Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cancer Res ; 84(12): 1978-1995, 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38635895

RESUMEN

T-cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domains (TIGIT) is an inhibitory receptor on immune cells that outcompetes an activating receptor, CD226, for shared ligands. Tumor-infiltrating lymphocytes express TIGIT and CD226 on regulatory T cells (Treg) and on CD8+ T cells with tumor-reactive or exhausted phenotypes, supporting the potential of therapeutically targeting TIGIT to enhance antitumor immunity. To optimize the efficacy of therapeutic antibodies against TIGIT, it is necessary to understand IgG Fc (Fcγ) receptor binding for therapeutic benefit. In this study, we showed that combining Fc-enabled (Fce) or Fc-silent (Fcs) anti-TIGIT with antiprogrammed cell death protein 1 in mice resulted in enhanced control of tumors by differential mechanisms: Fce anti-TIGIT promoted the depletion of intratumoral Treg, whereas Fcs anti-TIGIT did not. Despite leaving Treg numbers intact, Fcs anti-TIGIT potentiated the activation of tumor-specific exhausted CD8+ populations in a lymph node-dependent manner. Fce anti-TIGIT induced antibody-dependent cell-mediated cytotoxicity against human Treg in vitro, and significant decreases in Treg were measured in the peripheral blood of patients with phase I solid tumor cancer treated with Fce anti-TIGIT. In contrast, Fcs anti-TIGIT did not deplete human Treg in vitro and was associated with anecdotal objective clinical responses in two patients with phase I solid tumor cancer whose peripheral Treg frequencies remained stable on treatment. Collectively, these data provide evidence for pharmacologic activity and antitumor efficacy of anti-TIGIT antibodies lacking the ability to engage Fcγ receptor. SIGNIFICANCE: Fcs-silent anti-TIGIT antibodies enhance the activation of tumor-specific pre-exhausted T cells and promote antitumor efficacy without depleting T regulatory cells.


Asunto(s)
Receptores Inmunológicos , Linfocitos T Reguladores , Animales , Linfocitos T Reguladores/inmunología , Ratones , Receptores Inmunológicos/inmunología , Receptores Inmunológicos/antagonistas & inhibidores , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Femenino , Linfocitos T CD8-positivos/inmunología , Ratones Endogámicos C57BL , Línea Celular Tumoral , Neoplasias/inmunología , Neoplasias/tratamiento farmacológico
2.
Nat Chem Biol ; 20(3): 365-372, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37828400

RESUMEN

Stimulator of interferon genes (STING) is a dimeric transmembrane adapter protein that plays a key role in the human innate immune response to infection and has been therapeutically exploited for its antitumor activity. The activation of STING requires its high-order oligomerization, which could be induced by binding of the endogenous ligand, cGAMP, to the cytosolic ligand-binding domain. Here we report the discovery through functional screens of a class of compounds, named NVS-STGs, that activate human STING. Our cryo-EM structures show that NVS-STG2 induces the high-order oligomerization of human STING by binding to a pocket between the transmembrane domains of the neighboring STING dimers, effectively acting as a molecular glue. Our functional assays showed that NVS-STG2 could elicit potent STING-mediated immune responses in cells and antitumor activities in animal models.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas de la Membrana , Animales , Humanos , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Bioensayo , Citosol , Inmunidad Innata , Ligandos , Proteínas de la Membrana/metabolismo
3.
Mol Cancer Ther ; 21(6): 948-959, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35405741

RESUMEN

T cells play a critical role in the control of cancer. The development of immune checkpoint blockers (ICB) aimed at enhancing antitumor T-cell responses has revolutionized cancer treatment. However, durable clinical benefit is observed in only a subset of patients, prompting research efforts to focus on strategies that target multiple inhibitory signals within the tumor microenvironment (TME) to limit tumor evasion and improve patient outcomes. Adenosine has emerged as a potent immune suppressant within the TME, and CD73 is the major enzyme responsible for its extracellular production. CD73 can be co-opted within the TME to impair T-cell-mediated antitumor immunity and promote tumor growth. To target this pathway and block the formation of adenosine, we designed a novel, selective, and potent class of small-molecule inhibitors of CD73, including AB680 (quemliclustat), which is currently being tested in patients with cancer. AB680 effectively restored T-cell proliferation, cytokine secretion, and cytotoxicity that were dampened by the formation of immunosuppressive adenosine by CD73. Furthermore, in an allogeneic mixed lymphocyte reaction where CD73-derived adenosine had a dominant suppressive effect in the presence of PD-1 blockade, AB680 restored T-cell activation and function. Finally, in a preclinical mouse model of melanoma, AB680 inhibited CD73 in the TME and increased the antitumor activity of PD-1 blockade. Collectively, these data provide a rationale for the inhibition of CD73 with AB680 in combination with ICB, such as anti-PD-1, to improve cancer patient outcomes.


Asunto(s)
Melanoma , Receptor de Muerte Celular Programada 1 , Adenosina/metabolismo , Adenosina/farmacología , Adenosina/uso terapéutico , Animales , Humanos , Inhibidores de Puntos de Control Inmunológico , Melanoma/tratamiento farmacológico , Ratones , Receptor de Muerte Celular Programada 1/metabolismo , Microambiente Tumoral
5.
J Immunol ; 202(4): 1153-1162, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30642979

RESUMEN

Endothelial dysfunction and vascular leak, pathogenic hallmarks of severe dengue disease, are directly triggered by dengue virus (DENV) nonstructural protein 1 (NS1). Previous studies have shown that immunization with NS1, as well as passive transfer of NS1-immune serum or anti-NS1 mAb, prevent NS1-mediated lethality in vivo. In this study, we evaluated the immunogenicity and protective capacity of recombinant DENV NS1 administered with cyclic dinucleotides (CDNs), potent activators of innate immune pathways and highly immunogenic adjuvants. Using both wild-type C57BL/6 mice and IFN-α/ß receptor-deficient mice, we show that NS1-CDN immunizations elicit serotype-specific and cross-reactive Ab and T cell responses. Furthermore, NS1-CDN vaccinations conferred significant homotypic and heterotypic protection from DENV2-induced morbidity and mortality. In addition, we demonstrate that high anti-NS1 Ab titers are associated with protection, supporting the role of humoral responses against DENV NS1 as correlates of protection. These findings highlight the potential of CDN-based adjuvants for inducing Ab and T cell responses and validate NS1 as an important candidate for dengue vaccine development.


Asunto(s)
Adyuvantes Inmunológicos , Anticuerpos Antivirales/inmunología , Virus del Dengue/inmunología , Nucleótidos Cíclicos/inmunología , Linfocitos T/inmunología , Proteínas no Estructurales Virales/inmunología , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
6.
Cell Rep ; 25(11): 3074-3085.e5, 2018 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-30540940

RESUMEN

Intratumoral (IT) STING activation results in tumor regression in preclinical models, yet factors dictating the balance between innate and adaptive anti-tumor immunity are unclear. Here, clinical candidate STING agonist ADU-S100 (S100) is used in an IT dosing regimen optimized for adaptive immunity to uncover requirements for a T cell-driven response compatible with checkpoint inhibitors (CPIs). In contrast to high-dose tumor ablative regimens that result in systemic S100 distribution, low-dose immunogenic regimens induce local activation of tumor-specific CD8+ effector T cells that are responsible for durable anti-tumor immunity and can be enhanced with CPIs. Both hematopoietic cell STING expression and signaling through IFNAR are required for tumor-specific T cell activation, and in the context of optimized T cell responses, TNFα is dispensable for tumor control. In a poorly immunogenic model, S100 combined with CPIs generates a survival benefit and durable protection. These results provide fundamental mechanistic insights into STING-induced anti-tumor immunity.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Inmunidad , Proteínas de la Membrana/metabolismo , Neoplasias/inmunología , Animales , Antígeno CTLA-4/metabolismo , Línea Celular Tumoral , Citocinas/metabolismo , Relación Dosis-Respuesta Inmunológica , Resistencia a Antineoplásicos , Hematopoyesis , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neoplasias/patología , Receptor de Muerte Celular Programada 1/metabolismo , Proteínas S100/administración & dosificación , Proteínas S100/inmunología
7.
Cell Rep ; 23(5): 1435-1447, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29719256

RESUMEN

There are a limited number of adjuvants that elicit effective cell-based immunity required for protection against intracellular bacterial pathogens. Here, we report that STING-activating cyclic dinucleotides (CDNs) formulated in a protein subunit vaccine elicit long-lasting protective immunity to Mycobacterium tuberculosis in the mouse model. Subcutaneous administration of this vaccine provides equivalent protection to that of the live attenuated vaccine strain Bacille Calmette-Guérin (BCG). Protection is STING dependent but type I IFN independent and correlates with an increased frequency of a recently described subset of CXCR3-expressing T cells that localize to the lung parenchyma. Intranasal delivery results in superior protection compared with BCG, significantly boosts BCG-based immunity, and elicits both Th1 and Th17 immune responses, the latter of which correlates with enhanced protection. Thus, a CDN-adjuvanted protein subunit vaccine has the capability of eliciting a multi-faceted immune response that results in protection from infection by an intracellular pathogen.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Vacuna BCG/farmacología , Proteínas de la Membrana/inmunología , Mycobacterium tuberculosis/inmunología , Células Th17/inmunología , Tuberculosis Pulmonar/prevención & control , Animales , Vacuna BCG/inmunología , Modelos Animales de Enfermedad , Inmunidad Celular/efectos de los fármacos , Ratones , Ratones Noqueados , Células TH1/inmunología , Células TH1/patología , Células Th17/patología , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/patología , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/farmacocinética
8.
Cancer Immunol Res ; 6(4): 422-433, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29472271

RESUMEN

The cGAS-STING cytosolic DNA sensing pathway may play an integral role in the initiation of antitumor immune responses. Studies evaluating the immunogenicity of various cyclic dinucleotide (CDN) STING agonists administered by intratumoral (i.t.) injection showed potent induction of inflammation, tumor necrosis, and, in some cases, durable tumor-specific adaptive immunity. However, the specific immune mechanisms underlying these responses remain incompletely defined. The majority of these studies have focused on the effect of CDNs on immune cells but have not conclusively interrogated the role of stromal cells in the acute rejection of the CDN-injected tumor. Here, we revealed a mechanism of STING agonist-mediated tumor response that relied on both stromal and immune cells to achieve tumor regression and clearance. Using knockout and bone marrow chimeric mice, we showed that although bone marrow-derived TNFα was necessary for CDN-induced necrosis, STING signaling in radioresistant stromal cells was also essential for CDN-mediated tumor rejection. These results provide evidence for crosstalk between stromal and hematopoietic cells during CDN-mediated tumor collapse after i.t. administration. These mechanistic insights may prove critical in the clinical development of STING agonists. Cancer Immunol Res; 6(4); 422-33. ©2018 AACR.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de la Membrana/agonistas , Neoplasias/etiología , Neoplasias/metabolismo , Nucleótidos Cíclicos/farmacología , Tolerancia a Radiación , Células del Estroma/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Médula Ósea/metabolismo , Línea Celular Tumoral , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunidad Innata , Interferón beta/metabolismo , Melanoma Experimental , Ratones , Ratones Noqueados , Necrosis/metabolismo , Necrosis/patología , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Tolerancia a Radiación/efectos de los fármacos , Tolerancia a Radiación/genética , Transducción de Señal/efectos de los fármacos , Células del Estroma/patología , Células del Estroma/efectos de la radiación , Carga Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
10.
Cell Rep ; 11(7): 1018-30, 2015 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-25959818

RESUMEN

Spontaneous tumor-initiated T cell priming is dependent on IFN-ß production by tumor-resident dendritic cells. On the basis of recent observations indicating that IFN-ß expression was dependent upon activation of the host STING pathway, we hypothesized that direct engagement of STING through intratumoral (IT) administration of specific agonists would result in effective anti-tumor therapy. After proof-of-principle studies using the mouse STING agonist DMXAA showed a potent therapeutic effect, we generated synthetic cyclic dinucleotide (CDN) derivatives that activated all human STING alleles as well as murine STING. IT injection of STING agonists induced profound regression of established tumors in mice and generated substantial systemic immune responses capable of rejecting distant metastases and providing long-lived immunologic memory. Synthetic CDNs have high translational potential as a cancer therapeutic.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de la Membrana/antagonistas & inhibidores , Neoplasias Experimentales/inmunología , Nucleótidos Cíclicos/farmacología , Microambiente Tumoral/inmunología , Animales , Antineoplásicos/síntesis química , Western Blotting , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática , Técnicas de Inactivación de Genes , Humanos , Macrófagos , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neoplasias Experimentales/tratamiento farmacológico , Nucleótidos Cíclicos/síntesis química , Reacción en Cadena de la Polimerasa , Transfección , Xantonas/farmacología
11.
Sci Transl Med ; 7(283): 283ra52, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25877890

RESUMEN

Stimulator of interferon genes (STING) is a cytosolic receptor that senses both exogenous and endogenous cytosolic cyclic dinucleotides (CDNs), activating TBK1/IRF3 (interferon regulatory factor 3), NF-κB (nuclear factor κB), and STAT6 (signal transducer and activator of transcription 6) signaling pathways to induce robust type I interferon and proinflammatory cytokine responses. CDN ligands were formulated with granulocyte-macrophage colony-stimulating factor (GM-CSF)-producing cellular cancer vaccines--termed STINGVAX--that demonstrated potent in vivo antitumor efficacy in multiple therapeutic models of established cancer. We found that rationally designed synthetic CDN derivative molecules, including one with an Rp,Rp dithio diastereomer and noncanonical c[A(2',5')pA(3',5')p] phosphate bridge structure, enhanced antitumor efficacy of STINGVAX in multiple aggressive therapeutic models of established cancer in mice. Antitumor activity was STING-dependent and correlated with increased activation of dendritic cells and tumor antigen-specific CD8(+) T cells. Tumors from STINGVAX-treated mice demonstrated marked PD-L1 (programmed death ligand 1) up-regulation, which was associated with tumor-infiltrating CD8(+)IFNγ(+) T cells. When combined with PD-1 (programmed death 1) blockade, STINGVAX induced regression of palpable, poorly immunogenic tumors that did not respond to PD-1 blockade alone.


Asunto(s)
Antineoplásicos/química , Vacunas contra el Cáncer/química , Proteínas de la Membrana/agonistas , Receptor de Muerte Celular Programada 1/metabolismo , Animales , Linfocitos T CD8-positivos/citología , Línea Celular Tumoral , Citosol/metabolismo , Células Dendríticas/citología , Femenino , Humanos , Interferón gamma/metabolismo , Ligandos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Monocitos/citología , FN-kappa B/metabolismo , Trasplante de Neoplasias , Fosfatos/química , Proteínas Serina-Treonina Quinasas/metabolismo , Factor de Transcripción STAT6/metabolismo
12.
Cell Host Microbe ; 15(2): 203-13, 2014 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-24528866

RESUMEN

Pathogens utilize features of the host response as cues to regulate virulence gene expression. Salmonella enterica serovar Typhimurium (ST) sense Toll-like receptor (TLR)-dependent signals to induce Salmonella Pathogenicity Island 2 (SPI2), a locus required for intracellular replication. To examine pathogenicity in the absence of such cues, we evaluated ST virulence in mice lacking all TLR function (Tlr2(-/-)xTlr4(-/-)xUnc93b1(3d/3d)). When delivered systemically to TLR-deficient mice, ST do not require SPI2 and maintain virulence by replicating extracellularly. In contrast, SPI2 mutant ST are highly attenuated after oral infection of the same mice, revealing a role for SPI2 in the earliest stages of infection, even when intracellular replication is not required. This early requirement for SPI2 is abolished in MyD88(-/-)xTRIF(-/-) mice lacking both TLR- and other MyD88-dependent signaling pathways, a potential consequence of compromised intestinal permeability. These results demonstrate how pathogens use plasticity in virulence strategies to respond to different host immune environments.


Asunto(s)
Interacciones Huésped-Patógeno , Inmunidad Innata , Salmonella typhimurium/inmunología , Salmonella typhimurium/patogenicidad , Transducción de Señal , Receptores Toll-Like/inmunología , Animales , Ratones , Ratones Noqueados , Receptores Toll-Like/deficiencia , Virulencia
13.
Cell ; 144(5): 675-88, 2011 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-21376231

RESUMEN

Toll-like receptors (TLRs) contribute to host resistance to microbial pathogens and can drive the evolution of virulence mechanisms. We have examined the relationship between host resistance and pathogen virulence using mice with a functional allele of the nramp-1 gene and lacking combinations of TLRs. Mice deficient in both TLR2 and TLR4 were highly susceptible to the intracellular bacterial pathogen Salmonella typhimurium, consistent with reduced innate immune function. However, mice lacking additional TLRs involved in S. typhimurium recognition were less susceptible to infection. In these TLR-deficient cells, bacteria failed to upregulate Salmonella pathogenicity island 2 (SPI-2) genes and did not form a replicative compartment. We demonstrate that TLR signaling enhances the rate of acidification of the Salmonella-containing phagosome, and inhibition of this acidification prevents SPI-2 induction. Our results indicate that S. typhimurium requires cues from the innate immune system to regulate virulence genes necessary for intracellular survival, growth, and systemic infection.


Asunto(s)
Interacciones Huésped-Patógeno , Inmunidad Innata , Salmonella typhimurium/inmunología , Salmonella typhimurium/patogenicidad , Transducción de Señal , Receptores Toll-Like/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Línea Celular , Humanos , Macrófagos/inmunología , Macrófagos/microbiología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Receptores Toll-Like/inmunología
14.
J Immunol ; 184(4): 2065-75, 2010 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-20083670

RESUMEN

Uropathogenic Escherichia coli is the causative agent for >80% of uncomplicated urinary tract infections (UTIs). Uropathogenic E. coli strains express a number of virulence and fitness factors that allow successful colonization of the mammalian bladder. To combat this, the host has distinct mechanisms to prevent adherence to the bladder wall and to detect and kill uropathogenic E. coli in the event of colonization. In this study, we investigated the role of IL-17A, an innate-adaptive immunomodulatory cytokine, during UTI using a murine model. Splenocytes isolated from mice infected by the transurethral route robustly expressed IL-17A in response to in vitro stimulation with uropathogenic E. coli Ags. Transcript expression of IL-17A in the bladders of infected mice correlated with a role in the innate immune response to UTI, and gammadelta cells seem to be a key source of IL-17A production. Although IL-17A seems to be dispensable for the generation of a protective response to uropathogenic E. coli, its importance in innate immunity is demonstrated by a defect in acute clearance of uropathogenic E. coli in IL-17A(-/-) mice. This clearance defect is likely a result of deficient cytokine and chemokine transcripts and impaired macrophage and neutrophil influx during infection. These results show that IL-17A is a key mediator for the innate immune response to UTIs.


Asunto(s)
Infecciones por Escherichia coli/inmunología , Escherichia coli/inmunología , Inmunidad Innata , Interleucina-17/fisiología , Infecciones Urinarias/inmunología , Animales , Modelos Animales de Enfermedad , Escherichia coli/patogenicidad , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Interleucina-17/deficiencia , Interleucina-17/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T gamma-delta/deficiencia , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Infecciones Urinarias/metabolismo , Infecciones Urinarias/microbiología
15.
Infect Immun ; 78(2): 568-85, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19917708

RESUMEN

Urinary tract infection (UTI) caused by uropathogenic Escherichia coli (UPEC) is a substantial economic and societal burden-a formidable public health issue. Symptomatic UTI causes significant discomfort in infected patients, results in lost productivity, predisposes individuals to more serious infections, and usually necessitates antibiotic therapy. There is no licensed vaccine available for prevention of UTI in humans in the United States, likely due to the challenge of targeting a relatively heterogeneous group of pathogenic strains in a unique physiological niche. Despite significant advances in the understanding of UPEC biology, mechanistic details regarding the host response to UTI and full comprehension of genetic loci that influence susceptibility require additional work. Currently, there is an appreciation for the role of classic innate immune responses-from pattern receptor recognition to recruitment of phagocytic cells-that occur during UPEC-mediated UTI. There is, however, a clear disconnect regarding how factors involved in the innate immune response to UPEC stimulate acquired immunity that facilitates enhanced clearance upon reinfection. Unraveling the molecular details of this process is vital in the development of a successful vaccine for prevention of human UTI. Here, we survey the current understanding of host responses to UPEC-mediated UTI with an eye on molecular and cellular factors whose activity may be harnessed by a vaccine that stimulates lasting and sterilizing immunity.


Asunto(s)
Infecciones por Escherichia coli , Infecciones Urinarias/microbiología , Animales , Escherichia coli , Humanos
16.
PLoS Pathog ; 5(9): e1000586, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19806177

RESUMEN

Uncomplicated infections of the urinary tract, caused by uropathogenic Escherichia coli, are among the most common diseases requiring medical intervention. A preventive vaccine to reduce the morbidity and fiscal burden these infections have upon the healthcare system would be beneficial. Here, we describe the results of a large-scale selection process that incorporates bioinformatic, genomic, transcriptomic, and proteomic screens to identify six vaccine candidates from the 5379 predicted proteins encoded by uropathogenic E. coli strain CFT073. The vaccine candidates, ChuA, Hma, Iha, IreA, IroN, and IutA, all belong to a functional class of molecules that is involved in iron acquisition, a process critical for pathogenesis in all microbes. Intranasal immunization of CBA/J mice with these outer membrane iron receptors elicited a systemic and mucosal immune response that included the production of antigen-specific IgM, IgG, and IgA antibodies. The cellular response to vaccination was characterized by the induction and secretion of IFN-gamma and IL-17. Of the six potential vaccine candidates, IreA, Hma, and IutA provided significant protection from experimental infection. In immunized animals, class-switching from IgM to IgG and production of antigen-specific IgA in the urine represent immunological correlates of protection from E. coli bladder colonization. These findings are an important first step toward the development of a subunit vaccine to prevent urinary tract infections and demonstrate how targeting an entire class of molecules that are collectively required for pathogenesis may represent a fundamental strategy to combat infections.


Asunto(s)
Antígenos Bacterianos/inmunología , Infecciones por Escherichia coli/prevención & control , Proteínas de Escherichia coli/inmunología , Vacunas contra Escherichia coli/inmunología , Infecciones Urinarias/prevención & control , Administración Intranasal , Animales , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/genética , Antígenos Bacterianos/metabolismo , Células Cultivadas , Escherichia coli/inmunología , Infecciones por Escherichia coli/inmunología , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Vacunas contra Escherichia coli/administración & dosificación , Femenino , Inmunoglobulina A Secretora/metabolismo , Inmunoglobulina A Secretora/orina , Cambio de Clase de Inmunoglobulina/inmunología , Interferón gamma/metabolismo , Interleucina-17/metabolismo , Ratones , Ratones Endogámicos CBA , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Bazo/citología , Bazo/inmunología , Bazo/metabolismo , Estadísticas no Paramétricas , Infecciones Urinarias/inmunología
18.
Proc Natl Acad Sci U S A ; 105(29): 10191-6, 2008 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-18632558

RESUMEN

To gain insight into the interaction of intracellular pathogens with host innate immune pathways, we performed an unbiased genetic screen of Listeria monocytogenes mutants that induced an enhanced or diminished host innate immune response. Here, we show that the major facilitator superfamily of bacterial multidrug resistance transporters (MDRs) controlled the magnitude of a host cytosolic surveillance pathway, leading to the production of several cytokines, including type I IFN. Mutations mapping to repressors of MDRs resulted in ectopic expression of their cognate transporters, leading to host responses that were increased up to 20-fold over wild-type bacteria, and a 20-fold decrease in bacterial growth in vivo. Mutation of one of the MDRs, MdrM, led to a 3-fold reduction in the IFN-beta response to L. monocytogenes infection, indicating a pivotal role for MdrM in activation of the host cytosolic surveillance system. Bacterial MDRs had previously been associated with resistance to antibiotics and other toxic compounds. This report links bacterial MDRs and host immunity. Understanding the mechanisms through which live pathogens activate innate immune signaling pathways should lead to the discovery of adjuvants, vaccines, and perhaps new classes of therapeutics. Indeed, we show that the mutants identified in this screen induced vastly altered type I IFN response in vivo as well.


Asunto(s)
Subfamilia B de Transportador de Casetes de Unión a ATP/inmunología , Proteínas Bacterianas/inmunología , Inmunidad Innata , Listeria monocytogenes/inmunología , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Animales , Proteínas Bacterianas/genética , Citosol/inmunología , Genes Bacterianos , Genes MDR , Interferón beta/biosíntesis , Listeria monocytogenes/genética , Listeria monocytogenes/patogenicidad , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/genética
19.
Infect Immun ; 74(11): 6124-34, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16954394

RESUMEN

The secreted autotransporter toxin (Sat), found predominantly in uropathogenic Escherichia coli, is a member of the SPATE (serine protease autotransporters of Enterobacteriaceae) family and, as such, has serine protease activity and causes cytopathic effects on various cell types. To assess the contribution of the serine protease active site to the mechanism of action of Sat, mutations were made in the first (S256I), in the second (S258A), or in both (S256I/S258A) serine residues within the active site motif. Mutations in the first or both serines reduced protease activity to background levels (P<0.001); a single mutation in the second serine reduced activity by 60% compared to wild type (P<0.001). After reversion of the S256I mutation to wild type (I256S), we confirmed S256 as the catalytically active serine. None of these mutations affected secretion of the mature passenger domain or release into the supernatant. The S256I mutation, however, abrogated the cytotoxicity of Sat on human bladder (UM-UC-3) and kidney (HEK 293) epithelial cells, characterized by rounding and elongation, respectively, and a high level of cell detachment. Moreover, S256 is essential for Sat to mediate cytoskeletal contraction and actin loss in host cells as well as to degrade specific membrane/cytoskeletal (fodrin and leukocyte function-associated molecule 1) and nuclear [microtubule-associated proteins, LIM domain-only protein 7, Rap GTPase-activating protein, poly(ADP-ribose) polymerase] proteins in vitro. Lastly, Sat was internalized by host cells and localized to the cytoskeletal fraction where membrane/cytoskeletal target proteins reside.


Asunto(s)
Toxinas Bacterianas/toxicidad , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/toxicidad , Escherichia coli/enzimología , Escherichia coli/patogenicidad , Riñón/citología , Serina Endopeptidasas/metabolismo , Serina Endopeptidasas/toxicidad , Vejiga Urinaria/citología , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Línea Celular , Células Cultivadas , Escherichia coli/genética , Infecciones por Escherichia coli/enzimología , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli/genética , Humanos , Riñón/enzimología , Riñón/microbiología , Mutagénesis Sitio-Dirigida , Serina Endopeptidasas/genética , Vejiga Urinaria/enzimología , Vejiga Urinaria/microbiología , Urotelio/citología , Urotelio/enzimología , Urotelio/microbiología
20.
J Cell Biol ; 173(6): 853-9, 2006 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-16785320

RESUMEN

Misfolded proteins in the endoplasmic reticulum (ER) are retained in the organelle or retrotranslocated to the cytosol for proteasomal degradation. ER chaperones that guide these opposing processes are largely unknown. We developed a semipermeabilized cell system to study the retrotranslocation of cholera toxin (CT), a toxic agent that crosses the ER membrane to reach the cytosol during intoxication. We found that protein disulfide isomerase (PDI) facilitates CT retrotranslocation, whereas ERp72, a PDI-like protein, mediates its ER retention. In vitro analysis revealed that PDI and ERp72 alter CT's conformation in a manner consistent with their roles in retrotranslocation and ER retention. Moreover, we found that PDI's and ERp72's opposing functions operate on endogenous ER misfolded proteins. Thus, our data identify PDI family proteins that play opposing roles in ER quality control and establish an assay to further delineate the mechanism of CT retrotranslocation.


Asunto(s)
Retículo Endoplásmico/metabolismo , Glicoproteínas de Membrana/fisiología , Proteína Disulfuro Isomerasas/fisiología , Transporte de Proteínas/fisiología , Técnicas de Cultivo de Célula , Toxina del Cólera/química , Toxina del Cólera/metabolismo , Regulación hacia Abajo , Retículo Endoplásmico/fisiología , Retículo Endoplásmico/ultraestructura , Células HeLa , Humanos , Glicoproteínas de Membrana/genética , Pliegue de Proteína , Vibrio cholerae/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...