Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 145
Filter
1.
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
2.
Cell Microbiol ; 22(1): e13128, 2020 01.
Article in English | MEDLINE | ID: mdl-31652371

ABSTRACT

Leprosy neuropathy is a chronic degenerative infectious disorder of the peripheral nerve caused by the intracellular obligate pathogen Mycobacterium leprae (M. leprae). Among all nonneuronal cells that constitute the nerve, Schwann cells are remarkable in supporting M. leprae persistence intracellularly. Notably, the success of leprosy infection has been attributed to its ability in inducing the demyelination phenotype after contacting myelinated fibres. However, the exact role M. leprae plays during the ongoing process of myelin breakdown is entirely unknown. Here, we provided evidence showing an unexpected predilection of leprosy pathogen for degenerating myelin ovoids inside Schwann cells. In addition, M. leprae infection accelerated the rate of myelin breakdown and clearance leading to increased formation of lipid droplets, by modulating a set of regulatory genes involved in myelin maintenance, autophagy, and lipid storage. Remarkably, the blockage of myelin breakdown significantly reduced M. leprae content, demonstrating a new unpredictable role of myelin dismantling favouring M. leprae physiology. Collectively, our study provides novel evidence that may explain the demyelination phenotype as an evolutionarily conserved mechanism used by leprosy pathogen to persist longer in the peripheral nerve.


Subject(s)
Mycobacterium leprae/physiology , Myelin Sheath/metabolism , Schwann Cells/microbiology , Animals , Cells, Cultured , Humans , Leprosy/complications , Leprosy/microbiology , Male , Mice , Mice, Inbred BALB C , Mycobacterium leprae/pathogenicity , Myelin Sheath/microbiology
3.
Infect Immun ; 88(4)2020 03 23.
Article in English | MEDLINE | ID: mdl-31964742

ABSTRACT

Neisseria meningitidis, a common cause of sepsis and bacterial meningitis, infects the meninges and central nervous system (CNS), primarily via paracellular traversal across the blood-brain barrier (BBB) or blood-cerebrospinal fluid barrier. N. meningitidis is often present asymptomatically in the nasopharynx, and the nerves extending between the nasal cavity and the brain constitute an alternative route by which the meningococci may reach the CNS. To date, the cellular mechanisms involved in nerve infection are not fully understood. Peripheral nerve glial cells are phagocytic and are capable of eliminating microorganisms, but some pathogens may be able to overcome this protection mechanism and instead infect the glia, causing cell death or pathology. Here, we show that N. meningitidis readily infects trigeminal Schwann cells (the glial cells of the trigeminal nerve) in vitro in both two-dimensional and three-dimensional cell cultures. Infection of trigeminal Schwann cells may be one mechanism by which N. meningitidis is able to invade the CNS. Infection of the cells led to multinucleation and the appearance of atypical nuclei, with the presence of horseshoe nuclei and the budding of nuclei increasing over time. Using sequential window acquisition of all theoretical mass spectra (SWATH-MS) proteomics followed by bioinformatics pathway analysis, we showed that N. meningitidis induced protein alterations in the glia that were associated with altered intercellular signaling, cell-cell interactions, and cellular movement. The analysis also suggested that the alterations in protein levels were consistent with changes occurring in cancer. Thus, infection of the trigeminal nerve by N. meningitidis may have ongoing adverse effects on the biology of Schwann cells, which may lead to pathology.


Subject(s)
Host-Pathogen Interactions , Neisseria meningitidis/growth & development , Neisseria meningitidis/pathogenicity , Schwann Cells/microbiology , Schwann Cells/pathology , Trigeminal Nerve/cytology , Animals , Cells, Cultured , Mice, Transgenic , Proteome/analysis , Proteomics
4.
Int J Exp Pathol ; 100(2): 83-93, 2019 04.
Article in English | MEDLINE | ID: mdl-31090128

ABSTRACT

Schwann cells (SCs) critically maintain the plasticity of the peripheral nervous system. Peripheral nerve injuries and infections stimulate SCs in order to retrieve homeostasis in neural tissues. Previous studies indicate that Mycobacterium leprae (ML) regulates the expression of key factors related to SC identity, suggesting that alterations in cell phenotype may be involved in the pathogenesis of neural damage in leprosy. To better understand whether ML restricts the plasticity of peripheral nerves, the present study sought to determine the expression of Krox-20, Sox-10, c-Jun and p75NTR in SC culture and mice sciatic nerves, both infected by ML Thai-53 strain. Primary SC cultures were stimulated with two different multiplicities of infection (MOI 100:1; MOI 50:1) and assessed after 7 and 14 days. Sciatic nerves of nude mice (NU-Foxn1nu ) infected with ML were evaluated after 6 and 9 months. In vitro results demonstrate downregulation of Krox-20 and Sox-10 along with the increase in p75NTR-immunolabelled cells. Concurrently, sciatic nerves of infected mice showed a significant decrease in Krox-20 and increase in p75NTR. Our results corroborate previous findings on the interference of ML in the expression of factors involved in cell maturation, favouring the maintenance of a non-myelinating phenotype in SCs, with possible implications for the repair of adult peripheral nerves.


Subject(s)
Down-Regulation , Early Growth Response Protein 2/biosynthesis , Leprosy/metabolism , Schwann Cells/metabolism , Sciatic Nerve/metabolism , Animals , Cell Differentiation/physiology , Cells, Cultured , Disease Models, Animal , Leprosy/microbiology , Leprosy/pathology , Mice, Nude , Mycobacterium leprae/isolation & purification , Neuronal Plasticity/physiology , Receptors, Nerve Growth Factor/metabolism , Schwann Cells/microbiology , Schwann Cells/pathology , Sciatic Nerve/microbiology , Sciatic Nerve/pathology , Tissue Culture Techniques
5.
J Biol Chem ; 291(41): 21375-21387, 2016 Oct 07.
Article in English | MEDLINE | ID: mdl-27555322

ABSTRACT

Mycobacterium leprae, the intracellular etiological agent of leprosy, infects Schwann promoting irreversible physical disabilities and deformities. These cells are responsible for myelination and maintenance of axonal energy metabolism through export of metabolites, such as lactate and pyruvate. In the present work, we observed that infected Schwann cells increase glucose uptake with a concomitant increase in glucose-6-phosphate dehydrogenase (G6PDH) activity, the key enzyme of the oxidative pentose pathway. We also observed a mitochondria shutdown in infected cells and mitochondrial swelling in pure neural leprosy nerves. The classic Warburg effect described in macrophages infected by Mycobacterium avium was not observed in our model, which presented a drastic reduction in lactate generation and release by infected Schwann cells. This effect was followed by a decrease in lactate dehydrogenase isoform M (LDH-M) activity and an increase in cellular protection against hydrogen peroxide insult in a pentose phosphate pathway and GSH-dependent manner. M. leprae infection success was also dependent of the glutathione antioxidant system and its main reducing power source, the pentose pathway, as demonstrated by a 50 and 70% drop in intracellular viability after treatment with the GSH synthesis inhibitor buthionine sulfoximine, and aminonicotinamide (6-ANAM), an inhibitor of G6PDH 6-ANAM, respectively. We concluded that M. leprae could modulate host cell glucose metabolism to increase the cellular reducing power generation, facilitating glutathione regeneration and consequently free-radical control. The impact of this regulation in leprosy neuropathy is discussed.


Subject(s)
Energy Metabolism , Glucose/metabolism , Glucosephosphate Dehydrogenase/metabolism , Lactic Acid/metabolism , Leprosy, Tuberculoid/metabolism , Mycobacterium leprae/metabolism , Schwann Cells/metabolism , Cell Line , Humans , Methionine/analogs & derivatives , Methionine/pharmacology , Mitochondria/metabolism , Schwann Cells/microbiology
6.
Curr Opin Infect Dis ; 30(3): 309-315, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28134679

ABSTRACT

PURPOSE OF REVIEW: We wished to overview recent data on a subset of epigenetic changes elicited by intracellular bacteria in human cells. Reprogramming the gene expression pattern of various host cells may facilitate bacterial growth, survival, and spread. RECENT FINDINGS: DNA-(cytosine C5)-methyltransferases of Mycoplasma hyorhinis targeting cytosine-phosphate-guanine (CpG) dinucleotides and a Mycobacterium tuberculosis methyltransferase targeting non-CpG sites methylated the host cell DNA and altered the pattern of gene expression. Gene silencing by CpG methylation and histone deacetylation, mediated by cellular enzymes, also occurred in M. tuberculosis-infected macrophages. M. tuberculosis elicited cell type-specific epigenetic changes: it caused increased DNA methylation in macrophages, but induced demethylation, deposition of euchromatic histone marks and activation of immune-related genes in dendritic cells. A secreted transposase of Acinetobacter baumannii silenced a cellular gene, whereas Mycobacterium leprae altered the epigenotype, phenotype, and fate of infected Schwann cells. The 'keystone pathogen' oral bacterium Porphyromonas gingivalis induced local DNA methylation and increased the level of histone acetylation in host cells. These epigenetic changes at the biofilm-gingiva interface may contribute to the development of periodontitis. SUMMARY: Epigenetic regulators produced by intracellular bacteria alter the epigenotype and gene expression pattern of host cells and play an important role in pathogenesis.


Subject(s)
Bacteria/pathogenicity , Epigenesis, Genetic , Gene Expression Regulation, Bacterial/genetics , Acinetobacter baumannii/enzymology , Acinetobacter baumannii/pathogenicity , Bacteria/enzymology , DNA Methylation , Dendritic Cells/metabolism , Dendritic Cells/microbiology , Humans , Macrophages/metabolism , Macrophages/microbiology , Mycobacterium leprae/enzymology , Mycobacterium leprae/pathogenicity , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/pathogenicity , Mycoplasma hyorhinis/enzymology , Mycoplasma hyorhinis/pathogenicity , Porphyromonas gingivalis/enzymology , Porphyromonas gingivalis/pathogenicity , Schwann Cells/metabolism , Schwann Cells/microbiology
7.
J Infect Dis ; 214(2): 311-20, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27190175

ABSTRACT

Cytosolic detection of nucleic acids elicits a type I interferon (IFN) response and plays a critical role in host defense against intracellular pathogens. Herein, a global gene expression profile of Mycobacterium leprae-infected primary human Schwann cells identified the genes differentially expressed in the type I IFN pathway. Among them, the gene encoding 2'-5' oligoadenylate synthetase-like (OASL) underwent the greatest upregulation and was also shown to be upregulated in M. leprae-infected human macrophage cell lineages, primary monocytes, and skin lesion specimens from patients with a disseminated form of leprosy. OASL knock down was associated with decreased viability of M. leprae that was concomitant with upregulation of either antimicrobial peptide expression or autophagy levels. Downregulation of MCP-1/CCL2 release was also observed during OASL knock down. M. leprae-mediated OASL expression was dependent on cytosolic DNA sensing mediated by stimulator of IFN genes signaling. The addition of M. leprae DNA enhanced nonpathogenic Mycobacterium bovis bacillus Calmette-Guerin intracellular survival, downregulated antimicrobial peptide expression, and increased MCP-1/CCL2 secretion. Thus, our data uncover a promycobacterial role for OASL during M. leprae infection that directs the host immune response toward a niche that permits survival of the pathogen.


Subject(s)
2',5'-Oligoadenylate Synthetase/metabolism , Host-Pathogen Interactions , Membrane Proteins/metabolism , Microbial Viability , Mycobacterium leprae/physiology , Schwann Cells/microbiology , Cells, Cultured , Epithelial Cells/microbiology , Gene Expression Profiling , Gene Knockdown Techniques , Humans , Leprosy/microbiology , Leprosy/pathology , Macrophages/microbiology , Mycobacterium bovis/physiology
8.
Ann Clin Microbiol Antimicrob ; 15(1): 33, 2016 May 21.
Article in English | MEDLINE | ID: mdl-27209077

ABSTRACT

Leprosy, a chronic mycobacterial infection caused by Mycobacterium leprae, is an infectious disease that has ravaged human societies throughout millennia. This ancestral pathogen causes disfiguring cutaneous lesions, peripheral nerve injury, ostearticular deformity, limb loss and dysfunction, blindness and stigma. Despite ongoing efforts in interrupting leprosy transmission, large numbers of new cases are persistently identified in many endemic areas. Moreover, at the time of diagnosis, most newly identified cases have considerable neurologic disability. Many challenges remain in our understanding of the epidemiology of leprosy including: (a) the precise mode and route of transmission; (b) the socioeconomic, environmental, and behavioral factors that promote its transmission; and


Subject(s)
Blindness/epidemiology , Endemic Diseases , Leprosy/epidemiology , Mycobacterium leprae/pathogenicity , Peripheral Nerve Injuries/epidemiology , Skin/microbiology , Blindness/diagnosis , Blindness/etiology , Blindness/pathology , Cartilage, Articular/microbiology , Cartilage, Articular/pathology , Disabled Persons , Histiocytes/microbiology , Histiocytes/pathology , Human Migration , Humans , Leprosy/complications , Leprosy/diagnosis , Leprosy/transmission , Mycobacterium leprae/genetics , Mycobacterium leprae/growth & development , Peripheral Nerve Injuries/diagnosis , Peripheral Nerve Injuries/etiology , Peripheral Nerve Injuries/pathology , Peripheral Nerves/microbiology , Peripheral Nerves/pathology , Schwann Cells/microbiology , Schwann Cells/pathology , Skin/pathology , Socioeconomic Factors
9.
J Transl Med ; 13: 296, 2015 Sep 11.
Article in English | MEDLINE | ID: mdl-26362198

ABSTRACT

BACKGROUND: Peripheral nerve injury and bone lesions, well known leprosy complications, lead to deformities and incapacities. The phosphate-regulating gene with homologies to endopeptidase on the X chromosome (PHEX) encodes a homonymous protein (PHEX) implicated in bone metabolism. PHEX/PHEX alterations may result in bone and cartilage lesions. PHEX expression is downregulated by intracellular Mycobacterium leprae (M. leprae) in cultures of human Schwann cells and osteoblasts. M. leprae in vivo effect on PHEX/PHEX is not known. METHODS: Cross-sectional observational study of 36 leprosy patients (22 lepromatous and 14 borderline-tuberculoid) and 20 healthy volunteers (HV). The following tests were performed: PHEX flow cytometric analysis on blood mononuclear cells, cytokine production in culture supernatant, 25-hydroxyvitamin D (OHvitD) serum levels and (99m)Tc-MDP three-phase bone scintigraphy, radiography of upper and lower extremities and blood and urine biochemistry. RESULTS: Significantly lower PHEX expression levels were observed in lepromatous patients than in the other groups (χ(2) = 16.554, p < 0.001 for lymphocytes and χ(2) = 13.933, p = 0.001 for monocytes). Low levels of 25-(OHvitD) were observed in HV (median = 23.0 ng/mL) and BT patients (median = 27.5 ng/mL) and normal serum levels were found in LL patients (median = 38.6 ng/mL). Inflammatory cytokines, such as TNF, a PHEX transcription repressor, were lower after stimulation with M. leprae in peripheral blood mononuclear cells from lepromatous in comparison to BT patients and HV (χ(2) = 10.820, p < 0.001). CONCLUSION: Downregulation of PHEX may constitute an important early component of bone loss and joint damage in leprosy. The present results suggest a direct effect produced by M. leprae on the osteoarticular system that may use this mechanism.


Subject(s)
Down-Regulation , Leprosy, Borderline/metabolism , Leprosy, Multibacillary/metabolism , PHEX Phosphate Regulating Neutral Endopeptidase/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Bone and Bones/microbiology , Cartilage/microbiology , Cross-Sectional Studies , Cytokines/metabolism , Female , Flow Cytometry , Healthy Volunteers , Humans , Inflammation/metabolism , Inflammation/microbiology , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Osteoblasts/microbiology , Schwann Cells/microbiology , Technetium Tc 99m Medronate , Young Adult
10.
BMC Microbiol ; 14: 211, 2014 Aug 02.
Article in English | MEDLINE | ID: mdl-25085553

ABSTRACT

BACKGROUND: The ability of S. pneumoniae to generate infections depends on the restrictions imposed by the host's immunity, in order to prevent the bacterium from spreading from the nasopharynx to other tissues, such as the brain. Some authors claim that strains of S. pneumoniae, which fail to survive in the bloodstream, can enter the brain directly from the nasal cavity by axonal transport through the olfactory and/or trigeminal nerves. However, from the immunological point of view, glial cells are far more responsive to bacterial infections than are neurons. This hypothesis is consistent with several recent reports showing that bacteria can infect glial cells from the olfactory bulb and trigeminal ganglia. Since our group previously demonstrated that Schwann cells (SCs) express a functional and appropriately regulated mannose receptor (MR), we decided to test whether SCs are involved in the internalization of S. pneumoniae via MR. RESULTS: Immediately after the interaction step, as well as 3 h later, the percentage of association was approximately 56.5%, decreasing to 47.2% and 40.8% after 12 and 24 h, respectively. Competition assays by adding a 100-fold excess of mannan prior to the S. pneumoniae infection reduced the number of infected cells at 3 and 24 h. A cytochemistry assay with Man/BSA-FITC binding was performed in order to verify a possible overlap between mannosylated ligands and internalized bacteria. Incubation of the SCs with Man/BSA-FITC resulted in a large number of intracellular S. pneumoniae, with nearly complete loss of the capsule. Moreover, the anti-pneumococcal antiserum staining colocalized with the internalized man/BSA-FITC, suggesting that both markers are present within the same endocytic compartment of the SC. CONCLUSIONS: Our data offer novel evidence that SCs could be essential for pneumococcal cells to escape phagocytosis and killing by innate immune cells. On the other hand, the results also support the idea that SCs are immunocompetent cells of the PNS that can mediate an efficient immune response against pathogens via MR.


Subject(s)
Endocytosis , Host-Pathogen Interactions , Lectins, C-Type/metabolism , Mannose-Binding Lectins/metabolism , Receptors, Cell Surface/metabolism , Schwann Cells/immunology , Schwann Cells/microbiology , Streptococcus pneumoniae/immunology , Animals , Cells, Cultured , Mannose Receptor , Rats, Wistar
11.
Ultrastruct Pathol ; 38(2): 86-92, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24460757

ABSTRACT

A systematic ultrastructure of peripheral nerves across the spectrum of leprosy was studied with an aim to better understanding the pathogenesis of nerve involvement in leprosy using light and electron microscope. The pathogenesis of nerve destruction varies in leprosy considerably along the spectrum. The study has begun to shed new light on some aspects of the infection of Mycobacterium leprae (M. lepare) and phenomenon has opened new avenue of research and possible mechanism of pathogenesis in TT/BT/BL/LL leprosy. In tuberculoid type (TT) and borderline tuberculoid (BT) leprosy, the degenerative changes of Schwann cells (SCs) and presence of perineural and perivascular cuffing by mononuclear cells. The endoneurial blood vessel (EBV) showed thickening of basement membrane with hypertrophy of EC leading to narrowing or complete occlusion of lumen and causing ischemia. However, borderline lepromatous (BL) and lepromatous leprosy (LL) foamy macrophages and vacuolated SC contain numerous small dense materials, irregular in shape and size was prominent and, considered to be degenerated and fragmented M. Leprae. The dense materials were also found in the cytoplasm of vascular EC. It was revealed that besides SC, the EC of EBV frequently harbor M. leprae in LL. The lumen of the EBV was wide open with enlarged nucleus. In the present study, the ultrastructural characteristics suggest that hypersensitivity mechanisms are possibly responsible for nerve damage in TT/BT leprosy. However, the study indicates that the mechanisms of nerve damage in BL/LL are basically different wherein hypersensitivity appears to play a very limited role.


Subject(s)
Endothelial Cells/ultrastructure , Leprosy/pathology , Peripheral Nerves/ultrastructure , Schwann Cells/ultrastructure , Endothelial Cells/microbiology , Humans , Microscopy, Electron, Transmission , Peripheral Nerves/microbiology , Schwann Cells/microbiology
12.
Gut Microbes ; 16(1): 2363015, 2024.
Article in English | MEDLINE | ID: mdl-38845453

ABSTRACT

Gut microbiota is responsible for essential functions in human health. Several communication axes between gut microbiota and other organs via neural, endocrine, and immune pathways have been described, and perturbation of gut microbiota composition has been implicated in the onset and progression of an emerging number of diseases. Here, we analyzed peripheral nerves, dorsal root ganglia (DRG), and skeletal muscles of neonatal and young adult mice with the following gut microbiota status: a) germ-free (GF), b) gnotobiotic, selectively colonized with 12 specific gut bacterial strains (Oligo-Mouse-Microbiota, OMM12), or c) natural complex gut microbiota (CGM). Stereological and morphometric analyses revealed that the absence of gut microbiota impairs the development of somatic median nerves, resulting in smaller diameter and hypermyelinated axons, as well as in smaller unmyelinated fibers. Accordingly, DRG and sciatic nerve transcriptomic analyses highlighted a panel of differentially expressed developmental and myelination genes. Interestingly, the type III isoform of Neuregulin1 (NRG1), known to be a neuronal signal essential for Schwann cell myelination, was overexpressed in young adult GF mice, with consequent overexpression of the transcription factor Early Growth Response 2 (Egr2), a fundamental gene expressed by Schwann cells at the onset of myelination. Finally, GF status resulted in histologically atrophic skeletal muscles, impaired formation of neuromuscular junctions, and deregulated expression of related genes. In conclusion, we demonstrate for the first time a gut microbiota regulatory impact on proper development of the somatic peripheral nervous system and its functional connection to skeletal muscles, thus suggesting the existence of a novel 'Gut Microbiota-Peripheral Nervous System-axis.'


Subject(s)
Ganglia, Spinal , Gastrointestinal Microbiome , Neuromuscular Junction , Animals , Neuromuscular Junction/microbiology , Mice , Ganglia, Spinal/metabolism , Ganglia, Spinal/microbiology , Germ-Free Life , Peripheral Nerves/microbiology , Peripheral Nerves/growth & development , Muscle, Skeletal/microbiology , Mice, Inbred C57BL , Neuregulin-1/metabolism , Neuregulin-1/genetics , Male , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Bacteria/metabolism , Schwann Cells/microbiology , Schwann Cells/metabolism
13.
J Immunol ; 187(5): 2548-58, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21813774

ABSTRACT

The mechanisms responsible for nerve injury in leprosy need further elucidation. We recently demonstrated that the foamy phenotype of Mycobacterium leprae-infected Schwann cells (SCs) observed in nerves of multibacillary patients results from the capacity of M. leprae to induce and recruit lipid droplets (LDs; also known as lipid bodies) to bacterial-containing phagosomes. In this study, we analyzed the parameters that govern LD biogenesis by M. leprae in SCs and how this contributes to the innate immune response elicited by M. leprae. Our observations indicated that LD formation requires the uptake of live bacteria and depends on host cell cytoskeleton rearrangement and vesicular trafficking. TLR6 deletion, but not TLR2, completely abolished the induction of LDs by M. leprae, as well as inhibited the bacterial uptake in SCs. M. leprae-induced LD biogenesis correlated with increased PGE(2) and IL-10 secretion, as well as reduced IL-12 and NO production in M. leprae-infected SCs. Analysis of nerves from lepromatous leprosy patients showed colocalization of M. leprae, LDs, and cyclooxygenase-2 in SCs, indicating that LDs are sites for PGE(2) synthesis in vivo. LD biogenesis Inhibition by the fatty acid synthase inhibitor C-75 abolished the effect of M. leprae on SC production of immunoinflammatory mediators and enhanced the mycobacterial-killing ability of SCs. Altogether, our data indicated a critical role for TLR6-dependent signaling in M. leprae-SC interactions, favoring phagocytosis and subsequent signaling for induction of LD biogenesis in infected cells. Moreover, our observations reinforced the role of LDs favoring mycobacterial survival and persistence in the nerve. These findings give further support to a critical role for LDs in M. leprae pathogenesis in the nerve.


Subject(s)
Leprosy/pathology , Schwann Cells/microbiology , Schwann Cells/pathology , Toll-Like Receptor 6/immunology , Animals , Humans , Immunohistochemistry , Inclusion Bodies/immunology , Inclusion Bodies/metabolism , Inclusion Bodies/pathology , Inflammation/immunology , Inflammation/microbiology , Inflammation/pathology , Leprosy/immunology , Lipid Metabolism/physiology , Lipids/immunology , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Mycobacterium leprae/immunology , Schwann Cells/immunology , Toll-Like Receptor 6/metabolism
14.
Cell Microbiol ; 13(2): 259-73, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20955239

ABSTRACT

The predilection of Mycobacterium leprae (ML) for Schwann cells (SCs) leads to peripheral neuropathy, a major concern in leprosy. Highly infected SCs in lepromatous leprosy nerves show a foamy, lipid-laden appearance; but the origin and nature of these lipids, as well as their role in leprosy, have remained unclear. The data presented show that ML has a pronounced effect on host-cell lipid homeostasis through regulation of lipid droplet (lipid bodies, LD) biogenesis and intracellular distribution. Electron microscopy and immunohistochemical analysis of lepromatous leprosy nerves for adipose differentiation-related protein expression, a classical LD marker, revealed accumulating LDs in close association to ML in infected SCs. The capacity of ML to induce LD formation was confirmed in in vitro studies with human SCs. Moreover, via confocal and live-cell analysis, it was found that LDs are promptly recruited to bacterial phagosomes and that this process depends on cytoskeletal reorganization and PI3K signalling. ML-induced LD biogenesis and recruitment were found to be independent of TLR2 bacterial sensing. Notably, LD recruitment impairment by cytoskeleton drugs decreased intracellular bacterial survival. Altogether, our data revealed SC lipid accumulation in ML-containing phagosomes, which may represent a fundamental aspect of bacterial pathogenesis in the nerve.


Subject(s)
Lipid Metabolism , Mycobacterium leprae/pathogenicity , Phagosomes/microbiology , Schwann Cells/microbiology , Cells, Cultured , Cytoplasm/chemistry , Cytoplasm/ultrastructure , Cytoskeleton/metabolism , Humans , Immunohistochemistry , Membrane Proteins/analysis , Microbial Viability , Microscopy , Mycobacterium leprae/metabolism , Perilipin-2 , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction
15.
Mem Inst Oswaldo Cruz ; 107 Suppl 1: 156-66, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23283467

ABSTRACT

Leprosy is an infectious disease caused by Mycobacterium leprae that affects the skin and nerves, presenting a singular clinical picture. Across the leprosy spectrum, lepromatous leprosy (LL) exhibits a classical hallmark: the presence of a collection of M. leprae-infected foamy macrophages/Schwann cells characterised by their high lipid content. The significance of this foamy aspect in mycobacterial infections has garnered renewed attention in leprosy due to the recent observation that the foamy aspect represents cells enriched in lipid droplets (LD) (also known as lipid bodies). Here, we discuss the contemporary view of LD as highly regulated organelles with key functions in M. leprae persistence in the LL end of the spectrum. The modern methods of studying this ancient disease have contributed to recent findings that describe M. leprae-triggered LD biogenesis and recruitment as effective mycobacterial intracellular strategies for acquiring lipids, sheltering and/or dampening the immune response and favouring bacterial survival, likely representing a fundamental aspect of M. leprae pathogenesis. The multifaceted functions attributed to the LD in leprosy may contribute to the development of new strategies for adjunctive anti-leprosy therapies.


Subject(s)
Leprosy, Lepromatous/pathology , Mycobacterium leprae/immunology , Schwann Cells/microbiology , Humans , Inclusion Bodies/immunology , Inclusion Bodies/metabolism , Inclusion Bodies/pathology , Leprosy, Lepromatous/immunology , Lipids/immunology , Organelles/immunology , Schwann Cells/immunology
16.
J Biol Chem ; 285(44): 34086-96, 2010 Oct 29.
Article in English | MEDLINE | ID: mdl-20739294

ABSTRACT

Mycobacterium leprae (ML), the etiologic agent of leprosy, mainly affects the skin and peripheral nerves, leading to demyelization and loss of axonal conductance. Schwann cells (SCs) are the main cell population infected by ML in the nerves, and infection triggers changes in the SC phenotype from a myelinated to a nonmyelinated state. In the present study, we show that expression of 9-O-acetyl GD3, a ganglioside involved in cellular anti-apoptotic signaling and nerve regeneration, increases in SCs following infection with ML. Observation by confocal microscopy together with coimmunoprecipitation suggested that this ganglioside participates in ML attachment and internalization by SC. Immunoblockage of 9-O-acetyl GD3 in vitro significantly reduced adhesion of ML to SC surfaces. Finally, we show that activation of the MAPK (ERK 1/2) pathway and SC proliferation, two known effects of ML on SCs that result in demyelization, are significantly reduced when the 9-O-acetyl GD3 ganglioside is immunoblocked. Taken together, these data suggest the involvement of 9-O-acetyl GD3 in ML infection on SCs.


Subject(s)
Gangliosides/metabolism , Leprosy/microbiology , Mycobacterium leprae/metabolism , Schwann Cells/metabolism , Schwann Cells/microbiology , Animals , Apoptosis , Humans , Integrin beta1/metabolism , Leprosy/metabolism , Male , Mice , Mice, Nude , Models, Biological , Myelin Sheath/chemistry , Neurons/metabolism , Signal Transduction
17.
Cell Microbiol ; 12(1): 42-54, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19732058

ABSTRACT

Peripheral nerve lesions are considered the most relevant symptoms of leprosy, a chronic infectious disease caused by Mycobacterium leprae. The strategies employed by M. leprae to infect and multiply inside Schwann cells (SCs), however, remain poorly understood. In this study, it is shown that treatment of SCs with M. leprae significantly decreased cell death induced by serum deprivation. Not displayed by Mycobacterium smegmatis or Mycobacterium bovis BCG, the M. leprae survival effect was both dose dependent and specific. The conditioned medium (CM) of M. leprae-treated cultures was seen to mimic the protective effect of the bacteria, suggesting that soluble factors secreted by SCs in response to M. leprae were involved in cell survival. Indeed, by quantitative RT-PCR and dot blot/ELISA, it was demonstrated that M. leprae induced the expression and secretion of the SC survival factor insulin-like growth factor-I. Finally, the involvement of this hormone in M. leprae-induced SC survival was confirmed in experiments with neutralizing antibodies. Taken together, the results of this study delineate an important strategy for the successful colonization of M. leprae in the nerve based on the survival maintenance of the host cell through induction of IGF-I production.


Subject(s)
Culture Media, Serum-Free/pharmacology , Insulin-Like Growth Factor I/physiology , Mycobacterium leprae/physiology , Schwann Cells/metabolism , Schwann Cells/microbiology , Apoptosis/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Culture Media, Conditioned/pharmacology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunochemistry , Insulin-Like Growth Factor I/metabolism , Membrane Potential, Mitochondrial , Reverse Transcriptase Polymerase Chain Reaction , Schwann Cells/cytology , Schwann Cells/drug effects
18.
Infect Immun ; 78(11): 4634-43, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20713631

ABSTRACT

The ability of microbial pathogens to target specific cell types is a key aspect of the pathogenesis of infectious disease. Mycobacterium leprae, by infecting Schwann cells, contributes to nerve injury in patients with leprosy. Here, we investigated mechanisms of host-pathogen interaction in the peripheral nerve lesions of leprosy. We found that the expression of the C-type lectin, CD209, known to be expressed on tissue macrophages and to mediate the uptake of M. leprae, was present on Schwann cells, colocalizing with the Schwann cell marker, CNPase (2',3'-cyclic nucleotide 3'-phosphodiesterase), along with the M. leprae antigen PGL-1 in the peripheral nerve biopsy specimens. In vitro, human CD209-positive Schwann cells, both from primary cultures and a long-term line, have a higher binding of M. leprae compared to CD209-negative Schwann cells. Interleukin-4, known to be expressed in skin lesions from multibacillary patients, increased CD209 expression on human Schwann cells and subsequent Schwann cell binding to M. leprae, whereas Th1 cytokines did not induce CD209 expression on these cells. Therefore, the regulated expression of CD209 represents a common mechanism by which Schwann cells and macrophages bind and take up M. leprae, contributing to the pathogenesis of leprosy.


Subject(s)
Cell Adhesion Molecules/metabolism , Host-Pathogen Interactions , Interleukin-4/metabolism , Lectins, C-Type/metabolism , Leprosy, Tuberculoid/pathology , Mycobacterium leprae/physiology , Receptors, Cell Surface/metabolism , Schwann Cells/microbiology , Cell Line, Tumor , Humans , Interleukin-4/immunology , Leprosy, Tuberculoid/immunology , Leprosy, Tuberculoid/microbiology , Mycobacterium leprae/pathogenicity , Schwann Cells/immunology , Schwann Cells/metabolism , Schwann Cells/pathology , Up-Regulation
19.
Sci Rep ; 10(1): 12648, 2020 07 28.
Article in English | MEDLINE | ID: mdl-32724108

ABSTRACT

Leprosy, which is caused by the human pathogen Mycobacterium leprae, causes nerve damage, deformity and disability in over 200,000 people every year. Because of the long doubling time of M. leprae (13 days) and the delayed onset of detectable symptoms, which is estimated to be approximately 3-7 years after infection, there is always a large percentage of subclinically infected individuals in the population who will eventually develop the disease, mainly in endemic countries. piRNAs comprise the largest group of small noncoding RNAs found in humans, and they are distinct from microRNAs (miRNAs) and small interfering RNAs (siRNAs). piRNAs function in transposon silencing, epigenetic regulation, and germline development. The functional role of piRNAs and their associated PIWI proteins have started to emerge in the development of human cancers and viral infections, but their relevance to bacterial diseases has not been investigated. The present study reports the piRNome of human skin, revealing that all but one of the piRNAs examined are downregulated in leprosy skin lesions. Considering that one of the best characterized functions of piRNAs in humans is posttranscriptional mRNA silencing, their functions are similar to what we have described for miRNAs, including acting on apoptosis, M. leprae recognition and engulfment, Schwann cell (SC) demyelination, epithelial-mesenchymal transition (EMT), loss of sensation and neuropathic pain. In addition to new findings on leprosy physiopathology, the discovery of relevant piRNAs involved in disease processes in human skin may provide new clues for therapeutic targets, specifically to control nerve damage, a prominent feature of leprosy that has no currently available pharmaceutical treatment.


Subject(s)
Epithelial-Mesenchymal Transition , Leprosy/genetics , Leprosy/pathology , Mycobacterium leprae/pathogenicity , Neuralgia/pathology , RNA, Small Interfering/genetics , Schwann Cells/pathology , Case-Control Studies , Demyelinating Diseases , Epigenesis, Genetic , Humans , Leprosy/microbiology , Neuralgia/metabolism , Neuralgia/microbiology , Schwann Cells/metabolism , Schwann Cells/microbiology
20.
Science ; 162(3849): 126-7, 1968 Oct 04.
Article in English | MEDLINE | ID: mdl-5675180

ABSTRACT

Herpes simplex virus rapidly infected the trigeminal nerves of mice after intranasal inoculation. Centripetal neural spread was suggested by histologic evidence of encephalitis in the area of attachment of the trigeminal nerve. Furthermore, electron microscopy revealed virus replication primarily within Schwann cells of the trigeminal nerve, and neurons of the gasserian ganglion.


Subject(s)
Encephalitis/microbiology , Herpes Simplex/pathology , Trigeminal Nerve/microbiology , Animals , Animals, Newborn , Mice , Microscopy, Electron , Schwann Cells/microbiology , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL