1
|
Zhao X, Jacob C. Mechanisms of Demyelination and Remyelination Strategies for Multiple Sclerosis. Int J Mol Sci 2023; 24:ijms24076373. [PMID: 37047344 PMCID: PMC10093908 DOI: 10.3390/ijms24076373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/19/2023] [Accepted: 03/25/2023] [Indexed: 03/31/2023] Open
Abstract
All currently licensed medications for multiple sclerosis (MS) target the immune system. Albeit promising preclinical results demonstrated disease amelioration and remyelination enhancement via modulating oligodendrocyte lineage cells, most drug candidates showed only modest or no effects in human clinical trials. This might be due to the fact that remyelination is a sophistically orchestrated process that calls for the interplay between oligodendrocyte lineage cells, neurons, central nervous system (CNS) resident innate immune cells, and peripheral immune infiltrates and that this process may somewhat differ in humans and rodent models used in research. To ensure successful remyelination, the recruitment and activation/repression of each cell type should be regulated in a highly organized spatio–temporal manner. As a result, drug candidates targeting one single pathway or a single cell population have difficulty restoring the optimal microenvironment at lesion sites for remyelination. Therefore, when exploring new drug candidates for MS, it is instrumental to consider not only the effects on all CNS cell populations but also the optimal time of administration during disease progression. In this review, we describe the dysregulated mechanisms in each relevant cell type and the disruption of their coordination as causes of remyelination failure, providing an overview of the complex cell interplay in CNS lesion sites.
Collapse
|
2
|
Grassi S, Cabitta L, Prioni S, Mauri L, Ciampa MG, Yokoyama N, Iwabuchi K, Zorina Y, Prinetti A. Identification of the Lipid Antigens Recognized by rHIgM22, a Remyelination-Promoting Antibody. Neurochem Res 2023; 48:1783-1797. [PMID: 36695984 DOI: 10.1007/s11064-023-03859-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 11/22/2022] [Accepted: 01/09/2023] [Indexed: 01/26/2023]
Abstract
Failure of the immune system to discriminate myelin components from foreign antigens plays a critical role in the pathophysiology of multiple sclerosis. In fact, the appearance of anti-myelin autoantibodies, targeting both proteins and glycolipids, is often responsible for functional alterations in myelin-producing cells in this disease. Nevertheless, some of these antibodies were reported to be beneficial for remyelination. Recombinant human IgM22 (rHIgM22) binds to myelin and to the surface of O4-positive oligodendrocytes, and promotes remyelination in mouse models of chronic demyelination. Interestingly, the identity of the antigen recognized by this antibody remains to be elucidated. The preferential binding of rHIgM22 to sulfatide-positive cells or tissues suggests that sulfatide might be part of the antigen pattern recognized by the antibody, however, cell populations lacking sulfatide expression are also responsive to rHIgM22. Thus, we assessed the binding of rHIgM22 in vitro to purified lipids and lipid extracts from various sources to identify the antigen(s) recognized by this antibody. Our results show that rHIgM22 is indeed able to bind both sulfatide and its deacylated form, whereas no significant binding for other myelin sphingolipids has been detected. Remarkably, binding of rHIgM22 to sulfatide in lipid monolayers can be positively or negatively regulated by the presence of other lipids. Moreover, rHIgM22 also binds to phosphatidylinositol, phosphatidylserine and phosphatidic acid, suggesting that not only sulfatide, but also other membrane lipids might play a role in the binding of rHIgM22 to oligodendrocytes and to other cell types not expressing sulfatide.
Collapse
Affiliation(s)
- Sara Grassi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Fratelli Cervi 93, Segrate, 20090, Milan, Italy.
| | - Livia Cabitta
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Fratelli Cervi 93, Segrate, 20090, Milan, Italy
| | - Simona Prioni
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Fratelli Cervi 93, Segrate, 20090, Milan, Italy
| | - Laura Mauri
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Fratelli Cervi 93, Segrate, 20090, Milan, Italy
| | - Maria Grazia Ciampa
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Fratelli Cervi 93, Segrate, 20090, Milan, Italy
| | - Noriko Yokoyama
- Institute for Environmental and Gender Specific Medicine, Graduate School of Medicine, Juntendo University, Urayasu, Chiba, Japan
| | - Kazuhisa Iwabuchi
- Institute for Environmental and Gender Specific Medicine, Graduate School of Medicine, Juntendo University, Urayasu, Chiba, Japan
| | | | - Alessandro Prinetti
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Fratelli Cervi 93, Segrate, 20090, Milan, Italy
| |
Collapse
|
3
|
Calahorra L, Camacho-Toledano C, Serrano-Regal MP, Ortega MC, Clemente D. Regulatory Cells in Multiple Sclerosis: From Blood to Brain. Biomedicines 2022; 10:335. [PMID: 35203544 PMCID: PMC8961785 DOI: 10.3390/biomedicines10020335] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 01/26/2022] [Accepted: 01/27/2022] [Indexed: 02/01/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic, autoimmune, and neurodegenerative disease of the central nervous system (CNS) that affects myelin. The etiology of MS is unclear, although a variety of environmental and genetic factors are thought to increase the risk of developing the disease. Historically, T cells were considered to be the orchestrators of MS pathogenesis, but evidence has since accumulated implicating B lymphocytes and innate immune cells in the inflammation, demyelination, and axonal damage associated with MS disease progression. However, more recently the importance of the protective role of immunoregulatory cells in MS has become increasingly evident, such as that of myeloid-derived suppressor cells (MDSCs), regulatory T (Treg) and B (Breg) cells, or CD56bright natural killer cells. In this review, we will focus on how peripheral regulatory cells implicated in innate and adaptive immune responses are involved in the physiopathology of MS. Moreover, we will discuss how these cells are thought to act and contribute to MS histopathology, also addressing their promising role as promoters of successful remyelination within the CNS. Finally, we will analyze how understanding these protective mechanisms may be crucial in the search for potential therapies for MS.
Collapse
Affiliation(s)
| | | | | | | | - Diego Clemente
- Grupo de Neuroinmuno-Reparación, Hospital Nacional de Parapléjicos, Finca La Peraleda s/n, 45071 Toledo, Spain; (L.C.); (C.C.-T.); (M.P.S.-R.); (M.C.O.)
| |
Collapse
|
4
|
Mohammedsaleh ZM. The use of patient-specific stem cells in different autoimmune diseases. Saudi J Biol Sci 2022; 29:3338-3346. [PMID: 35844404 PMCID: PMC9280249 DOI: 10.1016/j.sjbs.2022.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/27/2022] [Accepted: 02/06/2022] [Indexed: 10/31/2022] Open
Abstract
Autoimmune diseases are developed when the immune system mistakenly attacks the body’s cells. These inflammatory disorders can be inherited or triggered by external forces, such as type 1 diabetes, which is caused by the immune system's destruction of pancreatic beta cells. So far, stem cells such as hESC and iPSC have been used to treat autoimmune disorders such as type 1 diabetes, rheumatoid arthritis (RA), multiple sclerosis (MS), and systemic lupus erythematosus (SLE), although these procedures have certain ethical concerns. On the other hand, bone marrow-derived mesenchymal stem cells (BM-MSC) are thought to be the best source of stem cells. Later, it was shown that mesenchymal stem cells produced from autologous adipose tissues have a great potential for producing huge volumes of stem cells. In-vitro and in-vivo investigations using autologous hematopoietic stem cells and autologous mesenchymal stem cells have been carried out on various rodent and human models, while clinical trials for inflammatory diseases such as multiple sclerosis and diabetes mellitus have yielded promising results. We attempted to summarise the usage of diverse stem cells in the therapy of various autoimmune disorders in this review. Shortly, we expect that the use of autologous stem cells will provide a new perspective on the treatment of autoimmune disorders.
Collapse
|
5
|
Kanhai KMS, Goulooze SC, van der Grond J, Harms AC, Hankemeier T, Verma A, Dent G, Chavez J, Meijering H, Groeneveld GJ. Kinetics of myelin breakdown products: A labeling study in patients with progressive multiple sclerosis. Clin Transl Sci 2021; 15:638-648. [PMID: 34799987 PMCID: PMC8932820 DOI: 10.1111/cts.13181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 05/10/2021] [Accepted: 10/06/2021] [Indexed: 11/26/2022] Open
Abstract
The majority of disease modifying therapies for multiple sclerosis (MS) reduce inflammation, but do no’t target remyelination. Development of remyelinating therapies will benefit from a method to quantify myelin kinetics in patients with MS. We labeled myelin in vivo with deuterium, and modeled kinetics of myelin breakdown products β‐galactosylceramide (β‐GalC) and N‐Octadecanoyl‐sulfatide (NO‐Sulf). Five patients with MS received 120 ml 70% D2O daily for 70 days and were compared with six healthy subjects who previously received the same procedure. Mass spectrometry and compartmental modeling were used to quantify the turnover rate of β‐GalC and NO‐Sulf in cerebrospinal fluid (CSF). Turnover rate constants of the fractions of β‐GalC and NO‐Sulf with non‐negligible turnover were 0.00186 and 0.00714, respectively, in both healthy subjects and patients with MS. The turnover half‐life of β‐GalC and NO‐Sulf was calculated as 373 days and 96.5 days, respectively. The effect of MS on the NO‐Sulf (49.4% lower fraction with non‐negligible turnover) was more pronounced compared to the effect on β‐GalC turnover (18.3% lower fraction with non‐negligible turnover). Kinetics of myelin breakdown products in the CSF are different in patients with MS compared with healthy subjects. This may be caused by slower myelin production in these patients, by a higher level of degradation of a more stable component of myelin, or, most likely, by a combination of these two processes. Labeling myelin breakdown products is a useful method that can be used to quantify myelin turnover in patients with progressive MS and can therefore be used in proof‐of‐concept studies with remyelination therapies.
Collapse
Affiliation(s)
- Kawita M S Kanhai
- Centre for Human Drug Research, Leiden, The Netherlands.,Prothya Biosolutions, Amsterdam, The Netherlands
| | - Sebastiaan C Goulooze
- Centre for Human Drug Research, Leiden, The Netherlands.,Department of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | | | - Amy C Harms
- Prothya Biosolutions, Amsterdam, The Netherlands.,Radiology Department, Leiden University Medical Center, Leiden, The Netherlands
| | - Thomas Hankemeier
- Department of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands.,Netherlands Metabolomics Centre, Leiden, The Netherlands
| | - Ajay Verma
- Yumanity Pharmaceuticals, Boston, Massachusetts, USA
| | | | | | | | | |
Collapse
|
6
|
Gingele S, Stangel M. Emerging myelin repair agents in preclinical and early clinical development for the treatment of multiple sclerosis. Expert Opin Investig Drugs 2020; 29:583-594. [PMID: 32348161 DOI: 10.1080/13543784.2020.1762567] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Remyelination is a highly effective regenerative process that can restore axon function, prevent axonal loss, and reverse clinical deficits after demyelination. Hence, the promotion of remyelination is a logical goal in patients with multiple sclerosis (MS) in which remyelination is often insufficient. However, despite great progress regarding the development of immunomodulatory therapies for MS and an abundance of promising evidence from preclinical experiments so far, no therapy has convincingly demonstrated clinically significant remyelination properties. Therefore, enhancing myelin repair is an urgent and unmet need in MS. AREAS COVERED We searched clinicaltrials.gov and pubmed.ncbi.nlm.nih.gov and focused on therapeutic agents in development from the preclinical stage to clinical phase II. We selected agents for which data are available from in vitro experiments and at least one toxic demyelination animal model that reached at least phase I in clinical development in MS patients. EXPERT OPINION The evidence to promote remyelination is very promising for several agents, some of which possess anti-muscarinergic properties. Since remyelination is a complex process that involves various coordinated steps, a combination of different therapeutic approaches addressing different aspects of this regenerative mechanism may be reasonable. Furthermore, suitable surrogate markers of remyelination are necessary for proof-of-concept clinical trials.
Collapse
Affiliation(s)
- Stefan Gingele
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School , Hannover, Germany
| | - Martin Stangel
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School , Hannover, Germany
| |
Collapse
|
7
|
Grassi S, Mauri L, Prioni S, Cabitta L, Sonnino S, Prinetti A, Giussani P. Sphingosine 1-Phosphate Receptors and Metabolic Enzymes as Druggable Targets for Brain Diseases. Front Pharmacol 2019; 10:807. [PMID: 31427962 PMCID: PMC6689979 DOI: 10.3389/fphar.2019.00807] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 06/21/2019] [Indexed: 12/12/2022] Open
Abstract
The central nervous system is characterized by a high content of sphingolipids and by a high diversity in terms of different structures. Stage- and cell-specific sphingolipid metabolism and expression are crucial for brain development and maintenance toward adult age. On the other hand, deep dysregulation of sphingolipid metabolism, leading to altered sphingolipid pattern, is associated with the majority of neurological and neurodegenerative diseases, even those totally lacking a common etiological background. Thus, sphingolipid metabolism has always been regarded as a promising pharmacological target for the treatment of brain disorders. However, any therapeutic hypothesis applied to complex amphipathic sphingolipids, components of cellular membranes, has so far failed probably because of the high regional complexity and specificity of the different biological roles of these structures. Simpler sphingosine-based lipids, including ceramide and sphingosine 1-phosphate, are important regulators of brain homeostasis, and, thanks to the relative simplicity of their metabolic network, they seem a feasible druggable target for the treatment of brain diseases. The enzymes involved in the control of the levels of bioactive sphingoids, as well as the receptors engaged by these molecules, have increasingly allured pharmacologists and clinicians, and eventually fingolimod, a functional antagonist of sphingosine 1-phosphate receptors with immunomodulatory properties, was approved for the therapy of relapsing-remitting multiple sclerosis. Considering the importance of neuroinflammation in many other brain diseases, we would expect an extension of the use of such analogs for the treatment of other ailments in the future. Nevertheless, many aspects other than neuroinflammation are regulated by bioactive sphingoids in healthy brain and dysregulated in brain disease. In this review, we are addressing the multifaceted possibility to address the metabolism and biology of bioactive sphingosine 1-phosphate as novel targets for the development of therapeutic paradigms and the discovery of new drugs.
Collapse
Affiliation(s)
- Sara Grassi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Laura Mauri
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Simona Prioni
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Livia Cabitta
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Sandro Sonnino
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Alessandro Prinetti
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Paola Giussani
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| |
Collapse
|
8
|
Abe H, Kajitani N, Okada-Tsuchioka M, Omori W, Yatsumoto M, Takebayashi M. Antidepressant amitriptyline-induced matrix metalloproteinase-9 activation is mediated by Src family tyrosine kinase, which leads to glial cell line-derived neurotrophic factor mRNA expression in rat astroglial cells. Neuropsychopharmacol Rep 2019; 39:156-163. [PMID: 31025529 PMCID: PMC7292280 DOI: 10.1002/npr2.12055] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/04/2019] [Accepted: 02/08/2019] [Indexed: 12/25/2022] Open
Abstract
Background Astrocytes have been implicated in the pathophysiology of mood disorders and in the mechanism of the pharmacological effects of antidepressant drugs by the production of neurotrophic/growth factors. Previous studies have identified astrocyte‐expressed Gαi/o‐coupled lysophosphatidic acid receptor 1 (LPAR1), as being involved in antidepressant‐induced production of glial cell line‐derived neurotrophic factor (GDNF) and matrix metalloproteinase‐9 (MMP‐9) activation, an important step in the production of GNDF. However, the precise mechanism of MMP‐9 activation by antidepressants has yet to be identified, in particular the intracellular signaling pathway between LPAR1/Gαi/o and MMP‐9. Methods and Results Treatment of rat C6 astroglial cells (C6 cells) with amitriptyline increased Src family tyrosine kinase phosphorylation in a time and concentration‐dependent manner. Amitriptyline‐induced GDNF mRNA expression was blocked by Src family tyrosine kinase inhibitors. In addition, inhibiting Src family tyrosine kinase blocked amitriptyline‐induced zymographic MMP‐9 activation in C6 cells. The amitriptyline‐induced zymographic MMP‐9 activity was completely blocked by selective inhibition of Gαi/o protein and LPAR1. Furthermore, the amitriptyline‐induced Src family tyrosine kinase phosphorylation was blocked by LPAR1, but not MMP‐9 inhibition, indicating that Src family tyrosine kinase involvement is downstream of LPAR1. Conclusions The current findings suggest that the pharmacological effect of antidepressant such as amitriptyline is mediated through an intracellular signaling pathway via the LPAR1/Gαi/o/Src family tyrosine kinase, which leads to MMP‐9 activation and GDNF production. Treatment of rat C6 astroglial cells (C6 cells) with amitriptyline increased Src family tyrosine kinase phosphorylation in a time‐ and concentration‐dependent manner. The current findings suggest that the pharmacological effect of antidepressant such as amitriptyline is mediated through an intracellular signaling pathway via the LPAR1/Gαi/o/Src family tyrosine kinase, which leads to MMP‐9 activation and glial cell line‐derived neurotrophic factor production.![]()
Collapse
Affiliation(s)
- Hiromi Abe
- Division of Psychiatry and Neuroscience, Institute for Clinical Research, National Hospital Organization (NHO) Kure Medical Center and Chugoku Cancer Center, Kure, Japan.,Department of Pharmacy, National Hospital Organization (NHO) Kure Medical Center and Chugoku Cancer Center, Kure, Japan
| | - Naoto Kajitani
- Division of Psychiatry and Neuroscience, Institute for Clinical Research, National Hospital Organization (NHO) Kure Medical Center and Chugoku Cancer Center, Kure, Japan
| | - Mami Okada-Tsuchioka
- Division of Psychiatry and Neuroscience, Institute for Clinical Research, National Hospital Organization (NHO) Kure Medical Center and Chugoku Cancer Center, Kure, Japan
| | - Wataru Omori
- Division of Psychiatry and Neuroscience, Institute for Clinical Research, National Hospital Organization (NHO) Kure Medical Center and Chugoku Cancer Center, Kure, Japan
| | - Masahide Yatsumoto
- Department of Pharmacy, National Hospital Organization (NHO) Kure Medical Center and Chugoku Cancer Center, Kure, Japan
| | - Minoru Takebayashi
- Division of Psychiatry and Neuroscience, Institute for Clinical Research, National Hospital Organization (NHO) Kure Medical Center and Chugoku Cancer Center, Kure, Japan.,Department of Neuropsychiatry, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
9
|
Fereidan-Esfahani M, Nayfeh T, Warrington A, Howe CL, Rodriguez M. IgM Natural Autoantibodies in Physiology and the Treatment of Disease. Methods Mol Biol 2019; 1904:53-81. [PMID: 30539466 DOI: 10.1007/978-1-4939-8958-4_3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Antibodies are vital components of the adaptive immune system for the recognition and response to foreign antigens. However, some antibodies recognize self-antigens in healthy individuals. These autoreactive antibodies may modulate innate immune functions. IgM natural autoantibodies (IgM-NAAs) are a class of primarily polyreactive immunoglobulins encoded by germline V-gene segments which exhibit low affinity but broad specificity to both foreign and self-antigens. Historically, these autoantibodies were closely associated with autoimmune disease. Nevertheless, not all human autoantibodies are pathogenic and compelling evidence indicates that IgM-NAAs may exert a spectrum of effects from injurious to protective depending upon cellular and molecular context. In this chapter, we review the current state of knowledge regarding the potential physiological and therapeutic roles of IgM-NAAs in different disease conditions such as atherosclerosis, cancer, and autoimmune disease. We also describe the discovery of two reparative IgM-NAAs by our laboratory and delineate their proposed mechanisms of action in central nervous system (CNS) disease.
Collapse
Affiliation(s)
| | - Tarek Nayfeh
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | | | - Charles L Howe
- Department of Neurology, Mayo Clinic, Rochester, MN, USA.
| | | |
Collapse
|
10
|
Human Remyelination Promoting Antibody Stimulates Astrocytes Proliferation Through Modulation of the Sphingolipid Rheostat in Primary Rat Mixed Glial Cultures. Neurochem Res 2018; 44:1460-1474. [PMID: 30569280 DOI: 10.1007/s11064-018-2701-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Revised: 11/22/2018] [Accepted: 12/12/2018] [Indexed: 01/17/2023]
Abstract
Remyelination promoting human IgMs effectively increase the number of myelinated axons in animal models of multiple sclerosis. Hence, they ultimately stimulate myelin production by oligodendrocytes (OLs); however, their exact mechanism of action remains to be elucidated, and in particular, it remains unclear whether they are directly targeting OLs, or their action is mediated by effects on other cell types. We assessed the effect of remyelination promoting antibody rHIgM22 on the proliferative response and on the ceramide/sphingosine 1-phosphate rheostat in mixed glial cell cultures (MGCs). rHIgM22 treatment caused a time-dependent increase in PDGFαR protein in MGCs. Forty-eight hours of treatment with rHIgM22 induced a dose-dependent proliferative response (evaluated as total cell number and as EdU(+) cell number) in MGCs. When the proliferation response of MGCs to rHIgM22 was analyzed as a function of the cell types, the most significant proliferative response was associated with GLAST(+) cells, i.e., astrocytes. In many cell types, the balance between different sphingolipid mediators (the "sphingolipid rheostat"), in particular ceramide and sphingosine 1-phosphate, is critical in determining the cell fate. rHIgM22 treatment in MGCs induced a moderate but significant inhibition of total acidic sphingomyelinase activity (measured in vitro on cell lysates), the main enzyme responsible for the stimulus-mediated production of ceramide, when treatment was performed in serum containing medium, but no significant differences were observed when antibody treatment was performed in the absence of serum. Moreover, rHIgM22 treatment, either in the presence or in absence of serum, had no effects on ceramide levels. On the other hand, rHIgM22 treatment for 24 h induced increased production and release of sphingosine 1-phosphate in the extracellular milieu of MGC. Release of sphingosine 1-phosphate upon rHIgM22 treatment was strongly reduced by a selective inhibitor of PDGFαR. Increased sphingosine 1-phosphate production does not seem to be mediated by regulation of the biosynthetic enzymes, sphingosine kinase 1 and 2, since protein levels of these enzymes and phosphorylation of sphingosine kinase 1 were unchanged upon rHIgM22 treatment. Instead, we observed a significant reduction in the levels of sphingosine 1-phosphate lyase 1, one of the key catabolic enzymes. Remarkably, rHIgM22 treatment under the same experimental conditions did not induce changes in the production and/or release of sphingosine 1-phosphate in pure astrocyte cultures. Taken together, these data suggest that rHIgM22 indirectly influences the proliferation of astrocytes in MGCs, by affecting the ceramide/sphingosine 1-phosphate balance. The specific cell population directly targeted by rHIgM22 remains to be identified, however our study unveils another aspect of the complexity of rHIgM22-induced remyelinating effect.
Collapse
|
11
|
LoPresti P. Silent Free Fall at Disease Onset: A Perspective on Therapeutics for Progressive Multiple Sclerosis. Front Neurol 2018; 9:973. [PMID: 30542317 PMCID: PMC6277889 DOI: 10.3389/fneur.2018.00973] [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: 07/25/2018] [Accepted: 10/29/2018] [Indexed: 01/08/2023] Open
Abstract
Central nervous system (CNS) degeneration occurs during multiple sclerosis (MS) following several years of reversible autoimmune demyelination. Progressive CNS degeneration appears later during the course of relapsing-remitting MS (RRMS), although it starts insidiously at disease onset. We propose that there is an early subclinical phase also for primary-progressive (PP) MS. Consensus exists that many different cell types are involved during disease onset. Furthermore, the response to the initial damage, which is specific for each individual, would result in distinct pathological pathways that add complexity to the disease and the mechanisms underlying progressive CNS degeneration. Progressive MS is classified as either active or not active, as well as with or without progression. Different forms of progressive MS might reflect distinct or overlapping pathogenetic pathways. Disease mechanisms should be determined for each patient at diagnosis and the time of treatment. Until individualized and time-sensitive treatments that specifically target the molecular mechanisms of the progressive aspect of the disease are identified, combined therapies directed at anti-inflammation, regeneration, and neuroprotection are the most effective for preventing MS progression. This review presents selected therapeutics in support of the overall idea of a multidimensional therapy applied early in the disease. This approach could limit damage and increase CNS repair. By targeting several cellular populations (i.e., microglia, astrocytes, neurons, oligodendrocytes, and lymphocytes) and multiple pathological processes (e.g., inflammation, demyelination, synaptopathy, and excitatory/inhibitory imbalance) progressive MS could be attenuated. Early timing for such multidimensional therapy is proposed as the prerequisite for effectively halting progressive MS.
Collapse
Affiliation(s)
- Patrizia LoPresti
- Department of Psychology, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
12
|
Lemus HN, Warrington AE, Rodriguez M. Multiple Sclerosis: Mechanisms of Disease and Strategies for Myelin and Axonal Repair. Neurol Clin 2018; 36:1-11. [PMID: 29157392 PMCID: PMC7125639 DOI: 10.1016/j.ncl.2017.08.002] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Hernan Nicolas Lemus
- Department of Neurology, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| | - Arthur E Warrington
- Department of Neurology, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA.
| | - Moses Rodriguez
- Department of Neurology, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA; Department of Immunology, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| |
Collapse
|
13
|
Perwein MK, Smestad JA, Warrington AE, Heider RM, Kaczor MW, Maher LJ, Wootla B, Kunbaz A, Rodriguez M. A comparison of human natural monoclonal antibodies and aptamer conjugates for promotion of CNS remyelination: where are we now and what comes next? Expert Opin Biol Ther 2018; 18:545-560. [PMID: 29460650 DOI: 10.1080/14712598.2018.1441284] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Multiple sclerosis (MS) is a chronic and progressive inflammatory demyelinating disease of the human central nervous system (CNS) and is the most common disabling neurological condition in young adults, resulting in severe neurological defects. No curative or long-term progression-inhibiting therapy has yet been developed. However, recent investigation has revealed potential strategies that do not merely modulate potentially pathogenic autoimmune responses, but stimulate remyelination within CNS lesions. AREAS COVERED We discuss the history and development of natural human IgM-isotype immunoglobulins (HIgMs) and recently-identified aptamer-conjugates that have been shown to enhance endogenous myelin repair in animal models of demyelination by acting on myelin-producing oligodendrocytes (OLs) or oligodendrocyte progenitor cells (OPCs) within CNS lesions. We also discuss future development aims and applications for these important novel technologies. EXPERT OPINION Aptamer conjugate Myaptavin-3064 and recombinant human IgM-isotype antibody rHIgM22 regenerate CNS myelin, thereby reducing axonal degeneration and offering the potential of recovery from MS relapses, reversal of disability and prevention of disease progression. Advancement of these technologies into the clinic for MS treatment is therefore a top priority. It remains unclear to what extent the therapeutic modalities of remyelinating antibodies and aptamers may synergize with other currently-approved therapies to yield enhanced therapeutic effects.
Collapse
Affiliation(s)
- Maria K Perwein
- a Department of Neurology , Mayo Clinic College of Medicine and Science , Rochester , MN , USA
| | - John A Smestad
- b Medical Scientist Training Program , Mayo Clinic College of Medicine and Science , Rochester , MN , USA.,c Department of Biochemistry and Molecular Biology , Mayo Clinic College of Medicine and Science , Rochester , MN , USA
| | - Arthur E Warrington
- a Department of Neurology , Mayo Clinic College of Medicine and Science , Rochester , MN , USA
| | - Robin M Heider
- c Department of Biochemistry and Molecular Biology , Mayo Clinic College of Medicine and Science , Rochester , MN , USA
| | - Mark W Kaczor
- a Department of Neurology , Mayo Clinic College of Medicine and Science , Rochester , MN , USA
| | - Louis J Maher
- c Department of Biochemistry and Molecular Biology , Mayo Clinic College of Medicine and Science , Rochester , MN , USA
| | - Bharath Wootla
- a Department of Neurology , Mayo Clinic College of Medicine and Science , Rochester , MN , USA
| | - Ahmad Kunbaz
- a Department of Neurology , Mayo Clinic College of Medicine and Science , Rochester , MN , USA
| | - Moses Rodriguez
- a Department of Neurology , Mayo Clinic College of Medicine and Science , Rochester , MN , USA.,d Department of Immunology , Mayo Clinic College of Medicine and Science , Rochester , MN , USA
| |
Collapse
|
14
|
Abstract
We have witnessed major successes in the development of effective immunomodulatory therapies capable of reducing adaptive immune-mediated myelin damage in MS over the last 30 years. However, until it is possible to prevent MS or initiate treatment before it has already caused lesions there is a need to repair myelin damage to prevent further axonal loss. The past decade has brought remarkable advances in our understanding of oligodendrocyte biology and the related search for remyelinating therapies in humans. In this review, we first outline the basic biology of central nervous system myelin and remyelination, including a discussion of the major identified pathways and targets that might help yield CNS remyelinating drugs. In conjunction, we provide an overview of techniques that have helped identify compounds capable of promoting oligodendrocyte precursor cell differentiation and myelination. This includes the methods for both initial in vitro screening and subsequent in vivo confirmation of the target. We then review methods proposed to quantify human remyelination in vivo, including visual evoked potentials and putative imaging modalities. As the remyelination era approaches, with the announcement of the first positive trial in remyelination, we are now tasked with answering new questions regarding patient-specific factors (e.g., age) that may influence the extent and optimal therapeutic window for remyelination.
Collapse
Affiliation(s)
- Riley M Bove
- Department of Neurology Weill Institute for the Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
| | - Ari J Green
- Department of Neurology Weill Institute for the Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
15
|
Timing of Future Remyelination Therapies and Their Potential to Stop Multiple Sclerosis Progression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 958:161-170. [PMID: 28093713 DOI: 10.1007/978-3-319-47861-6_10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Prior to the onset of demyelination in multiple sclerosis (MS), early oligodendrocyte injury, axonal degeneration and astroglial scarring occur. The irreversible progressive phase of MS begins when the axonal loss threshold is reached. Progressive disease onset has the highest impact on a poor prognosis in MS. Conversion to progressive disease is essentially an age-dependent process independent of disease duration and initial disease course. Although prevention of relapses has been the primary approach in the disease management, incomplete recovery from even the first relapse correlates with the long-term neurodegenerative phenotype of progressive MS onset. Therefore, the provider should review each patient's potential for relapse-related disability and start DMDs with the goal of preventing relapses. Existing immunomodulatory medications used to prevent MS relapses do not prevent long-term disability, which requires agents focused on remyelination and axonal repair. If applied immediately after a relapse rather than during the progressive phase of MS, remyelination-stimulating strategies may result in full recovery and prevention of long-term neurodegeneration and progressive disease course.
Collapse
|
16
|
Murphy NA, Franklin RJM. Recruitment of endogenous CNS stem cells for regeneration in demyelinating disease. PROGRESS IN BRAIN RESEARCH 2017; 231:135-163. [PMID: 28554395 DOI: 10.1016/bs.pbr.2016.12.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Demyelinating diseases, such as multiple sclerosis (MS), are responsible for a significant portion of the neurological disability burden worldwide, especially in young adults. Demyelination can be followed by a spontaneous regenerative process called remyelination, in which new myelin sheaths are restored to denuded axons. However, in chronic demyelinating disease such as MS, this process becomes progressively less efficient. This chapter reviews the biology of remyelination and the rationale and strategies by which it can be enhanced therapeutically in acquired demyelinating disease.
Collapse
Affiliation(s)
- Natalia A Murphy
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, Cambridge, United Kingdom; University of Cambridge, Cambridge, United Kingdom
| | - Robin J M Franklin
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, Cambridge, United Kingdom; University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
17
|
Lemus HN, Warrington AE, Denic A, Wootla B, Rodriguez M. Treatment with a recombinant human IgM that recognizes PSA-NCAM preserves brain pathology in MOG-induced experimental autoimmune encephalomyelitis. Hum Antibodies 2017; 25:121-129. [PMID: 28269761 DOI: 10.3233/hab-170313] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
A single peripheral dose of CNS-binding IgMs promote remyelination and preserve axons in a number of animal models of neurologic disease. A myelin-binding recombinant human IgM (rHIgM22) is presently in a safety trial in MS patients following an acute MS exacerbation. rHIgM22 (directed against oligodendrocytes) or rHIgM12 (directed against neurons) were administered to mice with MOG-induced experimental autoimmune encephalomyelitis (EAE) with study endpoints: clinical deficits and brain and spinal cord pathology. IgMs were administered at a therapeutic dose of 100 μ g intra peritoneal at the time of immunization (day -1, 0, +$1), disease onset (15 days) or peak of the disease (28 days). Disease course was not worsened by either human IgM regardless of the time of treatment. Of note, the human IgM that recognizes a carbohydrate epitope on gangliosides and NCAM, rHIgM12, reduced brain pathology when given at time of immunization or at onset of disease, but did not reduce clinical deficits or spinal cord disease burden. Hence, treatment with rHIgM12 resulted in marked reduction in meningeal inflammation. Data consistent with the hypothesis that in the EAE model this molecule has an immune-modulatory effect. Treatment with an anti-CD4 blocking IgG prevented both clinical course and CNS pathology. This pre-clinical study further supports the safety of therapeutic CNS-binding human IgMs in the presence of autoimmunity and clearly differentiates them from IgGs directed against MOG or aquaporin-4 that worsen neurologic disease.
Collapse
MESH Headings
- Animals
- Brain/drug effects
- Brain/immunology
- Brain/pathology
- Cognitive Dysfunction/chemically induced
- Cognitive Dysfunction/drug therapy
- Cognitive Dysfunction/immunology
- Cognitive Dysfunction/pathology
- Demyelinating Diseases/chemically induced
- Demyelinating Diseases/drug therapy
- Demyelinating Diseases/immunology
- Demyelinating Diseases/pathology
- Drug Administration Schedule
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Freund's Adjuvant/administration & dosage
- Humans
- Immunoglobulin M/pharmacology
- Immunologic Factors/pharmacology
- Injections, Intraperitoneal
- Mice
- Mice, Inbred C57BL
- Myelin Sheath/drug effects
- Myelin Sheath/immunology
- Myelin Sheath/pathology
- Myelin-Oligodendrocyte Glycoprotein/administration & dosage
- Neural Cell Adhesion Molecule L1/immunology
- Neural Cell Adhesion Molecule L1/metabolism
- Neurons/drug effects
- Neurons/immunology
- Neurons/pathology
- Neuroprotective Agents/pharmacology
- Oligodendroglia/drug effects
- Oligodendroglia/immunology
- Oligodendroglia/pathology
- Peptide Fragments/administration & dosage
- Protein Binding
- Recombinant Proteins/pharmacology
- Sialic Acids/immunology
- Sialic Acids/metabolism
- Spinal Cord/drug effects
- Spinal Cord/immunology
- Spinal Cord/pathology
Collapse
Affiliation(s)
| | | | | | - Bharath Wootla
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Moses Rodriguez
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
18
|
Abstract
Demyelination of central nervous system axons, associated with traumatic injury and demyelinating diseases such as multiple sclerosis, causes impaired neural transmission and ultimately axon degeneration. Consequently, extensive research has focused on signaling systems that promote myelinating activity of oligodendrocytes or promote production of new oligodendrocytes from oligodendrocyte progenitor cells. Many receptor systems, notably including growth factor receptors and G protein-coupled receptors, control myelination. A number of recent clinical trials target these receptor signaling pathways.
Collapse
Affiliation(s)
- Mark Bothwell
- Department of Physiology and Biophysics, University of Washington, Seattle, Washington 98195;
| |
Collapse
|
19
|
Fernandez-Castaneda A, Gaultier A. Adult oligodendrocyte progenitor cells - Multifaceted regulators of the CNS in health and disease. Brain Behav Immun 2016; 57:1-7. [PMID: 26796621 PMCID: PMC4940337 DOI: 10.1016/j.bbi.2016.01.005] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 12/28/2015] [Accepted: 01/11/2016] [Indexed: 01/17/2023] Open
Abstract
Oligodendrocyte progenitor cells (OPCs) are the often-overlooked fourth glial cell type in the central nervous system (CNS), comprising about 5% of the CNS. For a long time, our vision of OPC function was limited to the generation of mature oligodendrocytes. However, new studies have highlighted the multifaceted nature of OPCs. During homeostatic and pathological conditions, OPCs are the most proliferative cell type in the CNS, a property not consistent with the need to generate new oligodendrocytes. Indeed, OPCs modulate neuronal activity and OPC depletion in the brain can trigger depressive-like behavior. More importantly, OPCs are actively recruited to injury sites, where they orchestrate glial scar formation and contribute to the immune response. The following is a comprehensive analysis of the literature on OPC function beyond myelination, in the context of the healthy and diseased adult CNS.
Collapse
Affiliation(s)
- Anthony Fernandez-Castaneda
- Center for Brain Immunology and Glia, Department of Neuroscience, Graduate Program in Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Alban Gaultier
- Center for Brain Immunology and Glia, Department of Neuroscience, Graduate Program in Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
20
|
Wootla B, Denic A, Watzlawik JO, Warrington AE, Rodriguez M. Antibody-Mediated Oligodendrocyte Remyelination Promotes Axon Health in Progressive Demyelinating Disease. Mol Neurobiol 2016; 53:5217-28. [PMID: 26409478 PMCID: PMC5012151 DOI: 10.1007/s12035-015-9436-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 09/10/2015] [Indexed: 02/03/2023]
Abstract
Demyelination underlies early neurological symptoms in multiple sclerosis (MS); however, axonal damage is considered critical for permanent chronic deficits. The precise mechanisms by which axonal injury occurs in MS are unclear; one hypothesis is the absence or failure of remyelination, suggesting that promoting remyelination may protect axons from death. This report provides direct evidence that promoting oligodendrocyte remyelination protects axons and maintains transport function. Persistent Theiler's virus infection of Swiss Jim Lambert (SJL)/J mice was used as a model of MS to assess the effects of remyelination on axonal injury following demyelination in the spinal cord. Remyelination was induced using an oligodendrocyte/myelin-specific recombinant human monoclonal IgM, rHIgM22. The antibody is endowed with strong anti-apoptotic and pro-proliferative effects on oligodendrocyte progenitor cells. We used (1)H-magnetic resonance spectroscopy (MRS) at the brainstem to measure N-acetyl-aspartate (NAA) as a surrogate of neuronal health and spinal cord integrity. We found increased brainstem NAA concentrations at 5 weeks post-treatment with rHIgM22, which remained stable out to 10 weeks. Detailed spinal cord morphology studies revealed enhanced remyelination in the rHIgM22-treated group but not in the isotype control antibody- or saline-treated groups. Importantly, we found rHIgM22-mediated remyelination protected small- and medium-caliber mid-thoracic spinal cord axons from damage despite similar demyelination and inflammation across all experimental groups. The most direct confirmation of remyelination-mediated protection of descending neurons was an improvement in retrograde transport. Treatment with rHIgM22 significantly increased the number of retrograde-labeled neurons in the brainstem, indicating that preserved axons are functionally competent. This is direct validation that remyelination preserves spinal cord axons and protects functional axon integrity.
Collapse
Affiliation(s)
- Bharath Wootla
- Departments of Neurology, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
- Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Center for Regenerative Medicine, Neuroregeneration, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Aleksandar Denic
- Departments of Neurology, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
- Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Jens O Watzlawik
- Departments of Neurology, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
- Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Center for Regenerative Medicine, Neuroregeneration, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Arthur E Warrington
- Departments of Neurology, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
- Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Moses Rodriguez
- Departments of Neurology, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA.
- Departments of Immunology, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA.
- Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
21
|
Dasari H, Wootla B, Warrington AE, Rodriguez M. Concomitant Use of Neuroprotective Drugs in Neuro Rehabilitation of Multiple Sclerosis. ACTA ACUST UNITED AC 2016; 4. [PMID: 27595123 PMCID: PMC5006625 DOI: 10.4172/2329-9096.1000348] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We provide an overview of rehabilitation in neurological diseases. A large amount of literature available on neurorehabilitation is based from the rehabilitative work on stroke and spinal cord injuries. After a brief description of rehabilitation, the potential application of neurorehabilitation in neurodegenerative diseases specifically multiple sclerosis (MS) is summarized. Since MS causes a wide variety of symptoms, the rehabilitation in MS patients may benefit from an interdisciplinary approach that encloses physiotherapy, cognitive rehabilitation, psychological therapy, occupational therapy, and other methods to improve fatigue. Neurorehabilitation helps patients to reach and maintain their optimal physical, psychological and intellectual, levels but it does not reverse long-term disabilities that arise from neurological disorders. This calls for the need of better neuroregenerative and neuroprotective treatment strategies in addition to neurorehabilitation. We discuss neuroprotective drugs aimed at preventing axonal, neuronal, myelin and oligodendrocyte damage and cell death that are approved and others that are currently in clinical trials, with an emphasis on human derived natural antibodies with remyleination potential. Our investigative group developed recombinant natural human IgM antibodies against oligodendrocytes and neurons with a potential for CNS repair and remyleination. One such recombinant antibody, rHIgM22 completed a phase 1 clinical trial with no toxicity and with an objective of promoting remyleination in multiple sclerosis. Inclusion of these drugs as a multifaceted approach may further enhance the efficacy of neurorehabilitation in neuroinflammatory and neurodegenerative disorders.
Collapse
Affiliation(s)
- Harika Dasari
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA; Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Bharath Wootla
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA; Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Arthur E Warrington
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA; Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Moses Rodriguez
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA; Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA; Department of Immunology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| |
Collapse
|
22
|
Wootla B, Watzlawik JO, Stavropoulos N, Wittenberg NJ, Dasari H, Abdelrahim MA, Henley JR, Oh SH, Warrington AE, Rodriguez M. Recent Advances in Monoclonal Antibody Therapies for Multiple Sclerosis. Expert Opin Biol Ther 2016; 16:827-839. [PMID: 26914737 DOI: 10.1517/14712598.2016.1158809] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Multiple sclerosis (MS) is the most common chronic inflammatory, demyelinating disease of the CNS and results in neurological disability. Existing immunomodulatory and immunosuppressive approaches lower the number of relapses but do not cure or reverse existing deficits nor improve long-term disability in MS patients. AREAS COVERED Monogenic antibodies were described as treatment options for MS, however the immunogenicity of mouse antibodies hampered the efficacy of potential therapeutics in humans. Availability of improved antibody production technologies resulted in a paradigm shift in MS treatment strategies. In this review, an overview of immunotherapies for MS that use conventional monoclonal antibodies reactive to immune system and their properties and mechanisms of action will be discussed, including recent advances in MS therapeutics and highlight natural autoantibodies (NAbs) that directly target CNS cells. EXPERT OPINION Recent challenges for MS therapy are the identification of relevant molecular and cellular targets, time frame of treatment, and antibody toxicity profiles to identify safe treatment options for MS patients. The application of monoclonal antibody therapies with better biological efficacy associated with minimum side effects possesses huge clinical potential. Advances in monoclonal antibody technologies that directly target cells of nervous system may promote the CNS regeneration field from bench to bedside.
Collapse
Affiliation(s)
- Bharath Wootla
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.,Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.,Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Jens O Watzlawik
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road S, Jacksonville, FL 32224, USA
| | - Nikolaos Stavropoulos
- Department of General Medicine, Charles University in Prague, Faculty of Medicine in Hradec Kralove, Simkova 870, Hradec Kralove 1, 500 38, Czech Republic
| | - Nathan J Wittenberg
- Department of Electrical and Computer Engineering, University of Minnesota, 200 Union Street SE, 4-174 Keller Hall Minneapolis, MN 55455, USA.,Department of Biomedical Engineering, University of Minnesota, 200 Union Street SE, 4-174 Keller Hall Minneapolis, MN 55455, USA
| | - Harika Dasari
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.,Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Murtada A Abdelrahim
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.,Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - John R Henley
- Department of Neurologic Surgery, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.,Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.,Center for Regenerative Medicine, Neuroregeneration, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Sang-Hyun Oh
- Department of Electrical and Computer Engineering, University of Minnesota, 200 Union Street SE, 4-174 Keller Hall Minneapolis, MN 55455, USA.,Department of Biomedical Engineering, University of Minnesota, 200 Union Street SE, 4-174 Keller Hall Minneapolis, MN 55455, USA
| | - Arthur E Warrington
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.,Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Moses Rodriguez
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.,Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.,Department of Immunology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| |
Collapse
|
23
|
Grebing M, Nielsen HH, Fenger CD, T Jensen K, von Linstow CU, Clausen BH, Söderman M, Lambertsen KL, Thomassen M, Kruse TA, Finsen B. Myelin-specific T cells induce interleukin-1beta expression in lesion-reactive microglial-like cells in zones of axonal degeneration. Glia 2015; 64:407-24. [PMID: 26496662 DOI: 10.1002/glia.22937] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 10/07/2015] [Indexed: 12/18/2022]
Abstract
Infiltration of myelin-specific T cells into the central nervous system induces the expression of proinflammatory cytokines in patients with multiple sclerosis (MS). We have previously shown that myelin-specific T cells are recruited into zones of axonal degeneration, where they stimulate lesion-reactive microglia. To gain mechanistic insight, we used RNA microarray analysis to compare the transcript profile in hippocampi from perforant pathway axonal-lesioned mice with and without adoptively transferred myelin-specific T cells 2 days postlesion, when microglia are clearly lesion reactive. Pathway analysis revealed that, among the 1,447 differently expressed transcripts, the interleukin (IL)-1 pathway including all IL-1 receptor ligands was upregulated in the presence of myelin-specific T cells. Quantitative polymerase chain reaction showed increased mRNA levels of IL-1β, IL-1α, and IL-1 receptor antagonist in the T-cell-infiltrated hippocampi from axonal-lesioned mice. In situ hybridization and immunohistochemistry showed a T-cell-enhanced lesion-specific expression of IL-1β mRNA and protein, respectively, and induction of the apoptosis-associated speck-like protein, ASC, in CD11b(+) cells. Double in situ hybridization showed colocalization of IL-1β mRNA in a subset of CD11b mRNA(+) cells, of which many were part of cellular doublets or clusters, characteristic of proliferating, lesion-reactive microglia. Double-immunofluorescence showed a T-cell-enhanced colocalization of IL-1β to CD11b(+) cells, including lesion-reactive CD11b(+) ramified microglia. These results suggest that myelin-specific T cells stimulate lesion-reactive microglial-like cells to produce IL-1β. These findings are relevant to understand the consequences of T-cell infiltration in white and gray matter lesions in patients with MS.
Collapse
Affiliation(s)
- Manuela Grebing
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
| | - Helle H Nielsen
- Department of Neurology, Odense University Hospital, Odense C, Denmark
| | - Christina D Fenger
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
| | - Katrine T Jensen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
| | - Christian U von Linstow
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
| | - Bettina H Clausen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
| | - Martin Söderman
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
| | - Kate L Lambertsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
| | - Mads Thomassen
- Department of Clinical Genetics, Odense University Hospital, Odense C, Denmark
| | - Torben A Kruse
- Department of Clinical Genetics, Odense University Hospital, Odense C, Denmark
| | - Bente Finsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
| |
Collapse
|
24
|
Watzlawik JO, Kahoud RJ, Ng S, Painter MM, Papke LM, Zoecklein L, Wootla B, Warrington AE, Carey WA, Rodriguez M. Polysialic acid as an antigen for monoclonal antibody HIgM12 to treat multiple sclerosis and other neurodegenerative disorders. J Neurochem 2015; 134:865-78. [PMID: 25866077 DOI: 10.1111/jnc.13121] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 03/27/2015] [Accepted: 04/07/2015] [Indexed: 01/15/2023]
Abstract
CNS regeneration is a desirable goal for diseases of brain and spinal cord. Current therapeutic strategies for the treatment of multiple sclerosis (MS) aim to eliminate detrimental effects of the immune system, so far without reversing disability or affecting long-term prognosis in patients. Approachable molecular targets that stimulate CNS repair are not part of the clinical praxis or have not been identified yet. The purpose of this study was to identify the molecular target of the human monoclonal antibody HIgM12. HIgM12 reverses motor deficits in chronically demyelinated mice, a model of MS. Here, we identified polysialic acid (PSA) attached to the neural cell adhesion molecule (NCAM) as the antigen for HIgM12 by using different NCAM knockout strains and through PSA removal from the NCAM protein core. Antibody binding to CNS tissue and primary cells, antibody-mediated cell adhesion, and neurite outgrowth on HIgM12-coated nitrocellulose was detected only in the presence of PSA as assessed by western blotting, immunoprecipitation, immunocytochemistry, and histochemistry. We conclude that HIgM12 mediates its in vivo and in vitro effects through binding to PSA and has the potential to be an effective therapy for MS and neurodegenerative diseases. The human antibody HIgM12 stimulates neurite outgrowth in vitro and promotes function in chronically demyelinated mice, a model of multiple sclerosis. The cellular antigen for HIgM12 was undetermined. Here, we identified polysialic acid attached to NCAM (neural cell adhesion molecule) as the cellular target for HIgM12. This includes glial fibrillary acidic protein (GFAP)-positive mouse astrocytes (GFAP, red; HIgM12, green; DAPI, blue) among other cell types of the central nervous system. These findings indicate a new strategy for the treatment of neuro-motor disorders including multiple sclerosis.
Collapse
Affiliation(s)
- Jens O Watzlawik
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Robert J Kahoud
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.,Department of Pediatric and Adolescent Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Shermayne Ng
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Meghan M Painter
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Louisa M Papke
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Laurie Zoecklein
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Bharath Wootla
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Arthur E Warrington
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - William A Carey
- Department of Pediatric and Adolescent Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Moses Rodriguez
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| |
Collapse
|
25
|
Nadeem M, Sklover L, Sloane JA. Targeting remyelination treatment for multiple sclerosis. World J Neurol 2015; 5:5-16. [DOI: 10.5316/wjn.v5.i1.5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 11/29/2014] [Accepted: 12/17/2014] [Indexed: 02/06/2023] Open
Abstract
Since disability in multiple sclerosis (MS) is a product of neurodegeneration and deficient remyelination, the ability to enhance neuroregeneration and myelin regeneration in MS is an enticing goal for MS drug development. In particular, remyelination treatments could promote return of neurological function and also prevent further axonal loss and neurodegeneration in MS due to trophic effects of myelin. The study of remyelination has advanced dramatically in the last several years such that a number of pathways inhibiting remyelination have been discovered, including those involving LINGO-1, Notch-1, hyaluronan, retinoid X receptor, and wnt/ß-catenin. Other approaches such as high throughput drug screening for remyelination drugs have caught fire, with identification of dozens of known drugs with oligodendrocyte maturation stimulatory effects. Several drugs identified through screens and other mechanisms are in the process of being further evaluated for remyelination in MS and MS models. We discuss the potential molecular targets and the variety of mechanisms towards drug identification and development in remyelination for MS.
Collapse
|
26
|
Aureli M, Grassi S, Prioni S, Sonnino S, Prinetti A. Lipid membrane domains in the brain. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1851:1006-16. [PMID: 25677824 DOI: 10.1016/j.bbalip.2015.02.001] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 01/29/2015] [Accepted: 02/01/2015] [Indexed: 12/28/2022]
Abstract
The brain is characterized by the presence of cell types with very different functional specialization, but with the common trait of a very high complexity of structures originated by their plasma membranes. Brain cells bear evident membrane polarization with the creation of different morphological and functional subcompartments, whose formation, stabilization and function require a very high level of lateral order within the membrane. In other words, the membrane specialization of brain cells implies the presence of distinct membrane domains. The brain is the organ with the highest enrichment in lipids like cholesterol, glycosphingolipids, and the most recently discovered brain membrane lipid, phosphatidylglucoside, whose collective behavior strongly favors segregation within the membrane leading to the formation of lipid-driven membrane domains. Lipid-driven membrane domains function as dynamic platforms for signal transduction, protein processing, and membrane turnover. Essential events involved in the development and in the maintenance of the functional integrity of the brain depend on the organization of lipid-driven membrane domains, and alterations in lipid homeostasis, leading to deranged lipid-driven membrane organization, are common in several major brain diseases. In this review, we summarize the forces behind the formation of lipid membrane domains and their biological roles in different brain cells. This article is part of a Special Issue entitled Brain Lipids.
Collapse
Affiliation(s)
- Massimo Aureli
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Italy
| | - Sara Grassi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Italy
| | - Simona Prioni
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Italy
| | - Sandro Sonnino
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Italy
| | - Alessandro Prinetti
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Italy.
| |
Collapse
|
27
|
Williams A. Remyelination in multiple sclerosis: what do we know and where are we going? Neurodegener Dis Manag 2015; 5:49-59. [PMID: 25711454 DOI: 10.2217/nmt.14.40] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Multiple sclerosis (MS) patients today have more hope of a good disease outcome with an ever-increasing choice of immunomodulatory therapies to reduce disease relapses, thought to be caused by inflammation within the CNS, leading to axonal demyelination. However, although there has been much progress in this disease phase, there has been little impact on the progressive phase of MS, when neurodegeneration dominates and patients accumulate disability over years. This failure of prevention of progressive disease has led to a frame-shift in research thinking, focusing on neuroprotective strategies such as promotion of remyelination, to be used alongside immunomodulatory therapies. This review discusses this unmet need in MS, in terms of pathology and current knowledge of remyelination and proremyelinating therapies.
Collapse
|
28
|
Xu X, Ng SM, Hassouna E, Warrington A, Oh SH, Rodriguez M. Human-derived natural antibodies: biomarkers and potential therapeutics. FUTURE NEUROLOGY 2015; 10:25-39. [PMID: 25678860 DOI: 10.2217/fnl.14.62] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The immune system generates antibodies and antigen-specific T-cells as basic elements of the immune networks that differentiate self from non-self in a finely tuned manner. The antigen-specific nature of immune responses ensures that normal immune activation contains non-self when tolerating self. Here we review the B-1 subset of lymphocytes which produce self-reactive antibodies. By analyzing the IgM class of natural antibodies that recognize antigens from the nervous system, we emphasize that natural antibodies are biomarkers of how the immune system monitors the host. The immune response activated against self can be detrimental when triggered in an autoimmune genetic background. In contrast, tuning immune activity with natural antibodies is a potential therapeutic strategy.
Collapse
Affiliation(s)
- Xiaohua Xu
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Sher May Ng
- School of Clinical Medicine, University Of Cambridge, Hills Rd, Cambridge CB2 0SP, UK
| | - Eamonn Hassouna
- Department of General Medicine, Charles University Hradec Kralove Faculty, Prague, Czech Republic
| | - Arthur Warrington
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Sang-Hyun Oh
- Laboratory of Nanostructures & Biosensing, Department of Electrical & Computer Engineering, University of Minnesota, Minneapolis, MN 55455, USA ; Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Moses Rodriguez
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA ; Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| |
Collapse
|
29
|
Abstract
We review the current state of knowledge of remyelination in multiple sclerosis (MS), concentrating on advances in the understanding of the pathology and the regenerative response, and we summarise progress on the development of new therapies to enhance remyelination aimed at reducing progressive accumulation of disability in MS. We discuss key target pathways identified in experimental models, as although most identified targets have not yet progressed to the stage of being tested in human clinical trials, they may provide treatment strategies for demyelinating diseases in the future. Finally, we discuss some of the problems associated with testing this class of drugs, where they might fit into the therapeutic arsenal and the gaps in our knowledge.
Collapse
Affiliation(s)
- E. Jolanda Münzel
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh Bioquarter, 5 Little France Drive, Edinburgh, EH16 4UU UK
| | - Anna Williams
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh Bioquarter, 5 Little France Drive, Edinburgh, EH16 4UU UK
| |
Collapse
|
30
|
Joshi A, Mahfooz S, Maurya VK, Kumar V, Basanna CS, Kaur G, Hanif K, Jha RK. PARP1 during embryo implantation and its upregulation by oestradiol in mice. Reproduction 2014; 147:765-80. [DOI: 10.1530/rep-13-0588] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Pregnancy requires successful implantation of an embryo, which occurs during a restricted period defined as ‘receptivity of the endometrium’ and is influenced by the ovarian steroids progesterone and oestradiol. The role of poly(ADP-ribose)polymerase-1 (PARP1) in apoptosis is well established. However, it is also involved in cell differentiation, proliferation and tissue remodelling. Previous studies have described the presence of PARP in the uterus, but its exact role in embryo implantation is not yet elucidated. Hence, in this study, we studied the expression of PARP1 in the uterus during embryo implantation and decidualisation, and its regulation by ovarian steroids. Our results show upregulation of the native form of PARP1 (∼116 kDa) in the cytosolic and nuclear compartments of implantation and non-implantation sites at day 5 (0500 h), followed by downregulation at day 5 (1000 h), during the embryo implantation period. The transcript level of Parp1 was also augmented during day 5 (0500 h). Inhibition of PARP1 activity by the drug EB-47 decreased the number of embryo implantation sites and blastocysts at day 5 (1000 h). Further, cleavage of native PARP1 was due to the activity of caspase-3 during the peri-implantation stage (day 5 (0500 h)), and is also required for embryo implantation, as inhibition of its activity compromised blastocyst implantation. The native (∼116 kDa) and cleaved (∼89 kDa) forms of PARP1 were both elevated during decidualisation of the uterus. Furthermore, the expression level of PARP1 in the uterus was found to be under the control of the hormone oestrogen. Our results clearly demonstrate that PARP1 participates in the process of embryo implantation.
Collapse
|
31
|
Watzlawik JO, Wootla B, Painter MM, Warrington AE, Rodriguez M. Cellular targets and mechanistic strategies of remyelination-promoting IgMs as part of the naturally occurring autoantibody repertoire. Expert Rev Neurother 2014; 13:1017-29. [PMID: 24053345 DOI: 10.1586/14737175.2013.835601] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Immunoglobulins with germline sequences occur in invertebrates and vertebrates and are named naturally occurring autoantibodies (NAbs). NAbs may target foreign antigens, self- or altered self-components and are part of the normal immunoglobulin repertoire. Accumulating evidence indicates that naturally occurring antibodies can act as systemic surveillance molecules, which tag, damaged or stressed cells, invading pathogens and toxic cellular debris for elimination by the immune system. In addition to acting as detecting molecules, certain types of NAbs actively signal in different cell types with a broad range of responses from induction of apoptosis in cancer cells to stimulation of remyelination in glial cells. This review emphasizes functions and characteristics of NAbs with focus on remyelination-promoting mouse and human antibodies. Human remyelination-promoting NAbs are potential therapeutics to combat a wide spectrum of disease processes including demyelinating diseases like multiple sclerosis. We will highlight the identified glycosphingolipid (SL) antigens of polyreactive remyelination-promoting antibodies and their proposed mechanism(s) of action. The nature of the identified antigens suggests a lipid raft-based mechanism for remyelination-promoting antibodies with SLs as most essential raft components. However, accumulating evidence also suggests involvement of other antigens in stimulation of remyelination, which will be discussed in the text.
Collapse
Affiliation(s)
- Jens O Watzlawik
- Departments of Neurology and Immunology, Mayo Clinic, College of Medicine, 200 First Street, S.W., Rochester, MN 55905, USA
| | | | | | | | | |
Collapse
|
32
|
Baron W, Bijlard M, Nomden A, de Jonge JC, Teunissen CE, Hoekstra D. Sulfatide-mediated control of extracellular matrix-dependent oligodendrocyte maturation. Glia 2014; 62:927-42. [DOI: 10.1002/glia.22650] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Revised: 01/08/2014] [Accepted: 02/05/2014] [Indexed: 01/16/2023]
Affiliation(s)
- Wia Baron
- Department of Cell Biology; University of Groningen, University Medical Center Groningen; Groningen The Netherlands
| | - Marjolein Bijlard
- Department of Cell Biology; University of Groningen, University Medical Center Groningen; Groningen The Netherlands
| | - Anita Nomden
- Department of Cell Biology; University of Groningen, University Medical Center Groningen; Groningen The Netherlands
| | - Jenny C. de Jonge
- Department of Cell Biology; University of Groningen, University Medical Center Groningen; Groningen The Netherlands
| | - Charlotte E. Teunissen
- Neurochemistry Laboratory and Biobank; Department of Clinical Chemistry; Neuroscience Campus Amsterdam; VU University Medical Center Amsterdam; Amsterdam The Netherlands
| | - Dick Hoekstra
- Department of Cell Biology; University of Groningen, University Medical Center Groningen; Groningen The Netherlands
| |
Collapse
|
33
|
Role of galactosylceramide and sulfatide in oligodendrocytes and CNS myelin: formation of a glycosynapse. ADVANCES IN NEUROBIOLOGY 2014; 9:263-91. [PMID: 25151383 DOI: 10.1007/978-1-4939-1154-7_12] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The two major glycosphingolipids of myelin, galactosylceramide (GalC) and sulfatide (SGC), interact with each other by trans carbohydrate-carbohydrate interactions in vitro. They face each other in the apposed extracellular surfaces of the multilayered myelin sheath produced by oligodendrocytes and could also contact each other between apposed oligodendrocyte processes. Multivalent galactose and sulfated galactose, in the form of GalC/SGC-containing liposomes or silica nanoparticles conjugated to galactose and galactose-3-sulfate, interact with GalC and SGC in the membrane sheets of oligodendrocytes in culture. This interaction causes transmembrane signaling, loss of the cytoskeleton and clustering of membrane domains, similar to the effects of cross-linking by anti-GalC and anti-SGC antibodies. These effects suggest that GalC and SGC could participate in glycosynapses, similar to neural synapses or the immunological synapse, between GSL-enriched membrane domains in apposed oligodendrocyte membranes or extracellular surfaces of mature myelin. Formation of such glycosynapses in vivo would be important for myelination and/or oligodendrocyte/myelin function.
Collapse
|
34
|
Abstract
Immunoglobulins (Ig) or antibodies are heavy plasma proteins, with sugar chains added to amino-acid residues by N-linked glycosylation and occasionally by O-linked glycosylation. The versatility of antibodies is demonstrated by the various functions that they mediate such as neutralization, agglutination, fixation with activation of complement and activation of effector cells. Naturally occurring antibodies protect the organism against harmful pathogens, viruses and infections. In addition, almost any organic chemical induces antibody production of antibodies that would bind specifically to the chemical. These antibodies are often produced from multiple B cell clones and referred to as polyclonal antibodies. In recent years, scientists have exploited the highly evolved machinery of the immune system to produce structurally and functionally complex molecules such as antibodies from a single B clone, heralding the era of monoclonal antibodies. Most of the antibodies currently in the clinic, target components of the immune system, are not curative and seek to alleviate symptoms rather than cure disease. Our group used a novel strategy to identify reparative human monoclonal antibodies distinct from conventional antibodies. In this chapter, we discuss the therapeutic relevance of both polyclonal and monoclonal antibodies in clinic.
Collapse
Affiliation(s)
- Bharath Wootla
- Departments of Neurology and Immunology, Mayo Clinic, Rochester, MN, USA
| | | | | |
Collapse
|
35
|
Novel immunomodulatory approaches for the management of multiple sclerosis. Clin Pharmacol Ther 2013; 95:32-44. [PMID: 24173041 DOI: 10.1038/clpt.2013.196] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 09/13/2013] [Indexed: 11/09/2022]
Abstract
We provide a focused review of novel immunomodulatory approaches for the treatment of multiple sclerosis, the most common acquired inflammatory demyelinating disease of humans. The requirement for such a review was stimulated by the emerging application of novel oral medications and the need for the practicing physician to place these within the treatment paradigm. We provide a conceptual diagram of our current view of the pathogenesis of demyelination and remyelination in this disorder. In addition, we include a working template on how to use a tier 1 and tier 2 approach to medications as the disease worsens in the individual. We emphasize the approach of treatment based on "individualized medicine," tailored to the specific needs of each patient. In the future, we envision new drugs to enhance remyelination and protect neurons and axons from death in order to promote central nervous system regeneration and repair.
Collapse
|
36
|
Rodgers JM, Robinson AP, Miller SD. Strategies for protecting oligodendrocytes and enhancing remyelination in multiple sclerosis. DISCOVERY MEDICINE 2013; 16:53-63. [PMID: 23911232 PMCID: PMC3970909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system (CNS) characterized by encephalitogenic leukocyte infiltration and multifocal plaques of demyelination. Patients present with debilitating clinical sequelae including motor, sensory, and cognitive deficits. For the past 30 years, immune modulating treatments have entered the marketplace and continue to improve in limiting the frequency and severity of relapses, but no cure has been found and no drug has successfully stopped chronic progressive disease. Recent work focusing on the oligodendrocyte, the myelin-producing cell, has provided needed insight into the process of demyelination, the spontaneous ability of the CNS to regenerate, and the inevitable failure of remyelination. From this a number of promising molecular targets have been identified to protect oligodendrocytes and promote remyelination. Combining immunomodulatory therapy with strategies to protect oligodendrocytes from further degeneration and enhance remyelination presents a very real means to improve clinical outcome for chronic progressive patients in the near future. Here we lay out a combination therapy approach to treating MS and survey the current literature on promising drug candidates potentially capable of mediating oligodendrocyte protection and enhancing remyelination.
Collapse
Affiliation(s)
- Jane M Rodgers
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Northwestern University Feinberg School of Medicine, 303 E. Chicago Ave., Chicago, Illinois 60611, USA
| | | | | |
Collapse
|
37
|
Wootla B, Watzlawik JO, Denic A, Rodriguez M. The road to remyelination in demyelinating diseases: current status and prospects for clinical treatment. Expert Rev Clin Immunol 2013; 9:535-49. [PMID: 23730884 DOI: 10.1586/eci.13.37] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Within CNS disorders, demyelinating diseases are among the most devastating and cost intensive due to long-term disabilities affecting relatively young patients. Multiple sclerosis, a chronic inflammatory demyelinating disease in which the persistent inhibitory microenvironment of the resident oligodendrocyte precursor cells abrogates regeneration of myelin sheaths, is the most prominent disease in the spectrum of demyelinating diseases. The essential goal is to stimulate creation of new myelin sheaths on the demyelinated axons, leading to restoration of saltatory conduction and resolving functional deficits. The past few decades witnessed significant efforts to understand the cellular interactions at the lesion site with studies suggesting efficient remyelination as a prerequisite for functional repair. Despite its proven efficacy in experimental models, immunosuppression has not had profound clinical consequences in multiple sclerosis, which argued for a paradigm shift in the design of therapeutics aiming to achieve remyelination. For example, targeting oligodendrocytes themselves may drive remyelination in the CNS. This group and others have demonstrated that natural autoreactive antibodies directed at oligodendrocyte progenitors participate in remyelination. Accordingly, the authors developed a recombinant autoreactive natural human IgM antibody with therapeutic potential for remyelination.
Collapse
Affiliation(s)
- Bharath Wootla
- Department of Neurology, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905, USA.
| | | | | | | |
Collapse
|
38
|
Watzlawik JO, Warrington AE, Rodriguez M. PDGF is required for remyelination-promoting IgM stimulation of oligodendrocyte progenitor cell proliferation. PLoS One 2013; 8:e55149. [PMID: 23383310 PMCID: PMC3562326 DOI: 10.1371/journal.pone.0055149] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 12/27/2012] [Indexed: 12/13/2022] Open
Abstract
Background Promotion of remyelination is a major goal in treating demyelinating diseases such as multiple sclerosis (MS). The recombinant human monoclonal IgM, rHIgM22, targets myelin and oligodendrocytes (OLs) and promotes remyelination in animal models of MS. It is unclear whether rHIgM22-mediated stimulation of lesion repair is due to promotion of oligodendrocyte progenitor cell (OPC) proliferation and survival, OPC differentiation into myelinating OLs or protection of mature OLs. It is also unknown whether astrocytes or microglia play a functional role in IgM-mediated lesion repair. Methods We assessed the effect of rHIgM22 on cell proliferation in mixed CNS glial and OPC cultures by tritiated-thymidine uptake and by double-label immunocytochemistry using the proliferation marker, Ki-67. Antibody-mediated signaling events, OPC differentiation and OPC survival were investigated and quantified by Western blots. Results rHIgM22 stimulates OPC proliferation in mixed glial cultures but not in purified OPCs. There is no proliferative response in astrocytes or microglia. rHIgM22 activates PDGFαR in OPCs in mixed glial cultures. Blocking PDGFR-kinase inhibits rHIgM22-mediated OPC proliferation in mixed glia. We confirm in isolated OPCs that rHIgM22-mediated anti-apoptotic signaling and inhibition of OPC differentiation requires PDGF and FGF-2. We observed no IgM-mediated effect in mature OLs in the absence of PDGF and FGF-2. Conclusion Stimulation of OPC proliferation by rHIgM22 depends on co-stimulatory astrocytic and/or microglial factors. We demonstrate that rHIgM22-mediated activation of PDGFαR is required for stimulation of OPC proliferation. We propose that rHIgM22 lowers the PDGF threshold required for OPC proliferation and protection, which can result in remyelination of CNS lesions.
Collapse
Affiliation(s)
- Jens O. Watzlawik
- Departments of Neurology and Immunology, Mayo Clinic College of Medicine, Rochester, Minnesota, United States of America
| | - Arthur E. Warrington
- Departments of Neurology and Immunology, Mayo Clinic College of Medicine, Rochester, Minnesota, United States of America
| | - Moses Rodriguez
- Departments of Neurology and Immunology, Mayo Clinic College of Medicine, Rochester, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
39
|
Xu X, Wittenberg NJ, Jordan LR, Kumar S, Watzlawik JO, Warrington AE, Oh SH, Rodriguez M. A patterned recombinant human IgM guides neurite outgrowth of CNS neurons. Sci Rep 2013; 3:2267. [PMID: 23881231 PMCID: PMC3721078 DOI: 10.1038/srep02267] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 06/26/2013] [Indexed: 11/13/2022] Open
Abstract
Matrix molecules convey biochemical and physical guiding signals to neurons in the central nervous system (CNS) and shape the trajectory of neuronal fibers that constitute neural networks. We have developed recombinant human IgMs that bind to epitopes on neural cells, with the aim of treating neurological diseases. Here we test the hypothesis that recombinant human IgMs (rHIgM) can guide neurite outgrowth of CNS neurons. Microcontact printing was employed to pattern rHIgM12 and rHIgM22, antibodies that were bioengineered to have variable regions capable of binding to neurons or oligodendrocytes, respectively. rHIgM12 promoted neuronal attachment and guided outgrowth of neurites from hippocampal neurons. Processes from spinal neurons followed grid patterns of rHIgM12 and formed a physical network. Comparison between rHIgM12 and rHIgM22 suggested the biochemistry that facilitates anchoring the neuronal surfaces is a prerequisite for the function of IgM, and spatial properties cooperate in guiding the assembly of neuronal networks.
Collapse
Affiliation(s)
- Xiaohua Xu
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, MN 55905 USA
- These authors contributed equally to this work
| | - Nathan J. Wittenberg
- Laboratory of Nanostructures and Biosensing, Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, MN 55455 USA
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455 USA
- These authors contributed equally to this work
| | - Luke R. Jordan
- Laboratory of Nanostructures and Biosensing, Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, MN 55455 USA
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455 USA
| | - Shailabh Kumar
- Laboratory of Nanostructures and Biosensing, Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, MN 55455 USA
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455 USA
| | - Jens O. Watzlawik
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, MN 55905 USA
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN 55905 USA
| | - Arthur E. Warrington
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, MN 55905 USA
| | - Sang-Hyun Oh
- Laboratory of Nanostructures and Biosensing, Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, MN 55455 USA
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455 USA
| | - Moses Rodriguez
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, MN 55905 USA
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN 55905 USA
| |
Collapse
|
40
|
Warrington AE, Van Keulen V, Pease LR, Rodriguez M. Naturally occurring antibodies as therapeutics for neurologic disease: can human monoclonal IgMs replace the limited resource IVIG? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 750:44-55. [PMID: 22903665 DOI: 10.1007/978-1-4614-3461-0_4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Naturally occurring autoantibodies (NAbs) are common in normal humans. The majority of NAbs are IgMs, but a small proportion are IgGs. Therefore a certain portion of pooled whole human IgG (IVIG) can be considered NAbs. While the applications of IVIG to modulate human disease have increased dramatically, the use of IgMs as drugs has lagged. In fact, much of the contaminating IgM component of IVIG is disposed of as waste. However, a number of model studies, including those targeting Alzheimer and multiple sclerosis (MS) suggest that IgMs may better modulate disease at much lower doses than IVIG. Our own studies in a model of MS show that polyclonal human IgM promotes better remyelination than IVIG and that monoclonal IgMs promote greater remyelination than monoclonal IgGs containing identical variable region sequences. We propose that this difference is due to the ability of IgM to cross link cell surface antigens better than IgGs and induce signals in nervous system cells. Monoclonal antibodies (mAbs) that promote remyelination induce a transient Ca(2+) influx in myelin forming cells, whereas IgGs with identical variable sequences do not. MAbs that promote remyelination were identified in human serum and in EBV-immortalized human B-cell lines obtained from normal adults, fetal cord blood, and rheumatoid arthritis and MS patients. Therefore therapeutic mAbs are present and common in normal circulation. All therapeutic mAbs were IgMs and bound to nervous system cells, however, the tissue binding patterns suggest that binding any one of multiple antigens induces repair. An expression vector was constructed that can manufacture gram quantities of recombinant monoclonal human IgM. Therefore the technology exists to determine whether human monoclonal NAbs can modulate human disease. IVIG can modulate neurologic disease, but using IVIG to treat these chronic diseases is unsustainable. A long-term solution is to identify the functional component of IVIG and test whether a recombinant human monoclonal can replicate its efficacy.
Collapse
Affiliation(s)
- Arthur E Warrington
- Department of Neurology and Immunology, Mayo Clinic, Rochester, Minnesota, USA.
| | | | | | | |
Collapse
|
41
|
Colognato H, Tzvetanova ID. Glia unglued: how signals from the extracellular matrix regulate the development of myelinating glia. Dev Neurobiol 2012; 71:924-55. [PMID: 21834081 DOI: 10.1002/dneu.20966] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The health and function of the nervous system relies on glial cells that ensheath neuronal axons with a specialized plasma membrane termed myelin. The molecular mechanisms by which glial cells target and enwrap axons with myelin are only beginning to be elucidated, yet several studies have implicated extracellular matrix proteins and their receptors as being important extrinsic regulators. This review provides an overview of the extracellular matrix proteins and their receptors that regulate multiple steps in the cellular development of Schwann cells and oligodendrocytes, the myelinating glia of the PNS and CNS, respectively, as well as in the construction and maintenance of the myelin sheath itself. The first part describes the relevant cellular events that are influenced by particular extracellular matrix proteins and receptors, including laminins, collagens, integrins, and dystroglycan. The second part describes the signaling pathways and effector molecules that have been demonstrated to be downstream of Schwann cell and oligodendroglial extracellular matrix receptors, including FAK, small Rho GTPases, ILK, and the PI3K/Akt pathway, and the roles that have been ascribed to these signaling mediators. Throughout, we emphasize the concept of extracellular matrix proteins as environmental sensors that act to integrate, or match, cellular responses, in particular to those downstream of growth factors, to appropriate matrix attachment.
Collapse
Affiliation(s)
- Holly Colognato
- Department of Pharmacology, Stony Brook University, Stony Brook, New York 11794, USA.
| | | |
Collapse
|
42
|
Wootla B, Denic A, Warrington AE, Rodriguez M. Need for a paradigm shift in therapeutic approaches to CNS injury. Expert Rev Neurother 2012; 12:409-20. [PMID: 22449213 DOI: 10.1586/ern.12.24] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Irreversible damage to the nervous system can result from many causes including trauma, disruption of blood supply, pathogen infection or neurodegenerative disease. Common features following CNS injury include a disruption of axons, neuron death and injury, local B-cell and microglial activation, and the synthesis of pathogenic autoantibodies. CNS injury results in a pervasive inhibitory microenvironment that hinders regeneration. Current approaches to eliminate the inhibitory environment have met with limited success. These results argue for a paradigm shift in therapeutic approaches to CNS injury. Targeting CNS cells (neurons, oligodendrocytes and astrocytes) themselves may drive CNS repair. For example, our group and others have demonstrated that autoreactive antibodies can participate in aspects of CNS regeneration, including remyelination. We have developed recombinant autoreactive natural human IgM antibodies with the therapeutic potential for CNS repair in several neurologic diseases.
Collapse
Affiliation(s)
- Bharath Wootla
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | | | | | | |
Collapse
|
43
|
Kaveri SV, Silverman GJ, Bayry J. Natural IgM in immune equilibrium and harnessing their therapeutic potential. THE JOURNAL OF IMMUNOLOGY 2012; 188:939-45. [PMID: 22262757 DOI: 10.4049/jimmunol.1102107] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Natural IgM Abs are the constitutively secreted products of B1 cells (CD5(+) in mice and CD20(+)CD27(+)CD43(+)CD70(-) in humans) that have important and diverse roles in health and disease. Whereas the role of natural IgM as the first line of defense for protection against invading microbes has been extensively investigated, more recent reports have highlighted their potential roles in the maintenance of tissue homeostasis via clearance of apoptotic and altered cells through complement-dependent mechanisms, inhibition of inflammation, removal of misfolded proteins, and regulation of pathogenic autoreactive IgG Abs and autoantibody-producing B cells. These observations have provided the theoretical underpinnings for efforts that currently seek to harness the untapped therapeutic potential of natural IgM either by boosting in vivo natural IgM production or via therapeutic infusions of monoclonal and polyclonal IgM preparations.
Collapse
|
44
|
Robak T, Robak E. Tyrosine kinase inhibitors as potential drugs for B-cell lymphoid malignancies and autoimmune disorders. Expert Opin Investig Drugs 2012; 21:921-47. [PMID: 22612424 DOI: 10.1517/13543784.2012.685650] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION In the last few years, several tyrosine kinase inhibitors (TKIs) have been synthesized and become available for preclinical studies and clinical trials. This article summarizes recent achievements in the mechanism of action, pharmacological properties, and clinical activity and toxicity, as well as the emerging role of TKIs in lymphoid malignancies, allergic diseases, and autoimmune disorders. AREAS COVERED A literature review was conducted of the MEDLINE database PubMed for articles in English. Publications from 2000 through January 2012 were scrutinized. The search terms used were Bruton's tyrosine kinase (Btk) inhibitors, PCI-32765, GDC-0834, LFM-A13, AVL-101, AVL-292, spleen tyrosine kinase (Syk) inhibitors, R343, R406, R112, R788, fostamatinib, BAY-61-3606, C-61, piceatannol, Lyn, imatinib, nilotinib, bafetinib, dasatinib, GDC-0834, PP2, SU6656 in conjunction with lymphoid malignancy, NHL, CLL, autoimmune disease, allergic disease, asthma, and rheumatoid arthritis. Conference proceedings from the previous 5 years of the American Society of Hematology, European Hematology Association, American Society of Clinical Oncology, and ACR/ARHP Annual Scientific Meetings were searched manually. Additional relevant publications were obtained by reviewing the references from the chosen articles. EXPERT OPINION The use of TKIs, especially inhibitors of Btk, Syk, and Lyn, is a promising new strategy for targeted treatment of B-cell lymphoid malignancies, autoimmune disorders and allergic diseases. However, definitive data from ongoing and future clinical trials will aid in better defining the status of TKIs in the treatment of these disorders.
Collapse
Affiliation(s)
- Tadeusz Robak
- Medical University of Lodz, Department of Hematology, Lodz, Poland.
| | | |
Collapse
|
45
|
Is multiple sclerosis an autoimmune disease? Autoimmune Dis 2012; 2012:969657. [PMID: 22666554 PMCID: PMC3361990 DOI: 10.1155/2012/969657] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Revised: 03/05/2012] [Accepted: 03/15/2012] [Indexed: 01/26/2023] Open
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS) with varied clinical presentations and heterogeneous histopathological features. The underlying immunological abnormalities in MS lead to various neurological and autoimmune manifestations. There is strong evidence that MS is, at least in part, an immune-mediated disease. There is less evidence that MS is a classical autoimmune disease, even though many authors state this in the description of the disease. We show the evidence that both supports and refutes the autoimmune hypothesis. In addition, we present an alternate hypothesis based on virus infection to explain the pathogenesis of MS.
Collapse
|
46
|
Evidence for the role of B cells and immunoglobulins in the pathogenesis of multiple sclerosis. Neurol Res Int 2011; 2011:780712. [PMID: 21961063 PMCID: PMC3179868 DOI: 10.1155/2011/780712] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Accepted: 07/27/2011] [Indexed: 01/06/2023] Open
Abstract
The pathogenesis of multiple sclerosis (MS) remains elusive. Recent reports advocate greater involvement of B cells and immunoglobulins in the initiation and propagation of MS lesions at different stages of their ontogeny. The key role of B cells and immunoglobulins in pathogenesis was initially identified by studies in which patients whose fulminant attacks of demyelination did not respond to steroids experienced remarkable functional improvement following plasma exchange. The positive response to Rituximab in Phase II clinical trials of relapsing-remitting MS confirms the role of B cells. The critical question is how B cells contribute to MS. In this paper, we discuss both the deleterious and the beneficial roles of B cells and immunoglobulins in MS lesions. We provide alternative hypotheses to explain both damaging and protective antibody responses.
Collapse
|
47
|
Xu X, Warrington AE, Wright BR, Bieber AJ, Van Keulen V, Pease LR, Rodriguez M. A human IgM signals axon outgrowth: coupling lipid raft to microtubules. J Neurochem 2011; 119:100-12. [PMID: 21824142 DOI: 10.1111/j.1471-4159.2011.07416.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Mouse and human IgMs support neurite extension from primary cerebellar granule neurons. In this study using primary hippocampal and cortical neurons, we demonstrate that a recombinant human IgM, rHIgM12, promotes axon outgrowth by coupling membrane domains (lipid rafts) to microtubules. rHIgM12 binds to the surface of neuron and induces clustering of cholesterol and ganglioside GM1. After cell binding and membrane fractionation, rHIgM12 gets segregated into two pools, one associated with lipid raft fractions and the other with the detergent-insoluble cytoskeleton-containing pellet. Membrane-bound rHIgM12 co-localized with microtubules and co-immuno precipitated with β3-tubulin. rHIgM12-membrane interaction also enhanced the tyrosination of α-tubulin indicating a stabilization of new neurites. When presented as a substrate, rHIgM12 induced axon outgrowth from primary neurons. We now demonstrate that a recombinant human mAb can induce signals in neurons that regulate membrane lipids and microtubule dynamics required for axon extension. We propose that the pentameric structure of the IgM is critical to cross-link membrane lipids and proteins resulting in signaling cascades.
Collapse
Affiliation(s)
- Xiaohua Xu
- Department of Neurology, Mayo Clinic and Foundation, Rochester, MN 55905, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Xu X, Warrington AE, Bieber AJ, Rodriguez M. Enhancing CNS repair in neurological disease: challenges arising from neurodegeneration and rewiring of the network. CNS Drugs 2011; 25:555-73. [PMID: 21699269 PMCID: PMC3140701 DOI: 10.2165/11587830-000000000-00000] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Repair of the central nervous system (CNS) constitutes an integral part of treating neurological disease and plays a crucial role in restoring CNS architecture and function. Distinct strategies have been developed to reconstruct the damaged neural tissue, with many tested preclinically in animal models. We review cell replacement-based repair strategies. By taking spinal cord injury, cerebral ischaemia and degenerative CNS disorders as examples for CNS repair, we discuss progress and potential problems in utilizing embryonic stem cells and adult neural/non-neural stem cells to repair cell loss in the CNS. Nevertheless, CNS repair is not simply a matter of cell transplantation. The major challenge is to induce regenerating neural cells to integrate into the neural network and compensate for damaged neural function. The neural cells confront an environment very different from that of the developmental stage in which these cells differentiate to form interwoven networks. During the repair process, one of the challenges is neurodegeneration, which can develop from interrupted innervations to/from the targets, chronic inflammation, ischaemia, aging or idiopathic neural toxicity. Neurodegeneration, which occurs on the basis of a characteristic vascular and neural web, usually presents as a chronically progressive process with unknown aetiology. Currently, there is no effective treatment to stop or slow down neurodegeneration. Pathological changes from patients with Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis indicate a broken homeostasis in the CNS. We discuss how the blood-brain barrier and neural networks are formed to maintain CNS homeostasis and their contribution to neurodegeneration in diseased conditions. Another challenge is that some inhibitors produced by CNS injury do not facilitate the regenerating neural cells to incorporate into a pre-existing network. We review glial responses to CNS injury. Of note, the reactive astrocytes not only encompass the lesions/pathogens but may also form glial scars to impede regenerating axons from traversing the lesions. In addition, myelin debris can prevent axon growth. Myelination enables saltatory transduction of electrical impulses along axonal calibers and actually provides trophic support to stabilize the axons. Therefore, repair strategies should be designed to promote axonal growth, myelination and modulate astrocytic responses. Finally, we discuss recent progress in developing human monoclonal IgMs that regulate CNS homeostasis and promote neural regeneration.
Collapse
Affiliation(s)
- Xiaohua Xu
- Department of Neurology, Mayo Clinic and Foundation, Rochester, MN 55905
| | | | - Allan J. Bieber
- Department of Neurology, Mayo Clinic and Foundation, Rochester, MN 55905
| | - Moses Rodriguez
- Department of Neurology, Mayo Clinic and Foundation, Rochester, MN 55905, Department of Immunology, Mayo Clinic and Foundation, Rochester, MN 55905
| |
Collapse
|