Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Cell Rep Med ; 3(11): 100820, 2022 11 15.
Article in English | MEDLINE | ID: mdl-36384103

ABSTRACT

Ideal therapies for regenerative medicine or healthy aging require healthy organ growth and rejuvenation, but no organ-level approach is currently available. Using Mycobacterium leprae (ML) with natural partial cellular reprogramming capacity and its animal host nine-banded armadillos, we present an evolutionarily refined model of adult liver growth and regeneration. In infected armadillos, ML reprogram the entire liver and significantly increase total liver/body weight ratio by increasing healthy liver lobules, including hepatocyte proliferation and proportionate expansion of vasculature, and biliary systems. ML-infected livers are microarchitecturally and functionally normal without damage, fibrosis, or tumorigenesis. Bacteria-induced reprogramming reactivates liver progenitor/developmental/fetal genes and upregulates growth-, metabolism-, and anti-aging-associated markers with minimal change in senescence and tumorigenic genes, suggesting bacterial hijacking of homeostatic, regeneration pathways to promote de novo organogenesis. This may facilitate the unraveling of endogenous pathways that effectively and safely re-engage liver organ growth, with broad therapeutic implications including organ regeneration and rejuvenation.


Subject(s)
Armadillos , Cellular Reprogramming , Animals , Liver/metabolism , Carcinogenesis/metabolism , Fibrosis , Bacteria
2.
Microbiol Spectr ; 7(4)2019 07.
Article in English | MEDLINE | ID: mdl-31322104

ABSTRACT

The mammalian nervous system is invaded by a number of intracellular bacterial pathogens which can establish and progress infection in susceptible individuals. Subsequent clinical manifestation is apparent with the impairment of the functional units of the nervous system, i.e., the neurons and the supporting glial cells that produce myelin sheaths around axons and provide trophic support to axons and neurons. Most of these neurotrophic bacteria display unique features, have coevolved with the functional sophistication of the nervous system cells, and have adapted remarkably to manipulate neural cell functions for their own advantage. Understanding how these bacterial pathogens establish intracellular adaptation by hijacking endogenous pathways in the nervous system, initiating myelin damage and axonal degeneration, and interfering with myelin maintenance provides new knowledge not only for developing strategies to combat neurodegenerative conditions induced by these pathogens but also for gaining novel insights into cellular and molecular pathways that regulate nervous system functions. Since the pathways hijacked by bacterial pathogens may also be associated with other neurodegenerative diseases, it is anticipated that detailing the mechanisms of bacterial manipulation of neural systems may shed light on common mechanisms, particularly of early disease events. This chapter details a classic example of neurodegeneration, that caused by Mycobacterium leprae, which primarily infects glial cells of the peripheral nervous system (Schwann cells), and how it targets and adapts intracellularly by reprogramming Schwann cells to stem cells/progenitor cells. We also discuss implications of this host cell reprogramming by leprosy bacilli as a model in a wider context.


Subject(s)
Leprosy/microbiology , Mycobacterium leprae/physiology , Peripheral Nervous System/microbiology , Adaptation, Physiological , Animals , Humans , Mycobacterium leprae/genetics , Mycobacterium leprae/isolation & purification , Schwann Cells/microbiology
3.
Article in English | MEDLINE | ID: mdl-30533927

ABSTRACT

The draft genome sequences of three strains of Lawsonia intracellularis, an obligate intracellular animal pathogen responsible for causing proliferative enteropathy, obtained from swine in different prefectures in Japan revealed the absence of a genomic island previously reported to be linked to host adaptation and to high genomic diversity, despite geographical proximity.

4.
Curr Opin Microbiol ; 23: 179-88, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25541240

ABSTRACT

Bacterial pathogens employ a myriad of strategies to alter host tissue cell functions for bacterial advantage during infection. Recent advances revealed a fusion of infection biology with stem cell biology by demonstrating developmental reprogramming of lineage committed host glial cells to progenitor/stem cell-like cells by an intracellular bacterial pathogen Mycobacterium leprae. Acquisition of migratory and immunomodulatory properties of such reprogrammed cells provides an added advantage for promoting bacterial spread. This presents a previously unseen sophistication of cell manipulation by hijacking the genomic plasticity of host cells by a human bacterial pathogen. The rationale for such extreme fate conversion of host cells may be directly linked to the exceedingly passive obligate life style of M. leprae with a degraded genome and host cell dependence for both bacterial survival and dissemination, particularly the use of host-derived stem cell-like cells as a vehicle for spreading infection without being detected by immune cells. Thus, this unexpected link between cell reprogramming and infection opens up a new premise in host-pathogen interactions. Furthermore, such bacterial ingenuity could also be harnessed for developing natural ways of reprogramming host cells for repairing damaged tissues from infection, injury and diseases.


Subject(s)
Cellular Reprogramming , Host-Pathogen Interactions , Mycobacterium leprae/growth & development , Neuroglia/microbiology , Neuroglia/physiology , Stem Cells/microbiology , Stem Cells/physiology , Animals , Humans
5.
ILAR J ; 54(3): 304-14, 2014.
Article in English | MEDLINE | ID: mdl-24615444

ABSTRACT

Leprosy (also known as Hansen's Disease) is a chronic infectious disease caused by Mycobacterium leprae that primarily targets the peripheral nervous system; skin, muscle, and other tissues are also affected. Other than humans, nine-banded armadillos (Dasypus novemcinctus) are the only natural hosts of M. leprae, and they are the only laboratory animals that develop extensive neurological involvement with this bacterium. Infection in the armadillo closely recapitulates many of the structural, physiological, and functional aspects of leprosy seen in humans. Armadillos can be useful models of leprosy for basic scientific investigations into the pathogenesis of leprosy neuropathy and its associated myopathies, as well as for translational research studies in piloting new diagnostic methods or therapeutic interventions. Practical and ethical constraints often limit investigation into human neuropathies, but armadillos are an abundant source of leprotic neurologic fibers. Studies with these animals may provide new insights into the mechanisms involved in leprosy that also might benefit the understanding of other demyelinating neuropathies. Although there is only a limited supply of armadillo-specific reagents, the armadillo whole genomic sequence has been completed, and gene expression studies can be employed. Clinical procedures, such as electrophysiological nerve conduction testing, provide a functional assessment of armadillo nerves. A variety of standard histopathological and immunopathological procedures including Epidermal Nerve Fiber Density (ENFD) analysis, Schwann Cell Density, and analysis for other conserved cellular markers can be used effectively with armadillos and will be briefly reviewed in this text.


Subject(s)
Armadillos , Disease Models, Animal , Leprosy/complications , Peripheral Nervous System Diseases/etiology , Peripheral Nervous System Diseases/physiopathology , Animals , Cell Count , Electrophysiological Phenomena , Epidermis/innervation , Gene Expression Regulation/genetics , Humans , Leprosy/genetics , Schwann Cells/pathology
6.
Cell Reprogram ; 16(1): 9-17, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24279882

ABSTRACT

Recently, we showed a natural reprogramming process during infection with Mycobacterium leprae (ML), the causative organism of human leprosy. ML hijacks the notable plasticity of adult Schwann cells in the peripheral nervous system (PNS), bacteria's preferred nonimmune niche, to reprogram infected cells to progenitor/stem cell-like cells (pSLCs). Whereas ML appear to use this reprogramming process as a sophisticated bacterial strategy to spread infection to other tissues, understanding the mechanisms may shed new insights into the basic biology of cellular reprogramming and the development of new approaches for generating pSLC for therapeutic purposes as well as targeting bacterial infectious diseases at an early stage. Toward these goals, we extended our studies to identify other players that might be involved in this complex host cell reprogramming. Here we show that ML activates numerous immune-related genes mainly involved in innate immune responses and inflammation during early infection before downregulating Schwann cell lineage genes and reactivating developmental transcription factors. We validated these findings by demonstrating the ability of infected cells to secrete soluble immune factor proteins at early time points and their continued release during the course of reprogramming. By using time-lapse microscopy and a migration assay with reprogrammed Schwann cells (pSLCs) cultured with macrophages, we show that reprogrammed cells possess the ability to attract macrophages, providing evidence for a functional role of immune gene products during reprogramming. These findings suggest a potential role of innate immune response and the related signaling pathways in cellular reprogramming and the initiation of neuropathogenesis during ML infection.


Subject(s)
Cell Dedifferentiation/immunology , Down-Regulation/immunology , Immunity, Innate , Leprosy/immunology , Mycobacterium leprae/immunology , Schwann Cells/immunology , Animals , Humans , Inflammation/immunology , Inflammation/microbiology , Inflammation/pathology , Leprosy/pathology , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Inbred ICR , Schwann Cells/microbiology , Schwann Cells/pathology
7.
Cell ; 152(1-2): 51-67, 2013 Jan 17.
Article in English | MEDLINE | ID: mdl-23332746

ABSTRACT

Differentiated cells possess a remarkable genomic plasticity that can be manipulated to reverse or change developmental commitments. Here, we show that the leprosy bacterium hijacks this property to reprogram adult Schwann cells, its preferred host niche, to a stage of progenitor/stem-like cells (pSLC) of mesenchymal trait by downregulating Schwann cell lineage/differentiation-associated genes and upregulating genes mostly of mesoderm development. Reprogramming accompanies epigenetic changes and renders infected cells highly plastic, migratory, and immunomodulatory. We provide evidence that acquisition of these properties by pSLC promotes bacterial spread by two distinct mechanisms: direct differentiation to mesenchymal tissues, including skeletal and smooth muscles, and formation of granuloma-like structures and subsequent release of bacteria-laden macrophages. These findings support a model of host cell reprogramming in which a bacterial pathogen uses the plasticity of its cellular niche for promoting dissemination of infection and provide an unexpected link between cellular reprogramming and host-pathogen interaction.


Subject(s)
Host-Pathogen Interactions , Leprosy/microbiology , Leprosy/pathology , Mycobacterium leprae , Schwann Cells/pathology , Stem Cells/pathology , Animals , Cell Movement , Cell Survival , Epigenesis, Genetic , Epithelial-Mesenchymal Transition , Granuloma/microbiology , Humans , Leprosy/genetics , Macrophages/microbiology , Macrophages/pathology , Mice , Mice, Nude , Peripheral Nerves/pathology , Schwann Cells/microbiology
8.
F1000Res ; 2: 198, 2013.
Article in English | MEDLINE | ID: mdl-24358891

ABSTRACT

BACKGROUND: Bacterial pathogens can manipulate or subvert host tissue cells to their advantage at different stages during infection, from initial colonization in primary host niches to dissemination. Recently, we have shown that Mycobacterium leprae (ML), the causative agent of human leprosy, reprogrammed its preferred host niche de-differentiated adult Schwann cells to progenitor/stem cell-like cells (pSLC) which appear to facilitate bacterial spread. Here, we studied how this cell fate change influences bacterial retention and transfer properties of Schwann cells before and after reprogramming. RESULTS: Using primary fibroblasts as bacterial recipient cells, we showed that non-reprogrammed Schwann cells, which preserve all Schwann cell lineage and differentiation markers, possess high bacterial retention capacity when co-cultured with skin fibroblasts; Schwann cells failed to transfer bacteria to fibroblasts at higher numbers even after co-culture for 5 days. In contrast, pSLCs, which are derived from the same Schwann cells but have lost Schwann cell lineage markers due to reprogramming, efficiently transferred bacteria to fibroblasts within 24 hours. CONCLUSIONS: ML-induced reprogramming converts lineage-committed Schwann cells with high bacterial retention capacity to a cell type with pSLC stage with effective bacterial transfer properties. We propose that such changes in cellular properties may be associated with the initial intracellular colonization, which requires long-term bacterial retention within Schwann cells, in order to spread the infection to other tissues, which entails efficient bacterial transfer capacity to cells like fibroblasts which are abundant in many tissues, thereby potentially maximizing bacterial dissemination. These data also suggest how pathogens could take advantage of multiple facets of host cell reprogramming according to their needs during infection.

9.
PLoS One ; 6(6): e20985, 2011.
Article in English | MEDLINE | ID: mdl-21698192

ABSTRACT

Iron is an essential metal for living organisms but its level must be strictly controlled in cells, because ferrous ion induces toxicity by generating highly active reactive oxygen, hydroxyl radicals, through the Fenton reaction. In addition, ferric ion shows low solubility under physiological conditions. To overcome these obstacles living organisms possess Ferritin superfamily proteins that are distributed in all three domains of life: bacteria, archaea, and eukaryotes. These proteins minimize hydroxyl radical formation by ferroxidase activity that converts Fe(2+) into Fe(3+) and sequesters iron by storing it as a mineral inside a protein cage. In this study, we discovered that mycobacterial DNA-binding protein 1 (MDP1), a histone-like protein, has similar activity to ferritin superfamily proteins. MDP1 prevented the Fenton reaction and protects DNA by the ferroxidase activity. The K(m) values of the ferroxidase activity by MDP1 of Mycobacterium bovis bacillus Calmette-Guérin (BCG-3007c), Mycobacterium tuberculosis (Rv2986c), and Mycobacterium leprae (ML1683; ML-LBP) were 0.292, 0.252, and 0.129 mM, respectively. Furthermore, one MDP1 molecule directly captured 81.4±19.1 iron atoms, suggesting the role of this protein in iron storage. This study describes for the first time a ferroxidase-iron storage protein outside of the ferritin superfamily proteins and the protective role of this bacterial protein from DNA damage.


Subject(s)
DNA Damage , Ferritins/physiology , Histones/physiology , Mycobacterium/metabolism , Ceruloplasmin/metabolism , Mycobacterium/enzymology , Phylogeny , Protein Binding
10.
Prog Neurobiol ; 91(2): 102-7, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20005916

ABSTRACT

Multiple signaling pathways play key regulatory roles during the development of peripheral nervous system (PNS) and also in neuroregeneration process following nerve degeneration. Schwann cells, the glial cells of the PNS, by interacting with neuronal (axonal) ligands, mainly neuregulins via receptor tyrosine kinase (RTK) complex, ErbB2/ErbB3, initiate intracellular signaling pathways to drive proliferation and differentiation of Schwann cells, both during development and the process of regeneration and re-myelination after nerve injury. One of the major signaling kinases, extracellular signal-regulated kinase-1/2 (ERK1/2), that is also a downstream signaling pathway of neuregulin-ErbB2/ErbB3 activation, has been identified as a key regulator of Schwann cell proliferation, differentiation, demyelination and nerve regeneration. Recent studies have provided evidence that the bacterium that causes human leprosy, Mycobacterium leprae that has a unique capacity to invade Schwann cells of the adult PNS, utilizes the neuregulin-ErbB2/ErbB3 associated signaling network to the bacterial advantage. M. leprae directly bind to ErbB2 on myelinated Schwann cells and activate the RTK by a novel route that bypasses the classical neuregulin/growth factor-induced ErbB2-ErbB3 heterodimerization, and subsequently induce downstream the canonical Erk1/2 signaling, leading to myelin breakdown and subsequent axonal damage. This initial injury provides a survival advantage for M. leprae as it induces de-differentiation and generates myelin-free cells, which are highly susceptible to M. leprae invasion and promote bacterial survival. Once invaded M. leprae activate Erk1/2 via a non-canonical pathway and subsequently increase the cell proliferation and maintain the infected cells in de-differentiated state, thereby preventing remyelination. Therefore, by subverting major RTKs and signaling pathways in adult Schwann cells M. leprae appear to propagate the bacterial niche and maintain survival within the PNS. These studies may also provide new insights into our understanding of signaling mechanisms involve in both neurodegeneration and neuroregeneration.


Subject(s)
Mycobacterium leprae/physiology , Mycobacterium leprae/pathogenicity , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/microbiology , Peripheral Nerves/metabolism , Peripheral Nerves/microbiology , Regeneration , Signal Transduction , Adult , Animals , Humans , Leprosy/metabolism , Leprosy/microbiology , Models, Biological
11.
J Immunol ; 177(8): 5226-35, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-17015708

ABSTRACT

CD1d-restricted NKT cells expressing invariant TCR alpha-chains (iNKT cells) produce both proinflammatory and anti-inflammatory cytokines rapidly upon activation, and are believed to play an important role in both host defense and immunoregulation. To address the potential implications of iNKT cell responses for infectious or inflammatory diseases of the nervous system, we investigated the expression of CD1d in human peripheral nerve. We found that CD1d was expressed on the surface of Schwann cells in situ and on primary or immortalized Schwann cell lines in culture. Schwann cells activated iNKT cells in a CD1d-dependent manner in the presence of alpha-galactosylceramide. Surprisingly, the cytokine production of iNKT cells stimulated by alpha-galactosylceramide presented by CD1d+ Schwann cells showed a predominance of Th2-associated cytokines such as IL-5 and IL-13 with a marked deficiency of proinflammatory Th1 cytokines such as IFN-gamma or TNF-alpha. Our findings suggest a mechanism by which iNKT cells may restrain inflammatory responses in peripheral nerves, and raise the possibility that the expression of CD1d by Schwann cells could be relevant in the pathogenesis of infectious and inflammatory diseases of the peripheral nervous system.


Subject(s)
Antigens, CD1/analysis , Cell Communication/immunology , Killer Cells, Natural/immunology , Schwann Cells/immunology , Antigens, CD1d , Cells, Cultured , Coculture Techniques , Cytokines/analysis , Galactosylceramides/pharmacology , Humans , Immunity , Inflammation , Killer Cells, Natural/cytology , Nervous System Diseases/immunology , Nervous System Diseases/pathology , Schwann Cells/chemistry , Schwann Cells/cytology , T-Lymphocytes
12.
Nat Med ; 12(8): 961-6, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16892039

ABSTRACT

Demyelination is a common pathologic feature in many neurodegenerative diseases including infection with leprosy-causing Mycobacterium leprae. Because of the long incubation time and highly complex disease pathogenesis, the management of nerve damage in leprosy, as in other demyelinating diseases, is extremely difficult. Therefore, an important challenge in therapeutic interventions is to identify the molecular events that occur in the early phase before the progression of the disease. Here we provide evidence that M. leprae-induced demyelination is a result of direct bacterial ligation to and activation of ErbB2 receptor tyrosine kinase (RTK) signaling without ErbB2-ErbB3 heterodimerization, a previously unknown mechanism that bypasses the neuregulin-ErbB3-mediated ErbB2 phosphorylation. MEK-dependent Erk1 and Erk2 (hereafter referred to as Erk1/2) signaling is identified as a downstream target of M. leprae-induced ErbB2 activation that mediates demyelination. Herceptin (trastuzumab), a therapeutic humanized ErbB2-specific antibody, inhibits M. leprae binding to and activation of ErbB2 and Erk1/2 in human primary Schwann cells, and the blockade of ErbB2 activity by the small molecule dual ErbB1-ErbB2 kinase inhibitor PKI-166 (ref. 11) effectively abrogates M. leprae-induced myelin damage in in vitro and in vivo models. These results may have implications for the design of ErbB2 RTK-based therapies for both leprosy nerve damage and other demyelinating neurodegenerative diseases.


Subject(s)
Demyelinating Diseases/metabolism , Leprosy/metabolism , Mycobacterium leprae/metabolism , Receptor, ErbB-2/metabolism , Signal Transduction , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Butadienes/pharmacology , COS Cells , Cells, Cultured , Chlorocebus aethiops , Coculture Techniques , Demyelinating Diseases/pathology , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , HeLa Cells , Humans , Leprosy/microbiology , Mice , Mice, Knockout , Mice, Nude , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mycobacterium leprae/genetics , Nitriles/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Rats , Schwann Cells/enzymology , Schwann Cells/metabolism , Sciatic Nerve/metabolism , Sciatic Nerve/microbiology , Sciatic Nerve/ultrastructure , Trastuzumab
13.
s.l; s.n; 2006. 6 p. ilus, tab, graf.
Non-conventional in English | Sec. Est. Saúde SP, HANSEN, Hanseníase Leprosy, SESSP-ILSLACERVO, Sec. Est. Saúde SP | ID: biblio-1241807

ABSTRACT

Demyelination is a common pathologic feature in many neurodegenerative diseases including infection with leprosy-causing Mycobacterium leprae. Because of the long incubation time and highly complex disease pathogenesis, the management of nerve damage in leprosy, as in other demyelinating diseases, is extremely difficult. Therefore, an important challenge in therapeutic interventions is to identify the molecular events that occur in the early phase before the progression of the disease. Here we provide evidence that M. leprae-induced demyelination is a result of direct bacterial ligation to and activation of ErbB2 receptor tyrosine kinase (RTK) signaling without ErbB2-ErbB3 heterodimerization, a previously unknown mechanism that bypasses the neuregulin-ErbB3-mediated ErbB2 phosphorylation. MEK-dependent Erk1 and Erk2 (hereafter referred to as Erk1/2) signaling is identified as a downstream target of M. leprae-induced ErbB2 activation that mediates demyelination. Herceptin (trastuzumab), a therapeutic humanized ErbB2-specific antibody, inhibits M. leprae binding to and activation of ErbB2 and Erk1/2 in human primary Schwann cells, and the blockade of ErbB2 activity by the small molecule dual ErbB1-ErbB2 kinase inhibitor PKI-166 (ref. 11) effectively abrogates M. leprae-induced myelin damage in in vitro and in vivo models. These results may have implications for the design of ErbB2 RTK-based therapies for both leprosy nerve damage and other demyelinating neurodegenerative diseases.


Subject(s)
Rats , Mice , Humans , Animals , Antibodies, Monoclonal , Enzyme Activation , Butadienes , Mice, Knockout , Chlorocebus aethiops , COS Cells , Cells, Cultured , HeLa Cells , Schwann Cells , Demyelinating Diseases , Leprosy , Enzyme Inhibitors , Mycobacterium leprae , Sciatic Nerve , Nitriles , Pyrimidines , Pyrroles , Mitogen-Activated Protein Kinase 1 , Research Support, N.I.H., Extramural , Signal Transduction , Coculture Techniques
14.
Proc Natl Acad Sci U S A ; 102(26): 9188-93, 2005 Jun 28.
Article in English | MEDLINE | ID: mdl-15967991

ABSTRACT

Activation of extracellular signal-regulated kinase (Erk) 1/2, which plays a critical role in diverse cellular processes, including cell proliferation, is known to be mediated by the canonical Raf-mitogen-activated protein kinase kinase (MEK) kinase cascade. Alternative MEK-independent signaling pathways for Erk1/2 activation in mammalian cells are not known. During our studies of human primary Schwann cell response to long-term infection of Mycobacterium leprae, the causative organism of leprosy, we identified that intracellular M. leprae activated Erk1/2 directly by lymphoid cell kinase (p56Lck), a Src family member, by means of a PKCepsilon-dependent and MEK-independent signaling pathway. Activation of this signaling induced nuclear accumulation of cyclin D1, G1/S-phase progression, and continuous proliferation, but without transformation. Thus, our data reveal a previously unknown signaling mechanism of glial cell proliferation, which might play a role in dedifferentiation as well as nerve regeneration and degeneration. Our findings may also provide a potential mechanism by which an obligate intracellular bacterial pathogen like M. leprae subverts nervous system signaling to propagate its cellular niche for colonization and long-term bacterial survival.


Subject(s)
Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , MAP Kinase Kinase 1/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mycobacterium leprae/metabolism , Peripheral Nerves/metabolism , Schwann Cells/enzymology , Schwann Cells/metabolism , Blotting, Western , Bromodeoxyuridine/pharmacology , Cell Cycle , Cell Differentiation , Cell Nucleus/metabolism , Cell Proliferation , Cell Separation , Coloring Agents/pharmacology , Cyclin D1/metabolism , Enzyme Inhibitors/pharmacology , Flow Cytometry , G1 Phase , Humans , Leprosy/microbiology , Microscopy, Electron , Microscopy, Fluorescence , Models, Biological , Neuroglia/metabolism , Oligonucleotide Array Sequence Analysis , Phosphorylation , Protein Kinase C/metabolism , Protein Kinase C-epsilon , Reverse Transcriptase Polymerase Chain Reaction , S Phase , Signal Transduction , Time Factors , Transfection
15.
Curr Opin Immunol ; 16(4): 511-8, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15245748

ABSTRACT

The molecular events that occur at the early phase of many demyelinating neurodegenerative diseases are unknown. A recent demonstration of rapid demyelination and axonal injury induced by Mycobacterium leprae provides a model for elucidating the molecular events of early nerve degeneration which might be common to neurodegenerative diseases of both infectious origin and unknown etiology. The identification of the M. leprae-targeted Schwann cell receptor, dystroglycan, and its associated molecules in myelination, demyelination and axonal functions suggests a role for these molecules in early nerve degeneration.


Subject(s)
Demyelinating Autoimmune Diseases, CNS/immunology , Dystroglycans/immunology , Leprosy/immunology , Mycobacterium leprae/immunology , Myelin Sheath/immunology , Schwann Cells/immunology , Animals , Axons/immunology , Axons/pathology , Axons/ultrastructure , Demyelinating Autoimmune Diseases, CNS/etiology , Demyelinating Autoimmune Diseases, CNS/microbiology , Demyelinating Autoimmune Diseases, CNS/physiopathology , Dystrophin-Associated Protein Complex/immunology , Humans , Leprosy/complications , Mice , Schwann Cells/microbiology , Schwann Cells/pathology
16.
s.l; s.n; 2004. 8 p. ilus, tab.
Non-conventional in English | Sec. Est. Saúde SP, HANSEN, Hanseníase Leprosy, SESSP-ILSLACERVO, Sec. Est. Saúde SP | ID: biblio-1241810

ABSTRACT

The molecular events that occur at the early phase of many demyelinating neurodegenerative diseases are unknown. A recent demonstration of rapid demyelination and axonal injury induced by Mycobacterium leprae provides a model for elucidating the molecular events of early nerve degeneration which might be common to neurodegenerative diseases of both infectious origin and unknown etiology. The identification of the M. leprae-targeted Schwann cell receptor, dystroglycan, and its associated molecules in myelination, demyelination and axonal functions suggests a role for these molecules in early nerve degeneration.


Subject(s)
Humans , Animals , Mice , Axons , Myelin Sheath , Dystrophin-Associated Protein Complex , Schwann Cells , Dystroglycans , Demyelinating Autoimmune Diseases, CNS , Leprosy , Mycobacterium leprae
17.
Proc Natl Acad Sci U S A ; 100(26): 16071-6, 2003 Dec 23.
Article in English | MEDLINE | ID: mdl-14657400

ABSTRACT

Members of the arenavirus family, famous for their hemorrhagic syndromes, cause distinct neurological disorders; however, cellular and molecular targets as well as pathogenesis of peripheral nervous system disorders associated with these viruses are unknown. Using noncytolytic lymphocytic choriomeningitis virus, the prototype arenavirus, and pseudotyped Lassa fever virus, we showed that the Schwann cells, but not the neurons, were preferentially targeted and harbored the virus. This permissiveness was caused by the viral glycoprotein usage of its receptor alpha-dystroglycan, which was highly abundant on Schwann cell membranes. Persistent lymphocytic choriomeningitis virus infection rendered immature Schwann cells defective or incapable of forming compact myelin sheathes when they differentiated to myelinating phenotype in an in vitro differentiation model of Schwann cells. Persistent infection did not cause Schwann cell apoptosis or cytopathic effect. Defects in myelination coincided with the down-regulation of dystroglycan expression and disruption of the laminin-2 organization and basal lamina assembly on Schwann cell-axon units. The data provide evidence for a selective perturbation of laminin-2-laminin-2 receptor communication pathway in the peripheral nervous system by a nonlytic virus and the resulting myelin defects, which may partly contribute to neurological abnormalities associated with arenaviral infection.


Subject(s)
Arenaviridae Infections/physiopathology , Lymphocytic choriomeningitis virus , Myelin Sheath/physiology , Schwann Cells/physiology , Schwann Cells/virology , Animals , Binding, Competitive , Cells, Cultured , Coculture Techniques , Cytoskeletal Proteins/metabolism , Dystroglycans , Ganglia, Spinal/physiology , Ganglia, Spinal/virology , Humans , Laminin/metabolism , Lymphocytic choriomeningitis virus/physiology , Membrane Glycoproteins/metabolism , Moloney murine leukemia virus/physiology , Rats
18.
s.l; s.n; 2003. 7 p. ilus, graf.
Non-conventional in English | Sec. Est. Saúde SP, HANSEN, Hanseníase Leprosy, SESSP-ILSLACERVO, Sec. Est. Saúde SP | ID: biblio-1241010

ABSTRACT

Nerve damage is a clinical hallmark of leprosy and a major source of patient morbidity. We investigated the possibility that human Schwann cells are susceptible to cell death through the activation of Toll-like receptor 2 (TLR2), a pattern recognition receptor of the innate immune system. TLR2 was detected on the surface of human Schwann cell line ST88-14 and on cultured primary human Schwann cells. Activation of the human Schwann cell line and primary human Schwann cell cultures with a TLR2 agonist, a synthetic lipopeptide comprising the N-terminal portion of the putative Mycobacterium leprae 19-kDa lipoprotein, triggered an increase in the number of apoptotic cells. The lipopeptide-induced apoptosis of Schwann cells could be blocked by an anti-TLR2 monoclonal antibody. Schwann cells in skin lesions from leprosy patients were found to express TLR2. It was possible to identify in the lesions Schwann cells that had undergone apoptosis in vivo. The ability of M. leprae ligands to induce the apoptosis of Schwann cells through TLR2 provides a mechanism by which activation of the innate immune response contributes to nerve injury in leprosy.


Subject(s)
Humans , Apoptosis , Cells, Cultured , Schwann Cells/pathology , DNA Damage , Membrane Glycoproteins/analysis , Membrane Glycoproteins/physiology , Leprosy/immunology , Leprosy/pathology , Immunity, Innate , Lipoproteins/physiology , Receptors, Cell Surface/analysis , Receptors, Cell Surface/physiology
19.
Infect Immun ; 71(3): 1427-33, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12595460

ABSTRACT

Nerve damage is a clinical hallmark of leprosy and a major source of patient morbidity. We investigated the possibility that human Schwann cells are susceptible to cell death through the activation of Toll-like receptor 2 (TLR2), a pattern recognition receptor of the innate immune system. TLR2 was detected on the surface of human Schwann cell line ST88-14 and on cultured primary human Schwann cells. Activation of the human Schwann cell line and primary human Schwann cell cultures with a TLR2 agonist, a synthetic lipopeptide comprising the N-terminal portion of the putative Mycobacterium leprae 19-kDa lipoprotein, triggered an increase in the number of apoptotic cells. The lipopeptide-induced apoptosis of Schwann cells could be blocked by an anti-TLR2 monoclonal antibody. Schwann cells in skin lesions from leprosy patients were found to express TLR2. It was possible to identify in the lesions Schwann cells that had undergone apoptosis in vivo. The ability of M. leprae ligands to induce the apoptosis of Schwann cells through TLR2 provides a mechanism by which activation of the innate immune response contributes to nerve injury in leprosy.


Subject(s)
Apoptosis , Drosophila Proteins , Leprosy/pathology , Membrane Glycoproteins/physiology , Receptors, Cell Surface/physiology , Schwann Cells/pathology , Cells, Cultured , DNA Damage , Humans , Immunity, Innate , Leprosy/immunology , Lipoproteins/physiology , Membrane Glycoproteins/analysis , Receptors, Cell Surface/analysis , Toll-Like Receptor 2 , Toll-Like Receptors
20.
Science ; 296(5569): 927-31, 2002 May 03.
Article in English | MEDLINE | ID: mdl-11988579

ABSTRACT

Demyelination results in severe disability in many neurodegenerative diseases and nervous system infections, and it is typically mediated by inflammatory responses. Mycobacterium leprae, the causative organism of leprosy, induced rapid demyelination by a contact-dependent mechanism in the absence of immune cells in an in vitro nerve tissue culture model and in Rag1-knockout (Rag1-/-) mice, which lack mature B and T lymphocytes. Myelinated Schwann cells were resistant to M. leprae invasion but undergo demyelination upon bacterial attachment, whereas nonmyelinated Schwann cells harbor intracellular M. leprae in large numbers. During M. leprae-induced demyelination, Schwann cells proliferate significantly both in vitro and in vivo and generate a more nonmyelinated phenotype, thereby securing the intracellular niche for M. leprae.


Subject(s)
Antigens, Bacterial , Demyelinating Diseases/microbiology , Leprosy/microbiology , Mycobacterium leprae/pathogenicity , Myelin Sheath/physiology , Schwann Cells/microbiology , Schwann Cells/physiology , Animals , Apoptosis , Axons/microbiology , Axons/ultrastructure , B-Lymphocytes/immunology , Bacterial Adhesion , Cell Division , Coculture Techniques , Culture Techniques , Ganglia, Spinal/cytology , Genes, RAG-1 , Glycolipids/metabolism , Humans , Leprosy/immunology , Leprosy/pathology , Leprosy/physiopathology , Macrophages/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Mycobacterium leprae/physiology , Myelin Sheath/ultrastructure , Nerve Degeneration , Nerve Fibers, Myelinated/metabolism , Neurons/physiology , Sciatic Nerve/microbiology , Sciatic Nerve/pathology , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...