Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Int Immunol ; 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38567483

RESUMEN

Transforming growth factor-ß-activated kinase 1 (TAK1) plays a pivotal role in innate and adaptive immunity. TAK1 is essential for the activation of mitogen-activated protein kinases (MAPKs) and nuclear factor (NF)-κB pathways downstream of diverse immune receptors, including Toll-like receptors (TLRs). Upon stimulation with TLR ligands, TAK1 is activated via recruitment to lysine 63-linked polyubiquitin chain through TAK1-binding proteins (TAB) 2 and TAB3. However, the physiological importance of TAB2 and TAB3 in macrophages is still controversial. A previous study has shown that mouse bone marrow-derived macrophages (BMDMs) isolated from mice double deficient for TAB2 and TAB3 produced tumor necrosis factor (TNF)-α and interleukin (IL)-6 to the similar levels as control wild-type BMDMs in response to TLR ligands such as lipopolysaccharide (LPS) or Pam3CSK4, indicating that TAB2 and TAB3 are dispensable for TLR signaling. In this study, we revisited the role of TAB2 and TAB3 using an improved mouse model. We observed a significant impairment in the production of pro-inflammatory cytokines and chemokine in LPS- or Pam3CSK4-treated BMDMs deficient for both TAB2 and TAB3. Double deficiency of TAB2 and TAB3 resulted in the decreased activation of NF-κB and MAPK pathways as well as the slight decrease in TAK1 activation in response to LPS or Pam3CSK4. Notably, the TLR-mediated expression of inhibitor of NF-κB (IκB)ζ was severely compromised at the protein and mRNA levels in the TAB2/TAB3 double-deficient BMDMs, thereby impeding IL-6 production. Our results suggest that TAB2 and TAB3 play a redundant and indispensable role in TLR signaling pathway.

2.
Immunity ; 45(6): 1245-1257, 2016 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-27887882

RESUMEN

Phosphatidyl-inositol mannosides (PIM) are glycolipids unique to mycobacteria and other related bacteria that stimulate host immune responses and are implicated in mycobacteria pathogenicity. Here, we found that the FcRγ-coupled C-type lectin receptor DCAR (dendritic cell immunoactivating receptor; gene symbol Clec4b1) is a direct receptor for PIM. Mycobacteria activated reporter cells expressing DCAR, and delipidation of mycobacteria abolished this activity. Acylated PIMs purified from mycobacteria were identified as ligands for DCAR. DCAR was predominantly expressed in small peritoneal macrophages and monocyte-derived inflammatory cells in lungs and spleen. These cells produced monocyte chemoattractant protein-1 (MCP-1) upon PIM treatment, and absence of DCAR or FcRγ abrogated MCP-1 production. Upon mycobacterial infection, Clec4b1-deficient mice showed reduced numbers of monocyte-derived inflammatory cells at the infection site, impaired IFNγ production by T cells, and an increased bacterial load. Thus, DCAR is a critical receptor for PIM that functions to promote T cell responses against mycobacteria.


Asunto(s)
Proteínas Bacterianas/inmunología , Lectinas Tipo C/inmunología , Fosfatidilinositoles/inmunología , Receptores Inmunológicos/inmunología , Células TH1/inmunología , Animales , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mycobacterium/inmunología , Infecciones por Mycobacterium/inmunología
3.
FASEB J ; 36(1): e22096, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34907600

RESUMEN

Tuberculosis is a communicable disease caused by Mycobacterium tuberculosis which primarily infects macrophages and establishes intracellular parasitism. A mycobacterial virulence factor Zn2+ metalloprotease 1 (Zmp1) is known to suppress interleukin (IL)-1ß production by inhibiting caspase-1 resulting in phagosome maturation arrest. However, the molecular mechanism of caspase-1 inhibition by Zmp1 is still elusive. Here, we identified GRIM-19 (also known as NDUFA13), an essential subunit of mitochondrial respiratory chain complex I, as a novel Zmp1-binding protein. Using the CRISPR/Cas9 system, we generated GRIM-19 knockout murine macrophage cell line J774.1 and found that GRIM-19 is essential for IL-1ß production during mycobacterial infection as well as in response to NLRP3 inflammasome-activating stimuli such as extracellular ATP or nigericin. We also found that GRIM-19 is required for the generation of mitochondrial reactive oxygen species and NLRP3-dependent activation of caspase-1. Loss of GRIM-19 or forced expression of Zmp1 resulted in a decrease in mitochondrial membrane potential. Our study revealed a previously unrecognized role of GRIM-19 as an essential regulator of NLRP3 inflammasome and a molecular mechanism underlying Zmp1-mediated suppression of IL-1ß production during mycobacterial infection.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Inflamasomas/metabolismo , Macrófagos/metabolismo , Mycobacterium tuberculosis/metabolismo , NADH NADPH Oxidorreductasas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Bacterianas , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Inflamasomas/genética , Metaloproteasas , Ratones , Membranas Mitocondriales/metabolismo , Mycobacterium tuberculosis/genética , NADH NADPH Oxidorreductasas/genética , Proteína con Dominio Pirina 3 de la Familia NLR/genética
4.
Immunopharmacol Immunotoxicol ; 43(2): 203-211, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33541144

RESUMEN

INTRODUCTION: Multiple sclerosis (MS) is an autoimmune disease mediated by a pro-inflammatory immune response. Experimental autoimmune encephalomyelitis (EAE) induced by immunization of mice with a myelin oligodendrocyte glycoprotein (MOG) peptide emulsified in killed Mycobacterium tuberculosis-containing complete Freund's adjuvant (CFA-EAE) is used as a model of MS. Mycobacterium bovis BCG has been reported to ameliorate clinical symptoms of CFA-EAE, although the precise mechanism has not yet been documented. Since CFA-EAE uses adjuvant with mycobacterial antigens, mycobacterial antigen-specific T cells induced by CFA may cross-react with BCG and modulate EAE. METHODS: To exclude the influence of cross-reactivity, a modified murine EAE model (cell wall skeleton (CWS)-EAE) that does not induce mycobacterial antigen-specific T cells was established and used to reevaluate the therapeutic effects of BCG on EAE. RESULTS: Inoculation with BCG 6 d after CWS-EAE induction successfully ameliorated EAE symptoms, suggesting that the therapeutic effects of BCG are independent of the mycobacterial antigen-specific T cells induced by the CFA-EAE protocol. BCG inoculation into the CWS-EAE mice resulted in reduced levels of MOG-specific Th17 in the central nervous system (CNS) with reduced demyelinated lesions of the spinal cord. In the draining lymph nodes of the MOG-immunized sites, BCG inoculation resulted in an increase in MOG-specific Th17 and Th1 cells at an early stage of immune response. CONCLUSION: The results suggest that BCG inoculation suppresses the Th17 response in the CNS of EAE mice via a mechanism that may involve the suppression of egress of encephalitogenic T cells from lymphoid organs.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Vacuna BCG/administración & dosificación , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/inmunología , Mycobacterium bovis , Células Th17/inmunología , Animales , Células Cultivadas , Encefalomielitis Autoinmune Experimental/inducido químicamente , Femenino , Ratones , Ratones Endogámicos C57BL , Células Th17/efectos de los fármacos
5.
Microbiol Immunol ; 62(1): 1-13, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29205464

RESUMEN

Interleukin-17 family cytokines, consisting of six members, participate in immune response in infections and autoimmune and inflammatory diseases. The prototype cytokine of the family, IL-17A, was originally identified from CD4+ T cells which are now termed Th17 cells. Later, IL-17A-producing cells were expanded to include various hematopoietic cells, namely CD8+ T cells (Tc17), invariant NKT cells, γδ T cells, non-T non-B lymphocytes (termed type 3 innate lymphoid cells) and neutrophils. Some IL-17 family cytokines other than IL-17A are also expressed by CD4+ T cells: IL-17E by Th2 cells and IL-17F by Th17 cells. IL-17A and IL-17F induce expression of pro-inflammatory cytokines to induce inflammation and anti-microbial peptides to kill pathogens, whereas IL-17E induces allergic inflammation. However, the functions of other IL-17 family cytokines have been unclear. Recent studies have shown that IL-17B and IL-17C are expressed by epithelial rather than hematopoietic cells. Interestingly, expression of IL-17E and IL-17F by epithelial cells has also been reported and epithelial cell-derived IL-17 family cytokines shown to play important roles in immune responses to infections at epithelial sites. In this review, we summarize current information on hematopoietic cell-derived IL-17A and non-hematopoietic cell-derived IL-17B, IL-17C, IL-17D, IL-17E and IL-17F in infections and propose functional differences between these two categories of IL-17 family cytokines.


Asunto(s)
Citocinas/inmunología , Citocinas/metabolismo , Infecciones/inmunología , Interleucina-17/inmunología , Interleucina-17/metabolismo , Péptidos Catiónicos Antimicrobianos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Línea Celular , Células Epiteliales/inmunología , Humanos , Infecciones/microbiología , Infecciones/virología , Inflamación/inmunología , Interleucina-17/clasificación , Células T Asesinas Naturales , Neutrófilos/metabolismo , Receptores de Interleucina-17/inmunología , Receptores de Interleucina-17/metabolismo , Células Th17/inmunología , Células Th2/metabolismo
6.
Infect Immun ; 84(2): 573-9, 2016 02.
Artículo en Inglés | MEDLINE | ID: mdl-26644377

RESUMEN

Listeria monocytogenes is a bacterial pathogen which establishes intracellular parasitism in various cells, including macrophages and nonhematopoietic cells, such as hepatocytes. It has been reported that several proinflammatory cytokines have pivotal roles in innate protection against L. monocytogenes infection. We found that a proinflammatory cytokine, interleukin 22 (IL-22), was expressed by CD3(+) CD4(+) T cells at an early stage of L. monocytogenes infection in mice. To assess the influence of IL-22 on L. monocytogenes infection in hepatocytes, cells of a human hepatocellular carcinoma line, HepG2, were treated with IL-22 before L. monocytogenes infection in vitro. Gene expression analysis of the IL-22-treated HepG2 cells identified phospholipase A2 group IIA (PLA2G2A) as an upregulated antimicrobial molecule. Addition of recombinant PLA2G2A to the HepG2 culture significantly suppressed L. monocytogenes infection. Culture supernatant of the IL-22-treated HepG2 cells contained bactericidal activity against L. monocytogenes, and the activity was abrogated by a specific PLA2G2A inhibitor, demonstrating that HepG2 cells secreted PLA2G2A, which killed extracellular L. monocytogenes. Furthermore, colocalization of PLA2G2A and L. monocytogenes was detected in the IL-22-treated infected HepG2 cells, which suggests involvement of PLA2G2A in the mechanism of intracellular killing of L. monocytogenes by HepG2 cells. These results suggest that IL-22 induced at an early stage of L. monocytogenes infection enhances innate immunity against L. monocytogenes in the liver by stimulating hepatocytes to produce an antimicrobial molecule, PLA2G2A.


Asunto(s)
Fosfolipasas A2 Grupo II/metabolismo , Hepatocitos/enzimología , Inmunidad Innata , Interleucinas/inmunología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Hígado/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Fosfolipasas A2 Grupo II/genética , Células Hep G2 , Hepatocitos/inmunología , Hepatocitos/microbiología , Humanos , Interleucinas/genética , Listeria monocytogenes/efectos de los fármacos , Listeriosis/enzimología , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Análisis por Micromatrices , Interleucina-22
7.
Biochem Biophys Res Commun ; 467(2): 235-41, 2015 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-26435501

RESUMEN

Monocytes and macrophages are important effectors and regulators of inflammation, and both their differentiation and activation are regulated strictly in response to environmental cues. Angiopoietin-like protein 2 (Angptl2) is a multifaceted protein, displaying many physiological and pathological functions in inflammation, angiogenesis, hematopoiesis, and tumor development. Although recent studies implicate Angptl2 in chronic inflammation, the mechanisms of inflammation caused by Angptl2 remain unclear. The purpose of the present study was to elucidate the role of Angptl2 in inflammation by understanding the effects of Angptl2 on monocytes/macrophages. We showed that Angptl2 directly activates resident murine peritoneal monocytes and macrophages and induces a drastic upregulation of the transcription of several inflammatory genes including nitric oxide synthase 2 and prostaglandin-endoperoxide synthase 2, and several proinflammatory cytokine genes such as interleukin (IL)-1ß, IL-6, TNFα, and CSF2, along with activation of ERK, JNK, p38, and nuclear factor kappa B signaling pathways. Concordantly, proinflammatory cytokines IL-1ß, IL-6, TNFα, and GM-CSF, were rapidly elevated from murine peritoneal monocytes and macrophages. These results demonstrate a novel role for Angptl2 in inflammation via the direct activation of peritoneal monocytes and macrophages.


Asunto(s)
Angiopoyetinas/inmunología , Macrófagos Peritoneales/inmunología , Monocitos/inmunología , Proteína 2 Similar a la Angiopoyetina , Proteínas Similares a la Angiopoyetina , Angiopoyetinas/genética , Animales , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/inmunología , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/inmunología , Regulación de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Células HEK293 , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Macrófagos Peritoneales/patología , Ratones , Ratones Endogámicos C57BL , Monocitos/patología , FN-kappa B/genética , FN-kappa B/inmunología , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/inmunología , Cultivo Primario de Células , Transducción de Señal , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología
8.
Microbiol Immunol ; 59(12): 735-43, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26577130

RESUMEN

Although the BCG vaccine can prevent tuberculosis (TB) in infants, its ability to prevent adult pulmonary TB is reportedly limited. Therefore, development of a novel effective vaccine against pulmonary TB has become an international research priority. We have previously reported that intranasal vaccination of mice with a mycobacterial heparin-binding hemagglutinin adhesin (HBHA) plus mucosal adjuvant cholera toxin (CT) enhances production of IFN-γ and anti-HBHA antibody and suppresses extrapulmonary bacterial dissemination after intranasal infection with BCG. In the present study, the effects of intranasal HBHA + CT vaccine on murine pulmonary Mycobacterium tuberculosis (Mtb) infection were examined. Intranasal HBHA + CT vaccination alone failed to reduce the bacterial burden in the infected lung. However, a combination vaccine consisting of s.c. BCG priming and an intranasal HBHA + CT booster significantly enhanced protective immunity against pulmonary Mtb infection on day 14 compared with BCG vaccine alone. Further, it was found that intranasal HBHA + CT vaccine enhanced not only IFN-γ but also IL-17A production by HBHA-specific T cells in the lung after pulmonary Mtb infection. Therefore, this combination vaccine may be a good candidate for a new vaccine strategy against pulmonary TB.


Asunto(s)
Adhesinas Bacterianas/inmunología , Vacuna BCG/farmacología , Lectinas/inmunología , Pulmón/inmunología , Mycobacterium tuberculosis/inmunología , Células Th17/inmunología , Tuberculosis Pulmonar/prevención & control , Adyuvantes Inmunológicos/farmacología , Animales , Antígenos Bacterianos/inmunología , Vacuna BCG/inmunología , Toxina del Cólera/inmunología , Femenino , Interferón gamma/biosíntesis , Interleucina-17/biosíntesis , Interleucina-17/inmunología , Lectinas/genética , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Linfocitos T/metabolismo , Células TH1/inmunología , Tuberculosis Pulmonar/inmunología
9.
Microbiol Immunol ; 57(6): 470-7, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23773026

RESUMEN

An engineered bio-nanocapsule (BNC) comprising modified hepatitis B surface antigen L protein was used as a physical scaffold for envelope protein domain III (D3) of Japanese encephalitis virus (JEV). At the N terminus, the BNC contained a two-tandem repeat of the Z domain (ZZ) derived from Staphylococcus aureus protein A (ZZ-BNC). The Lys-rich ZZ moiety exposed on the surface of ZZ-BNC was used for chemical conjugation with the JEV D3 antigen, which had been expressed and purified from Escherichia coli. Immunization of mice with D3 loaded on the surface of ZZ-BNC (ZZ-BNC:D3) augmented serum IgG response against JEV and increased protection against lethal JEV infection. The present study suggests that innocuous recombinant antigens, when loaded on the surface of ZZ-BNC, can be transformed to immunogenic antigens.


Asunto(s)
Portadores de Fármacos/administración & dosificación , Virus de la Encefalitis Japonesa (Especie)/inmunología , Vacunas contra la Encefalitis Japonesa/inmunología , Nanocápsulas/administración & dosificación , Proteínas del Envoltorio Viral/inmunología , Animales , Anticuerpos Antivirales/sangre , Virus de la Encefalitis Japonesa (Especie)/genética , Escherichia coli/genética , Femenino , Inmunoglobulina G/sangre , Vacunas contra la Encefalitis Japonesa/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Unión Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteína Estafilocócica A/genética , Proteína Estafilocócica A/metabolismo , Análisis de Supervivencia , Vacunación/métodos , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología , Proteínas del Envoltorio Viral/genética
10.
Nihon Hansenbyo Gakkai Zasshi ; 82(3): 123-32, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24579460

RESUMEN

Pulmonary tuberculosis is an infectious disease caused by Mycobacterium tuberculosis, and continues to be a serious threat to human life. Since M. tuberculosis establishes intracellular parasitism in macrophages, host innate and acquired immune systems have to detect and enhance bactericidal activity against the intracellular bacteria. Understanding of interaction between pathogenic factors of M. tuberculosis and host is also important to understand how immune system copes with the pathogen. In this review, we shortly summarize the mechanisms how innate and acquired immunity recognize M. tuberculosis or M. tuberculosis-infected cells and protects hosts from the infection. Furthermore, IL-17A/IL-23 axis, a recently focused inflammatory cytokine system, is discussed in the context of anti-mycobacterial protective immunity.


Asunto(s)
Inmunidad Adaptativa/inmunología , Inmunidad Innata/inmunología , Interleucina-17/inmunología , Interleucina-23/inmunología , Mycobacterium tuberculosis/inmunología , Tuberculosis Pulmonar/inmunología , Humanos , Interleucina-23/fisiología , Mycobacterium tuberculosis/patogenicidad , Mycobacterium tuberculosis/fisiología , Vacunas contra la Tuberculosis , Tuberculosis Pulmonar/prevención & control
11.
J Immunol ; 184(8): 4414-22, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20212094

RESUMEN

Granulomas play an essential role in the sequestration and killing of mycobacteria in the lung; however, the mechanisms of their development and maturation are still not clearly understood. IL-17A is involved in mature granuloma formation in the mycobacteria-infected lung. Therefore, IL-17A gene-knockout (KO) mice fail to develop mature granulomas in the Mycobacterium bovis bacille Calmette-Guérin (BCG)-infected lung. This study analyzed the mechanism of IL-17A-dependent mature granuloma formation in the mycobacteria-infected lung. The IL-17A KO mice showed a normal level of nascent granuloma formation on day 14 but failed to develop mature granulomas on day 28 after the BCG infection in the lung. The observation implies that IL-17A is required for the maturation of granuloma from the nascent to mature stage. TCR gammadelta T cells expressing TCR Vgamma4 or Vgamma6 were identified as the major IL-17A-producing cells that resided in the BCG-induced lung granuloma. The adoptive transfer of the IL-17A-producing TCR gammadelta T cells reconstituted granuloma formation in the IL-17A KO mice. The expression of ICAM-1 and LFA-1, which are adhesion molecules important in granuloma formation, decreased in the lung of the BCG-infected IL-17A KO mice, and their expression was induced on BCG-infected macrophages in coculture with IL-17A-producing TCR gammadelta T cells. Furthermore, IL-17A KO mice showed not only an impaired mature granuloma formation, but also an impaired protective response to virulent Mycobacterium tuberculosis. Therefore, IL-17A produced by TCR gammadelta T cells plays a critical role in the prevention of M. tuberculosis infection through the induction of mature granuloma formation.


Asunto(s)
Granuloma/inmunología , Granuloma/patología , Interleucina-17/fisiología , Mycobacterium bovis/inmunología , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/patología , Animales , Movimiento Celular/inmunología , Células Cultivadas , Técnicas de Cocultivo , Granuloma/microbiología , Interleucina-17/deficiencia , Interleucina-17/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mycobacterium bovis/patogenicidad , Receptores de Antígenos de Linfocitos T gamma-delta/biosíntesis , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/trasplante , Tuberculosis Pulmonar/prevención & control , Virulencia/inmunología
12.
Infect Immun ; 79(10): 4260-75, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21807905

RESUMEN

The creation of subunit vaccines to prevent malaria infection has been hampered by the intrinsically weak immunogenicity of the recombinant antigens. We have developed a novel strategy to increase immune responses by creating genetic fusion proteins to target specific antigen-presenting cells (APCs). The fusion complex was composed of three physically linked molecular entities: (i) a vaccine antigen, (ii) a multimeric α-helical coiled-coil core, and (iii) an APC-targeting ligand linked to the core via a flexible linker. The vaccine efficacy of the tricomponent complex was evaluated using an ookinete surface protein of Plasmodium vivax, Pvs25, and merozoite surface protein-1 of Plasmodium yoelii. Immunization of mice with the tricomponent complex induced a robust antibody response and conferred substantial levels of P. vivax transmission blockade as evaluated by a membrane feed assay, as well as protection from lethal P. yoelii infection. The observed effect was strongly dependent on the presence of all three components physically integrated as a fusion complex. This system, designated the tricomponent immunopotentiating system (TIPS), onto which any recombinant protein antigens or nonproteinaceous substances could be loaded, may be a promising strategy for devising subunit vaccines or adjuvants against various infectious diseases, including malaria.


Asunto(s)
Diseño de Fármacos , Vacunas contra la Malaria/administración & dosificación , Malaria/prevención & control , Plasmodium vivax/inmunología , Plasmodium yoelii/inmunología , Vacunas de Subunidad/administración & dosificación , Vacunas Sintéticas/administración & dosificación , Secuencia de Aminoácidos , Animales , Células Presentadoras de Antígenos/inmunología , Antígenos de Protozoos/química , Antígenos de Protozoos/genética , Antígenos de Protozoos/inmunología , Antígenos de Superficie/química , Antígenos de Superficie/genética , Antígenos de Superficie/inmunología , Linfocitos B/inmunología , Secuencia de Bases , Proteínas de la Matriz Extracelular/química , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/inmunología , Femenino , Glicoproteínas/química , Glicoproteínas/genética , Glicoproteínas/inmunología , Ligandos , Activación de Linfocitos , Malaria/inmunología , Vacunas contra la Malaria/química , Vacunas contra la Malaria/genética , Vacunas contra la Malaria/inmunología , Malaria Vivax/inmunología , Malaria Vivax/prevención & control , Proteínas Matrilinas , Proteína 1 de Superficie de Merozoito/química , Proteína 1 de Superficie de Merozoito/genética , Proteína 1 de Superficie de Merozoito/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Plasmodium vivax/genética , Plasmodium yoelii/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/inmunología , Vacunas de Subunidad/química , Vacunas de Subunidad/genética , Vacunas de Subunidad/inmunología , Vacunas Sintéticas/química , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
13.
Int Immunol ; 22(4): 307-18, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20167585

RESUMEN

Although the importance of T(h)1-type immune response in protection against mycobacterial infection is well recognized, its regulatory mechanism in the Mycobacterium tuberculosis (Mtb)-infected lung is not well characterized. To address this issue, we analyzed kinetics of induction of mycobacterial antigen-specific CD4(+) T(h)1 T cells after mycobacterial infection in P25 TCR-transgenic (Tg) mice which express TCR alpha and beta chains from a mycobacterial Ag85B-specific MHC class II A(b)-restricted CD4(+) T-cell clone. To supply normal regulatory T-cell repertoire, we transferred normal spleen T cells into the P25 TCR-Tg mice before infection. High dose subcutaneous infection with Mtb or Mycobacterium bovis bacillus Calmette-Guérin (BCG) induced P25 TCR-Tg CD4(+) T(h)1 cells within a week. In contrast, high-dose Mtb or BCG infection into the lung failed to induce P25 TCR-Tg CD4(+) T(h)1 cells at the early stage of the infection. Furthermore, low-dose Mtb infection into the lung induced P25 TCR-Tg CD4(+) T(h)1 cells on day 21 in the mediastinal lymph node but not in the lung. IL-10 was partially involved in the suppression of T(h)1 induction in the lung because pretreatment of mice with anti-IL-10 antibody resulted in increase of P25 TCR-Tg CD4(+) T(h)1 cells in the Mtb-infected lung on day 21 of the infection, whereas neutralization of transforming growth factor-beta, another important suppressive cytokine in the lung, showed no effects on the T(h)1 induction. Our data suggest that induction of anti-mycobacterial CD4(+) T(h)1 cells is suppressed in the mycobacteria-infected lung partially by IL-10.


Asunto(s)
Antígenos Bacterianos/inmunología , Linfocitos T CD4-Positivos/inmunología , Pulmón/inmunología , Mycobacterium bovis/inmunología , Mycobacterium tuberculosis/inmunología , Células TH1/inmunología , Tuberculosis Pulmonar/inmunología , Animales , Regulación hacia Abajo , Interleucina-10/inmunología , Pulmón/microbiología , Ganglios Linfáticos/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Especificidad del Receptor de Antígeno de Linfocitos T
14.
Nihon Rinsho ; 69(8): 1368-72, 2011 Aug.
Artículo en Japonés | MEDLINE | ID: mdl-21838031

RESUMEN

Pulmonary tuberculosis is an infectious disease caused by Mycobacterium tuberculosis which establishes intracellular parasitism in phagosomes of macrophages as initial infection sites. When host immune system recognizes the bacteria, anti-mycobacterial immunity is activated by various cytokines including interleukins, tumor necrosis factor-alpha and interferon-gamma. The cytokines form a network of co-operative action and mutual regulation. In this review, role of the network of interleukins and cytokines in protection against pulmonary tuberculosis will be discussed.


Asunto(s)
Interleucinas/fisiología , Tuberculosis Pulmonar/inmunología , Humanos
15.
Biosci Microbiota Food Health ; 40(2): 84-91, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33996364

RESUMEN

Heat-killed Lactobacillus plantarum L-137 (HK L-137) has anti-allergic, antitumor, and antiviral effects in mice, as well as an anti-inflammatory effect in rats with metabolic syndrome through regulation of immunity. To evaluate the influence of HK L-137 on chronic inflammation in mice with diet-induced obesity, C57BL/6 J mice were fed a normal diet (16% of energy as fat) or a high-fat diet (62% of energy as fat) with or without 0.002% HK L-137 for 4 to 20 weeks. It was found that HK L-137 supplementation alleviated weight gain and elevation of plasma glucose, cholesterol, alanine aminotransferase, and aspartate transaminase levels in mice with diet-induced obesity. Expression of several inflammation-related genes, including F4/80, CD11c, and IL-1ß, in the epididymal adipose tissue of these mice was significantly downregulated by HK L-137. In addition, plasma levels of lipopolysaccharide-binding protein, a marker of endotoxemia, tended to be decreased by administration of HK L-137. These findings suggest that HK L-137 supplementation ameliorates obesity-induced metabolic abnormalities and adipose tissue inflammation, possibly through improvement of intestinal permeability.

16.
Nat Commun ; 12(1): 2299, 2021 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-33863908

RESUMEN

Mycobacterial cell-wall glycolipids elicit an anti-mycobacterial immune response via FcRγ-associated C-type lectin receptors, including Mincle, and caspase-recruitment domain family member 9 (CARD9). Additionally, mycobacteria harbor immuno-evasive cell-wall lipids associated with virulence and latency; however, a mechanism of action is unclear. Here, we show that the DAP12-associated triggering receptor expressed on myeloid cells 2 (TREM2) recognizes mycobacterial cell-wall mycolic acid (MA)-containing lipids and suggest a mechanism by which mycobacteria control host immunity via TREM2. Macrophages respond to glycosylated MA-containing lipids in a Mincle/FcRγ/CARD9-dependent manner to produce inflammatory cytokines and recruit inducible nitric oxide synthase (iNOS)-positive mycobactericidal macrophages. Conversely, macrophages respond to non-glycosylated MAs in a TREM2/DAP12-dependent but CARD9-independent manner to recruit iNOS-negative mycobacterium-permissive macrophages. Furthermore, TREM2 deletion enhances Mincle-induced macrophage activation in vitro and inflammation in vivo and accelerates the elimination of mycobacterial infection, suggesting that TREM2-DAP12 signaling counteracts Mincle-FcRγ-CARD9-mediated anti-mycobacterial immunity. Mycobacteria, therefore, harness TREM2 for immune evasion.


Asunto(s)
Evasión Inmune , Tuberculosis Latente/inmunología , Glicoproteínas de Membrana/metabolismo , Mycobacterium tuberculosis/inmunología , Ácidos Micólicos/metabolismo , Receptores Inmunológicos/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas Adaptadoras de Señalización CARD/genética , Proteínas Adaptadoras de Señalización CARD/metabolismo , Pared Celular/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Glucolípidos/metabolismo , Humanos , Tuberculosis Latente/microbiología , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Masculino , Glicoproteínas de Membrana/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/patogenicidad , Cultivo Primario de Células , Receptores de IgG/metabolismo , Receptores Inmunológicos/genética , Factores de Virulencia/metabolismo
17.
Infect Immun ; 78(9): 3773-82, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20584978

RESUMEN

The nontoxic cholera toxin B subunit (CTB) was evaluated as a potential delivery molecule for the Plasmodium vivax ookinete surface protein, Pvs25. Recombinant Pvs25 was expressed as a secreted protein in the yeast Pichia pastoris, as a mixture of isoforms including multimers and the A and B monomers. The A isoform with the presumed native protein fold was the most abundant, accounting for more than 40% of all expressed protein. The molecularly uniform A isoform was chemically conjugated to CTB via its primary amines, and the fusion protein, retaining GM1-ganglioside affinity, was administered to BALB/c mice by the subcutaneous (s.c.) or intranasal (i.n.) route. Immunization of mice with conjugated Pvs25 without supplemental adjuvant induced antisera that specifically recognized P. vivax ookinetes in vitro. Furthermore, the antisera, when mixed with parasitized blood isolated from P. vivax patients from Thailand, was found to reduce parasite transmission to mosquitoes, conferring a 93 to 98% (s.c.) or a 73 to 88% (i.n.) decrease in oocyst number. Unconjugated Pvs25 alone conferred only a 23 to 60% (s.c.) or a 0 to 6% (i.n.) decrease in oocyst number. Coadministration of extraneous adjuvants, however, further enhanced the vaccine efficacy up to complete blockade. Taken together, we conclude that a weakly immunogenic Pvs25 by itself, when linked to CTB, transforms into a potent transmission-blocking antigen in both i.n. and s.c. routes. In addition, the present study is, to the best of our knowledge, the first demonstration of the immune potentiating function of CTB for a vaccine antigen delivered by the s.c. route.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Antígenos de Protozoos/inmunología , Antígenos de Superficie/inmunología , Toxina del Cólera/administración & dosificación , Vacunas contra la Malaria/inmunología , Malaria Vivax/transmisión , Plasmodium vivax/inmunología , Administración Intranasal , Animales , Anopheles , Femenino , Humanos , Inyecciones Subcutáneas , Vacunas contra la Malaria/administración & dosificación , Malaria Vivax/prevención & control , Ratones , Ratones Endogámicos BALB C , Vacunas Sintéticas/inmunología
18.
Immunology ; 131(3): 377-85, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20575990

RESUMEN

Salmonella enterica serovar Typhimurium (S. typhimurium) causes a localized enteric infection and its elimination is dependent on a T helper type 1 immune response. However, the mechanism of the protective immune response against the pathogen in gut-associated lymphoid tissue (GALT) at an early stage of the infection is not yet clarified. Here, we show that interleukin-17A (IL-17A) was constitutively expressed in GALT; it was also detected on crypt and epithelial cells of the small intestine. Neutralization of the IL-17A in the intestinal lumen exacerbated epithelial damage induced by intestinal S. typhimurium infection at an early stage of the infection. The result suggests that IL-17A has a pivotal role in the immediate early stage of protection against bacterial infection at the intestinal mucosa. As IL-17A neutralization also suppressed the constitutive localization of ß-defensin 3 (BD3), an IL-17A-induced antimicrobial peptide, at the apical site of the intestinal mucosa, it is estimated that IL-17A constitutively induces the expression of the antimicrobial peptide to kill invading pathogens at the epithelial surface immediately after the infection. In contrast, interferon-γ is induced around 3 days after S. typhimurium infection, and its expression level increases thereafter. Taken together, the findings lead to the hypothesis that IL-17A participates in the immediate early stage of protection against S. typhimurium intestinal infection whereas interferon-γ is important at a later stage of the infection.


Asunto(s)
Interleucina-17/metabolismo , Mucosa Intestinal/metabolismo , Salmonella typhi/inmunología , Células TH1/metabolismo , Fiebre Tifoidea/inmunología , Animales , Anticuerpos Bloqueadores/administración & dosificación , Inmunidad Mucosa , Interferón gamma/genética , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-17/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Intestino Delgado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Salmonella typhi/patogenicidad , Células TH1/inmunología , Células TH1/microbiología , Células TH1/patología , beta-Defensinas/genética , beta-Defensinas/inmunología , beta-Defensinas/metabolismo
19.
J Immunol ; 181(5): 3456-63, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18714018

RESUMEN

IL-17A is originally identified as a proinflammatory cytokine that induces neutrophils. Although IL-17A production by CD4(+) Th17 T cells is well documented, it is not clear whether IL-17A is produced and participates in the innate immune response against infections. In the present report, we demonstrate that IL-17A is expressed in the liver of mice infected with Listeria monocytogenes from an early stage of infection. IL-17A is important in protective immunity at an early stage of listerial infection in the liver because IL-17A-deficient mice showed aggravation of the protective response. The major IL-17A-producing cells at the early stage were TCR gammadelta T cells expressing TCR Vgamma4 or Vgamma6. Interestingly, TCR gammadelta T cells expressing both IFN-gamma and IL-17A were hardly detected, indicating that the IL-17A-producing TCR gammadelta T cells are distinct from IFN-gamma-producing gammadelta T cells, similar to the distinction between Th17 and Th1 in CD4(+) T cells. All the results suggest that IL-17A is a newly discovered effector molecule produced by TCR gammadelta T cells, which is important in innate immunity in the liver.


Asunto(s)
Inmunidad Innata , Interleucina-17/inmunología , Listeriosis/inmunología , Hepatopatías/microbiología , Receptores de Antígenos de Linfocitos T gamma-delta , Linfocitos T/inmunología , Animales , Interferón gamma/biosíntesis , Interleucina-17/biosíntesis , Listeria monocytogenes , Ratones , Ratones Noqueados , Subgrupos de Linfocitos T/inmunología
20.
Anticancer Res ; 40(11): 6101-6113, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33109548

RESUMEN

BACKGROUND/AIM: Oral squamous cell carcinoma (OSCC) is a common malignancy with poor prognosis. Therefore, novel therapeutic options are needed to improve prognosis of OSCC. Recently, microRNAs (miRs) have received increasing attention as a potential therapeutic tool for carcinomas. However, no definitive miR-based drugs for patients with OSCC have been reported to date. The aim of this study was to identify new miRs potentially involved in cellular processes associated with OSCC malignancy, which could lead to novel therapeutic strategies. MATERIALS AND METHODS: We identified miRs that are modulated in OSCC and possibly regulate OSCC malignancy, using miR microarray on OSCC cell lines. RESULTS: miR-935 and miR-509-3p were down-regulated in OSCC cell lines and patient tissues. When miR-935 was overexpressed in HSC-3-M3 cells, proliferation, migration, and invasion of the cell line was suppressed, whereas apoptosis was increased. Moreover, we showed that the gene inositol polyphosphate-4-phosphatase type I A (INPP4A) is a potential target whose expression is positively regulated by miR-935. CONCLUSION: miR-935 may function as a tumor suppressor by inhibiting OSCC malignancy via INPP4A induction. Therefore, miR-935 can be a new therapeutic candidate for OSCC treatment.


Asunto(s)
Carcinoma de Células Escamosas/enzimología , Carcinoma de Células Escamosas/genética , MicroARNs/metabolismo , Neoplasias de la Boca/enzimología , Neoplasias de la Boca/genética , Monoéster Fosfórico Hidrolasas/metabolismo , Apoptosis/genética , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Regulación Neoplásica de la Expresión Génica , Humanos , MicroARNs/genética , Neoplasias de la Boca/patología , Invasividad Neoplásica , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda