1
|
Carver JJ, Bunner WP, Denbrock RR, Yin C, Huang W, Szatmari EM, Didonna A. Loss of ADAP1/CentA1 Protects Against Autoimmune Demyelination. FASEB J 2025; 39:e70604. [PMID: 40326762 PMCID: PMC12054340 DOI: 10.1096/fj.202403078r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 03/25/2025] [Accepted: 04/25/2025] [Indexed: 05/07/2025]
Abstract
ArfGAP with dual PH domain-containing protein 1 (ADAP1), also known as Centaurin alpha-1 (CentA1), is an actin-binding protein highly expressed in the central nervous system (CNS) that was previously shown to regulate dendritic spine density and plasticity. In the context of disease, ADAP1/CentA1 has been linked to Alzheimer's disease (AD) pathogenesis, cancer progression, and human immunodeficiency virus (HIV) reactivation. Here, we document that ADAP1/CentA1 is also mechanistically involved in CNS autoimmunity. We show that ADAP1/CentA1 deficient mice exhibit partial resistance to developing experimental autoimmune encephalomyelitis (EAE), an in vivo disease model recapitulating several features of multiple sclerosis (MS) pathogenesis. MS is a chronic autoimmune disorder of the CNS characterized by focal immune cell infiltration, demyelination, and axonal injury. Its etiology is still elusive, but genetic and environmental factors contribute to disease risk. By combining detailed immunophenotyping and single-cell RNA sequencing (scRNA-seq), we demonstrate that ADAP1/CentA1 is necessary for mounting a sufficient autoimmune response for EAE initiation and progression. In particular, the current study highlights that ADAP1/CentA1 expression in the immune system mainly targets the functioning of regulatory T cells (Tregs), monocytes, and natural killer (NK) cells. In summary, our study defines a novel function for ADAP/CentA1 outside of the CNS and helps elucidate the early molecular events taking place in the peripheral immune system in response to encephalitogenic challenges.
Collapse
MESH Headings
- Animals
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Mice
- Mice, Inbred C57BL
- Multiple Sclerosis/immunology
- Multiple Sclerosis/metabolism
- Mice, Knockout
- Female
- Microfilament Proteins/genetics
- Microfilament Proteins/metabolism
- Autoimmunity
Collapse
Affiliation(s)
- Jonathan J. Carver
- Department of Anatomy and Cell Biology, Brody School of MedicineEast Carolina UniversityGreenvilleNorth CarolinaUSA
| | - Wyatt P. Bunner
- Department of Physical Therapy, College of Allied Health SciencesEast Carolina UniversityGreenvilleNorth CarolinaUSA
- Center for Immunotherapy & Precision Immuno‐OncologyCleveland ClinicClevelandOhioUSA
| | - Rachael R. Denbrock
- Department of Anatomy and Cell Biology, Brody School of MedicineEast Carolina UniversityGreenvilleNorth CarolinaUSA
| | - Changhong Yin
- Department of Pathology and Laboratory Medicine, Brody School of MedicineEast Carolina UniversityGreenvilleNorth CarolinaUSA
| | - Weihua Huang
- Department of Pathology and Laboratory Medicine, Brody School of MedicineEast Carolina UniversityGreenvilleNorth CarolinaUSA
| | - Erzsebet M. Szatmari
- Department of Physical Therapy, College of Allied Health SciencesEast Carolina UniversityGreenvilleNorth CarolinaUSA
| | - Alessandro Didonna
- Department of Anatomy and Cell Biology, Brody School of MedicineEast Carolina UniversityGreenvilleNorth CarolinaUSA
| |
Collapse
|
2
|
Kim MW, Kipnis J. Glymphatics and meningeal lymphatics unlock the brain-immune code. Immunity 2025; 58:1040-1051. [PMID: 40324376 DOI: 10.1016/j.immuni.2025.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/06/2025] [Accepted: 03/06/2025] [Indexed: 05/07/2025]
Abstract
The central nervous system (CNS) was once perceived as entirely shielded from the immune system, protected behind the blood-brain barrier and thought to lack lymphatic drainage. However, recent evidence has challenged many dogmas in neuroimmunology. Indeed, by means of glymphatics, brain-derived "waste" from deep within the CNS mobilizes toward immunologically active brain borders, where meningeal lymphatic vessels are appropriately positioned to drain antigens from the brain to the periphery. Accordingly, the presentation of brain-derived self-peptides emerges at the brain's borders and drives T cell responses with suppressive properties, critical in allowing active immunosurveillance while limiting aberrant immune reactivity. Taking into consideration these concepts, we further discuss how inflammation, aging, and neurodegenerative diseases potentially reshape the repertoire of self-antigens and immune cells, disrupting the healthy dialogue between the CNS and immune system. Collectively, this evolving perspective unveils new therapeutic avenues for CNS pathologies.
Collapse
Affiliation(s)
- Min Woo Kim
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Medical Scientist Training Program, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.
| | - Jonathan Kipnis
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Medical Scientist Training Program, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
3
|
Vladymyrov M, Marchetti L, Aydin S, Soldati SGN, Mossu A, Pal A, Gueissaz L, Ariga A, Engelhardt B. UFMTrack, an Under-Flow Migration Tracker enabling analysis of the entire multi-step immune cell extravasation cascade across the blood-brain barrier in microfluidic devices. eLife 2025; 13:RP91150. [PMID: 40230092 PMCID: PMC11999694 DOI: 10.7554/elife.91150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025] Open
Abstract
The endothelial blood-brain barrier (BBB) strictly controls immune cell trafficking into the central nervous system (CNS). In neuroinflammatory diseases such as multiple sclerosis, this tight control is, however, disturbed, leading to immune cell infiltration into the CNS. The development of in vitro models of the BBB combined with microfluidic devices has advanced our understanding of the cellular and molecular mechanisms mediating the multistep T-cell extravasation across the BBB. A major bottleneck of these in vitro studies is the absence of a robust and automated pipeline suitable for analyzing and quantifying the sequential interaction steps of different immune cell subsets with the BBB under physiological flow in vitro. Here, we present the under-flow migration tracker (UFMTrack) framework for studying immune cell interactions with endothelial monolayers under physiological flow. We then showcase a pipeline built based on it to study the entire multistep extravasation cascade of immune cells across brain microvascular endothelial cells under physiological flow in vitro. UFMTrack achieves 90% track reconstruction efficiency and allows for scaling due to the reduction of the analysis cost and by eliminating experimenter bias. This allowed for an in-depth analysis of all behavioral regimes involved in the multistep immune cell extravasation cascade. The study summarizes how UFMTrack can be employed to delineate the interactions of CD4+ and CD8+ T cells with the BBB under physiological flow. We also demonstrate its applicability to the other BBB models, showcasing broader applicability of the developed framework to a range of immune cell-endothelial monolayer interaction studies. The UFMTrack framework along with the generated datasets is publicly available in the corresponding repositories.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Akitaka Ariga
- Laboratory for High Energy Physics, University of BernBernSwitzerland
| | | |
Collapse
|
4
|
Zahoor I, Mir S, Giri S. Profiling Blood-Based Neural Biomarkers and Cytokines in Experimental Autoimmune Encephalomyelitis Model of Multiple Sclerosis Using Single-Molecule Array Technology. Int J Mol Sci 2025; 26:3258. [PMID: 40244087 PMCID: PMC11989419 DOI: 10.3390/ijms26073258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 03/18/2025] [Accepted: 03/25/2025] [Indexed: 04/18/2025] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is a preclinical animal model widely used to study multiple sclerosis (MS). Blood-based analytes, including cytokines and neural biomarkers are the predictors of neurodegeneration, disease activity, and disability in patients with MS. However, understudied confounding factors cause variation in reports on EAE across animal strains/studies, limiting the utility of these biomarkers for predicting disease activity. In this study, we investigated blood-based analyte profiles, including neural markers (NFL and GFAP) and cytokines (IL-6, IL-17, IL-12p70, IL-10, and TNF-α), in two clinically distinct EAE models: relapsing-remitting (RR)-EAE and chronic-EAE. Ultrasensitive single-molecule array technology (SIMOA, Quanterix) was used to profile the analytes in the blood plasma of mice at the acute, chronic, and progressive phases of disease. In both models, NFL was substantially increased during post-disease onset across all phases, with a pronounced increase observed in chronic-EAE. The leakage of GFAP into peripheral blood was also greater after disease onset in both EAE models, especially in the acute phase of chronic-EAE. Among all cytokines, only IL-10 had consistently lower levels in both EAE models throughout the course of disease. This study suggests NFL, GFAP, and IL-10 as potential translational predictors of disease activity in EAE, making them potential candidates as surrogate markers for the preclinical testing of therapeutic interventions in animal models of MS.
Collapse
Affiliation(s)
- Insha Zahoor
- Department of Neurology, Henry Ford Health, Detroit, MI 48202, USA;
| | | | - Shailendra Giri
- Department of Neurology, Henry Ford Health, Detroit, MI 48202, USA;
| |
Collapse
|
5
|
Zahoor I, Mir S, Giri S. Profiling blood-based neural biomarkers and cytokines in experimental autoimmune encephalomyelitis model of multiple sclerosis using single-molecule array technology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.12.25.573285. [PMID: 38234812 PMCID: PMC10793409 DOI: 10.1101/2023.12.25.573285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Experimental autoimmune encephalomyelitis (EAE) is a preclinical animal model widely used to study multiple sclerosis (MS). Blood-based cytokines and neural biomarkers are predictors of neurodegeneration, disease activity, and disability in patients with MS. However, understudied confounding factors cause variation in reports on EAE across animal strains/studies, limiting the utility of these biomarkers for predicting disease activity. In this study, we investigated blood-based analyte profiles, including neural markers (NFL and GFAP) and cytokines (IL-6, IL-17, IL-12p70, IL-10, and TNF-α), in two clinically distinct EAE models: relapsing-remitting (RR)-EAE and chronic-EAE. Ultrasensitive single-molecule array technology (SIMOA, Quanterix) was used to profile the analytes in blood plasma of mice at the acute, chronic, and progressive phases of disease. In both models, NFL was substantially increased during post-disease onset at the peak, chronic, and progressive phases, with pronounced increase in chronic-EAE. Leakage of GFAP into peripheral blood was also greater after disease onset in both EAE models, especially in the acute phase of chronic-EAE. Among all cytokines, only IL-10 had consistently lower levels in both EAE models throughout the course of disease. This study suggests NFL, GFAP, and IL-10 as potential translational predictors of disease activity in EAE, making them potential candidates for surrogate markers for preclinical testing of therapeutic interventions in animal models of MS.
Collapse
Affiliation(s)
- Insha Zahoor
- Department of Neurology, Henry Ford Health, Detroit, MI 48202, USA
| | - Sajad Mir
- Department of Neurology, Henry Ford Health, Detroit, MI 48202, USA
| | - Shailendra Giri
- Department of Neurology, Henry Ford Health, Detroit, MI 48202, USA
| |
Collapse
|
6
|
García-Salas R, Cilleros-Holgado P, Di Spirito A, Gómez-Fernández D, Piñero-Pérez R, Romero-Domínguez JM, Álvarez-Córdoba M, Reche-López D, Romero-González A, López-Cabrera A, Sánchez-Alcázar JA. Mitochondrial dysfunction, iron accumulation, lipid peroxidation, and inflammasome activation in cellular models derived from patients with multiple sclerosis. Aging (Albany NY) 2025; 17:365-392. [PMID: 39918890 PMCID: PMC11892916 DOI: 10.18632/aging.206198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 01/21/2025] [Indexed: 02/09/2025]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS). Despite advancements in managing relapsing active illness, effective treatments for the irreversible progressive decline in MS remain limited. Research employing skin fibroblasts obtained from patients with neurological disorders revealed modifications in cellular stress pathways and bioenergetics. However, research using MS patient-derived cellular models is scarce. In this study, we collected fibroblasts from two MS patients to investigate cellular pathological alterations. We observed that MS fibroblasts showed a senescent morphology associated with iron/lipofuscin accumulation and altered expression of iron metabolism proteins. In addition, we found increased lipid peroxidation and downregulation of antioxidant enzymes expression levels in MS fibroblasts. When challenged against erastin, a ferroptosis inducer, MS fibroblasts showed decreased viability, suggesting increased sensitivity to ferroptosis. Furthermore, MS fibroblasts presented alterations in the expression levels of autophagy-related proteins. Interestingly, these alterations were associated with mitochondrial dysfunction and inflammasome activation. These findings were validated in 7 additional patient-derived cell lines. Our findings suggest that the underlying stress phenotype of MS fibroblasts may be disease-specific and recapitulate the main cellular pathological alterations found in the disease such as mitochondrial dysfunction, iron accumulation, lipid peroxidation, inflammasome activation, and pro-inflammatory cytokine production.
Collapse
Affiliation(s)
- Raquel García-Salas
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Sevilla 41013, Spain
| | - Paula Cilleros-Holgado
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Sevilla 41013, Spain
| | - Anna Di Spirito
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Sevilla 41013, Spain
| | - David Gómez-Fernández
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Sevilla 41013, Spain
| | - Rocío Piñero-Pérez
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Sevilla 41013, Spain
| | | | - Mónica Álvarez-Córdoba
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Sevilla 41013, Spain
| | - Diana Reche-López
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Sevilla 41013, Spain
| | - Ana Romero-González
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Sevilla 41013, Spain
| | - Alejandra López-Cabrera
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Sevilla 41013, Spain
| | | |
Collapse
|
7
|
Hashemi E, Srivastava IN, Aguirre A, Yoseph ET, Kaushal E, Awani A, Ryu JK, Akassoglou K, Talebian S, Chu P, Pisani L, Musolino P, Steinman L, Doyle K, Robinson WH, Sharpe O, Cayrol R, Orchard PJ, Lund T, Vogel H, Lenail M, Han MH, Bonkowsky JL, Van Haren KP. A Novel Mouse Model for Cerebral Inflammatory Demyelination in X-Linked Adrenoleukodystrophy: Insights into Pathogenesis and Potential Therapeutic Targets. Ann Neurol 2025; 97:296-312. [PMID: 39467011 PMCID: PMC11747894 DOI: 10.1002/ana.27117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 10/04/2024] [Accepted: 10/04/2024] [Indexed: 10/30/2024]
Abstract
OBJECTIVE X-linked adrenoleukodystrophy (ALD) is caused by mutations in ABCD1, a peroxisomal gene. More than half of males with an ABCD1 mutation develop inflammatory cerebral demyelination (cALD), but underlying mechanisms remain unknown and therapies are limited. We sought to develop and characterize a mouse model of cALD to facilitate study of disease mechanisms and therapy development. METHODS We used immunoassays and immunohistochemistry to assess novel (interleukin 18 [IL-18]) and established molecular markers in cerebrospinal fluid (CSF) and postmortem brain tissue from cALD patients. We generated a cALD phenotype in Abcd1-knockout mice using a 2-hit method that combines cuprizone and experimental autoimmune encephalomyelitis models. We then used magnetic resonance imaging (MRI) and immunohistochemistry to assess the fidelity of cALD molecular markers in the mice. RESULTS Human and mouse cALD lesions shared histologic features of myelin phagocytosis, myelin loss, abundant microglial activation, T and B-cell infiltration, and astrogliosis. Compared to wild-type controls, Abcd1-knockout mice displayed more cerebral demyelination, blood-brain barrier disruption, and perivascular immune cell infiltration. This enhanced inflammatory response was associated with higher levels of fibrin deposition, oxidative stress, demyelination, and axonal injury. IL-18 immunoreactivity co-localized with perivascular monocytes/macrophages in both human and mouse brain tissue. In cALD patients, CSF IL-18 levels correlated with MRI lesion severity. INTERPRETATION Our results suggest loss of Abcd1 function in mice predisposes to more severe blood-brain barrier disruption, cerebral inflammation driven by the infiltration of peripheral immune cells, demyelination, and axonal damage, replicating human cALD features. This novel mouse model could shed light on cALD mechanisms and accelerate cALD therapy development. ANN NEUROL 2025;97:296-312.
Collapse
Affiliation(s)
- Ezzat Hashemi
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Isha N. Srivastava
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Alejandro Aguirre
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Ezra T. Yoseph
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Esha Kaushal
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Avni Awani
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Jae Kyu Ryu
- Gladstone Institute for Neurological Disease; San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF; San Francisco, CA USA
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco; San Francisco, CA, USA
| | - Katerina Akassoglou
- Gladstone Institute for Neurological Disease; San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF; San Francisco, CA USA
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco; San Francisco, CA, USA
| | - Shahrzad Talebian
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Pauline Chu
- Stanford Human Research Histology Core, Stanford University School of Medicine, Stanford, CA, USA
| | - Laura Pisani
- Department of Radiology, Stanford University School of Medicine Stanford, CA, USA
| | - Patricia Musolino
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Lawrence Steinman
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Kristian Doyle
- Department of Immunobiology, University of Arizona, Tucson, AZ, USA
| | - William H Robinson
- Department of Immunology & Rheumatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Orr Sharpe
- Department of Immunology & Rheumatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Romain Cayrol
- Department of Pathology, Clinical Department of Laboratory Medicine, University of Montreal, Quebec, Canada
| | - Paul J. Orchard
- Division of Pediatric Blood & Marrow Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Troy Lund
- Division of Pediatric Blood & Marrow Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Hannes Vogel
- Departments of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Max Lenail
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - May H. Han
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Joshua L. Bonkowsky
- Division of Pediatric Neurology, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah
- Brain and Spine Center, Primary Children’s Hospital, Salt Lake City, Utah
- Primary Children’s Center for Personalized Medicine, Salt Lake City, Utah
| | - Keith P. Van Haren
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
8
|
Frank C, Salapa HE, Allen KJH, Levin MC, Dawicki W, Dadachova E. Antibody-Mediated Depletion of Autoreactive T Lymphocytes through PD-1 Improves Disease Outcomes and Visualizes T Cell Activation in Experimental Autoimmune Encephalomyelitis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1647-1657. [PMID: 38578274 DOI: 10.4049/jimmunol.2300751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 03/16/2024] [Indexed: 04/06/2024]
Abstract
Long-term therapeutic outcomes of multiple sclerosis (MS) remain hindered by the chronic nature of immune cell stimulation toward self-antigens. Development of novel methods to target and deplete autoreactive T lymphocytes remains an attractive target for therapeutics for MS. We developed a programmed cell death 1 (PD-1)-targeted radiolabeled mAb and assessed its ability to deplete activated PD-1+ T lymphocytes in vitro and its ability to reduce disease burden of the myelin oligodendrocyte glycoprotein 35-55 experimental autoimmune encephalomyelitis (EAE) model in C57BL/6 mice. We also investigated the upregulation of PD-1 on infiltrating lymphocytes in an animal model of MS. Finally, we demonstrate the (to our knowledge) first reported positron-emission tomography/computed tomography imaging of activated PD-1+ cells in the EAE animal model of MS. We found that the 177Lu radioisotope-labeled anti-PD-1 mAb demonstrated significant in vitro cytotoxicity toward activated CD4+PD-1+ T lymphocytes and led to significant reduction in overall disease progression in the EAE animal model. Our results show high expression of PD-1 on infiltrating lymphocytes in the spinal cords of EAE diseased animals. Positron-emission tomography/computed tomography imaging of the anti-PD-1 mAb demonstrated significant uptake in the cervical draining lymph nodes highlighting accumulation of activated lymphocytes. Targeted depletion of T lymphocytes using T cell activation markers such as PD-1 may present a novel method to reduce autoimmune attack and inflammation in autoimmune diseases such as MS. Development of multimodal nuclear theranostic agents may present the opportunity to monitor T cell activation via imaging radioisotopes and simultaneously treat MS using therapeutic radioisotopes.
Collapse
Affiliation(s)
- Connor Frank
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Hannah E Salapa
- Office of Saskatchewan Multiple Sclerosis Clinical Research Chair, Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Neurology Division, Department of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Kevin J H Allen
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Michael C Levin
- Office of Saskatchewan Multiple Sclerosis Clinical Research Chair, Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Neurology Division, Department of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Wojciech Dawicki
- Department of Biochemistry, Microbiology and Immunology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Ekaterina Dadachova
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
9
|
Muñoz-Jurado A, Escribano BM, Túnez I. Animal model of multiple sclerosis: Experimental autoimmune encephalomyelitis. Methods Cell Biol 2024; 188:35-60. [PMID: 38880527 DOI: 10.1016/bs.mcb.2024.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Multiple sclerosis (MS) is a very complex and heterogeneous disease, with an unknown etiology and which, currently, remains incurable. For this reason, animal models are crucial to investigate this disease, which has increased in prevalence in recent years, affecting 2.8 million people worldwide, and is the leading cause of non-traumatic disability in young adults between the ages of 20-30years. Of all the models developed to replicate MS, experimental autoimmune encephalomyelitis (EAE) best reflects the autoimmune pathogenesis of MS. There are different methods to induce it, which will give rise to different types of EAE, which will vary in clinical presentation and severity. Of the EAE models, the most widespread and used is the one induced in rodents due to its advantages over other species. Likewise, EAE has become a widely used model in the development of therapies for the treatment of MS. Likewise, it is very useful to define the cellular and molecular mechanisms involved in the pathogenesis of MS and to establish therapeutic targets for this disease. For all these reasons, the EAE model plays a key role in improving the understanding of MS.
Collapse
Affiliation(s)
- Ana Muñoz-Jurado
- Department of Cell Biology, Physiology and Immunology, Faculty of Veterinary Medicine, University of Cordoba, Cordoba, Spain; Maimonides Institute for Research in Biomedicine of Cordoba (IMIBIC), Cordoba, Spain.
| | - Begoña M Escribano
- Department of Cell Biology, Physiology and Immunology, Faculty of Veterinary Medicine, University of Cordoba, Cordoba, Spain; Maimonides Institute for Research in Biomedicine of Cordoba (IMIBIC), Cordoba, Spain
| | - Isaac Túnez
- Maimonides Institute for Research in Biomedicine of Cordoba (IMIBIC), Cordoba, Spain; Department of Biochemistry and Molecular Biology, Faculty of Medicine and Nursing, University of Cordoba, Cordoba, Spain.
| |
Collapse
|
10
|
Taherian N, Vaezi G, Neamati A, Hojjati V, Ghorbani-Taherdehi F, Sahebkar A, Gorji-Valokola M. The dose-dependent neuroprotective effect of norepinephrine in improving memory retrieval in an experimental model of multiple sclerosis, experimental autoimmune encephalomyelitis. Brain Res Bull 2024; 209:110907. [PMID: 38395110 DOI: 10.1016/j.brainresbull.2024.110907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/26/2024] [Accepted: 02/20/2024] [Indexed: 02/25/2024]
Abstract
Multiple sclerosis (MS) is considered an immune-mediated inflammatory disorder that causes cognitive impairments by damaging the hippocampal tissue. Conversely, norepinephrine (NEP) has anti-inflammatory and re-myelinating properties, which improve cognitive impairments. The aim of this study was to assess the neuroprotective effects of NEP on learning and memory disorders in an experimental animal model of MS. Two guide cannulas were bilaterally implanted in the rat hippocampal CA1 regions. After recovery, the animals received 3 μl of 0.01% ethidium bromide (EtB) in each of both hippocampal regions. After three days, the rats were randomly divided into 6 groups (8 rats/group), including control, sham 1, sham 2, and three groups of NEP 0.25, 0.5, and 1 mg/kg by intrahippocampal injection. Behavioral tests (e.g. shuttle box test and open-field test) were then performed. Finally, ROS, MDA, GSH, TNF-α, IL-6, and IL-1β concentrations in the left CA1 area, as well as using western-blot analysis, p-p38, p-JNK, p-AKT, p-ERK1/2, p-NMDA, p-AMPA, p-CREB, and BDNF proteins in the right CA1 region evaluated. The EtB injection increased ROS, MDA, TNF-α, IL-6, and IL-1β levels, as well as p-JNK and p-P38, except all other proteins, while decreasing GSH content, as well as step-through latency and locomotor activity in sham groups compared to the control group. Conversely, NEP (0.5 and 1 mg/kg, particularly at the dose of 1 mg/kg) counterbalanced all the alterations mentioned above in comparison to the sham groups. The EtB induced learning and memory impairment; however, NEP dose-dependently restored these impairments to normal levels.
Collapse
Affiliation(s)
- Narjes Taherian
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Gholamhassan Vaezi
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Ali Neamati
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Vida Hojjati
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Faezeh Ghorbani-Taherdehi
- Department of Anatomy and Cell Biology, School of Medicine, Esfahan University of Medical Sciences, Esfahan, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Gorji-Valokola
- Department of Food and Drug Administration, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Department of Pharmacology, Brain and Spinal Injury Repair Research Center, Tehran University of Medical Science, Tehran, Iran.
| |
Collapse
|
11
|
Tang S, Zhang J, Lou F, Zhou H, Cai X, Wang Z, Sun L, Sun Y, Li X, Fan L, Li Y, Jin X, Deng S, Yin Q, Bai J, Wang H, Wang H. A lncRNA Dleu2-encoded peptide relieves autoimmunity by facilitating Smad3-mediated Treg induction. EMBO Rep 2024; 25:1208-1232. [PMID: 38291338 PMCID: PMC10933344 DOI: 10.1038/s44319-024-00070-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 01/02/2024] [Accepted: 01/09/2024] [Indexed: 02/01/2024] Open
Abstract
Micropeptides encoded by short open reading frames (sORFs) within long noncoding RNAs (lncRNAs) are beginning to be discovered and characterized as regulators of biological and pathological processes. Here, we find that lncRNA Dleu2 encodes a 17-amino-acid micropeptide, which we name Dleu2-17aa, that is abundantly expressed in T cells. Dleu2-17aa promotes inducible regulatory T (iTreg) cell generation by interacting with SMAD Family Member 3 (Smad3) and enhancing its binding to the Foxp3 conserved non-coding DNA sequence 1 (CNS1) region. Importantly, the genetic deletion of Dleu2-17aa in mice by start codon mutation impairs iTreg generation and worsens experimental autoimmune encephalomyelitis (EAE). Conversely, the exogenous supplementation of Dleu2-17aa relieves EAE. Our findings demonstrate an indispensable role of Dleu2-17aa in maintaining immune homeostasis and suggest therapeutic applications for this peptide in treating autoimmune diseases.
Collapse
Affiliation(s)
- Sibei Tang
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201610, China
| | - Junxun Zhang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Fangzhou Lou
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201610, China
| | - Hong Zhou
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201610, China
| | - Xiaojie Cai
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201610, China
| | - Zhikai Wang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Libo Sun
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201610, China
| | - Yang Sun
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201610, China
| | - Xiangxiao Li
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201610, China
| | - Li Fan
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201610, China
| | - Yan Li
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201610, China
| | - Xinping Jin
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Siyu Deng
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201610, China
| | - Qianqian Yin
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jing Bai
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201610, China
| | - Hong Wang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Honglin Wang
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201610, China.
| |
Collapse
|
12
|
Oertel FC, Hastermann M, Paul F. Delimiting MOGAD as a disease entity using translational imaging. Front Neurol 2023; 14:1216477. [PMID: 38333186 PMCID: PMC10851159 DOI: 10.3389/fneur.2023.1216477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 08/23/2023] [Indexed: 02/10/2024] Open
Abstract
The first formal consensus diagnostic criteria for myelin oligodendrocyte glycoprotein antibody-associated disease (MOGAD) were recently proposed. Yet, the distinction of MOGAD-defining characteristics from characteristics of its important differential diagnoses such as multiple sclerosis (MS) and aquaporin-4 antibody seropositive neuromyelitis optica spectrum disorder (NMOSD) is still obstructed. In preclinical research, MOG antibody-based animal models were used for decades to derive knowledge about MS. In clinical research, people with MOGAD have been combined into cohorts with other diagnoses. Thus, it remains unclear to which extent the generated knowledge is specifically applicable to MOGAD. Translational research can contribute to identifying MOGAD characteristic features by establishing imaging methods and outcome parameters on proven pathophysiological grounds. This article reviews suitable animal models for translational MOGAD research and the current state and prospect of translational imaging in MOGAD.
Collapse
Affiliation(s)
- Frederike Cosima Oertel
- Experimental and Clinical Research Center, Max-Delbrück-Centrum für Molekulare Medizin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Neuroscience Clinical Research Center, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Department of Neurology, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Maria Hastermann
- Experimental and Clinical Research Center, Max-Delbrück-Centrum für Molekulare Medizin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Neuroscience Clinical Research Center, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Friedemann Paul
- Experimental and Clinical Research Center, Max-Delbrück-Centrum für Molekulare Medizin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Neuroscience Clinical Research Center, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Department of Neurology, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
13
|
Guo M, Wang X, Li Y, Luo A, Zhao Y, Luo X, Li S. Intermittent Fasting on Neurologic Diseases: Potential Role of Gut Microbiota. Nutrients 2023; 15:4915. [PMID: 38068773 PMCID: PMC10707790 DOI: 10.3390/nu15234915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 11/13/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
As the global population ages, the prevalence of neurodegenerative diseases is surging. These disorders have a multifaceted pathogenesis, entwined with genetic and environmental factors. Emerging research underscores the profound influence of diet on the development and progression of health conditions. Intermittent fasting (IF), a dietary pattern that is increasingly embraced and recommended, has demonstrated potential in improving neurophysiological functions and mitigating pathological injuries with few adverse effects. Although the precise mechanisms of IF's beneficial impact are not yet completely understood, gut microbiota and their metabolites are believed to be pivotal in mediating these effects. This review endeavors to thoroughly examine current studies on the shifts in gut microbiota and metabolite profiles prompted by IF, and their possible consequences for neural health. It also highlights the significance of dietary strategies as a clinical consideration for those with neurological conditions.
Collapse
Affiliation(s)
- Mingke Guo
- Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Department of Anesthesiology, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (M.G.); (X.W.); (Y.L.); (A.L.); (Y.Z.)
| | - Xuan Wang
- Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Department of Anesthesiology, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (M.G.); (X.W.); (Y.L.); (A.L.); (Y.Z.)
| | - Yujuan Li
- Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Department of Anesthesiology, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (M.G.); (X.W.); (Y.L.); (A.L.); (Y.Z.)
| | - Ailin Luo
- Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Department of Anesthesiology, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (M.G.); (X.W.); (Y.L.); (A.L.); (Y.Z.)
| | - Yilin Zhao
- Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Department of Anesthesiology, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (M.G.); (X.W.); (Y.L.); (A.L.); (Y.Z.)
| | - Xiaoxiao Luo
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shiyong Li
- Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Department of Anesthesiology, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (M.G.); (X.W.); (Y.L.); (A.L.); (Y.Z.)
| |
Collapse
|
14
|
Hashemi E, Narain Srivastava I, Aguirre A, Tilahan Yoseph E, Kaushal E, Awani A, Kyu. Ryu J, Akassoglou K, Talebian S, Chu P, Pisani L, Musolino P, Steinman L, Doyle K, Robinson WH, Sharpe O, Cayrol R, Orchard P, Lund T, Vogel H, Lenail M, Han MH, Bonkowsky JL, Van Haren KP. A novel mouse model of cerebral adrenoleukodystrophy highlights NLRP3 activity in lesion pathogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.07.564025. [PMID: 37986739 PMCID: PMC10659266 DOI: 10.1101/2023.11.07.564025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Objective We sought to create and characterize a mouse model of the inflammatory, cerebral demyelinating phenotype of X-linked adrenoleukodystrophy (ALD) that would facilitate the study of disease pathogenesis and therapy development. We also sought to cross-validate potential therapeutic targets such as fibrin, oxidative stress, and the NLRP3 inflammasome, in post-mortem human and murine brain tissues. Background ALD is caused by mutations in the gene ABCD1 encoding a peroxisomal transporter. More than half of males with an ABCD1 mutation develop the cerebral phenotype (cALD). Incomplete penetrance and absence of a genotype-phenotype correlation imply a role for environmental triggers. Mechanistic studies have been limited by the absence of a cALD phenotype in the Abcd1-null mouse. Methods We generated a cALD phenotype in 8-week-old, male Abcd1-null mice by deploying a two-hit method that combines cuprizone (CPZ) and experimental autoimmune encephalomyelitis (EAE) models. We employed in vivo MRI and post-mortem immunohistochemistry to evaluate myelin loss, astrogliosis, blood-brain barrier (BBB) disruption, immune cell infiltration, fibrin deposition, oxidative stress, and Nlrp3 inflammasome activation in mice. We used bead-based immunoassay and immunohistochemistry to evaluate IL-18 in CSF and post-mortem human cALD brain tissue. Results MRI studies revealed T2 hyperintensities and post-gadolinium enhancement in the medial corpus callosum of cALD mice, similar to human cALD lesions. Both human and mouse cALD lesions shared common histologic features of myelin phagocytosis, myelin loss, abundant microglial activation, T and B-cell infiltration, and astrogliosis. Compared to wild-type controls, Abcd1-null mice had more severe cerebral inflammation, demyelination, fibrin deposition, oxidative stress, and IL-18 activation. IL-18 immunoreactivity co-localized with macrophages/microglia in the perivascular region of both human and mouse brain tissue. Interpretation This novel mouse model of cALD suggests loss of Abcd1 function predisposes to more severe cerebral inflammation, oxidative stress, fibrin deposition, and Nlrp3 pathway activation, which parallels the findings seen in humans with cALD. We expect this model to enable long-sought investigations into cALD mechanisms and accelerate development of candidate therapies for lesion prevention, cessation, and remyelination.
Collapse
Affiliation(s)
- Ezzat Hashemi
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Isha Narain Srivastava
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Alejandro Aguirre
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Ezra Tilahan Yoseph
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Esha Kaushal
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Avni Awani
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Jae Kyu. Ryu
- Gladstone Institute for Neurological Disease; San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF; San Francisco, CA USA
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco; San Francisco, CA, USA
| | - Katerina Akassoglou
- Gladstone Institute for Neurological Disease; San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF; San Francisco, CA USA
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco; San Francisco, CA, USA
| | - Shahrzad Talebian
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Pauline Chu
- Stanford Human Research Histology Core, Stanford University School of Medicine, Stanford, CA, USA
| | - Laura Pisani
- Department of Radiology, Stanford University School of Medicine Stanford, CA, USA
| | - Patricia Musolino
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Lawrence Steinman
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Kristian Doyle
- Department of Immunobiology, University of Arizona, Tucson, AZ, USA
| | - William H Robinson
- Department of Immunology & Rheumatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Orr Sharpe
- Department of Immunology & Rheumatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Romain Cayrol
- Department of Pathology, Clinical Department of Laboratory Medicine, University of Montreal, Quebec, Canada
| | - Paul Orchard
- Division of Pediatric Blood & Marrow Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Troy Lund
- Division of Pediatric Blood & Marrow Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Hannes Vogel
- Departments of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Max Lenail
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - May Htwe Han
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Joshua Leith Bonkowsky
- Division of Pediatric Neurology, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah
- Brain and Spine Center, Primary Children’s Hospital, Salt Lake City, Utah
- Primary Children’s Center for Personalized Medicine, Salt Lake City, Utah
| | - Keith P. Van Haren
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
15
|
Dedoni S, Scherma M, Camoglio C, Siddi C, Dazzi L, Puliga R, Frau J, Cocco E, Fadda P. An overall view of the most common experimental models for multiple sclerosis. Neurobiol Dis 2023:106230. [PMID: 37453561 DOI: 10.1016/j.nbd.2023.106230] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 07/01/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023] Open
Abstract
Multiple sclerosis (MS) is a complex chronic disease with an unknown etiology. It is considered an inflammatory demyelinating and neurodegenerative disorder of the central nervous system (CNS) characterized, in most cases, by an unpredictable onset of relapse and remission phases. The disease generally starts in subjects under 40; it has a higher incidence in women and is described as a multifactorial disorder due to the interaction between genetic and environmental risk factors. Unfortunately, there is currently no definitive cure for MS. Still, therapies can modify the disease's natural history, reducing the relapse rate and slowing the progression of the disease or managing symptoms. The limited access to human CNS tissue slows down. It limits the progression of research on MS. This limit has been partially overcome over the years by developing various experimental models to study this disease. Animal models of autoimmune demyelination, such as experimental autoimmune encephalomyelitis (EAE) and viral and toxin or transgenic MS models, represent the most significant part of MS research approaches. These models have now been complemented by ex vivo studies, using organotypic brain slice cultures and in vitro, through induced Pluripotent Stem cells (iPSCs). We will discuss which clinical features of the disorders might be reproduced and investigated in vivo, ex vivo, and in vitro in models commonly used in MS research to understand the processes behind the neuropathological events occurring in the CNS of MS patients. The primary purpose of this review is to give the reader a global view of the main paradigms used in MS research, spacing from the classical animal models to transgenic mice and 2D and 3D cultures.
Collapse
Affiliation(s)
- S Dedoni
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy.
| | - M Scherma
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy.
| | - C Camoglio
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy.
| | - C Siddi
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy
| | - L Dazzi
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato (Cagliari), Italy.
| | - R Puliga
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato (Cagliari), Italy.
| | - J Frau
- Regional Multiple Sclerosis Center, ASSL Cagliari, ATS Sardegna, Italy
| | - E Cocco
- Regional Multiple Sclerosis Center, ASSL Cagliari, ATS Sardegna, Italy; Department Medical Science and Public Health, University of Cagliari, Italy.
| | - P Fadda
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy; Neuroscience Institute, Section of Cagliari, National Research Council of Italy (CNR), Cagliari, Italy.
| |
Collapse
|
16
|
Shafit-Zagardo B, Sidoli S, Goldman JE, DuBois JC, Corboy JR, Strittmatter SM, Guzik H, Edema U, Arackal AG, Botbol YM, Merheb E, Nagra RM, Graff S. TMEM106B Puncta Is Increased in Multiple Sclerosis Plaques, and Reduced Protein in Mice Results in Delayed Lipid Clearance Following CNS Injury. Cells 2023; 12:1734. [PMID: 37443768 PMCID: PMC10340176 DOI: 10.3390/cells12131734] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 06/12/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
During inflammatory, demyelinating diseases such as multiple sclerosis (MS), inflammation and axonal damage are prevalent early in the course. Axonal damage includes swelling, defects in transport, and failure to clear damaged intracellular proteins, all of which affect recovery and compromise neuronal integrity. The clearance of damaged cell components is important to maintain normal turnover and restore homeostasis. In this study, we used mass spectrometry to identify insoluble proteins within high-speed/mercaptoethanol/sarcosyl-insoluble pellets from purified white matter plaques isolated from the brains of individuals with relapsing-remitting MS (RRMS). We determined that the transmembrane protein 106B (TMEM106B), normally lysosome-associated, is insoluble in RRMS plaques relative to normal-appearing white matter from individuals with Alzheimer's disease and non-neurologic controls. Relative to wild-type mice, hypomorphic mice with a reduction in TMEM106B have increased axonal damage and lipid droplet accumulation in the spinal cord following myelin-oligodendrocyte-glycoprotein-induced experimental autoimmune encephalomyelitis. Additionally, the corpora callosa from cuprizone-challenged hypomorphic mice fail to clear lipid droplets efficiently during remyelination, suggesting that when TMEM106B is compromised, protein and lipid clearance by the lysosome is delayed. As TMEM106B contains putative lipid- and LC3-binding sites, further exploration of these sites is warranted.
Collapse
Affiliation(s)
| | - Simone Sidoli
- Department of Biochemistry, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - James E Goldman
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Juwen C DuBois
- Department of Pathology, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - John R Corboy
- Rocky Mountain MS Brain Bank, Department of Neurology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Stephen M Strittmatter
- Departments of Neurology and Neuroscience, Yale School of Medicine, Boyer Center for Molecular Medicine, New Haven, CT 06510, USA
| | - Hillary Guzik
- Analytic Imaging Facility, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Ukuemi Edema
- Department of Anatomic and Clinical Pathology, Montefiore Medical Center, Bronx, NY 10467, USA
| | - Anita G Arackal
- Department of Anatomic and Clinical Pathology, Montefiore Medical Center, Bronx, NY 10467, USA
| | - Yair M Botbol
- Department of Pathology, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Emilio Merheb
- Department of Biochemistry, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Rashed M Nagra
- UCLA Brain Bank, VA Healthcare System, Los Angeles, CA 90073, USA
| | - Sarah Graff
- Department of Biochemistry, Albert Einstein College of Medicine, New York, NY 10461, USA
| |
Collapse
|
17
|
Ahmad SF, Ansari MA, Nadeem A, Bakheet SA, Al-Mazroua HA, Alomar HA, Al-Hamamah MA, Attia SM. S3I-201, a selective stat3 inhibitor, ameliorates clinical symptoms in a mouse model of experimental autoimmune encephalomyelitis through the regulation of multiple intracellular signalling in Th1, Th17, and treg cells. Mult Scler Relat Disord 2023; 73:104658. [PMID: 36989705 DOI: 10.1016/j.msard.2023.104658] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/13/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023]
Abstract
CD4+ T cells, specifically Th cells (Th1 and Th17) and regulatory T cells (Tregs), play a pivotal role in the pathogenesis of multiple sclerosis (MS), a demyelinating autoimmune disease of the CNS. STAT3 inhibitors are potential therapeutic targets for several immune disorders. In this study, we investigated the role of a well-known STAT3 inhibitor, S3I-201, in experimental autoimmune encephalomyelitis (EAE), a model of MS. Following induction of EAE, mice were intraperitoneally administered S3I-201 (10 mg/kg) each day, beginning on day 14 and continuing till day 35 and were evaluated for clinical signs. Flow cytometry was used to investigate further the effect of S3I-201 on Th1 (IFN-γ, STAT1, pSTAT1, and T-bet), Th17 (IL-17A, STAT3, pSTAT3, and RORγt), and regulatory T cells (Treg, IL-10, TGF-β1, and FoxP3) expressed in splenic CD4+ T cells. Moreover, we analyzed the effects of S3I-201 on mRNA and protein expression of IFN-γ, T-bet, IL-17A, STAT1, STAT3, pSTAT1, pSTAT3, RORγ, IL-10, TGF-β1, and FoxP3 in the brains of EAE mice. The severity of clinical scores decreased in S3I-201-treated EAE mice compared to vehicle-treated EAE mice. S3I-201 treatment significantly decreased CD4+IFN-γ+, CD4+STAT1+, CD4+pSTAT1+, CD4+T-bet+, CD4+IL-17A+, CD4+STAT3+, CD4+pSTAT3+, and CD4+RORγt+ and increased CD4+IL-10+, CD4+TGF-β1+, and CD4+FoxP3+ in the spleens of EAE mice. Additionally, S3I-201 administration in EAE mice significantly decreased the mRNA and protein expression of Th1 and Th17 and increased those of Treg. These results suggest that S3I-201 may have novel therapeutic potential against MS.
Collapse
|
18
|
Liu W, Yu Z, Wang Z, Waubant EL, Zhai S, Benet LZ. Using an animal model to predict the effective human dose for oral multiple sclerosis drugs. Clin Transl Sci 2023; 16:467-477. [PMID: 36419359 PMCID: PMC10014696 DOI: 10.1111/cts.13458] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 11/02/2022] [Accepted: 11/04/2022] [Indexed: 11/25/2022] Open
Abstract
The objective of this study was to determine the potential usefulness of an animal model to predict the appropriate dose of newly developed drugs for treating relapsing remitting multiple sclerosis (RRMS). Conversion of the lowest effective dose (LEffD) for mice and rats in the experimental autoimmune encephalomyelitis (EAE) model was used to predict the human effective dose utilizing the body surface area correction factor found in the 2005 US Food and Drug Administration (FDA) Guidance for Industry in selecting safe starting doses for clinical trials. Predictions were also tested by comparison with doses estimated by scaling up the LEffD in the model by the human to animal clearance ratio. Although initial proof-of-concept studies of oral fingolimod tested the efficacy and safety of 1.25 and 5 mg in treating RRMS, the EAE animal model predicted the approved dose of this drug, 0.5 mg daily. This approach would have also provided useful predictions of the approved human oral doses for cladribine, dimethyl fumarate, ozanimod, ponesimod, siponimod, and teriflunomide, drugs developed with more than one supposed mechanism of action. The procedure was not useful for i.v. dosed drugs, including monoclonal antibodies. We maintain that drug development scientists should always examine a simple allometric method to predict the therapeutic effective dose in humans. Then, following clinical studies, we believe that the animal model might be expected to yield useful predictions of other drugs developed to treat the same condition. The methodology may not always be predictive, but the approach is so simple it should be investigated.
Collapse
Affiliation(s)
- Wei Liu
- Department of PharmacyPeking University Third HospitalBeijingChina
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and MedicineUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Zhiheng Yu
- Department of PharmacyPeking University Third HospitalBeijingChina
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and MedicineUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Ziyu Wang
- Department of PharmacyPeking University Third HospitalBeijingChina
- School of Basic Medical Sciences and Clinical PharmacyChina Pharmaceutical UniversityNanjingChina
| | - Emmanuelle L. Waubant
- Weill Institute for Neurosciences and San Francisco Multiple Sclerosis CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Suodi Zhai
- Department of PharmacyPeking University Third HospitalBeijingChina
| | - Leslie Z. Benet
- Department of PharmacyPeking University Third HospitalBeijingChina
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and MedicineUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| |
Collapse
|
19
|
Duesman SJ, Ortega-Francisco S, Olguin-Alor R, Acevedo-Dominguez NA, Sestero CM, Chellappan R, De Sarno P, Yusuf N, Salgado-Lopez A, Segundo-Liberato M, de Oca-Lagunas SM, Raman C, Soldevila G. Transforming growth factor receptor III (Betaglycan) regulates the generation of pathogenic Th17 cells in EAE. Front Immunol 2023; 14:1088039. [PMID: 36855628 PMCID: PMC9968395 DOI: 10.3389/fimmu.2023.1088039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 01/23/2023] [Indexed: 02/10/2023] Open
Abstract
The transforming growth factor receptor III (TβRIII) is commonly recognized as a co-receptor that promotes the binding of TGFβ family ligands to type I and type II receptors. Within the immune system, TβRIII regulates T cell development in the thymus and is differentially expressed through activation; however, its function in mature T cells is unclear. To begin addressing this question, we developed a conditional knock-out mouse with restricted TβRIII deletion in mature T cells, necessary because genomic deletion of TβRIII results in perinatal mortality. We determined that TβRIII null mice developed more severe autoimmune central nervous neuroinflammatory disease after immunization with myelin oligodendrocyte peptide (MOG35-55) than wild-type littermates. The increase in disease severity in TβRIII null mice was associated with expanded numbers of CNS infiltrating IFNγ+ CD4+ T cells and cells that co-express both IFNγ and IL-17 (IFNγ+/IL-17+), but not IL-17 alone expressing CD4 T cells compared to Tgfbr3fl/fl wild-type controls. This led us to speculate that TβRIII may be involved in regulating conversion of encephalitogenic Th17 to Th1. To directly address this, we generated encephalitogenic Th17 and Th1 cells from wild type and TβRIII null mice for passive transfer of EAE into naïve mice. Remarkably, Th17 encephalitogenic T cells from TβRIII null induced EAE of much greater severity and earlier in onset than those from wild-type mice. The severity of EAE induced by encephalitogenic wild-type and Tgfbr3fl/fl.dLcKCre Th1 cells were similar. Moreover, in vitro restimulation of in vivo primed Tgfbr3fl/fl.dLcKCre T cells, under Th17 but not Th1 polarizing conditions, resulted in a significant increase of IFNγ+ T cells. Altogether, our data indicate that TβRIII is a coreceptor that functions as a key checkpoint in controlling the pathogenicity of autoreactive T cells in neuroinflammation probably through regulating plasticity of Th17 T cells into pathogenic Th1 cells. Importantly, this is the first demonstration that TβRIII has an intrinsic role in T cells.
Collapse
Affiliation(s)
- Samuel J. Duesman
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Sandra Ortega-Francisco
- Department of Immunology, Biomedical Research Institute, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
- National Laboratory of Flow Cytometry, Biomedical Research Institute, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| | - Roxana Olguin-Alor
- National Laboratory of Flow Cytometry, Biomedical Research Institute, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| | - Naray A. Acevedo-Dominguez
- Department of Immunology, Biomedical Research Institute, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| | - Christine M. Sestero
- Department of Biology, Chemistry, Mathematics and Computer Science, University of Montevallo, Montevello, AL, United States
| | - Rajeshwari Chellappan
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Patrizia De Sarno
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Nabiha Yusuf
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Adrian Salgado-Lopez
- Department of Immunology, Biomedical Research Institute, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| | - Marisol Segundo-Liberato
- Department of Immunology, Biomedical Research Institute, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
- National Laboratory of Flow Cytometry, Biomedical Research Institute, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| | - Selina Montes de Oca-Lagunas
- Department of Immunology, Biomedical Research Institute, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| | - Chander Raman
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Gloria Soldevila
- Department of Immunology, Biomedical Research Institute, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
- National Laboratory of Flow Cytometry, Biomedical Research Institute, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| |
Collapse
|
20
|
Toward a Combination of Biomarkers for Molecular Characterization of Multiple Sclerosis. Int J Mol Sci 2022; 23:ijms232214000. [PMID: 36430480 PMCID: PMC9695566 DOI: 10.3390/ijms232214000] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 11/16/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease affecting the central nervous system associated with chronic inflammation, demyelination, and axonal damage. MS is a highly heterogeneous disease that leads to discrepancies regarding the clinical appearance, progression, and therapy response of patients. Therefore, there is a strong unmet need for clinically relevant biomarkers capable of recapitulating the features of the disease. Experimental autoimmune encephalomyelitis (EAE) is a valuable model for studying the pathophysiology of MS as it recapitulates the main hallmarks of the disease: inflammation, blood-brain barrier (BBB) disruption, gliosis, myelin damage, and repair mechanisms. In this study, we used the EAE-PLP animal model and established a molecular RNA signature for each phase of the disease (onset, peak, remission). We compared variances of expression of known biomarkers by RT-qPCR in the brain and spinal cord of sham and EAE animals monitoring each of the five hallmarks of the disease. Using magnetic cell isolation technology, we isolated microglia and oligodendrocytes of mice of each category, and we compared the RNA expression variations. We identify genes deregulated during a restricted time frame, and we provide insight into the timing and interrelationships of pathological disease processes at the organ and cell levels.
Collapse
|
21
|
Voskuhl RR, MacKenzie-Graham A. Chronic experimental autoimmune encephalomyelitis is an excellent model to study neuroaxonal degeneration in multiple sclerosis. Front Mol Neurosci 2022; 15:1024058. [PMID: 36340686 PMCID: PMC9629273 DOI: 10.3389/fnmol.2022.1024058] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 09/30/2022] [Indexed: 08/19/2023] Open
Abstract
Animal models of multiple sclerosis (MS), specifically experimental autoimmune encephalomyelitis (EAE), have been used extensively to develop anti-inflammatory treatments. However, the similarity between MS and one particular EAE model does not end at inflammation. MS and chronic EAE induced in C57BL/6 mice using myelin oligodendrocyte glycoprotein (MOG) peptide 35-55 share many neuropathologies. Beyond both having white matter lesions in spinal cord, both also have widespread neuropathology in the cerebral cortex, hippocampus, thalamus, striatum, cerebellum, and retina/optic nerve. In this review, we compare neuropathologies in each of these structures in MS with chronic EAE in C57BL/6 mice, and find evidence that this EAE model is well suited to study neuroaxonal degeneration in MS.
Collapse
Affiliation(s)
- Rhonda R. Voskuhl
- UCLA MS Program, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | | |
Collapse
|
22
|
ELBini-Dhouib I, Manai M, Neili NE, Marzouki S, Sahraoui G, Ben Achour W, Zouaghi S, BenAhmed M, Doghri R, Srairi-Abid N. Dual Mechanism of Action of Curcumin in Experimental Models of Multiple Sclerosis. Int J Mol Sci 2022; 23:ijms23158658. [PMID: 35955792 PMCID: PMC9369178 DOI: 10.3390/ijms23158658] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 12/16/2022] Open
Abstract
Background: Multiple sclerosis (MS) is characterized by a combination of inflammatory and demyelination processes in the spinal cord and brain. Conventional drugs generally target the autoimmune response, without any curative effect. For that reason, there is a great interest in identifying novel agents with anti-inflammatory and myelinating effects, to counter the inflammation and cell death distinctive of the disease. Methods and results: An in vitro assay showed that curcumin (Cur) at 10 µM enhanced the proliferation of C8-D1A cells and modulated the production of Th1/Th2/Th17 cytokines in the cells stimulated by LPS. Furthermore, two in vivo pathophysiological experimental models were used to assess the effect of curcumin (100 mg/kg). The cuprizone model mimics the de/re-myelination aspect in MS, and the experimental autoimmune encephalomyelitis model (EAE) reflects immune-mediated events. We found that Cur alleviated the neurological symptomatology in EAE and modulated the expression of lymphocytes CD3 and CD4 in the spinal cord. Interestingly, Cur restored motor and behavioral deficiencies, as well as myelination, in demyelinated mice, as indicated by the higher index of luxol fast blue (LFB) and the myelin basic protein (MBP) intensity in the corpus callosum. Conclusions: Curcumin is a potential therapeutic agent that can diminish the MS neuroimmune imbalance and demyelination through its anti-inflammatory and antioxidant effects.
Collapse
Affiliation(s)
- Ines ELBini-Dhouib
- Laboratoire des Biomolécules, Venins et Applications Théranostiques (LR20IPT01), Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis 1002, Tunisia
- Correspondence: or
| | - Maroua Manai
- Laboratoire de Génétique Humaine (LR99ES10), Faculté de Médecine de Tunis, Université de Tunis El Manar, Tunis 2092, Tunisia or
| | - Nour-elhouda Neili
- Laboratoire des Biomolécules, Venins et Applications Théranostiques (LR20IPT01), Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis 1002, Tunisia
| | - Soumaya Marzouki
- Laboratoire de Transmission, Contrôle et Immunobiologie des Infections (LR11IPT02), Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis 1002, Tunisia
| | - Ghada Sahraoui
- Laboratoire de Médecine de Précision, Médecine Personnalisée et Investigation en Oncologie (LR21SP01), Service d’Anatomie Pathologique, Institut Salah Azaiez, Bab Saadoun, Tunis 1006, Tunisia
- Faculté de Médecine de Tunis, Université de Tunis El Manar, Tunis 1068, Tunisia
| | - Warda Ben Achour
- Laboratoire des Biomolécules, Venins et Applications Théranostiques (LR20IPT01), Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis 1002, Tunisia
| | - Sondes Zouaghi
- Laboratoire des Biomolécules, Venins et Applications Théranostiques (LR20IPT01), Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis 1002, Tunisia
| | - Melika BenAhmed
- Laboratoire de Transmission, Contrôle et Immunobiologie des Infections (LR11IPT02), Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis 1002, Tunisia
- Faculté de Médecine de Tunis, Université de Tunis El Manar, Tunis 1068, Tunisia
| | - Raoudha Doghri
- Laboratoire de Médecine de Précision, Médecine Personnalisée et Investigation en Oncologie (LR21SP01), Service d’Anatomie Pathologique, Institut Salah Azaiez, Bab Saadoun, Tunis 1006, Tunisia
- Faculté de Médecine de Tunis, Université de Tunis El Manar, Tunis 1068, Tunisia
| | - Najet Srairi-Abid
- Laboratoire des Biomolécules, Venins et Applications Théranostiques (LR20IPT01), Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis 1002, Tunisia
| |
Collapse
|
23
|
Osorio-Querejeta I, Alberro A, Suárez J, Sáenz-Cuesta M, Oregi A, Moles L, Muñoz-Culla M, Otaegui D. The innovative animal monitoring device for experimental autoimmune encephalomyelitis (“I AM D EAE”): A more detailed evaluation for improved results. Mult Scler Relat Disord 2022; 63:103836. [DOI: 10.1016/j.msard.2022.103836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/22/2022] [Accepted: 04/28/2022] [Indexed: 10/18/2022]
|
24
|
Sadeghi Hassanabadi N, Broux B, Marinović S, Gotthardt D. Innate Lymphoid Cells - Neglected Players in Multiple Sclerosis. Front Immunol 2022; 13:909275. [PMID: 35784374 PMCID: PMC9247827 DOI: 10.3389/fimmu.2022.909275] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/20/2022] [Indexed: 12/29/2022] Open
Abstract
Multiple sclerosis (MS) is a highly debilitating autoimmune disease affecting millions of individuals worldwide. Although classically viewed as T-cell mediated disease, the role of innate lymphoid cells (ILC) such as natural killer (NK) cells and ILC 1-3s has become a focal point as several findings implicate them in the disease pathology. The role of ILCs in MS is still not completely understood as controversial findings have been reported assigning them either a protective or disease-accelerating role. Recent findings in experimental autoimmune encephalomyelitis (EAE) suggest that ILCs infiltrate the central nervous system (CNS), mediate inflammation, and have a disease exacerbating role by influencing the recruitment of autoreactive T-cells. Elucidating the detailed role of ILCs and altered signaling pathways in MS is essential for a more complete picture of the disease pathology and novel therapeutic targets. We here review the current knowledge about ILCs in the development and progression of MS and preclinical models of MS and discuss their potential for therapeutic applications.
Collapse
Affiliation(s)
| | - Bieke Broux
- University MSCenter; Campus Diepenbeek, Diepenbeek, Belgium
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, UHasselt, Diepenbeek, Belgium
| | - Sonja Marinović
- Division of Molecular Medicine, Laboratory of Personalized Medicine, Ruder Boskovic Institute, Zagreb, Croatia
| | - Dagmar Gotthardt
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
- *Correspondence: Dagmar Gotthardt,
| |
Collapse
|
25
|
Scalabrino G. Newly Identified Deficiencies in the Multiple Sclerosis Central Nervous System and Their Impact on the Remyelination Failure. Biomedicines 2022; 10:biomedicines10040815. [PMID: 35453565 PMCID: PMC9026986 DOI: 10.3390/biomedicines10040815] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/18/2022] [Accepted: 03/21/2022] [Indexed: 12/14/2022] Open
Abstract
The pathogenesis of multiple sclerosis (MS) remains enigmatic and controversial. Myelin sheaths in the central nervous system (CNS) insulate axons and allow saltatory nerve conduction. MS brings about the destruction of myelin sheaths and the myelin-producing oligodendrocytes (ODCs). The conundrum of remyelination failure is, therefore, crucial in MS. In this review, the roles of epidermal growth factor (EGF), normal prions, and cobalamin in CNS myelinogenesis are briefly summarized. Thereafter, some findings of other authors and ourselves on MS and MS-like models are recapitulated, because they have shown that: (a) EGF is significantly decreased in the CNS of living or deceased MS patients; (b) its repeated administration to mice in various MS-models prevents demyelination and inflammatory reaction; (c) as was the case for EGF, normal prion levels are decreased in the MS CNS, with a strong correspondence between liquid and tissue levels; and (d) MS cobalamin levels are increased in the cerebrospinal fluid, but decreased in the spinal cord. In fact, no remyelination can occur in MS if these molecules (essential for any form of CNS myelination) are lacking. Lastly, other non-immunological MS abnormalities are reviewed. Together, these results have led to a critical reassessment of MS pathogenesis, partly because EGF has little or no role in immunology.
Collapse
Affiliation(s)
- Giuseppe Scalabrino
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| |
Collapse
|
26
|
Shi C, Cha J, Gong J, Wang S, Zeng P, Lian J, Zhang B, Hua Q, Lv J, Du C, Xie X, Zhang R. Amelioration of Experimental Autoimmune Encephalomyelitis in Alzheimer’s Disease Mouse Models: A Potential Role for Aβ. Cells 2022; 11:cells11061004. [PMID: 35326455 PMCID: PMC8946952 DOI: 10.3390/cells11061004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/05/2022] [Accepted: 03/14/2022] [Indexed: 11/16/2022] Open
Abstract
Emerging data have highlighted the coexistence of multiple sclerosis (MS) and Alzheimer’s disease (AD), both of which are common central nervous system degenerative diseases with a heavy burden on patients, their families, and society. However, it is unclear how MS progresses under an AD pathological background. We aimed to address the question of how MS progresses under an AD pathological background. We induced the experimental autoimmune encephalomyelitis (EAE) model of MS in two types of AD mouse models, Tg6799 and APP/PS1 mice. We found that, compared with wild-type mice, the clinical symptoms of EAE were significantly ameliorated in APP/PS1 mice but not in Tg6799 mice. Moreover, a much lower level of serum Aβ was observed in Tg6799 mice. EAE clinical symptoms in Tg6799 and C57BL/6J mice were ameliorated by intraperitoneal injection of Aβ42. Peripheral administration of Aβ42 peptides was able to inhibit Th17 development in vivo, which is likely to occur through the inhibition of IL-6 production in dendritic cells. Our findings revealed that AD and EAE could coexist in the same mouse, and Aβ residing in peripheral circulation likely plays an anti-inflammatory role in preventing EAE progression. These findings reveal the potential benefit of Aβ, one of the supervillains of AD, at least in certain contexts.
Collapse
Affiliation(s)
- Changjie Shi
- Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-Based Bio-Medicine, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; (C.S.); (J.C.); (J.G.); (S.W.); (P.Z.); (J.L.); (B.Z.); (Q.H.); (J.L.); (C.D.)
| | - Jiaxue Cha
- Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-Based Bio-Medicine, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; (C.S.); (J.C.); (J.G.); (S.W.); (P.Z.); (J.L.); (B.Z.); (Q.H.); (J.L.); (C.D.)
| | - Junyuan Gong
- Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-Based Bio-Medicine, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; (C.S.); (J.C.); (J.G.); (S.W.); (P.Z.); (J.L.); (B.Z.); (Q.H.); (J.L.); (C.D.)
| | - Shaodeng Wang
- Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-Based Bio-Medicine, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; (C.S.); (J.C.); (J.G.); (S.W.); (P.Z.); (J.L.); (B.Z.); (Q.H.); (J.L.); (C.D.)
| | - Peng Zeng
- Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-Based Bio-Medicine, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; (C.S.); (J.C.); (J.G.); (S.W.); (P.Z.); (J.L.); (B.Z.); (Q.H.); (J.L.); (C.D.)
| | - Junjiang Lian
- Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-Based Bio-Medicine, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; (C.S.); (J.C.); (J.G.); (S.W.); (P.Z.); (J.L.); (B.Z.); (Q.H.); (J.L.); (C.D.)
| | - Bowen Zhang
- Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-Based Bio-Medicine, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; (C.S.); (J.C.); (J.G.); (S.W.); (P.Z.); (J.L.); (B.Z.); (Q.H.); (J.L.); (C.D.)
| | - Qiuhong Hua
- Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-Based Bio-Medicine, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; (C.S.); (J.C.); (J.G.); (S.W.); (P.Z.); (J.L.); (B.Z.); (Q.H.); (J.L.); (C.D.)
| | - Jie Lv
- Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-Based Bio-Medicine, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; (C.S.); (J.C.); (J.G.); (S.W.); (P.Z.); (J.L.); (B.Z.); (Q.H.); (J.L.); (C.D.)
| | - Changsheng Du
- Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-Based Bio-Medicine, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; (C.S.); (J.C.); (J.G.); (S.W.); (P.Z.); (J.L.); (B.Z.); (Q.H.); (J.L.); (C.D.)
| | - Xin Xie
- CAS Key Laboratory of Receptor Research, The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China;
| | - Ru Zhang
- Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-Based Bio-Medicine, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; (C.S.); (J.C.); (J.G.); (S.W.); (P.Z.); (J.L.); (B.Z.); (Q.H.); (J.L.); (C.D.)
- Correspondence: ; Tel.: +86-216-598-6852
| |
Collapse
|
27
|
Matsuyama S, Tanaka Y, Hasebe R, Hojyo S, Murakami M. Gateway Reflex and Mechanotransduction. Front Immunol 2022; 12:780451. [PMID: 35003096 PMCID: PMC8728022 DOI: 10.3389/fimmu.2021.780451] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/30/2021] [Indexed: 12/18/2022] Open
Abstract
The gateway reflex explains how autoreactive CD4+ T cells cause inflammation in tissues that have blood-barriers, such as the central nervous system and retina. It depends on neural activations in response to specific external stimuli, such as gravity, pain, stress, and light, which lead to the secretion of noradrenaline at specific vessels in the tissues. Noradrenaline activates NFkB at these vessels, followed by an increase of chemokine expression as well as a reduction of tight junction molecules to accumulate autoreactive CD4+ T cells, which breach blood-barriers. Transient receptor potential vanilloid 1 (TRPV1) molecules on sensory neurons are critical for the gateway reflex, indicating the importance of mechano-sensing. In this review, we overview the gateway reflex with a special interest in mechanosensory transduction (mechanotransduction).
Collapse
Affiliation(s)
- Shiina Matsuyama
- Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yuki Tanaka
- Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan.,Group of Quantum Immunology, Institute for Quantum Life Science, National Institute for Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Rie Hasebe
- Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Shintaro Hojyo
- Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan.,Group of Quantum Immunology, Institute for Quantum Life Science, National Institute for Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Masaaki Murakami
- Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan.,Group of Quantum Immunology, Institute for Quantum Life Science, National Institute for Quantum and Radiological Science and Technology (QST), Chiba, Japan.,Division of Neurommunology, National Institute for Physiological Sciences, Okazaki, Japan
| |
Collapse
|
28
|
Treatment of Experimental Autoimmune Encephalomyelitis with an Inhibitor of Phosphodiesterase-8 (PDE8). Cells 2022; 11:cells11040660. [PMID: 35203312 PMCID: PMC8870644 DOI: 10.3390/cells11040660] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 02/04/2022] [Accepted: 02/09/2022] [Indexed: 11/17/2022] Open
Abstract
After decades of development, inhibitors targeting cyclic nucleotide phosphodiesterases (PDEs) expressed in leukocytes have entered clinical practice for the treatment of inflammatory disorders, with three PDE4 inhibitors being in clinical use as therapeutics for psoriasis, psoriatic arthritis, chronic obstructive pulmonary disease and atopic dermatitis. In contrast, the PDE8 family that is upregulated in pro-inflammatory T cells is a largely unexplored therapeutic target. We have previously demonstrated a role for the PDE8A-Raf-1 kinase complex in the regulation of myelin oligodendrocyte glycoprotein peptide 35–55 (MOG35–55) activated CD4+ effector T cell adhesion and locomotion by a mechanism that differs from PDE4 activity. In this study, we explored the in vivo treatment of experimental autoimmune encephalomyelitis (EAE), a model for multiple sclerosis (MS) induced in mice immunized with MOG using the PDE8-selective inhibitor PF-04957325. For treatment in vivo, mice with EAE were either subcutaneously (s.c.) injected three times daily (10 mg/kg/dose), or were implanted subcutaneously with Alzet mini-osmotic pumps to deliver the PDE8 inhibitor (15.5 mg/kg/day). The mice were scored daily for clinical signs of paresis and paralysis which were characteristic of EAE. We observed the suppression of the clinical signs of EAE and a reduction of inflammatory lesion formation in the CNS by histopathological analysis through the determination of the numbers of mononuclear cells isolated from the spinal cord of mice with EAE. The PDE8 inhibitor treatment reduces the accumulation of both encephalitogenic Th1 and Th17 T cells in the CNS. Our study demonstrates the efficacy of targeting PDE8 as a treatment of autoimmune inflammation in vivo by reducing the inflammatory lesion load.
Collapse
|
29
|
Watanabe H, Maekawa R, Iikuni S, Kakae M, Matsuo N, Shirakawa H, Kaneko S, Ono M. Characterization of Radioiodinated Diaryl Oxadiazole Derivatives as SPECT Probes for Detection of Myelin in Multiple Sclerosis. ACS Chem Neurosci 2022; 13:363-369. [PMID: 35019269 DOI: 10.1021/acschemneuro.1c00753] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Multiple sclerosis (MS) is an intractable disease of the central nervous system that results from destruction of the myelin sheath. Direct measurement of de- and remyelination is required for monitoring the disease stage of MS, but no useful method has been established. In this study, we characterized four diaryl oxadiazole derivatives as novel myelin-imaging probes for single photon emission computed tomography (SPECT). All the diaryl oxadiazole derivatives penetrated the blood-brain barrier in normal mice. Among them, the highest ratio of radioactivity accumulation in the white matter (myelin-rich region) against the gray matter (myelin-deficient region) was observed at 60 min postinjection of [125I]1,3,4-PODP-DM in ex vivo autoradiography using normal mice. In the blocking study with ex vivo autoradiography, the radioactivity accumulation of [125I]1,3,4-PODP-DM in the white matter markedly reduced. [125I]1,3,4-PODP-DM detected demyelination in the ex vivo autoradiographic images of not only the spinal cord of the experimental autoimmune encephalomyelitis mice but also the brain after lysophosphatidylcholine (LPC) injection. In addition, [123I]1,3,4-PODP-DM could image LPC-induced demyelination in the mouse brain with SPECT. These results suggest that [123I]1,3,4-PODP-DM may be a potential SPECT probe for imaging myelin in MS.
Collapse
Affiliation(s)
- Hiroyuki Watanabe
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Rinka Maekawa
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Shimpei Iikuni
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Masashi Kakae
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho,
Sakyo-ku, Kyoto 606-8501, Japan
| | - Nagisa Matsuo
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho,
Sakyo-ku, Kyoto 606-8501, Japan
| | - Hisashi Shirakawa
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho,
Sakyo-ku, Kyoto 606-8501, Japan
| | - Shuji Kaneko
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho,
Sakyo-ku, Kyoto 606-8501, Japan
| | - Masahiro Ono
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
30
|
Lindsay SL, Molęda AM, MacLellan LM, Keh SM, McElroy DE, Linington C, Goodyear CS, Barnett SC. Human olfactory mesenchymal stromal cell transplantation ameliorates experimental autoimmune encephalomyelitis revealing an inhibitory role for IL16 on myelination. Acta Neuropathol Commun 2022; 10:12. [PMID: 35093166 PMCID: PMC8800340 DOI: 10.1186/s40478-022-01316-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 11/26/2022] Open
Abstract
One of the therapeutic approaches for the treatment of the autoimmune demyelinating disease, multiple sclerosis (MS) is bone marrow mesenchymal stromal cell (hBM-MSCs) transplantation. However, given their capacity to enhance myelination in vitro, we hypothesised that human olfactory mucosa-derived MSCs (hOM-MSCs) may possess additional properties suitable for CNS repair. Herein, we have examined the efficacy of hOM-MSCs versus hBM-MSCs using the experimental autoimmune encephalomyelitis (EAE) model. Both MSC types ameliorated disease, if delivered during the initial onset of symptomatic disease. Yet, only hOM-MSCs improved disease outcome if administered during established disease when animals had severe neurological deficits. Histological analysis of spinal cord lesions revealed hOM-MSC transplantation reduced blood–brain barrier disruption and inflammatory cell recruitment and enhanced axonal survival. At early time points post-hOM-MSC treatment, animals had reduced levels of circulating IL-16, which was reflected in both the ability of immune cells to secrete IL-16 and the level of IL-16 in spinal cord inflammatory lesions. Further in vitro investigation revealed an inhibitory role for IL-16 on oligodendrocyte differentiation and myelination. Moreover, the availability of bioactive IL-16 after demyelination was reduced in the presence of hOM-MSCs. Combined, our data suggests that human hOM-MSCs may have therapeutic benefit in the treatment of MS via an IL-16-mediated pathway, especially if administered during active demyelination and inflammation.
Collapse
|
31
|
Mirabelli E, Elkabes S. Neuropathic Pain in Multiple Sclerosis and Its Animal Models: Focus on Mechanisms, Knowledge Gaps and Future Directions. Front Neurol 2022; 12:793745. [PMID: 34975739 PMCID: PMC8716468 DOI: 10.3389/fneur.2021.793745] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 11/17/2021] [Indexed: 12/22/2022] Open
Abstract
Multiple sclerosis (MS) is a multifaceted, complex and chronic neurological disease that leads to motor, sensory and cognitive deficits. MS symptoms are unpredictable and exceedingly variable. Pain is a frequent symptom of MS and manifests as nociceptive or neuropathic pain, even at early disease stages. Neuropathic pain is one of the most debilitating symptoms that reduces quality of life and interferes with daily activities, particularly because conventional pharmacotherapies do not adequately alleviate neuropathic pain. Despite advances, the mechanisms underlying neuropathic pain in MS remain elusive. The majority of the studies investigating the pathophysiology of MS-associated neuropathic pain have been performed in animal models that replicate some of the clinical and neuropathological features of MS. Experimental autoimmune encephalomyelitis (EAE) is one of the best-characterized and most commonly used animal models of MS. As in the case of individuals with MS, rodents affected by EAE manifest increased sensitivity to pain which can be assessed by well-established assays. Investigations on EAE provided valuable insights into the pathophysiology of neuropathic pain. Nevertheless, additional investigations are warranted to better understand the events that lead to the onset and maintenance of neuropathic pain in order to identify targets that can facilitate the development of more effective therapeutic interventions. The goal of the present review is to provide an overview of several mechanisms implicated in neuropathic pain in EAE by summarizing published reports. We discuss current knowledge gaps and future research directions, especially based on information obtained by use of other animal models of neuropathic pain such as nerve injury.
Collapse
Affiliation(s)
- Ersilia Mirabelli
- Reynolds Family Spine Laboratory, Department of Neurosurgery, New Jersey Medical School, Rutgers the State University of New Jersey, Newark, NJ, United States.,Department of Biology and Chemistry, School of Health Sciences, Liberty University, Lynchburg, VA, United States
| | - Stella Elkabes
- Reynolds Family Spine Laboratory, Department of Neurosurgery, New Jersey Medical School, Rutgers the State University of New Jersey, Newark, NJ, United States
| |
Collapse
|
32
|
Scalabrino G. New Epidermal-Growth-Factor-Related Insights Into the Pathogenesis of Multiple Sclerosis: Is It Also Epistemology? Front Neurol 2021; 12:754270. [PMID: 34899572 PMCID: PMC8664554 DOI: 10.3389/fneur.2021.754270] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/05/2021] [Indexed: 11/23/2022] Open
Abstract
Recent findings showing that epidermal growth factor (EGF) is significantly decreased in the cerebrospinal fluid (CSF) and spinal cord (SC) of living or deceased multiple sclerosis (MS) patients, and that its repeated administration to rodents with chemically- or virally-induced demyelination of the central nervous system (CNS) or experimental allergic encephalomyelitis (EAE) prevents demyelination and inflammatory reactions in the CNS, have led to a critical reassessment of the MS pathogenesis, partly because EGF is considered to have little or no role in immunology. EGF is the only myelinotrophic factor that has been tested in the CSF and spinal cord of MS patients, and it has been shown there is a good correspondence between liquid and tissue levels. This review: (a) briefly summarises the positive EGF effects on neural stem cells, oligodendrocyte cell lineage, and astrocytes in order to explain, at least in part, the biological basis of the myelin loss and remyelination failure in MS; and (b) after a short analysis of the evolution of the principle of cause-effect in the history of Western philosophy, highlights the lack of any experimental immune-, toxin-, or virus-mediated model that precisely reproduces the histopathological features and “clinical” symptoms of MS, thus underlining the inapplicability of Claude Bernard's crucial sequence of “observation, hypothesis, and hypothesis testing.” This is followed by a discussion of most of the putative non-immunologically-linked points of MS pathogenesis (abnormalities in myelinotrophic factor CSF levels, oligodendrocytes (ODCs), astrocytes, extracellular matrix, and epigenetics) on the basis of Popper's falsification principle, and the suggestion that autoimmunity and phologosis reactions (surely the most devasting consequences of the disease) are probably the last links in a chain of events that trigger the reactions. As it is likely that there is a lack of other myelinotrophic growth factors because myelinogenesis is controlled by various CNS and extra-CNS growth factors and other molecules within and outside ODCs, further studies are needed to investigate the role of non-immunological molecules at the time of the onset of the disease. In the words of Galilei, the human mind should be prepared to understand what nature has created.
Collapse
Affiliation(s)
- Giuseppe Scalabrino
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| |
Collapse
|
33
|
Hassan A, Merghany N, Ouchkat F, Regragui W, Kedah H, Hamdy SM, Abdel-Naseer M, Shehata HS, Shalaby NM, Kishk NA, Nada MAF, Hegazy MI, Farghaly M, Ahmed SM, Hussein M. Impact of Ramadan fasting on disease activity in patients with multiple sclerosis: a multicenter study. Nutr Neurosci 2021:1-10. [PMID: 34842062 DOI: 10.1080/1028415x.2021.2006955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND The safety of Ramadan fasting for Muslim patients suffering from multiple sclerosis (MS) is still a matter of debate. This work aimed to study the clinical course of MS during Ramadan fasting and to clarify the predictors of relapses and symptoms exacerbation. METHODS This retrospective study included 153 Muslim patients with MS. Data related to the disease course before Ramadan were obtained from patients' files, whereas data related to the disease activity during Ramadan, were collected from patients over the two months following Ramadan. RESULTS Patients with MS who experienced relapses, exacerbation of symptoms and development of new symptoms during Ramadan had a statistically significant longer disease duration compared to those who did not experience (P < 0.001, <0.001, 0.01 respectively). Also, patients who experienced relapses, exacerbation of symptoms and development of new symptoms during Ramadan had a statistically significant higher expanded disability status scale (EDSS) compared to those who did not experience (P <0.001, <0.001,0.01, respectively). The occurrence of relapses, exacerbation of symptoms and development of new symptoms during Ramadan, were significantly higher in patients who experienced relapses in the preceding year compared to those who did not (P= 0.002, 0.002, 0.01, respectively). Binary logistic regression revealed that each score elevation of EDSS increased the odds of relapse during Ramadan by 1.02 (P-value = 0.04). Also, each month's increase in disease duration increased the odds of relapse during Ramadan by 1.87 (P-value = 0.046). CONCLUSION High EDSS and long disease duration are independent predictors of relapse during Ramadan.
Collapse
Affiliation(s)
- Amr Hassan
- Neurology Department-Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Nahla Merghany
- Neurology Department-Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Fatima Ouchkat
- Department of Neurology and Neurogenetics, Hospital of Specialties, University Mohammed V - Souissi Rabat, Rabat, Morocco
| | - Wafa Regragui
- Department of Neurology and Neurogenetics, Hospital of Specialties, University Mohammed V - Souissi Rabat, Rabat, Morocco
| | - Hanaa Kedah
- Neurology Department, Hera Hospital, Makkah, KSA
| | - Sherif M Hamdy
- Neurology Department-Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Maged Abdel-Naseer
- Neurology Department-Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Hatem S Shehata
- Neurology Department-Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Nevin M Shalaby
- Neurology Department-Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Nirmeen A Kishk
- Neurology Department-Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mona A F Nada
- Neurology Department-Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mohamed I Hegazy
- Neurology Department-Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Marwa Farghaly
- Neurology Department-Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Sandra M Ahmed
- Neurology Department-Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mona Hussein
- Neurology Department, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| |
Collapse
|
34
|
Dai H, Wang L, Li L, Huang Z, Ye L. Metallothionein 1: A New Spotlight on Inflammatory Diseases. Front Immunol 2021; 12:739918. [PMID: 34804020 PMCID: PMC8602684 DOI: 10.3389/fimmu.2021.739918] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 10/18/2021] [Indexed: 01/15/2023] Open
Abstract
MT1 has been demonstrated to be an essential stress protein in maintaining physiological balance and regulating immune homeostasis. While the immunological involvement of MT1 in central nervous system disorders and cancer has been extensively investigated, mounting evidence suggests that MT1 has a broader role in inflammatory diseases and can shape innate and adaptive immunity. In this review, we will first summarize the biological features of MT1 and the regulators that influence MT1 expression, emphasizing metal, inflammation, and immunosuppressive factors. We will then focus on the immunoregulatory function of MT1 on diverse immune cells and the signaling pathways regulated by MT1. Finally, we will discuss recent advances in our knowledge of the biological role of MT1 in several inflammatory diseases to develop novel therapeutic strategies.
Collapse
Affiliation(s)
- Hanying Dai
- Department of Immunology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Lu Wang
- Respiratory Medicine Department, Shenzhen University General Hospital, Shenzhen, China
| | - Lingyun Li
- Department of Immunology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Zhong Huang
- Department of Immunology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Liang Ye
- Department of Immunology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| |
Collapse
|
35
|
Balasa R, Barcutean L, Mosora O, Manu D. Reviewing the Significance of Blood-Brain Barrier Disruption in Multiple Sclerosis Pathology and Treatment. Int J Mol Sci 2021; 22:ijms22168370. [PMID: 34445097 PMCID: PMC8395058 DOI: 10.3390/ijms22168370] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/19/2021] [Accepted: 07/31/2021] [Indexed: 12/27/2022] Open
Abstract
The disruption of blood–brain barrier (BBB) for multiple sclerosis (MS) pathogenesis has a double effect: early on during the onset of the immune attack and later for the CNS self-sustained ‘inside-out’ demyelination and neurodegeneration processes. This review presents the characteristics of BBB malfunction in MS but mostly highlights current developments regarding the impairment of the neurovascular unit (NVU) and the metabolic and mitochondrial dysfunctions of the BBB’s endothelial cells. The hypoxic hypothesis is largely studied and agreed upon recently in the pathologic processes in MS. Hypoxia in MS might be produced per se by the NVU malfunction or secondary to mitochondria dysfunction. We present three different but related terms that denominate the ongoing neurodegenerative process in progressive forms of MS that are indirectly related to BBB disruption: progression independent of relapses, no evidence of disease activity and smoldering demyelination or silent progression. Dimethyl fumarate (DMF), modulators of S1P receptor, cladribine and laquinimode are DMTs that are able to cross the BBB and exhibit beneficial direct effects in the CNS with very different mechanisms of action, providing hope that a combined therapy might be effective in treating MS. Detailed mechanisms of action of these DMTs are described and also illustrated in dedicated images. With increasing knowledge about the involvement of BBB in MS pathology, BBB might become a therapeutic target in MS not only to make it impenetrable against activated immune cells but also to allow molecules that have a neuroprotective effect in reaching the cell target inside the CNS.
Collapse
Affiliation(s)
- Rodica Balasa
- Department of Neurology, University of Medicine, Pharmacy, Sciences and Technology “George Emil Palade”, 540136 Targu Mures, Romania;
- Neurology 1 Clinic, Emergency Clinical County Hospital Mures, 540136 Targu Mures, Romania;
| | - Laura Barcutean
- Department of Neurology, University of Medicine, Pharmacy, Sciences and Technology “George Emil Palade”, 540136 Targu Mures, Romania;
- Neurology 1 Clinic, Emergency Clinical County Hospital Mures, 540136 Targu Mures, Romania;
- Correspondence: ; Tel.: +40-745-373947
| | - Oana Mosora
- Neurology 1 Clinic, Emergency Clinical County Hospital Mures, 540136 Targu Mures, Romania;
| | - Doina Manu
- Advanced Research Center Medical and Pharmaceutical, University of Medicine, Pharmacy, Sciences and Technology “George Emil Palade”, 540142 Targu Mures, Romania;
| |
Collapse
|
36
|
Lourbopoulos A, Mourouzis I, Xinaris C, Zerva N, Filippakis K, Pavlopoulos A, Pantos C. Translational Block in Stroke: A Constructive and "Out-of-the-Box" Reappraisal. Front Neurosci 2021; 15:652403. [PMID: 34054413 PMCID: PMC8160233 DOI: 10.3389/fnins.2021.652403] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 04/06/2021] [Indexed: 12/13/2022] Open
Abstract
Why can we still not translate preclinical research to clinical treatments for acute strokes? Despite > 1000 successful preclinical studies, drugs, and concepts for acute stroke, only two have reached clinical translation. This is the translational block. Yet, we continue to routinely model strokes using almost the same concepts we have used for over 30 years. Methodological improvements and criteria from the last decade have shed some light but have not solved the problem. In this conceptual analysis, we review the current status and reappraise it by thinking "out-of-the-box" and over the edges. As such, we query why other scientific fields have also faced the same translational failures, to find common denominators. In parallel, we query how migraine, multiple sclerosis, and hypothermia in hypoxic encephalopathy have achieved significant translation successes. Should we view ischemic stroke as a "chronic, relapsing, vascular" disease, then secondary prevention strategies are also a successful translation. Finally, based on the lessons learned, we propose how stroke should be modeled, and how preclinical and clinical scientists, editors, grant reviewers, and industry should reconsider their routine way of conducting research. Translational success for stroke treatments may eventually require a bold change with solutions that are outside of the box.
Collapse
Affiliation(s)
- Athanasios Lourbopoulos
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Department of Neurointensive Care Unit, Schoen Klinik Bad Aibling, Bad Aibling, Germany
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig Maximilian University, Munich, Germany
| | - Iordanis Mourouzis
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christodoulos Xinaris
- IRCCS – Istituto di Ricerche Farmacologiche ‘Mario Negri’, Centro Anna Maria Astori, Bergamo, Italy
- University of Nicosia Medical School, Nicosia, Cyprus
| | - Nefeli Zerva
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Filippakis
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Angelos Pavlopoulos
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Constantinos Pantos
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
37
|
das Neves SP, Delivanoglou N, Da Mesquita S. CNS-Draining Meningeal Lymphatic Vasculature: Roles, Conundrums and Future Challenges. Front Pharmacol 2021; 12:655052. [PMID: 33995074 PMCID: PMC8113819 DOI: 10.3389/fphar.2021.655052] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 04/13/2021] [Indexed: 12/11/2022] Open
Abstract
A genuine and functional lymphatic vascular system is found in the meninges that sheath the central nervous system (CNS). This unexpected (re)discovery led to a reevaluation of CNS fluid and solute drainage mechanisms, neuroimmune interactions and the involvement of meningeal lymphatics in the initiation and progression of neurological disorders. In this manuscript, we provide an overview of the development, morphology and unique functional features of meningeal lymphatics. An outline of the different factors that affect meningeal lymphatic function, such as growth factor signaling and aging, and their impact on the continuous drainage of brain-derived molecules and meningeal immune cells into the cervical lymph nodes is also provided. We also highlight the most recent discoveries about the roles of the CNS-draining lymphatic vasculature in different pathologies that have a strong neuroinflammatory component, including brain trauma, tumors, and aging-associated neurodegenerative diseases like Alzheimer's and Parkinson's. Lastly, we provide a critical appraisal of the conundrums, challenges and exciting questions involving the meningeal lymphatic system that ought to be investigated in years to come.
Collapse
Affiliation(s)
| | | | - Sandro Da Mesquita
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
| |
Collapse
|
38
|
Liang X, Cao Y, Li C, Yu H, Yang C, Liu H. MALT1 as a promising target to treat lymphoma and other diseases related to MALT1 anomalies. Med Res Rev 2021; 41:2388-2422. [PMID: 33763890 DOI: 10.1002/med.21799] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 12/23/2020] [Accepted: 03/03/2021] [Indexed: 12/25/2022]
Abstract
Mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1) is a key adaptor protein that regulates the NF-κB pathway, in which MALT1 functions as a scaffold protein and protease to trigger downstream signals. The abnormal expression of MALT1 is closely associated with lymphomagenesis and other diseases, including solid tumors and autoimmune diseases. MALT1 is the only protease in the underlying pathogenesis of these diseases, and its proteolytic activity can be pharmacologically regulated. Therefore, MALT1 is a potential and promising target for anti-lymphoma and other MALT1-related disease treatments. Currently, the development of MALT1 inhibitors is still in its early stages. This review presents an overview of MALT1, particularly its X-ray structures and biological functions, and elaborates on the pathogenesis of diseases associated with its dysregulation. We then summarize previously reported MALT1 inhibitors, focusing on their molecular structure, biological activity, structure-activity relationship, and limitations. Finally, we propose future research directions to accelerate the discovery of novel MALT1 inhibitors with clinical applications. Overall, this review provides a comprehensive and systematic overview of MALT1-related research advances and serves as a theoretical basis for drug discovery and research.
Collapse
Affiliation(s)
- Xuewu Liang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - YiChun Cao
- School of Pharmacy, Fudan University, Shanghai, China
| | - Chunpu Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Haolan Yu
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Chenghua Yang
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Hong Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
39
|
Wang P, Jiang LL, Wang C, Zhu Z, Lai C. Neurofilament Protein as a Potential Biomarker of Axonal Degeneration in Experimental Autoimmune Encephalomyelitis. Neurol India 2021; 68:364-367. [PMID: 32189712 DOI: 10.4103/0028-3886.280651] [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: 11/04/2022]
Abstract
Background Neurofilament proteins as biomarkers of axonal degeneration have the potential to improve our capacity to predict and monitor neurological outcome in experimental autoimmune encephalitis (EAE), a model of multiple sclerosis (MS). We urgently need more accurate early predictive markers to direct the clinician when to provide neuroprotective therapy. Aims To highlight the possible roles of neurofilament proteins in physiological and pathophysiological processes in the MS. Materials and Methods Fifty female Wistar rats with MOG35-55 peptide induced EAE were randomly divided into two parts: control group and EAE group. All of them were along with expanded disability status scale (EDSS). The mice were sacrificed on day 0, 1, 3, 7, 14, and 28 after the first immunization. Supernatant and pellet were separated at the same time. The degradation rates of NF in the brain nerve and spinal cord of each rat were measured by Western Blotting. Statistical Analysis The data were expressed as mean ± SD. Statistical analysis was performed with one-way analysis of variance (ANOVA), followed by LSD's post-hoc tests, which was provided by SPSS 23.0 statistical software. Results and Conclusions Neurofilament light protein may be more useful as a measure of ongoing neurodegenerative activity in EAE, which would make this protein a potential candidate for use as a surrogate marker for assessment of treatments aimed at reducing axonal injury. Future studies are warranted to support or refute the value neurofilament in clinical practice.
Collapse
Affiliation(s)
- Pin Wang
- Department of Neurology, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Lu L Jiang
- Department of Neurotoxicology, Institute of Toxicology, School of Public Health, Shandong University, Shandong, China
| | - Cunfu Wang
- Department of Neurology, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Zhengyu Zhu
- Department of Neurology, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Chao Lai
- Department of Neurology, The Second Hospital of Shandong University, Jinan, Shandong, China
| |
Collapse
|
40
|
Ramasamy R, Smith PP. Animal modeling of lower urinary tract dysfunction associated with multiple sclerosis: Part I: Justification of the mouse model for MS research. Neurourol Urodyn 2021; 40:950-957. [PMID: 33719097 DOI: 10.1002/nau.24649] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/23/2020] [Accepted: 01/16/2021] [Indexed: 12/20/2022]
Abstract
Lower urinary tract symptoms and dysfunction (LUTS/LUTD) contribute to loss of quality of life, morbidity, and need for medical intervention in most patients with multiple sclerosis (MS). Although MS is an inflammatory neurodegenerative disease, clinical manifestations including continence control disorders have traditionally been attributed to the loss of neural signaling due to neurodegeneration. Clinical approaches to MS-LUTS/LUTD have focused on addressing symptoms in the context of urodynamic dysfunctions as pathophysiologic understandings are incomplete. The mouse model provides a useful research platform for the discovery of more detailed molecular, cellular, and tissue-level knowledge of the disease and its clinical manifestations. The aim of this two-part review is to provide a state-of-the-art update on the use of the mouse model for MS research, with a focus on lower urinary tract symptoms. Part I presents a summary of the current understanding of MS pathophysiology, the impact on lower urinary tract symptoms, and briefly introduces the types of mouse models available to study MS. Part II presents the common animal models that are currently available to study MS, their mechanism, relevance to MS-LUTS/LUTD and their urinary pathophysiology, advantages, and disadvantages.
Collapse
Affiliation(s)
- Ramalakshmi Ramasamy
- UConn Center on Aging, UConn Health, Farmington, Connecticut, USA.,Department of Neuroscience, University of Connecticut Graduate School, Farmington, Connecticut, USA.,CT Institute for the Brain and Cognitive Sciences, University of Connecticut, Storrs, Connecticut, USA
| | - Phillip P Smith
- UConn Center on Aging, UConn Health, Farmington, Connecticut, USA.,Department of Neuroscience, University of Connecticut Graduate School, Farmington, Connecticut, USA.,CT Institute for the Brain and Cognitive Sciences, University of Connecticut, Storrs, Connecticut, USA.,Department of Surgery, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| |
Collapse
|
41
|
Orian JM, D'Souza CS, Kocovski P, Krippner G, Hale MW, Wang X, Peter K. Platelets in Multiple Sclerosis: Early and Central Mediators of Inflammation and Neurodegeneration and Attractive Targets for Molecular Imaging and Site-Directed Therapy. Front Immunol 2021; 12:620963. [PMID: 33679764 PMCID: PMC7933211 DOI: 10.3389/fimmu.2021.620963] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 01/27/2021] [Indexed: 12/20/2022] Open
Abstract
Platelets are clearly central to thrombosis and hemostasis. In addition, more recently, evidence has emerged for non-hemostatic roles of platelets including inflammatory and immune reactions/responses. Platelets express immunologically relevant ligands and receptors, demonstrate adhesive interactions with endothelial cells, monocytes and neutrophils, and toll-like receptor (TLR) mediated responses. These properties make platelets central to innate and adaptive immunity and potential candidate key mediators of autoimmune disorders. Multiple sclerosis (MS) is the most common chronic autoimmune central nervous system (CNS) disease. An association between platelets and MS was first indicated by the increased adhesion of platelets to endothelial cells. This was followed by reports identifying structural and functional changes of platelets, their chronic activation in the peripheral blood of MS patients, platelet presence in MS lesions and the more recent revelation that these structural and functional abnormalities are associated with all MS forms and stages. Investigations based on the murine experimental autoimmune encephalomyelitis (EAE) MS model first revealed a contribution to EAE pathogenesis by exacerbation of CNS inflammation and an early role for platelets in EAE development via platelet-neuron and platelet-astrocyte associations, through sialated gangliosides in lipid rafts. Our own studies refined and extended these findings by identifying the critical timing of platelet accumulation in pre-clinical EAE and establishing an initiating and central rather than merely exacerbating role for platelets in disease development. Furthermore, we demonstrated platelet-neuron associations in EAE, coincident with behavioral changes, but preceding the earliest detectable autoreactive T cell accumulation. In combination, these findings establish a new paradigm by asserting that platelets play a neurodegenerative as well as a neuroinflammatory role in MS and therefore, that these two pathological processes are causally linked. This review will discuss the implications of these findings for our understanding of MS, for future applications for imaging toward early detection of MS, and for novel strategies for platelet-targeted treatment of MS.
Collapse
Affiliation(s)
- Jacqueline M Orian
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Claretta S D'Souza
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Pece Kocovski
- Department of Psychology and Counselling, School of Psychology and Public Health, College of Science, Health and Engineering, La Trobe University, Melbourne, VIC, Australia
| | - Guy Krippner
- Medicinal Chemistry, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Matthew W Hale
- Department of Psychology and Counselling, School of Psychology and Public Health, College of Science, Health and Engineering, La Trobe University, Melbourne, VIC, Australia
| | - Xiaowei Wang
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia.,Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Physiology, Anatomy and Microbiology, School of Life Science, La Trobe University, Melbourne, VIC, Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia.,Department of Physiology, Anatomy and Microbiology, School of Life Science, La Trobe University, Melbourne, VIC, Australia
| |
Collapse
|
42
|
Liu R, Li Y, Zhou H, Wang H, Liu D, Wang H, Wang Z. OIP5-AS1 facilitates Th17 differentiation and EAE severity by targeting miR-140-5p to regulate RhoA/ROCK2 signaling pathway. Life Sci 2021:119108. [PMID: 33515560 DOI: 10.1016/j.lfs.2021.119108] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 01/04/2021] [Accepted: 01/21/2021] [Indexed: 12/18/2022]
Abstract
AIMS Multiple sclerosis (MS) is one of the commonest neurologic disorders globally. LncRNA OIP5-AS1 has been found to be implicated in the etiology of MS. This study was to explore the roles and molecular mechanisms of OIP5-AS1 in the development of MS. MATERIALS AND METHODS RT-qPCR assay was used to measure expressions of OIP5-AS1, miR-140-5p, IL-17A mRNA and RhoA mRNA. CD4+IL-17+ cell proportion was determined by flow cytometry. IL-17A secretion was examined by ELISA assay. Cell inflammatory infiltration and demyelination were assessed by histological analyses. The interaction between miR-140-5p and OIP5-AS1 or RhoA 3'UTR was validated by bioinformatical analysis and luciferase reporter assay. Western blot assay was performed to detect protein expressions of ROCK2 and RhoA. An experimental autoimmune encephalomyelitis (EAE) models was established to explore the role of OIP5-AS1 in MS in vivo. KEY FINDINGS OIP5-AS1 expression was enhanced in MS patients. Also, elevated OIP5-AS1 level was observed during T-helper 17 (Th17) differentiation. Moreover, OIP5-AS1 promoted Th17 differentiation in vitro and contributed to the development of EAE in vivo. Mechanical explorations revealed that OIP5-AS1 targeted miR-140-5p to regulate Th17 differentiation. Moreover, RhoA was a target of miR-140-5p and miR-140-5p inhibited the activation of RhoA/ROCK2 signaling. Also, RhoA upregulation abrogated the inhibitory effects of miR-140-5p on Th17 differentiation. SIGNIFICANCE OIP5-AS1 contributed to EAE development by targeting miR-140-5p/RhoA and activating RhoA/ROCK2 signaling pathway, shedding light on the roles and molecular mechanisms of OIP5-AS1 in the development of MS and providing some candidate targets for the diagnose and treatment of MS.
Collapse
Affiliation(s)
- Ruihua Liu
- Department of Neurology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China.
| | - Yan Li
- Department of Neurology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Haitao Zhou
- Department of Neurology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Hao Wang
- Department of Neurology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Dequan Liu
- Department of Neurology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Huilin Wang
- Department of Neurology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Zhenghua Wang
- Department of Laboratory, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China
| |
Collapse
|
43
|
Ramos-Martinez E, Ramos-Martínez I, Molina-Salinas G, Zepeda-Ruiz WA, Cerbon M. The role of prolactin in central nervous system inflammation. Rev Neurosci 2021; 32:323-340. [PMID: 33661585 DOI: 10.1515/revneuro-2020-0082] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 10/25/2020] [Indexed: 12/14/2022]
Abstract
Prolactin has been shown to favor both the activation and suppression of the microglia and astrocytes, as well as the release of inflammatory and anti-inflammatory cytokines. Prolactin has also been associated with neuronal damage in diseases such as multiple sclerosis, epilepsy, and in experimental models of these diseases. However, studies show that prolactin has neuroprotective effects in conditions of neuronal damage and inflammation and may be used as neuroprotector factor. In this review, we first discuss general information about prolactin, then we summarize recent findings of prolactin function in inflammatory and anti-inflammatory processes and factors involved in the possible dual role of prolactin are described. Finally, we review the function of prolactin specifically in the central nervous system and how it promotes a neuroprotective effect, or that of neuronal damage, particularly in experimental autoimmune encephalomyelitis and during excitotoxicity. The overall studies indicated that prolactin may be a promising molecule for the treatment of some neurological diseases.
Collapse
Affiliation(s)
- Edgar Ramos-Martinez
- Escuela de Ciencias, Universidad Autónoma "Benito Juárez" de Oaxaca, Oaxaca68120, Mexico
- Instituto de Cómputo Aplicado en Ciencias, Oaxaca68000, Mexico
| | - Ivan Ramos-Martínez
- Glycobiology, Cell Growth and Tissue Repair Research Unit (Gly-CRRET), Université Paris Est Créteil (UPEC), 94010Créteil, France
| | - Gladys Molina-Salinas
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, CDMX, 04510Coyoacan, Mexico
| | - Wendy A Zepeda-Ruiz
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, CDMX, 04510Coyoacan, Mexico
| | - Marco Cerbon
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, CDMX, 04510Coyoacan, Mexico
| |
Collapse
|
44
|
Liu B, Ding Y, Li P, Wang T, He S, Jia Z, Yang J. MicroRNA-219c-5p regulates bladder fibrosis by targeting FN1. BMC Urol 2020; 20:193. [PMID: 33287818 PMCID: PMC7720614 DOI: 10.1186/s12894-020-00765-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 11/30/2020] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND We found that the bladders of multiple sclerosis mice were significantly fibrotic. This study aimed to investigate the relationship between fibronectin 1 (FN1) and bladder fibrosis, as well as the microRNAs involved in FN1 regulation. METHODS The degree of bladder smooth muscle fibrosis was observed by immunohistochemistry. In addition, we used quantitative real-time polymerase chain reaction (RT-qPCR) and Western blotting to determine FN1 expression in bladders with different grades of fibrosis. Bioinformatics analysis revealed that miR-199a-3p, miR-219c-5p and miR-3572-3p could inhibit FN1 synthesis. Therefore, miR-199a-3p, miR-219c-5p and miR-3572-3p were overexpressed or knocked down in bladder smooth muscle cells (BSMCs), and the respective transfection and FN1 knockdown efficiencies were detected by RT-qPCR. Only miR-219c-5p overexpression and knockdown produced the expected results. A dual luciferase reporter assay was used to determine the targeting relationship between miR-219c-5p and FN1. Flow cytometry and Cell Counting Kit 8 (CCK8) experiments confirmed that miR-219c-5p reduced FN1 expression and affected the biological activity of smooth muscle cells. Agomir and anagomir of miR-219c-5p were transfected in vivo to observe the change of bladder fibrosis in mice. RESULTS With increasing bladder fibrosis, FN1 expression increased, while miR-199a-3p, miR-219c-5p, and miR-3572-3p expression levels decreased. The RT-qPCR results after transfection showed that only miR-219c-5p could regulate FN1. Indeed, the dual luciferase reporter assay results indicated that miR-219c-5p targeted FN1 directly. CCK8 and cell cycle assays showed that miR-219c-5p overexpression inhibited BSMC proliferation, while miR-219c-5p knockdown promoted BSMC proliferation. An apoptosis assay showed that miR-219c-5p overexpression promoted apoptosis, while miR-219c-5p knockdown inhibited BSMC apoptosis. The agomir and anagomir transfected with miR-219c-5p in vivo found that the bladder fibrosis of the mice in the agomir group was reduced, and the anagomir group was worse. CONCLUSIONS Our findings indicate that FN1 up-regulation and miR-219c-5p down-regulation play an important role in the development of bladder fibrosis, and miR-219c-5p participates in bladder fibrosis by regulating FN1 expression. Thus, a novel antifibrotic function of miR-219c-5p is proposed, which may represent a potential target for the diagnosis and treatment of bladder fibrosis.
Collapse
Affiliation(s)
- Bowen Liu
- Department of Urology, Zhengzhou University First Affiliated Hospital, Zhengzhou, 450052, China.,Zhengzhou Institute of Urology, Zhengzhou University First Affiliated Hospital, Zhengzhou, 450052, China
| | - Yafei Ding
- Department of Urology, Zhengzhou University First Affiliated Hospital, Zhengzhou, 450052, China
| | - Peng Li
- Department of Urology, Zhengzhou University First Affiliated Hospital, Zhengzhou, 450052, China
| | - Tao Wang
- Department of Urology, Zhengzhou University First Affiliated Hospital, Zhengzhou, 450052, China
| | - Siyuan He
- Department of Urology, Zhengzhou University First Affiliated Hospital, Zhengzhou, 450052, China
| | - Zhankui Jia
- Department of Urology, Zhengzhou University First Affiliated Hospital, Zhengzhou, 450052, China.,Zhengzhou Institute of Urology, Zhengzhou University First Affiliated Hospital, Zhengzhou, 450052, China
| | - Jinjian Yang
- Department of Urology, Zhengzhou University First Affiliated Hospital, Zhengzhou, 450052, China. .,Zhengzhou Institute of Urology, Zhengzhou University First Affiliated Hospital, Zhengzhou, 450052, China.
| |
Collapse
|
45
|
Watanabe H, Sakai S, Iikuni S, Shimizu Y, Shirakawa H, Kaneko S, Ono M. Synthesis and biological evaluation of radioiodinated 3-phenylcoumarin derivatives targeting myelin in multiple sclerosis. Bioorg Med Chem Lett 2020; 30:127562. [PMID: 32971260 DOI: 10.1016/j.bmcl.2020.127562] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/11/2020] [Accepted: 09/16/2020] [Indexed: 02/06/2023]
Abstract
Myelin is a lipid multilayer involved in the rate of nerve transmission, and its loss is a pathological feature of multiple sclerosis in brains. Since in vivo imaging of myelin may be useful for drug development, early diagnosis, and monitoring the disease stage, we designed, synthesized, and evaluated eight novel radioiodinated 3-phenylcoumarin derivatives as imaging probes targeting myelin. In the biodistribution study using normal mice, all compounds displayed sufficient brain uptake, ranging from 2.5 to 5.0% ID/g, at 2 min postinjection. On ex vivo autoradiography, [125I]18 and [125I]21, which have a dimethylamino group, showed high binding affinity for myelin in the normal mouse brain. In addition, the radioactivity accumulation of [125I]21 in the white matter of the spinal cord in the experimental autoimmune encephalomyelitis mice was lower than that in naive mice. These results suggest that [123I]21 shows potential as a single photon emission computed tomography probe targeting myelin.
Collapse
Affiliation(s)
- Hiroyuki Watanabe
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan.
| | - Shiori Sakai
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Shimpei Iikuni
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yoichi Shimizu
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan; Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Hisashi Shirakawa
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Shuji Kaneko
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Masahiro Ono
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan.
| |
Collapse
|
46
|
Bai M, Wang Y, Han R, Xu L, Huang M, Zhao J, Lin Y, Song S, Chen Y. Intermittent caloric restriction with a modified fasting-mimicking diet ameliorates autoimmunity and promotes recovery in a mouse model of multiple sclerosis. J Nutr Biochem 2020; 87:108493. [PMID: 32920091 DOI: 10.1016/j.jnutbio.2020.108493] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 08/07/2020] [Accepted: 08/12/2020] [Indexed: 12/13/2022]
Abstract
Dietary interventions such as fasting have been proved to be effective in the prevention of metabolic and autoimmune diseases as well as aging-related conditions. The complicated interaction between nutrition and immunity has drawn wide attention in recent years. In this study, we investigated the therapeutic effect of intermittent caloric restriction on autoimmune encephalomyelitis (EAE), a model of multiple sclerosis, in mice. EAE was induced by immunization of C57BL/6 mice with myelin oligodendrocyte glycoprotein 35-55 peptide. After the EAE symptoms became obvious at the 4th week post-immunization, the mice were administered with a modified fasting-mimicking diet (FMD) at 1/3 cal of control for 3 days, followed by ad libitum with normal chow for 4 days. A total of two cycles of FMD was applied. Compared with the mice without receiving caloric restriction, the mice using FMD had significant decreases in EAE severity, immune cell infiltration in spinal cord and CNS demyelination. FMD administration also reversed EAE-mediated CNS accumulation of total CD4+ T cells and in particular, IFN-γ-producing CD4+ T cells. Moreover, FMD application elevated the cell proliferation rate in CNS and enhanced expression of brain-derived neurotrophic factor (BDNF) and remyelination markers. In conclusion, our results indicate that intermittent caloric restriction using the modified FMD was effective in the treatment of EAE through ameliorating inflammatory response and promoting recovery of the damaged tissue.
Collapse
Affiliation(s)
- Meijuan Bai
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China, 200031
| | - Yan Wang
- CAS Key Laboratory of Tumor and Microenvironment, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China, 200031
| | - Ruomei Han
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China, 200031; School of Life Sciences and Technology, Shanghai Tech University, Shanghai, China 200031
| | - Lijiao Xu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China, 200031; School of Life Sciences and Technology, Shanghai Tech University, Shanghai, China 200031
| | - Meiqin Huang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China, 200031
| | - Jingyu Zhao
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China, 200031
| | - Yijun Lin
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China, 200031
| | - Shuo Song
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China, 200031; School of Life Sciences and Technology, Shanghai Tech University, Shanghai, China 200031
| | - Yan Chen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China, 200031; School of Life Sciences and Technology, Shanghai Tech University, Shanghai, China 200031.
| |
Collapse
|
47
|
Nasrollahzadeh Sabet M, Biglari S, Khorram Khorshid HR, Esmaeilzadeh E. Shikonin ameliorates experimental autoimmune encephalomyelitis (EAE) via immunomodulatory, anti-apoptotic and antioxidative activity. J Pharm Pharmacol 2020; 72:1970-1976. [PMID: 32892382 DOI: 10.1111/jphp.13364] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 07/25/2020] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Multiple sclerosis is a common autoimmune inflammatory disease of the central nervous system. There are several underlying mechanisms for the pathogenesis of the disease, including inflammation, oligodendrocyte apoptosis and oxidative stress. METHODS The mechanism of action of shikonin was investigated in the C57BL/6 experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis. KEY FINDINGS The results revealed that EAE induction significantly increased the extent of demyelination in the corpus callosum tissues of the animals, while treatment of the mice with shikonin significantly decreased the extent of demyelination. Real-time polymerase chain reaction-based analysis of the brain samples from the EAE mice revealed significant enhancement in the expression levels of tumour necrosis factor-α (TNF-α), interferon-γ (IFN-γ) and Bax genes as well as a reduction in the expression levels of transforming growth factor-ß (TGF-β) and Bcl2. But, shikonin treatment significantly reduced the expression levels of TNF-α, IFN-γ and Bax. On the other hand, the expression levels of TGF-β and Bcl2 as well as the activity of glutathione peroxidase-1 (GPX-1) enzyme were significantly increased following the shikonin treatment. CONCLUSIONS This study emphasized the immune-modulatory and antioxidative effects of shikonin, which may have an important healing effect on the severity of EAE.
Collapse
Affiliation(s)
| | - Sajjad Biglari
- School of Medicine, Aja University of Medical Science, Tehran, Iran
| | | | | |
Collapse
|
48
|
Karpus WJ. Cytokines and Chemokines in the Pathogenesis of Experimental Autoimmune Encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2020; 204:316-326. [PMID: 31907274 DOI: 10.4049/jimmunol.1900914] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 11/13/2019] [Indexed: 12/25/2022]
Abstract
Experimental autoimmune encephalomyelitis is a CD4+ T cell-mediated demyelinating disease of the CNS that serves as a model for multiple sclerosis. Cytokines and chemokines shape Th1 and Th17 effector responses as well as regulate migration of leukocytes to the CNS during disease. The CNS cellular infiltrate consists of Ag-specific and nonspecific CD4+ and CD8+ T cells, neutrophils, B cells, monocytes, macrophages, and dendritic cells. The mechanism of immune-mediated inflammation in experimental autoimmune encephalomyelitis has been extensively studied in an effort to develop therapeutic modalities for multiple sclerosis and, indeed, has provided insight in modern drug discovery. The present Brief Review highlights critical pathogenic aspects of cytokines and chemokines involved in generation of effector T cell responses and migration of inflammatory cells to the CNS. Select cytokines and chemokines are certainly important in the regulatory response, which involves T regulatory, B regulatory, and myeloid-derived suppressor cells. However, that discussion is beyond the scope of this brief review.
Collapse
Affiliation(s)
- William J Karpus
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53706
| |
Collapse
|
49
|
Lisak RP, Nedelkoska L, Benjamins JA. Sigma-1 receptor agonists as potential protective therapies in multiple sclerosis. J Neuroimmunol 2020; 342:577188. [PMID: 32179326 DOI: 10.1016/j.jneuroim.2020.577188] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 01/17/2020] [Accepted: 02/07/2020] [Indexed: 01/25/2023]
Abstract
The sigma-1 receptor (σ-1R) is an endoplasmic reticulum (ER) chaperone upregulated during ER stress, and regulates calcium homeostasis. Agonists of σ-1R are neuroprotective. ANAVEX2-73, a new σ-1R agonist, is undergoing several clinical trials. We show that ANAVEX2-73 protects oligodendroglia (OL) and oligodendroglial precursors (OPC) from apoptosis, excitotoxicity, reactive oxygen species (ROS) and quinolinic acid (QA), associated with inflammation. ANAVEX2-73 stimulates OPC proliferation, but does not alter early maturation to OL. We previously reported that dextromethorphan (DM), another σ-1R agonist with a different structure, had similar effects. We now show that both DM and ANAVEX2-73 protect neurons from the four cytotoxic agents.
Collapse
Affiliation(s)
- Robert P Lisak
- Department of Neurology, Wayne State University School of Medicine, Detroit, MI, USA; Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Liljana Nedelkoska
- Department of Neurology, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Joyce A Benjamins
- Department of Neurology, Wayne State University School of Medicine, Detroit, MI, USA; Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI, USA.
| |
Collapse
|
50
|
Migotto MA, Mardon K, Orian J, Weckbecker G, Kneuer R, Bhalla R, Reutens DC. Efficient Distribution of a Novel Zirconium-89 Labeled Anti-cd20 Antibody Following Subcutaneous and Intravenous Administration in Control and Experimental Autoimmune Encephalomyelitis-Variant Mice. Front Immunol 2019; 10:2437. [PMID: 31681317 PMCID: PMC6813232 DOI: 10.3389/fimmu.2019.02437] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 09/30/2019] [Indexed: 11/13/2022] Open
Abstract
Objective: To investigate the imaging and biodistribution of a novel zirconium-89 (89Zr)-labeled mouse anti-cd20 monoclonal antibody (mAb) in control and experimental autoimmune encephalomyelitis (EAE) mice following subcutaneous (s. c.) and intravenous (i.v.) administration. Background: Anti-cd20-mediated B-cell depletion using mAbs is a promising therapy for multiple sclerosis. Recombinant human myelin oligodendrocyte glycoprotein (rhMOG)-induced EAE involves B-cell-mediated inflammation and demyelination in mice. Design/Methods: C57BL/6J mice (n = 39) were EAE-induced using rhMOG. On Day 14 post EAE induction, 89Zr-labeled-anti-cd20 mAb was injected in control and EAE mice in the right lower flank (s.c.) or tail vein (i.v.). Positron emission tomography/computed tomography (PET/CT) imaging and gamma counting (ex vivo) were performed on Days 1, 3, and 7 to quantify tracer accumulation in the major organs, lymphatics, and central nervous system (CNS). A preliminary study was conducted in healthy mice to elucidate full and early kinetics of the tracer that were subsequently applied in the EAE and control mice study. Results:89Zr-labeled anti-cd20 mAb was effectively absorbed from s.c. and i.v. injection sites and distributed to all major organs in the EAE and control mice. There was a good correlation between in vivo PET/CT data and ex vivo quantification of biodistribution of the tracer. From gamma counting studies, initial tracer uptake within the lymphatic system was found to be higher in the draining lymph nodes (inguinal or subiliac and sciatic) following s.c. vs. i.v. administration; within the CNS a significantly higher tracer uptake was observed at 24 h in the cerebellum, cerebrum, and thoracic spinal cord (p < 0.05 for all) following s.c. vs. i.v. administration. Conclusions: The preclinical data suggest that initial tracer uptake was significantly higher in the draining lymph nodes (subiliac and sciatic) and parts of CNS (the cerebellum and cerebrum) when administered s.c. compared with i.v in EAE mice.
Collapse
Affiliation(s)
- Mary-Anne Migotto
- Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia
| | - Karine Mardon
- Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia.,National Imaging Facility, The University of Queensland, Brisbane, QLD, Australia
| | - Jacqueline Orian
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Gisbert Weckbecker
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Rainer Kneuer
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Rajiv Bhalla
- Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia.,ARC Training Centre for Innovation in Biomedical Imaging Technology, Brisbane, QLD, Australia
| | - David C Reutens
- Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia.,ARC Training Centre for Innovation in Biomedical Imaging Technology, Brisbane, QLD, Australia
| |
Collapse
|