 15:610295. [PMID: 33642995 PMCID: PMC7902503 DOI: 10.3389/fncel.2021.610295] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 01/20/2021] [Indexed: 12/12/2022] Open
Abstract
Multiple Sclerosis (MS) is a complex and chronic disease of the central nervous system (CNS), characterized by both degenerative and inflammatory processes leading to axonal damage, demyelination, and neuronal loss. In the last decade, the traditional outside-in standpoint on MS pathogenesis, which identifies a primary autoimmune inflammatory etiology, has been challenged by a complementary inside-out theory. By focusing on the degenerative processes of MS, the axo-myelinic system may reveal new insights into the disease triggering mechanisms. Oxidative stress (OS) has been widely described as one of the means driving tissue injury in neurodegenerative disorders, including MS. Axonal mitochondria constitute the main energy source for electrically active axons and neurons and are largely vulnerable to oxidative injury. Consequently, axonal mitochondrial dysfunction might impair efficient axo-glial communication, which could, in turn, affect axonal integrity and the maintenance of axonal, neuronal, and synaptic signaling. In this review article, we argue that OS-derived mitochondrial impairment may underline the dysfunctional relationship between axons and their supportive glia cells, specifically oligodendrocytes and that this mechanism is implicated in the development of a primary cytodegeneration and a secondary pro-inflammatory response (inside-out), which in turn, together with a variably primed host's immune system, may lead to the onset of MS and its different subtypes.
Collapse
Affiliation(s)
| | | | - Geert J. Schenk
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam University Medical Center, Amsterdam MS Center, Amsterdam, Netherlands
| |
Collapse
|
17
|
Dehghan S, Aref E, Raoufy MR, Javan M. An optimized animal model of lysolecithin induced demyelination in optic nerve; more feasible, more reproducible, promising for studying the progressive forms of multiple sclerosis. J Neurosci Methods 2021; 352:109088. [PMID: 33508411 DOI: 10.1016/j.jneumeth.2021.109088] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 01/01/2021] [Accepted: 01/19/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Multiple Sclerosis (MS) is a demyelinating disease leading to long-term neurological deficit due to unsuccessful remyelination and axonal loss. Currently, there are no satisfactory treatments for progressive MS somewhat due to the lack of an adequate animal model for studying the mechanisms of disease progression and screening new drugs. NEW METHOD Lysolecithin (LPC) or agarose-gel loaded LPC (AL-LPC) were applied to mouse optic nerve behind the globe via a minor surgery. Agarose loading was used to achieve longer time of LPC exposure and subsequently long-lasting demyelination. RESULTS The lesion sites characterized by luxol fast blue (LFB), FluoroMyelin, Bielschowsky's staining, and immunostaining showed extensive demyelination and axonal damage. The loss of Retinal ganglion cells (RGCs) in the corresponding retinal layer was shown by immunostaining and H&E staining. Visual evoked potential (VEP) recordings showed a significant increase in the latency of the P1 wave and a decrease in the amplitude of the P1N1 wave. COMPARISON WITH EXISTING METHODS The new approach with a very minor surgery seems to be more feasible and reproducible compared to stereotaxic LPC injection to optic chiasm. Our data revealed prolonged demyelination, axonal degeneration and RGCs loss in both AL-LPC and LPC groups; however, these pathologies were more extensive in the AL-LPC group. CONCLUSION The optimized model provides a longer demyelination time frame and axonal damage followed by RGC degeneration; which is of exceptional interest in investigating axonal degeneration mechanisms and screening the new drugs for progressive MS.
Collapse
Affiliation(s)
- Samaneh Dehghan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, P.O. Box:14115-331, Tehran, Iran
| | - Ehsan Aref
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, P.O. Box:14115-331, Tehran, Iran
| | - Mohammad Reza Raoufy
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, P.O. Box:14115-331, Tehran, Iran; Institute for Brain and Cognition, Tarbiat Modares University, P.O. Box:14115-331, Tehran, Iran
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, P.O. Box:14115-331, Tehran, Iran; Institute for Brain and Cognition, Tarbiat Modares University, P.O. Box:14115-331, Tehran, Iran; Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, P.O. Box:14115-331, Tehran, Iran.
| |
Collapse
|
18
|
C-reactive protein/albumin ratio in patients with multiple sclerosis and its relationship with disease subtype and disability. JOURNAL OF SURGERY AND MEDICINE 2020. [DOI: 10.28982/josam.736846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
19
|
Kostyuk AI, Panova AS, Kokova AD, Kotova DA, Maltsev DI, Podgorny OV, Belousov VV, Bilan DS. In Vivo Imaging with Genetically Encoded Redox Biosensors. Int J Mol Sci 2020; 21:E8164. [PMID: 33142884 PMCID: PMC7662651 DOI: 10.3390/ijms21218164] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 12/13/2022] Open
Abstract
Redox reactions are of high fundamental and practical interest since they are involved in both normal physiology and the pathogenesis of various diseases. However, this area of research has always been a relatively problematic field in the context of analytical approaches, mostly because of the unstable nature of the compounds that are measured. Genetically encoded sensors allow for the registration of highly reactive molecules in real-time mode and, therefore, they began a new era in redox biology. Their strongest points manifest most brightly in in vivo experiments and pave the way for the non-invasive investigation of biochemical pathways that proceed in organisms from different systematic groups. In the first part of the review, we briefly describe the redox sensors that were used in vivo as well as summarize the model systems to which they were applied. Next, we thoroughly discuss the biological results obtained in these studies in regard to animals, plants, as well as unicellular eukaryotes and prokaryotes. We hope that this work reflects the amazing power of this technology and can serve as a useful guide for biologists and chemists who work in the field of redox processes.
Collapse
Affiliation(s)
- Alexander I. Kostyuk
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (A.I.K.); (A.S.P.); (A.D.K.); (D.A.K.); (D.I.M.); (O.V.P.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Anastasiya S. Panova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (A.I.K.); (A.S.P.); (A.D.K.); (D.A.K.); (D.I.M.); (O.V.P.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Aleksandra D. Kokova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (A.I.K.); (A.S.P.); (A.D.K.); (D.A.K.); (D.I.M.); (O.V.P.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Daria A. Kotova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (A.I.K.); (A.S.P.); (A.D.K.); (D.A.K.); (D.I.M.); (O.V.P.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Dmitry I. Maltsev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (A.I.K.); (A.S.P.); (A.D.K.); (D.A.K.); (D.I.M.); (O.V.P.); (V.V.B.)
- Federal Center for Cerebrovascular Pathology and Stroke, 117997 Moscow, Russia
| | - Oleg V. Podgorny
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (A.I.K.); (A.S.P.); (A.D.K.); (D.A.K.); (D.I.M.); (O.V.P.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Vsevolod V. Belousov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (A.I.K.); (A.S.P.); (A.D.K.); (D.A.K.); (D.I.M.); (O.V.P.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
- Federal Center for Cerebrovascular Pathology and Stroke, 117997 Moscow, Russia
- Institute for Cardiovascular Physiology, Georg August University Göttingen, D-37073 Göttingen, Germany
| | - Dmitry S. Bilan
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (A.I.K.); (A.S.P.); (A.D.K.); (D.A.K.); (D.I.M.); (O.V.P.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| |
Collapse
|
20
|
Zhang Y, Figueroa-Miranda G, Wu C, Willbold D, Offenhäusser A, Mayer D. Electrochemical dual-aptamer biosensors based on nanostructured multielectrode arrays for the detection of neuronal biomarkers. NANOSCALE 2020; 12:16501-16513. [PMID: 32729601 DOI: 10.1039/d0nr03421e] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Multielectrode arrays (MEAs) have been increasingly used for the development of biosensors due to their capability to record signals from multiple channels, fast mass transfer rates, and high spatial resolution. Alzheimer's disease (AD) is often associated with mitochondrial dysfunction, which is closely related to reduced levels of adenosine triphosphate (ATP). Therefore, simultaneous detection of ATP together with amyloid-β oligomers (AβO), a reliable biomarker for AD, can potentially advance the early detection of Alzheimer's disease. In this work, a dual-aptamer modified MEA chip was developed that consists of microelectrodes modified with electrodeposited 3D nanostructures (3D-GMEs). Electrodeposition methods, deposition potential, and deposition time were systematically altered and the active surface areas as well as the electrode morphologies were characterized by cyclic voltammetry and scanning electron microscopy. The nanostructured microelectrodes were sequentially modified with AβO and ATP specific aptamer receptors. To achieve the modification of different aptamer receptors at different 3D-GMEs of the same MEA chip, electrochemical cleaning was applied to individual 3D-GMEs. Ferrocene labels were attached to the aptamer receptors to enable amperometric signaling after target-aptamer binding. The developed aptasensor showed a linear detection range from 1 pM to 200 nM for the detection of AβO and from 0.01 nM to 1000 nM for the detection of ATP. Finally, ATP and AβO were detected simultaneously in the same analyte solution by the same sensor chip, which could support the early detection of AD, provide comprehensive information about the health status of the patient, and be helpful for pathological studies of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yuting Zhang
- Institute of Biological Information Processing, Bioelectronics (IBI-3), Forschungszentrum Jülich GmbH, 52428 Jülich, Germany.
| | | | | | | | | | | |
Collapse
|
21
|
Zhou KQ, Draghi V, Lear CA, Dean JM, Ashton JL, Hou Y, Bennet L, Gunn AJ, Davidson JO. Protection of axonal integrity with 48 or 72 h of cerebral hypothermia in near-term fetal sheep. Pediatr Res 2020; 88:48-56. [PMID: 31234193 DOI: 10.1038/s41390-019-0475-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 06/06/2019] [Accepted: 06/08/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Therapeutic hypothermia is partially protective for neonatal hypoxic-ischemic encephalopathy (HIE). Damage to the white matter tracts is highly associated with adverse outcomes after HIE, but the effectiveness and optimal duration of hypothermia to attenuate axonal injury are unclear. METHODS Near-term fetal sheep were randomized to sham control or cerebral ischemia for 30 min with normothermia or cerebral hypothermia from 3 to either 48 or 72 h. Sheep were killed after 7 days. SMI-312-labeled axons and myelin basic protein were quantified in the intragyral white matter of the first and second parasagittal gyri. RESULTS Ischemia was associated with reduced axonal and myelin area fraction (p < 0.05); loss of axonal and myelin linearity (p < 0.05); and thin, sparse axons, with spheroids, compared to dense, linear morphology in sham controls and associated with induction of microglia in an amoeboid morphology. Both ischemia-48 h hypothermia and ischemia-72 h hypothermia improved axonal area fraction and linearity (p < 0.05), although abnormal morphological features were seen in a subset. Microglial induction was partially suppressed by ischemia-48 h hypothermia, with a ramified morphology. CONCLUSIONS These data suggest that therapeutic hypothermia can alleviate post-ischemic axonopathy, in part by suppressing secondary inflammation.
Collapse
Affiliation(s)
- Kelly Q Zhou
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Vittoria Draghi
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Christopher A Lear
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Justin M Dean
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Jesse L Ashton
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Yufeng Hou
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Laura Bennet
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Alistair J Gunn
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand.
| | - Joanne O Davidson
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
22
|
Morales Pantoja IE, Smith MD, Rajbhandari L, Cheng L, Gao Y, Mahairaki V, Venkatesan A, Calabresi PA, Fitzgerald KC, Whartenby KA. iPSCs from people with MS can differentiate into oligodendrocytes in a homeostatic but not an inflammatory milieu. PLoS One 2020; 15:e0233980. [PMID: 32511247 PMCID: PMC7279569 DOI: 10.1371/journal.pone.0233980] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Accepted: 05/15/2020] [Indexed: 11/19/2022] Open
Abstract
Multiple sclerosis (MS) is an inflammatory and demyelinating disease of the central nervous system (CNS) that results in variable severities of neurodegeneration. The understanding of MS has been limited by the inaccessibility of the affected cells and the lengthy timeframe of disease development. However, recent advances in stem cell technology have facilitated the bypassing of some of these challenges. Towards gaining a greater understanding of the innate potential of stem cells from people with varying degrees of disability, we generated induced pluripotent stem cells (iPSCs) from peripheral blood mononuclear cells derived from stable and progressive MS patients, and then further differentiated them into oligodendrocyte (OL) lineage cells. We analyzed differentiation under both homeostatic and inflammatory conditions via sustained exposure to low-dose interferon gamma (IFNγ), a prominent cytokine in MS. We found that all iPSC lines differentiated into mature myelinating OLs, but chronic exposure to IFNγ dramatically inhibited differentiation in both MS groups, particularly if exposure was initiated during the pre-progenitor stage. Low-dose IFNγ was not toxic but led to an early upregulation of interferon response genes in OPCs followed by an apparent redirection in lineage commitment from OL to a neuron-like phenotype in a significant portion of the treated cells. Our results reveal that a chronic low-grade inflammatory environment may have profound effects on the efficacy of regenerative therapies.
Collapse
Affiliation(s)
- Itzy E. Morales Pantoja
- Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Matthew D. Smith
- Department of Neurology, Johns Hopkins University, School of Medicine, Baltimore, Maryland, United States of America
| | - Labchan Rajbhandari
- Department of Neurology, Johns Hopkins University, School of Medicine, Baltimore, Maryland, United States of America
| | - Linzhao Cheng
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Yongxing Gao
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Vasiliki Mahairaki
- Department of Neurology, Johns Hopkins University, School of Medicine, Baltimore, Maryland, United States of America
| | - Arun Venkatesan
- Department of Neurology, Johns Hopkins University, School of Medicine, Baltimore, Maryland, United States of America
| | - Peter A. Calabresi
- Department of Neurology, Johns Hopkins University, School of Medicine, Baltimore, Maryland, United States of America
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Kathryn C. Fitzgerald
- Department of Neurology, Johns Hopkins University, School of Medicine, Baltimore, Maryland, United States of America
- Department of Epidemiology Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Katharine A. Whartenby
- Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Neurology, Johns Hopkins University, School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
23
|
Winiarska-Mieczan A, Baranowska-Wójcik E, Kwiecień M, Grela ER, Szwajgier D, Kwiatkowska K, Kiczorowska B. The Role of Dietary Antioxidants in the Pathogenesis of Neurodegenerative Diseases and Their Impact on Cerebral Oxidoreductive Balance. Nutrients 2020; 12:nu12020435. [PMID: 32046360 PMCID: PMC7071337 DOI: 10.3390/nu12020435] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 01/26/2020] [Accepted: 02/04/2020] [Indexed: 12/12/2022] Open
Abstract
Neurodegenerative diseases are progressive diseases of the nervous system that lead to neuron loss or functional disorders. Neurodegenerative diseases require long-term, sometimes life-long pharmacological treatment, which increases the risk of adverse effects and a negative impact of pharmaceuticals on the patients’ general condition. One of the main problems related to the treatment of this type of condition is the limited ability to deliver drugs to the brain due to their poor solubility, low bioavailability, and the effects of the blood-brain barrier. Given the above, one of the main objectives of contemporary scientific research focuses on the prevention of neurodegenerative diseases. As disorders related to the competence of the antioxidative system are a marker in all diseases of this type, the primary prophylactics should entail the use of exogenous antioxidants, particularly ones that can be used over extended periods, regardless of the patient’s age, and that are easily available, e.g., as part of a diet or as diet supplements. The paper analyzes the significance of the oxidoreductive balance in the pathogenesis of neurodegenerative diseases. Based on information published globally in the last 10 years, an analysis is also provided with regard to the impact of exogenous antioxidants on brain functions with respect to the prevention of this type of diseases.
Collapse
Affiliation(s)
- Anna Winiarska-Mieczan
- Department of Bromatology and Food Physiology, University of Life Sciences in Lublin, 20-950 Lublin, Poland; (M.K.); (E.R.G.); (K.K.); (B.K.)
- Correspondence: ; Tel.: +48-81-445-67-44; Fax: +48-81-53-335-49
| | - Ewa Baranowska-Wójcik
- Department of Biotechnology, Microbiology and Human Nutrition, University of Life Sciences in Lublin, 20-950 Lublin, Poland; (E.B.-W.); (D.S.)
| | - Małgorzata Kwiecień
- Department of Bromatology and Food Physiology, University of Life Sciences in Lublin, 20-950 Lublin, Poland; (M.K.); (E.R.G.); (K.K.); (B.K.)
| | - Eugeniusz R. Grela
- Department of Bromatology and Food Physiology, University of Life Sciences in Lublin, 20-950 Lublin, Poland; (M.K.); (E.R.G.); (K.K.); (B.K.)
| | - Dominik Szwajgier
- Department of Biotechnology, Microbiology and Human Nutrition, University of Life Sciences in Lublin, 20-950 Lublin, Poland; (E.B.-W.); (D.S.)
| | - Katarzyna Kwiatkowska
- Department of Bromatology and Food Physiology, University of Life Sciences in Lublin, 20-950 Lublin, Poland; (M.K.); (E.R.G.); (K.K.); (B.K.)
| | - Bożena Kiczorowska
- Department of Bromatology and Food Physiology, University of Life Sciences in Lublin, 20-950 Lublin, Poland; (M.K.); (E.R.G.); (K.K.); (B.K.)
| |
Collapse
|
24
|
Hu H, Jiang H, Gameiro GR, Hernandez J, Delgado S, Wang J. Focal Thickness Reduction of the Ganglion Cell-Inner Plexiform Layer Best Discriminates Prior Optic Neuritis in Patients With Multiple Sclerosis. Invest Ophthalmol Vis Sci 2020; 60:4257-4269. [PMID: 31618762 PMCID: PMC6996667 DOI: 10.1167/iovs.19-27574] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Purpose The goal was to visualize topographic thickness maps of the intraretinal layers and evaluate their discrimination abilities and relationships with clinical manifestations in patients with multiple sclerosis (MS) and a history of optic neuritis (ON). Methods Thirty patients with relapsing-remitting MS (34 eyes with a history of ON [MSON] and 26 non-ON fellow eyes [MSFE]) were recruited together with 63 age- and sex-matched controls (HC). Ultrahigh resolution optical coherence tomography was used to image the macula and the volumetric data set was segmented to yield six intraretinal layers. Topographic thickness maps were aligned and averaged for the visualization. The thickness maps were partitioned using the Early Treatment Diabetic Retinopathy Study (ETDRS) and related to Sloan low-contrast letter acuity (LCLA), Expanded Disability Status Scale (EDSS), and disease duration. Results Focal thickness reduction occurred in the macular retinal nerve fiber layer (mRNFL) and ganglion cell-inner plexiform layer (GCIPL), with the most profound reduction occurring in MSON eyes (P < 0.05). A horseshoe-like thickness reduction pattern (U Zone) in the GCIPL appeared in MSON. The thickness of the U Zone had better discrimination power than the ETDRS partitions (area under the curve = 0.97) and differentiated 96% of MSON from HC. The thickness of the U Zone was positively correlated to 2.5% LCLA (r = 0.38, P < 0.05) and 1.25% LCLA (r = 0.57, P < 0.05). Conclusions The horseshoe-like thickness reduction of the GCIPL appeared to be an ON-specific focal thickness alteration with the highest discrimination power of prior ON.
Collapse
Affiliation(s)
- Huiling Hu
- Shenzhen Key Laboratory of Ophthalmology, Shenzhen Eye Hospital, Jinan University, Shenzhen, China.,Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Hong Jiang
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, United States.,Department of Neurology, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Giovana Rosa Gameiro
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Jeffrey Hernandez
- Department of Neurology, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Silvia Delgado
- Department of Neurology, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Jianhua Wang
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
| |
Collapse
|
25
|
Tobore TO. Towards a comprehensive etiopathogenetic and pathophysiological theory of multiple sclerosis. Int J Neurosci 2019; 130:279-300. [PMID: 31588832 DOI: 10.1080/00207454.2019.1677648] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background: Multiple sclerosis (MS) is a neurodegenerative disease caused by dysfunction of the immune system that affects the central nervous system (CNS). It is characterized by demyelination, chronic inflammation, neuronal and oligodendrocyte loss and reactive astrogliosis. It can result in physical disability and acute neurological and cognitive problems. Despite the gains in knowledge of immunology, cell biology, and genetics in the last five decades, the ultimate etiology or specific elements that trigger MS remain unknown. The objective of this review is to propose a theoretical basis for MS etiopathogenesis.Methods: Search was done by accessing PubMed/Medline, EBSCO, and PsycINFO databases. The search string used was "(multiple sclerosis* OR EAE) AND (pathophysiology* OR etiopathogenesis)". The electronic databases were searched for titles or abstracts containing these terms in all published articles between January 1, 1960, and June 30, 2019. The search was filtered down to 362 articles which were included in this review.Results: A framework to better understand the etiopathogenesis and pathophysiology of MS can be derived from four essential factors; mitochondria dysfunction (MtD) & oxidative stress (OS), vitamin D (VD), sex hormones and thyroid hormones. These factors play a direct role in MS etiopathogenesis and have a modulatory effect on many other factors involved in the disease.Conclusions: For better MS prevention and treatment outcomes, efforts should be geared towards treating thyroid problems, sex hormone alterations, VD deficiency, sleep problems and melatonin alterations. MS patients should be encouraged to engage in activities that boost total antioxidant capacity (TAC) including diet and regular exercise and discouraged from activities that promote OS including smoking and alcohol consumption.
Collapse
|
26
|
Tobore TO. On elucidation of the role of mitochondria dysfunction and oxidative stress in multiple sclerosis. ACTA ACUST UNITED AC 2019. [DOI: 10.1111/ncn3.12335] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
27
|
Gonzalo H, Nogueras L, Gil-Sánchez A, Hervás JV, Valcheva P, González-Mingot C, Martin-Gari M, Canudes M, Peralta S, Solana MJ, Pamplona R, Portero-Otin M, Boada J, Serrano JCE, Brieva L. Impairment of Mitochondrial Redox Status in Peripheral Lymphocytes of Multiple Sclerosis Patients. Front Neurosci 2019; 13:938. [PMID: 31551694 PMCID: PMC6738270 DOI: 10.3389/fnins.2019.00938] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/21/2019] [Indexed: 11/13/2022] Open
Abstract
Literature suggests that oxidative stress (OS) may be involved in the pathogenesis of multiple sclerosis (MS), in which the immune system is known to play a key role. However, to date, the OS in peripheral lymphocytes and its contribution to the disease remain unknown. The aim of the present study was to explore the influence of OS in peripheral lymphocytes of MS patients. To that end, a cross-sectional, observational pilot study was conducted [n = 58: 34 MS and 24 healthy subjects (control group)]. We have measured superoxide production and protein mitochondrial complex levels in peripheral blood mononuclear cells (PBMCs) isolated from MS patients and control. Lactate levels and the antioxidant capacity were determined in plasma. We adjusted the comparisons between study groups by age, sex and cell count according to case. Results demonstrated that PBMCs, specifically T cells, from MS patients exhibited significantly increased superoxide anion production compared to control group (p = 0.027 and p = 0.041, respectively). Increased superoxide production in PBMCs was maintained after the adjustment (p = 0.044). Regarding mitochondrial proteins, we observe a significant decrease in the representative protein content of the mitochondrial respiratory chain complexes I-V in PBMCs of MS patients (p = 0.002, p = 0.037, p = 0.03, p = 0.044, and p = 0.051, respectively), which was maintained for complexes I, III, and V after the adjustment (p = 0.026; p = 0.033; p = 0.033, respectively). In MS patients, a trend toward increased plasma lactate concentration was detected [8.04 mg lactate/dL (5.25, 9.49) in the control group, 11.36 mg lactate/dL (5.41, 14.81) in MS patients] that was statistically significant after the adjustment (p = 0.013). This might be indicative of compromised mitochondrial function. Finally, antioxidant capacity was also decreased in plasma from MS patients, both before (p = 0.027) and after adjusting for sex and age (p = 0.006). Our findings demonstrate that PBMCs of MS patients show impaired mitochondrial redox status and deficient antioxidant capacity. These results demonstrate for the first time the existence of mitochondrial alterations in the cells immune cells of MS patients already at the peripheral level.
Collapse
Affiliation(s)
- Hugo Gonzalo
- Institut de Recerca Biomèdica de Lleida, Lleida, Spain.,Clinical University Hospital of Valladolid (HCUV), Department of Research and Innovation, SACYL/IECSCYL, Valladolid, Spain
| | - Lara Nogueras
- Universitat de Lleida, Departament de Medicina Experimental, Lleida, Spain
| | | | | | | | | | | | - Marc Canudes
- Institut de Recerca Biomèdica de Lleida, Lleida, Spain
| | | | | | - Reinald Pamplona
- Universitat de Lleida, Departament de Medicina Experimental, Lleida, Spain
| | | | - Jordi Boada
- Universitat de Lleida, Departament de Medicina Experimental, Lleida, Spain
| | | | - Luis Brieva
- Hospital Universitario Arnau de Vilanova, Lleida, Spain
| |
Collapse
|
28
|
Shi C, Jiang H, Gameiro GR, Hu H, Hernandez J, Delgado S, Wang J. Visual Function and Disability Are Associated With Focal Thickness Reduction of the Ganglion Cell-Inner Plexiform Layer in Patients With Multiple Sclerosis. Invest Ophthalmol Vis Sci 2019; 60:1213-1223. [PMID: 30913293 PMCID: PMC6892386 DOI: 10.1167/iovs.18-25809] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Purpose The purpose of this study was to visualize the topographic thickness patterns of the intraretinal layers and their associations with clinical manifestations in patients with multiple sclerosis (MS). Methods Ninety-four eyes of 47 relapsing-remitting MS patients without history of optic neuritis were imaged using optical coherence tomography and compared with 134 eyes of 67 healthy subjects. Volumetric data centered on the fovea were segmented to obtain the thickness maps of six intraretinal layers. The thickness measurements partitioned using the Early Treatment Diabetic Retinopathy Study (ETDRS) partition were correlated to the Expanded Disability State Scale (EDSS) and Sloan low contrast visual acuity (LCVA). The receiver-operating characteristics (ROC) curves were calculated to obtain the area under the ROC curves (AUCs). Results The ganglion cell-inner plexiform layer (GCIPL) showed horseshoe-like thickness reduction profoundly at the nasal sector. The most profound thickness reduction zone (circular area, diameter = 1 mm) was located at 2 mm in the nasal sector and 0.4 mm inferior from the fovea, named the “M zone.” The thickness reduction of the M zone was −7.3 μm in MS eyes, which was the most profound alteration, compared to any ETDRS sectors. The AUC of the M zone was 0.75. The relationship between the thickness of the M zone and EDSS (r = −0.59, P < 0.001) or 2.5% LCVA (r = 0.51, P < 0.001) were ranked as the strongest relation compared to any ETDRS sectors. Conclusions This is the first study, to our knowledge, to visualize focal thickness alteration of GCIPL and reveal its relationship to visual function and disability in patients with MS without history of optic neuritis.
Collapse
Affiliation(s)
- Ce Shi
- School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Hong Jiang
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, United States.,Department of Neurology, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Giovana Rosa Gameiro
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Huiling Hu
- School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Shenzhen Key Laboratory of Ophthalmology, Shenzhen Eye Hospital, Jinan University, Shenzhen, China
| | - Jeffrey Hernandez
- Department of Neurology, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Silvia Delgado
- Department of Neurology, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Jianhua Wang
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
| |
Collapse
|
29
|
van Hameren G, Campbell G, Deck M, Berthelot J, Gautier B, Quintana P, Chrast R, Tricaud N. In vivo real-time dynamics of ATP and ROS production in axonal mitochondria show decoupling in mouse models of peripheral neuropathies. Acta Neuropathol Commun 2019; 7:86. [PMID: 31186069 PMCID: PMC6558672 DOI: 10.1186/s40478-019-0740-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 05/16/2019] [Indexed: 12/31/2022] Open
Abstract
Mitochondria are critical for the function and maintenance of myelinated axons notably through Adenosine triphosphate (ATP) production. A direct by-product of this ATP production is reactive oxygen species (ROS), which are highly deleterious for neurons. While ATP shortage and ROS levels increase are involved in several neurodegenerative diseases, it is still unclear whether the real-time dynamics of both ATP and ROS production in axonal mitochondria are altered by axonal or demyelinating neuropathies. To answer this question, we imaged and quantified mitochondrial ATP and hydrogen peroxide (H2O2) in resting or stimulated peripheral nerve myelinated axons in vivo, using genetically-encoded fluorescent probes, two-photon time-lapse and CARS imaging. We found that ATP and H2O2 productions are intrinsically higher in nodes of Ranvier even in resting conditions. Axonal firing increased both ATP and H2O2 productions but with different dynamics: ROS production peaked shortly and transiently after the stimulation while ATP production increased gradually for a longer period of time. In neuropathic MFN2R94Q mice, mimicking Charcot-Marie-Tooth 2A disease, defective mitochondria failed to upregulate ATP production following axonal activity. However, elevated H2O2 production was largely sustained. Finally, inducing demyelination with lysophosphatidylcholine resulted in a reduced level of ATP while H2O2 level soared. Taken together, our results suggest that ATP and ROS productions are decoupled under neuropathic conditions, which may compromise axonal function and integrity.
Collapse
Affiliation(s)
- Gerben van Hameren
- Institut des Neurosciences de Montpellier, INSERM U1051, Université de Montpellier, 34091, Montpellier, France.
| | - Graham Campbell
- Institut des Neurosciences de Montpellier, INSERM U1051, Université de Montpellier, 34091, Montpellier, France
| | - Marie Deck
- Institut des Neurosciences de Montpellier, INSERM U1051, Université de Montpellier, 34091, Montpellier, France
| | - Jade Berthelot
- Institut des Neurosciences de Montpellier, INSERM U1051, Université de Montpellier, 34091, Montpellier, France
| | - Benoit Gautier
- Institut des Neurosciences de Montpellier, INSERM U1051, Université de Montpellier, 34091, Montpellier, France
| | - Patrice Quintana
- Institut des Neurosciences de Montpellier, INSERM U1051, Université de Montpellier, 34091, Montpellier, France
| | - Roman Chrast
- Department of Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
- Department of Clinical Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Nicolas Tricaud
- Institut des Neurosciences de Montpellier, INSERM U1051, Université de Montpellier, 34091, Montpellier, France.
| |
Collapse
|
30
|
Turner MP, Hubbard NA, Sivakolundu DK, Himes LM, Hutchison JL, Hart J, Spence JS, Frohman EM, Frohman TC, Okuda DT, Rypma B. Preserved canonicality of the BOLD hemodynamic response reflects healthy cognition: Insights into the healthy brain through the window of Multiple Sclerosis. Neuroimage 2019; 190:46-55. [PMID: 29454932 DOI: 10.1016/j.neuroimage.2017.12.081] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 12/18/2017] [Accepted: 12/22/2017] [Indexed: 10/18/2022] Open
Abstract
The hemodynamic response function (HRF), a model of brain blood-flow changes in response to neural activity, reflects communication between neurons and the vasculature that supplies these neurons in part by means of glial cell intermediaries (e.g., astrocytes). Intact neural-vascular communication might play a central role in optimal cognitive performance. This hypothesis can be tested by comparing healthy individuals to those with known white-matter damage and impaired performance, as seen in Multiple Sclerosis (MS). Glial cell intermediaries facilitate the ability of neurons to adequately convey metabolic needs to cerebral vasculature for sufficient oxygen and nutrient perfusion. In this study, we isolated measurements of the HRF that could quantify the extent to which white-matter affects neural-vascular coupling and cognitive performance. HRFs were modeled from multiple brain regions during multiple cognitive tasks using piecewise cubic spline functions, an approach that minimized assumptions regarding HRF shape that may not be valid for diseased populations, and were characterized using two shape metrics (peak amplitude and time-to-peak). Peak amplitude was reduced, and time-to-peak was longer, in MS patients relative to healthy controls. Faster time-to-peak was predicted by faster reaction time, suggesting an important role for vasodilatory speed in the physiology underlying processing speed. These results support the hypothesis that intact neural-glial-vascular communication underlies optimal neural and cognitive functioning.
Collapse
Affiliation(s)
- Monroe P Turner
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Nicholas A Hubbard
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Dinesh K Sivakolundu
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Lyndahl M Himes
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Joanna L Hutchison
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - John Hart
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA; Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jeffrey S Spence
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Elliot M Frohman
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Teresa C Frohman
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Darin T Okuda
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bart Rypma
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA; Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
31
|
Eisele P, Konstandin S, Szabo K, Ebert A, Roßmanith C, Paschke N, Kerschensteiner M, Platten M, Schoenberg SO, Schad LR, Gass A. Temporal evolution of acute multiple sclerosis lesions on serial sodium (23Na) MRI. Mult Scler Relat Disord 2019; 29:48-54. [DOI: 10.1016/j.msard.2019.01.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 01/11/2019] [Accepted: 01/16/2019] [Indexed: 10/27/2022]
|
32
|
De Santis S, Granberg T, Ouellette R, Treaba CA, Herranz E, Fan Q, Mainero C, Toschi N. Evidence of early microstructural white matter abnormalities in multiple sclerosis from multi-shell diffusion MRI. Neuroimage Clin 2019; 22:101699. [PMID: 30739842 PMCID: PMC6370560 DOI: 10.1016/j.nicl.2019.101699] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 12/07/2018] [Accepted: 01/28/2019] [Indexed: 12/29/2022]
Abstract
Irreversible white matter (WM) damage, including severe demyelination and axonal loss, is a main determinant of long-term disability in multiple sclerosis (MS). Non-invasive detection of changes in microstructural WM integrity in the disease is challenging since commonly used imaging metrics lack the necessary sensitivity, especially in the early phase of the disease. This study aims at assessing microstructural WM abnormalities in early-stage MS by using ultra-high gradient strength multi-shell diffusion MRI and the restricted signal fraction (FR) from the Composite Hindered and Restricted Model of Diffusion (CHARMED), a metric sensitive to the volume fraction of axons. In 22 early MS subjects (disease duration ≤5 years) and 15 age-matched healthy controls, restricted fraction estimates were obtained through the CHARMED model along with conventional Diffusion Tensor Imaging (DTI) metrics. All imaging parameters were compared cross-sectionally between the MS subjects and controls both in WM lesions and normal-appearing white matter (NAWM). We found a significant reduction in FR focally in WM lesions and widespread in the NAWM in MS patients relative to controls (corrected p < .05). Signal fraction changes in NAWM were not driven by perilesional tissue, nor were they influenced by proximity to the ventricles, challenging the hypothesis of an outside-in pathological process driven by CSF-mediated immune cytotoxic factors. No significant differences were found in conventional DTI parameters. In a cross-validated classification task, FR showed the largest effect size and outperformed all other diffusion imaging metrics in discerning lesions from contralateral NAWM. Taken together, our data provide evidence for the presence of widespread microstructural changes in the NAWM in early MS stages that are, at least in part, unrelated to focal demyelinating lesions. Interestingly, these pathological changes were not yet detectable by conventional diffusion imaging at this early disease stage, highlighting the sensitivity and value of multi-shell diffusion imaging for better characterizing axonal microstructure in MS.
Collapse
Affiliation(s)
- Silvia De Santis
- Instituto de Neurociencias de Alicante (CSIC-UMH), San Juan de Alicante, Spain; Cardiff University Brain Research Imaging Centre (CUBRIC), Cardiff University, Cardiff, UK
| | - Tobias Granberg
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Radiology, Karolinska University Hospital, Stockholm, Sweden; Athinoula A. Martinos Center for Biomedical Imaging and Harvard Medical School, Boston, MA, USA
| | - Russell Ouellette
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Athinoula A. Martinos Center for Biomedical Imaging and Harvard Medical School, Boston, MA, USA
| | - Constantina A Treaba
- Athinoula A. Martinos Center for Biomedical Imaging and Harvard Medical School, Boston, MA, USA
| | - Elena Herranz
- Athinoula A. Martinos Center for Biomedical Imaging and Harvard Medical School, Boston, MA, USA
| | - Qiuyun Fan
- Athinoula A. Martinos Center for Biomedical Imaging and Harvard Medical School, Boston, MA, USA
| | - Caterina Mainero
- Athinoula A. Martinos Center for Biomedical Imaging and Harvard Medical School, Boston, MA, USA
| | - Nicola Toschi
- Athinoula A. Martinos Center for Biomedical Imaging and Harvard Medical School, Boston, MA, USA.; Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
33
|
Lan M, Tang X, Zhang J, Yao Z. Insights in pathogenesis of multiple sclerosis: nitric oxide may induce mitochondrial dysfunction of oligodendrocytes. Rev Neurosci 2018; 29:39-53. [PMID: 28822986 DOI: 10.1515/revneuro-2017-0033] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 06/15/2017] [Indexed: 01/01/2023]
Abstract
Demyelinating diseases, such as multiple sclerosis (MS), are kinds of common diseases in the central nervous system (CNS), and originated from myelin loss and axonal damage. Oligodendrocyte dysfunction is the direct reason of demyelinating lesions in the CNS. Nitric oxide (NO) plays an important role in the pathological process of demyelinating diseases. Although the neurotoxicity of NO is more likely mediated by peroxynitrite rather than NO itself, NO can impair oligodendrocyte energy metabolism through mediating the damaging of mitochondrial DNA, mitochondrial membrane and mitochondrial respiratory chain complexes. In the progression of MS, NO can mainly mediate demyelination, axonal degeneration and cell death. Hence, in this review, we extensively discuss endangerments of NO in oligodendrocytes (OLs), which is suggested to be the main mediator in demyelinating diseases, e.g. MS. We hypothesize that NO takes part in MS through impairing the function of monocarboxylate transporter 1, especially causing axonal degeneration. Then, it further provides a new insight that NO for OLs may be a reliable therapeutic target to ameliorate the course of demyelinating diseases.
Collapse
Affiliation(s)
- Minghong Lan
- Department of Physiology, Third Military Medical University, Chongqing 400038, China
| | - Xiaoyi Tang
- Department of Physiology, Third Military Medical University, Chongqing 400038, China
| | - Jie Zhang
- Department of Physiology, Third Military Medical University, Chongqing 400038, China
| | - Zhongxiang Yao
- Department of Physiology, Third Military Medical University, Chongqing 400038, China
| |
Collapse
|
34
|
Murata H, Khine CC, Nishikawa A, Yamamoto KI, Kinoshita R, Sakaguchi M. c-Jun N-terminal kinase (JNK)-mediated phosphorylation of SARM1 regulates NAD + cleavage activity to inhibit mitochondrial respiration. J Biol Chem 2018; 293:18933-18943. [PMID: 30333228 DOI: 10.1074/jbc.ra118.004578] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 10/03/2018] [Indexed: 01/03/2023] Open
Abstract
Mitochondrial dysfunction is a key pathological feature of many different types of neurodegenerative disease. Sterile alpha and Toll/interleukin receptor motif-containing protein 1 (SARM1) has been attracting much attention as an important molecule for inducing axonal degeneration and neuronal cell death by causing loss of NAD (NADH). However, it has remained unclear what exactly regulates the SARM1 activity. Here, we report that NAD+ cleavage activity of SARM1 is regulated by its own phosphorylation at serine 548. The phosphorylation of SARM1 was mediated by c-jun N-terminal kinase (JNK) under oxidative stress conditions, resulting in inhibition of mitochondrial respiration concomitant with enhanced activity of NAD+ cleavage. Nonphosphorylatable mutation of Ser-548 or treatment with a JNK inhibitor decreased SARM1 activity. Furthermore, neuronal cells derived from a familial Parkinson's disease (PD) patient showed a congenitally increased level of SARM1 phosphorylation compared with that in neuronal cells from a healthy person and were highly sensitive to oxidative stress. These results indicate that JNK-mediated phosphorylation of SARM1 at Ser-548 is a regulator of SARM1 leading to inhibition of mitochondrial respiration. These findings suggest that an abnormal regulation of SARM1 phosphorylation is involved in the pathogenesis of Parkinson's disease and possibly other neurodegenerative diseases.
Collapse
Affiliation(s)
- Hitoshi Murata
- From the Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Cho Cho Khine
- From the Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Akane Nishikawa
- From the Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Ken-Ichi Yamamoto
- From the Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Rie Kinoshita
- From the Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Masakiyo Sakaguchi
- From the Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| |
Collapse
|
35
|
Rahim T, Becquart P, Baeva ME, Quandt J. Expression of the neuroprotective protein aryl hydrocarbon receptor nuclear translocator 2 correlates with neuronal stress and disability in models of multiple sclerosis. J Neuroinflammation 2018; 15:270. [PMID: 30231889 PMCID: PMC6145183 DOI: 10.1186/s12974-018-1290-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 08/23/2018] [Indexed: 01/07/2023] Open
Abstract
Background Axonal degeneration and neuronal loss have been described as the major causes of irreversible clinical disability in multiple sclerosis (MS). The aryl-hydrocarbon receptor nuclear translocator 2 (ARNT2) protein has been associated with neuroprotection in models of ischemia and neuronal responses to stressors. Methods To characterize its potential to influence inflammatory neurodegeneration, we examined ARNT2 expression in the experimental autoimmune encephalomyelitis (EAE) model of MS and characterized mediators that influence ARNT2 expression as well as plausible partners and targets. Results Arnt2 message and protein levels dropped significantly in EAE spinal cords as disease developed and were lowest at peak disability. ARNT2 expression is prominent in neuronal cell bodies within the gray matter with some staining in glial fibrillary acidic protein (GFAP)+ astrocytes in healthy animals. At peak disease, ARNT2 expression is reduced by 20–50% in gray matter neurons compared to healthy controls. ARNT2 intensity in neurons throughout the EAE spinal cord correlated inversely with the degree of immune cell infiltration (r = − 0.5085, p < 0.01) and axonal damage identified with SMI32 staining (r = − 0.376, p = 0.032). To understand the relationship between ARNT2 expression and neuronal health, we exposed enriched cortical cultures of neurons to hydrogen peroxide (H2O2) to mimic oxidative stress. H2O2 at lower doses rapidly increased ARNT2 protein levels which returned to baseline within 3–4 h. Exposure to higher doses of H2O2) dropped ARNT2 levels below baseline, preceding cytotoxicity measured by morphological changes and lactate dehydrogenase release from cells. Decreases in ARNT2 secondary to staurosporine and H2O2 preceded increases in cleaved caspase 3 and associated apoptosis. We also examined expression of neuronal pas 4 (Npas4), whose heterodimerization with ARNT2 drives expression of the neurotrophic factor brain-derived neurotrophic factor (Bdnf). Like ARNT2, Npas4 levels also decline at the onset of EAE and are linked to decreases in Bdnf. In vitro, H2O2 exposure drives Npas4 expression that is tied to increases in Bdnf. Conclusion Our data support ARNT2 as a neuronal transcription factor whose sustained expression is linked to neuronal and axonal health, protection that may primarily be driven through its partnering with Npas4 to influence BDNF expression. Electronic supplementary material The online version of this article (10.1186/s12974-018-1290-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tissa Rahim
- Department of Pathology and Laboratory Medicine, University of British Columbia, G227-2211 Wesbrook Mall, Vancouver, BC, V6T 2B5, Canada
| | - Pierre Becquart
- Department of Pathology and Laboratory Medicine, University of British Columbia, G227-2211 Wesbrook Mall, Vancouver, BC, V6T 2B5, Canada
| | - Maria-Elizabeth Baeva
- Department of Pathology and Laboratory Medicine, University of British Columbia, G227-2211 Wesbrook Mall, Vancouver, BC, V6T 2B5, Canada
| | - Jacqueline Quandt
- Department of Pathology and Laboratory Medicine, University of British Columbia, G227-2211 Wesbrook Mall, Vancouver, BC, V6T 2B5, Canada.
| |
Collapse
|
36
|
Abstract
The 1996 originally established multiple sclerosis (MS) subtypes, based solely on clinical impression and consensus, were revised in 2013 to review potential imaging and biological correlates and to reflect recently identified clinical aspects of MS. As a result, potential new disease phenotypes, radiologically isolated syndrome, and clinically isolated syndrome were considered along with the addition of two new descriptor subtypes: activity and progression applied to relapsing remitting and progressive MS phenotypes. In this way, the description of an individual patient's disease course is refined and provides temporal information about the ongoing disease process. There is still a lack of imaging and biological markers that would distinguish MS phenotypes and prognosticate the disease course on an individual patient's level, creating a pressing need for large collaborative research efforts in this field.
Collapse
Affiliation(s)
- Sylvia Klineova
- The CGD Center for Multiple Sclerosis, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Fred D Lublin
- The CGD Center for Multiple Sclerosis, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| |
Collapse
|
37
|
Correale J, Ysrraelit MC, Benarroch EE. Metabolic coupling of axons and glial cells. Neurology 2018; 90:737-744. [DOI: 10.1212/wnl.0000000000005339] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
38
|
|
39
|
Effect of mitochondrial apoptotic activation through the mitochondrial membrane permeability transition pore on yak meat tenderness during postmortem aging. Food Chem 2017; 234:323-331. [DOI: 10.1016/j.foodchem.2017.04.185] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 04/25/2017] [Accepted: 04/29/2017] [Indexed: 12/23/2022]
|
40
|
Qureshi M, Al-Suhaimi EA, Wahid F, Shehzad O, Shehzad A. Therapeutic potential of curcumin for multiple sclerosis. Neurol Sci 2017; 39:207-214. [PMID: 29079885 DOI: 10.1007/s10072-017-3149-5] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 10/07/2017] [Indexed: 01/08/2023]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune inflammatory disease of the central nervous system (CNS), characterized by demyelination, neuronal injury, and breaching of the blood-brain barrier (BBB). Epidemiological studies have shown that immunological, genetic, and environmental factors contribute to the progression and development of MS. T helper 17 (Th17) cells are crucial immunological participant in the pathophysiology of MS. The aberrant production of IL-17 and IL-22 by Th17 cells crosses BBB promotes its disruption and interferes with transmission of nerve signals through activation of neuroinflammation in the CNS. These inflammatory responses promote demyelination through transcriptional activation of signal transducers and activators of transcription-1 (STAT-1), nuclear factor kappa-B (NF-κB), matrix metalloproteinases (MMPs), interferon ϒ (IFNϒ), and Src homology region 2 domain-containing phosphatase-1 (SHP-1). B cells also contribute to disease progression through abnormal regulation of antibodies, cytokines, and antigen presentation. Additionally, oxidative stress has been known as a causative agent for the MS. Curcumin is a hydrophobic yellowish diphenolic component of turmeric, which can interact and modulate multiple cell signaling pathways and prevent the development of various autoimmune neurological diseases including MS. Studies have reported curcumin as a potent anti-inflammatory, antioxidant agent that could modulate cell cycle regulatory proteins, enzymes, cytokines, and transcription factors in CNS-related disorders including MS. The current study summarizes the reported knowledge on therapeutic potential of curcumin against MS, with future indication as neuroprotective and neuropharmacological drug.
Collapse
Affiliation(s)
- Munibah Qureshi
- Department of Biomedical Engineering and Sciences, School of Mechanical and Manufacturing Engineering, National University of Sciences and Technology, Islamabad, Pakistan
| | - Ebtesam A Al-Suhaimi
- Department of Biology, Sciences College, University of Dammam, Dammam, Saudi Arabia
| | - Fazli Wahid
- Biotechnology Program, Department of Environmental Sciences, COMSATS Institute of Information Technology, Abbottabad, 22060, Pakistan
| | - Omer Shehzad
- Department of pharmacy, Abdul Wali Khan University , Mardan, Khyber Pakhtunkhwa, Pakistan
| | - Adeeb Shehzad
- Department of Biomedical Engineering and Sciences, School of Mechanical and Manufacturing Engineering, National University of Sciences and Technology, Islamabad, Pakistan.
| |
Collapse
|
41
|
Chhablani PP, Ambiya V, Nair AG, Bondalapati S, Chhablani J. Retinal Findings on OCT in Systemic Conditions. Semin Ophthalmol 2017. [DOI: 10.1080/08820538.2017.1332233] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Preeti Patil Chhablani
- Srimati Kanuri Santhamma Centre for Vitreo Retinal Diseases, KAR Campus, L. V. Prasad Eye Institute, Hyderabad, Telangana, India
| | - Vikas Ambiya
- Srimati Kanuri Santhamma Centre for Vitreo Retinal Diseases, KAR Campus, L. V. Prasad Eye Institute, Hyderabad, Telangana, India
| | - Akshay G. Nair
- Srimati Kanuri Santhamma Centre for Vitreo Retinal Diseases, KAR Campus, L. V. Prasad Eye Institute, Hyderabad, Telangana, India
| | | | - Jay Chhablani
- Srimati Kanuri Santhamma Centre for Vitreo Retinal Diseases, KAR Campus, L. V. Prasad Eye Institute, Hyderabad, Telangana, India
| |
Collapse
|
42
|
Yong H, Chartier G, Quandt J. Modulating inflammation and neuroprotection in multiple sclerosis. J Neurosci Res 2017; 96:927-950. [PMID: 28580582 DOI: 10.1002/jnr.24090] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 04/17/2017] [Accepted: 05/04/2017] [Indexed: 12/13/2022]
Abstract
Multiple sclerosis (MS) is a neurological disorder of the central nervous system with a presentation and disease course that is largely unpredictable. MS can cause loss of balance, impaired vision or speech, weakness and paralysis, fatigue, depression, and cognitive impairment. Immunomodulation is a major target given the appearance of focal demyelinating lesions in myelin-rich white matter, yet progression and an increasing appreciation for gray matter involvement, even during the earliest phases of the disease, highlights the need to afford neuroprotection and limit neurodegenerative processes that correlate with disability. This review summarizes key aspects of MS pathophysiology and histopathology with a focus on neuroimmune interactions in MS, which may facilitate neurodegeneration through both direct and indirect mechanisms. There is a focus on processes thought to influence disease progression and the role of oxidative stress and mitochondrial dysfunction in MS. The goals and efficacy of current disease-modifying therapies and those in the pipeline are discussed, highlighting recent advances in our understanding of pathways mediating disease progression to identify and translate both immunomodulatory and neuroprotective therapeutics from the bench to the clinic.
Collapse
Affiliation(s)
- Heather Yong
- Department of Pathology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Gabrielle Chartier
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jacqueline Quandt
- Department of Pathology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
43
|
Neil S, Huh J, Baronas V, Li X, McFarland HF, Cherukuri M, Mitchell JB, Quandt JA. Oral administration of the nitroxide radical TEMPOL exhibits immunomodulatory and therapeutic properties in multiple sclerosis models. Brain Behav Immun 2017; 62:332-343. [PMID: 28238951 PMCID: PMC5496657 DOI: 10.1016/j.bbi.2017.02.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 02/13/2017] [Accepted: 02/22/2017] [Indexed: 01/01/2023] Open
Abstract
Therapies with both immunomodulatory and neuroprotective properties are thought to have the greatest promise in reducing the severity and progression of multiple sclerosis (MS). Several reactive oxygen (ROS) and reactive nitrogen species (RNS) are implicated in inflammatory-mediated damage to the central nervous system (CNS) in MS and its animal model, experimental autoimmune encephalomyelitis (EAE). TEMPOL (4-hydroxy-2,2,6,6-tetramethylpiperidine-N-oxyl) is a stable nitroxide radical with potent antioxidant activity. The goal of our studies was to investigate the immunomodulatory effects and therapeutic potential of orally-delivered TEMPOL in the mouse EAE model. Mice receiving TEMPOL chow ad libitum for 2weeks prior to induction of active EAE showed delayed onset and reduced incidence of disease compared to control-fed animals. Reduced disease severity was associated with limited microglial activation and fewer inflammatory infiltrates. TEMPOL's effects were immunomodulatory, not immunosuppressive: T cells produced less interferon-γ and tumor necrosis factor-α, and TEMPOL-fed mice exhibited a shift towards TH2-type antibody responses. Both myeloid and myeloid-dendritic cells of TEMPOL-fed EAE animals had significantly lower levels of MHC class II expression than controls; CD40 was also significantly reduced. TEMPOL administration was associated with an enrichment of CD8+ T cell populations and CD4+FoxP3+ regulatory populations. TEMPOL reduced the severity of clinical disease when administered after the induction of disease, and also after the onset of clinical symptoms. To exclude effects on T cell priming in vivo, TEMPOL was tested with the passive transfer of encephalitogenic T cells and was found to reduce the incidence and peak severity of disease. Protection was associated with reduced infiltrates and a relative sparing of neurofilaments and axons. The ability of oral TEMPOL to reduce inflammation and axonal damage and loss demonstrate both anti-inflammatory and protective properties, with significant promise for the treatment of MS and related neurological disorders.
Collapse
Affiliation(s)
- Sarah Neil
- University of British Columbia, Department of Pathology & Laboratory Medicine, Vancouver, Canada
| | - Jaebong Huh
- Neuroimmunology Branch, NINDS, NIH, Bethesda, MD 20892 USA
| | - Victoria Baronas
- University of British Columbia, Department of Pathology & Laboratory Medicine, Vancouver, Canada
| | - Xinhui Li
- Neuroimmunology Branch, NINDS, NIH, Bethesda, MD 20892 USA
| | | | | | | | - Jacqueline A. Quandt
- University of British Columbia, Department of Pathology & Laboratory Medicine, Vancouver, Canada,To whom correspondence should be addressed: University of British Columbia, Department of Pathology & Laboratory Medicine, G227-2211 Wesbrook Mall, Vancouver, B.C. V6T 2B5, Canada,
| |
Collapse
|
44
|
Varhaug KN, Vedeler CA, Tzoulis C, Bindoff LA. [Multiple sclerosis - a mitochondria-mediated disease?]. TIDSSKRIFT FOR DEN NORSKE LEGEFORENING 2017; 137:284-287. [PMID: 28225235 DOI: 10.4045/tidsskr.16.0210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
BACKGROUND Mitochondria play an important role in the pathogenesis of various neurodegenerative disorders, including Parkinson's disease. Neurodegenerative changes occur early in the course of multiple sclerosis (MS). This article aims to present information on a possible association between mitochondrial dysfunction and multiple sclerosis.MATERIAL AND METHOD The article is based on original and review articles selected following a literature search in PubMed, restricted to articles written in English, and concluded in May 2016. The literature search resulted in a total of 2276 articles. After a discretionary evaluation by the authors, 71 articles were read in full. Of these, 19 were used as references. In addition, we included 15 articles from reference lists and seven from the authors' own literature archive.RESULTS Mitochondrial changes have been demonstrated in affected areas of the brains of patients with MS. Although some of the changes may be attributed to mitochondrial damage that is secondary to inflammation, others may be compensatory due to the increased energy demands of demyelinated axons. The type of mitochondrial damage varies and is dependent on the pathology that triggers it.INTERPRETATION Mitochondrial damage secondary to inflammation, combined with increased energy demands secondary to demyelination, may result in a chronic energy deficiency in the central nervous system. This in turn may lead to neurodegeneration. Improved knowledge of the role of mitochondria in MS, both secondary to inflammation and possibly as a direct contributor to neurodegeneration, may provide a better understanding of the pathogenesis of the disease and perhaps contribute to new treatment options.
Collapse
Affiliation(s)
- Kristin N Varhaug
- Klinisk institutt 1 Universitetet i Bergen og Nevrologisk avdeling Haukeland universitetssykehus
| | - Christian A Vedeler
- Klinisk institutt 1 Universitetet i Bergen og Nevrologisk avdeling Haukeland universitetssykehus
| | - Charalampos Tzoulis
- Klinisk institutt 1 Universitetet i Bergen og Nevrologisk avdeling Haukeland universitetssykehus
| | - Laurence A Bindoff
- Klinisk institutt 1 Universitetet i Bergen og Nevrologisk avdeling Haukeland universitetssykehus
| |
Collapse
|
45
|
Gianfrancesco MA, Glymour MM, Walter S, Rhead B, Shao X, Shen L, Quach H, Hubbard A, Jónsdóttir I, Stefánsson K, Strid P, Hillert J, Hedström A, Olsson T, Kockum I, Schaefer C, Alfredsson L, Barcellos LF. Causal Effect of Genetic Variants Associated With Body Mass Index on Multiple Sclerosis Susceptibility. Am J Epidemiol 2017; 185:162-171. [PMID: 28073764 DOI: 10.1093/aje/kww120] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 06/02/2016] [Indexed: 12/23/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease with both genetic and environmental risk factors. Recent studies indicate that childhood and adolescent obesity double the risk of MS, but this association may reflect unmeasured confounders rather than causal effects of obesity. We used separate-sample Mendelian randomization to estimate the causal effect of body mass index (BMI) on susceptibility to MS. Using data from non-Hispanic white members of the Kaiser Permanente Medical Care Plan of Northern California (KPNC) (2006-2014; 1,104 cases of MS and 10,536 controls) and a replication data set from Sweden (the Epidemiological Investigation of MS (EIMS) and the Genes and Environment in MS (GEMS) studies, 2005-2013; 5,133 MS cases and 4,718 controls), we constructed a weighted genetic risk score using 97 variants previously established to predict BMI. Results were adjusted for birth year, sex, education, smoking status, ancestry, and genetic predictors of MS. Estimates in KPNC and Swedish data sets suggested that higher genetically induced BMI predicted greater susceptibility to MS (odds ratio = 1.13, 95% confidence interval: 1.04, 1.22 for the KPNC sample; odds ratio = 1.09, 95% confidence interval: 1.03, 1.15 for the Swedish sample). Although the mechanism remains unclear, to our knowledge, these findings support a causal effect of increased BMI on susceptibility to MS for the first time, and they suggest a role for inflammatory pathways that characterize both obesity and the MS disease process.
Collapse
Affiliation(s)
- Milena A Gianfrancesco
- Genetic Epidemiology and Genomics Lab, Division of Epidemiology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - M Maria Glymour
- Department of Epidemiology and Biostatistics, University of California, San Francisco, USA
| | - Stefan Walter
- Institute of Clinical and Experimental Medicine, Division of Gastroenterology, Linköping University, Linköping, Sweden
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| | - Brooke Rhead
- Computational Biology Graduate Group, University of California, Berkeley, CA, USA
| | - Xiaorong Shao
- Centre for Reproductive Medicine, Dalian Maternal and Children's Centre, Dalian, China
| | - Ling Shen
- Collaborative Innovation Center on Forecast and Evaluation of Meteorological Disasters, Jiangsu Key Laboratory of Agricultural Meteorology, Department of Agricultural Resource and Environment, College of Applied Meteorology, University of Information Science and Technology, Nanjing, 210044, China
| | - Hong Quach
- Human Evolutionary Genetics, Institut Pasteur, 75015 Paris, France
- Centre National de la Recherche Scientifique URA3012, 75015 Paris, France
- Center of Bioinformatics, Biostatistics, and Integrative Biology, Institut Pasteur, 75015 Paris, France
| | - Alan Hubbard
- Institute of Pharmaceutical Science, King's College London, London, UK; Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Ingileif Jónsdóttir
- deCODE Genetics/Amgen, Inc., Reykjavik, Iceland
- Department of Immunology, Landspitali, the National University Hospital of Iceland, Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Kári Stefánsson
- deCODE Genetics/Amgen, Inc., Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | | | - Jan Hillert
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Anna Hedström
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Tomas Olsson
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Ingrid Kockum
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden/Department of Neurology, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Catherine Schaefer
- Department of Soil Science, Federal University of Viçosa, Viçosa, MG, Brazil
| | - Lars Alfredsson
- Centre for Occupational and Environmental Medicine, Stockholm County Council, Sweden
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lisa F Barcellos
- Division of Epidemiology, School of Public Health, University of California, 324 Stanley Hall, Berkeley, CA, USA
| |
Collapse
|
46
|
Ischemic optic neuropathy as a model of neurodegenerative disorder: A review of pathogenic mechanism of axonal degeneration and the role of neuroprotection. J Neurol Sci 2016; 375:430-441. [PMID: 28320183 DOI: 10.1016/j.jns.2016.12.044] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 12/20/2016] [Accepted: 12/22/2016] [Indexed: 02/07/2023]
Abstract
Optic neuropathy is a neurodegenerative disease which involves optic nerve injury. It is caused by acute or intermittent insults leading to visual dysfunction. There are number of factors, responsible for optic neuropathy, and the optic nerve axon is affected in all type which causes the loss of retinal ganglion cells. In this review we will highlight various mechanisms involved in the cell loss cascades during axonal degeneration as well as ischemic optic neuropathy. These mechanisms include oxidative stress, excitotoxicity, angiogenesis, neuroinflammation and apoptosis following retinal ischemia. We will also discuss the effect of neuroprotective agents in attenuation of the negative effect of factors involve in the disease occurrence and progression.
Collapse
|
47
|
Eisele P, Alonso A, Szabo K, Gass A. Reduced diffusion in acute cervical cord multiple sclerosis lesions. Neurol Clin Pract 2016; 7:401-403. [PMID: 29620066 DOI: 10.1212/cpj.0000000000000309] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 09/06/2016] [Indexed: 11/15/2022]
Affiliation(s)
- Philipp Eisele
- Department of Neurology, Universitätsmedizin Mannheim, University of Heidelberg, Mannheim, Germany
| | - Angelika Alonso
- Department of Neurology, Universitätsmedizin Mannheim, University of Heidelberg, Mannheim, Germany
| | - Kristina Szabo
- Department of Neurology, Universitätsmedizin Mannheim, University of Heidelberg, Mannheim, Germany
| | - Achim Gass
- Department of Neurology, Universitätsmedizin Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
48
|
Alizadeh-Ghodsi M, Zavvari A, Ebrahimi-Kalan A, Shiri-Shahsavar MR, Yousefi B. The hypothetical roles of arsenic in multiple sclerosis by induction of inflammation and aggregation of tau protein: A commentary. Nutr Neurosci 2016; 21:92-96. [PMID: 27697018 DOI: 10.1080/1028415x.2016.1239399] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Multiple sclerosis (MS) is a disease which manifests demyelination of neuronal cells in the brain. Despite extensive research on the mechanisms of disease development and progression, the exact mechanism is not elucidated yet, which has hampered drug development and subsequent treatment of the disease. We have recently shown that the serum levels of arsenic and malondialdehyde, a lipid peroxidation marker, are high in MS patients. In this article, we would like to formulate the hypothesis that arsenic may cause MS by induction of inflammation, degeneration, and apoptosis in neuronal cells. The induction of ROS generation in cells upon exposure to arsenic as a heavy metal may be involved in the pathogenesis of MS. Tau protein, a member of the family of microtubule-associated proteins, is mainly expressed in neurons and contribute to the assembly of neuronal microtubules network. Arsenic may affect the hyperphosphorylation and aggregation of tau proteins and may be involved in the cascade leading to deregulation of tau function associated with neurodegeneration. For validation of this hypothesis, studies might be conducted to evaluate the association of arsenic levels and tau protein levels in MS patients. Further studies might also focus on the trafficking along microtubules in neurons of MS patient with regard to hyperphosphorylation of tau protein. This hypothesis may add a new dimension to the understanding of MS etiology and help to design novel therapeutic agents against potential targets that might be discovered. If this hypothesis proves to be true, tau phosphorylation inhibitors can be potential candidates for MS drug development.
Collapse
Affiliation(s)
- Mohammadreza Alizadeh-Ghodsi
- a Neurosciences Research Center , Tabriz University of Medical Sciences , Tabriz , Iran.,b Department of Biochemistry and Clinical Laboratories, Faculty of Medicine , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Ali Zavvari
- b Department of Biochemistry and Clinical Laboratories, Faculty of Medicine , Tabriz University of Medical Sciences , Tabriz , Iran.,c Immunology Research Center , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Abbas Ebrahimi-Kalan
- a Neurosciences Research Center , Tabriz University of Medical Sciences , Tabriz , Iran.,d Neurosciences Department, School of Advanced Medical Sciences , Tabriz University of Medical Sciences, Tabriz , Iran
| | - Mohammad Reza Shiri-Shahsavar
- e Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, International Campus , Tehran University of Medical Sciences , Tehran , Iran
| | - Bahman Yousefi
- c Immunology Research Center , Tabriz University of Medical Sciences , Tabriz , Iran.,f Students' Research Committee , Tabriz University of Medical Sciences , Tabriz , Iran
| |
Collapse
|
49
|
Biasutto L, Azzolini M, Szabò I, Zoratti M. The mitochondrial permeability transition pore in AD 2016: An update. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1863:2515-30. [PMID: 26902508 DOI: 10.1016/j.bbamcr.2016.02.012] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 02/04/2016] [Accepted: 02/05/2016] [Indexed: 12/13/2022]
Abstract
Over the past 30years the mitochondrial permeability transition - the permeabilization of the inner mitochondrial membrane due to the opening of a wide pore - has progressed from being considered a curious artifact induced in isolated mitochondria by Ca(2+) and phosphate to a key cell-death-inducing process in several major pathologies. Its relevance is by now universally acknowledged and a pharmacology targeting the phenomenon is being developed. The molecular nature of the pore remains to this day uncertain, but progress has recently been made with the identification of the FOF1 ATP synthase as the probable proteic substrate. Researchers sharing this conviction are however divided into two camps: these believing that only the ATP synthase dimers or oligomers can form the pore, presumably in the contact region between monomers, and those who consider that the ring-forming c subunits in the FO sector actually constitute the walls of the pore. The latest development is the emergence of a new candidate: Spastic Paraplegia 7 (SPG7), a mitochondrial AAA-type membrane protease which forms a 6-stave barrel. This review summarizes recent developments of research on the pathophysiological relevance and on the molecular nature of the mitochondrial permeability transition pore. This article is part of a Special Issue entitled: Mitochondrial Channels edited by Pierre Sonveaux, Pierre Maechler and Jean-Claude Martinou.
Collapse
Affiliation(s)
- Lucia Biasutto
- CNR Neuroscience Institute, Viale G. Colombo 3, 35121 Padova, Italy; University of Padova, Department of Biomedical Sciences, Viale G. Colombo 3, 35121 Padova, Italy
| | - Michele Azzolini
- CNR Neuroscience Institute, Viale G. Colombo 3, 35121 Padova, Italy; University of Padova, Department of Biomedical Sciences, Viale G. Colombo 3, 35121 Padova, Italy
| | - Ildikò Szabò
- CNR Neuroscience Institute, Viale G. Colombo 3, 35121 Padova, Italy; University of Padova, Department of Biology, Viale G. Colombo 3, 35121 Padova, Italy
| | - Mario Zoratti
- CNR Neuroscience Institute, Viale G. Colombo 3, 35121 Padova, Italy; University of Padova, Department of Biomedical Sciences, Viale G. Colombo 3, 35121 Padova, Italy.
| |
Collapse
|
50
|
Nimmagadda VKC, Makar TK, Chandrasekaran K, Sagi AR, Ray J, Russell JW, Bever CT. SIRT1 and NAD+ precursors: Therapeutic targets in multiple sclerosis a review. J Neuroimmunol 2016; 304:29-34. [PMID: 27474445 DOI: 10.1016/j.jneuroim.2016.07.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 07/06/2016] [Indexed: 12/16/2022]
Abstract
Neurodegeneration is an important determinant of disability in multiple sclerosis (MS) but while currently approved treatments reduce inflammation, they have not been shown to reduce neurodegeneration. SIRT1, a NAD dependent protein deacetylase, has been implicated in the pathogenesis of neurodegeneration in neurological diseases including MS. We have studied the role of SIRT1 in experimental autoimmune encephalomyelitis (EAE) and found evidence for a neuroprotective role. In this review we summarize the most recent findings from the use of SIRT1 activators and SIRT1 overexpression in transgenic mice. These data support provide a rational for the use of SIRT1 activators in MS.
Collapse
Affiliation(s)
- Vamshi K C Nimmagadda
- Department of Neurology, University of Maryland, Baltimore, MD 21201, USA; Research Service, VA Maryland Health Care System, Baltimore, MD 21201, USA
| | - Tapas K Makar
- Department of Neurology, University of Maryland, Baltimore, MD 21201, USA; Research Service, VA Maryland Health Care System, Baltimore, MD 21201, USA; VA Multiple Sclerosis Center of Excellence East, Baltimore, MD 21201, USA
| | | | - Avinash Rao Sagi
- Department of Neurology, University of Maryland, Baltimore, MD 21201, USA
| | - Jayanta Ray
- Department of Neurology, University of Maryland, Baltimore, MD 21201, USA
| | - James W Russell
- Department of Neurology, University of Maryland, Baltimore, MD 21201, USA; Research Service, VA Maryland Health Care System, Baltimore, MD 21201, USA
| | - Christopher T Bever
- Department of Neurology, University of Maryland, Baltimore, MD 21201, USA; Research Service, VA Maryland Health Care System, Baltimore, MD 21201, USA; VA Multiple Sclerosis Center of Excellence East, Baltimore, MD 21201, USA.
| |
Collapse
|