Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Microbiol ; 9(3): 727-736, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38374245

RESUMEN

In sub-Saharan Africa, multidrug-resistant non-typhoidal Salmonella serovars are a common cause of fatal bloodstream infection. Malnutrition is a predisposing factor, but the underlying mechanisms are unknown. Here we show that vitamin A deficiency, one of the most prevalent micronutrient deficits afflicting African children, increases susceptibility to disseminated non-typhoidal Salmonella disease in mice and impairs terminal neutrophil maturation. Immature neutrophils had reduced expression of Slc11a1, a gene that encodes a metal ion transporter generally thought to restrict pathogen growth in macrophages. Adoptive transfer of SLC11A1-proficient neutrophils, but not SLC11A1-deficient neutrophils, reduced systemic Salmonella burden in Slc11a1-/- mice or mice with vitamin A deficiency. Loss of terminal granulopoiesis regulator CCAAT/enhancer-binding protein ϵ (C/EBPϵ) also decreased neutrophil-mediated control of Salmonella, but not that mediated by peritoneal macrophages. Susceptibility to infection increased in Cebpe-/- Slc11a1+/+ mice compared with wild-type controls, in an Slc11a1-expression-dependent manner. These data suggest that SLC11A1 deficiency impairs Salmonella control in part by blunting neutrophil-mediated defence.


Asunto(s)
Salmonelosis Animal , Deficiencia de Vitamina A , Niño , Ratones , Humanos , Animales , Neutrófilos , Salmonella , Macrófagos
2.
PLoS Pathog ; 20(1): e1011914, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38166152

RESUMEN

Chlamydia vaccine approaches aspire to induce Th1 cells for optimal protection, despite the fact that there is no direct evidence demonstrating Th1-mediated Chlamydia clearance from the female reproductive tract (FRT). We recently reported that T-bet-deficient mice can resolve primary Chlamydia infection normally, undermining the potentially protective role of Th1 cells in Chlamydia immunity. Here, we show that T-bet-deficient mice develop robust Th17 responses and that mice deficient in Th17 cells exhibit delayed bacterial clearance, demonstrating that Chlamydia-specific Th17 cells represent an underappreciated protective population. Additionally, Th2-deficient mice competently clear cervicovaginal infection. Furthermore, we show that sensing of IFN-γ by non-hematopoietic cells is essential for Chlamydia immunity, yet bacterial clearance in the FRT does not require IFN-γ secretion by CD4 T cells. Despite the fact that Th1 cells are not necessary for Chlamydia clearance, protective immunity to Chlamydia is still dependent on MHC class-II-restricted CD4 T cells and IL-12p40. Together, these data point to IL-12p40-dependent CD4 effector maturation as essential for Chlamydia immunity, and Th17 cells to a lesser extent, yet neither Th1 nor Th2 cell development is critical. Future Chlamydia vaccination efforts will be more effective if they focus on induction of this protective CD4 T cell population.


Asunto(s)
Infecciones por Chlamydia , Chlamydia muridarum , Animales , Femenino , Ratones , Linfocitos T CD4-Positivos , Infecciones por Chlamydia/genética , Infecciones por Chlamydia/microbiología , Subunidad p40 de la Interleucina-12 , Ratones Endogámicos C57BL , Células TH1 , Células Th17 , Células Th2
3.
Vaccine ; 41(43): 6426-6433, 2023 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-37739887

RESUMEN

Salmonella infections cause a wide range of intestinal and systemic disease that affects global human health. While some vaccines are available, they do not mitigate the impact of Salmonella on endemic areas. Research using Salmonella mouse models has revealed the important role of CD4 T cells and antibody in the development of protective immunity against Salmonella infection. Recent work points to a critical role for hepatic tissue-resident memory lymphocytes in naturally acquired immunity to systemic infection. Thus, understanding the genesis and function of this Salmonella-specific population is an important objective and is the primary focus of this review. Greater understanding of how these memory lymphocytes contribute to bacterial elimination could suggest new approaches to vaccination against an important human pathogen.

4.
J Immunol ; 211(2): 180-185, 2023 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-37283516

RESUMEN

CD4 tissue-resident memory T cells (TRMs) allow robust protection of barrier surfaces against pathogens. We investigated the role of T-bet in the formation of liver CD4 TRMs using mouse models. T-bet-deficient CD4 T cells did not efficiently form liver TRMs when compared with wild-type (WT). In addition, ectopic expression of T-bet enhanced the formation of liver CD4 TRMs, but only when in competition with WT CD4 T cells. Liver TRMs also expressed higher levels of CD18, which was T-bet dependent. The WT competitive advantage was blocked by Ab neutralization of CD18. Taken together, our data show that activated CD4 T cells compete for entry to liver niches via T-bet-induced expression of CD18, allowing TRM precursors to access subsequent hepatic maturation signals. These findings uncover an essential role for T-bet in liver TRM CD4 formation and suggest targeted enhancement of this pathway could increase the efficacy of vaccines that require hepatic TRMs.


Asunto(s)
Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos , Animales , Ratones , Memoria Inmunológica , Hígado , Células T de Memoria , Antígenos CD18
5.
Proc Natl Acad Sci U S A ; 120(16): e2214699120, 2023 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-37040404

RESUMEN

Hepatic CD4 tissue-resident memory T cells (TRM) are required for robust protection against Salmonella infection; however, the generation of this T cell population is poorly understood. To interrogate the contribution of inflammation, we developed a simple Salmonella-specific T cell transfer system that allowed direct visualization of hepatic TRM formation. Salmonella-specific (SM1) T cell receptor (TCR) transgenic CD4 T cells were activated in vitro and adoptively transferred into C57BL/6 mice while hepatic inflammation was induced by acetaminophen overdose or L. monocytogenes infection. In both model systems, hepatic CD4 TRM formation was accentuated by local tissue responses. Liver inflammation also enhanced the suboptimal protection provided by a subunit Salmonella vaccine which typically induces circulating memory CD4 T cells. To further elucidate the mechanism of CD4 TRM formation in response to liver inflammation, various cytokines were examined by RNAseq, bone marrow chimeras, and in vivo neutralization. Surprisingly, IL-2 and IL-1 were found to enhance CD4 TRM formation. Thus, local inflammatory mediators enhance CD4 TRM populations and can boost the protective immunity provided by a suboptimal vaccine. This knowledge will be foundational for the development of a more effective vaccine against invasive nontyphoidal salmonellosis (iNTS).


Asunto(s)
Linfocitos T CD4-Positivos , Vacunas , Ratones , Animales , Interleucina-2 , Memoria Inmunológica , Células T de Memoria , Ratones Endogámicos C57BL , Hígado , Inflamación , Interleucina-1
6.
Cell Host Microbe ; 30(6): 836-847.e6, 2022 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-35568027

RESUMEN

Changes in the microbiota composition are associated with many human diseases, but factors that govern strain abundance remain poorly defined. We show that a commensal Escherichia coli strain and a pathogenic Salmonella enterica serovar Typhimurium isolate both utilize nitrate for intestinal growth, but each accesses this resource in a distinct biogeographical niche. Commensal E. coli utilizes epithelial-derived nitrate, whereas nitrate in the niche occupied by S. Typhimurium is derived from phagocytic infiltrates. Surprisingly, avirulent S. Typhimurium was shown to be unable to utilize epithelial-derived nitrate because its chemotaxis receptors McpB and McpC exclude the pathogen from the niche occupied by E. coli. In contrast, E. coli invades the niche constructed by S. Typhimurium virulence factors and confers colonization resistance by competing for nitrate. Thus, nutrient niches are not defined solely by critical resources, but they can be further subdivided biogeographically within the host into distinct microhabitats, thereby generating new niche opportunities for distinct bacterial species.


Asunto(s)
Microbioma Gastrointestinal , Salmonella typhimurium , Escherichia coli , Humanos , Nitratos , Nutrientes
7.
Immunohorizons ; 6(2): 184-190, 2022 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-35210292

RESUMEN

The presence of memory lymphocytes in nonlymphoid tissues reflects prior immunological experience and can provide nonspecific defense against infection. In this study, we used a mouse cohousing approach to examine the effect of prior immunological experience on Salmonella and Chlamydia infection. As expected, cohousing of "dirty mice" with specific pathogen-free laboratory mice increased the frequency of effector memory T cells in laboratory mice and enhanced protection against systemic Listeria infection. In contrast, the course of systemic infection with Salmonella and mucosal infection with Chlamydia was largely unaffected by cohousing, despite enhanced frequencies of memory T cells. Thus, cohousing of laboratory mice reliably increases the proportion of memory T cells in circulation, but can it have variable effects on pathogen clearance.


Asunto(s)
Infecciones por Chlamydia , Memoria Inmunológica , Animales , Linfocitos T CD8-positivos , Infecciones por Chlamydia/microbiología , Células T de Memoria , Ratones , Ratones Endogámicos C57BL
8.
PLoS Pathog ; 18(2): e1010333, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35196366

RESUMEN

Protective immune responses to Chlamydia infection within the female reproductive tract (FRT) are incompletely understood. MHC class II-restricted CD4 Th1 responses are believed to be vital for bacterial clearance due to their capacity to secrete IFN-γ, but an essential requirement for T-bet-expressing Th1 cells has yet to be demonstrated in the mouse model of Chlamydia infection. Here, we investigated the role of T-bet and IFN-γ in primary clearance of Chlamydia after FRT infection. Surprisingly, IFN-γ producing CD4 T cells from the FRT expressed low levels of T-bet throughout infection, suggesting that classical T-bet-expressing Th1 cells are inefficiently generated and therefore unlikely to participate in bacteria clearance. Furthermore, mice deficient in T-bet expression or with a CD4-specific T-bet deficiency cleared FRT infection similarly to wild-type controls. T-bet-deficient mice displayed significant skewing of FRT CD4 T cells towards Th17 responses, demonstrating that compensatory effector pathways are generated in the absence of Th1 cells. In marked contrast, IFN-γ-, and IFN-γR-deficient mice were able to reduce FRT bacterial burdens, but suffered systemic bacterial dissemination and 100% mortality. Together, these data demonstrate that IFN-γ signaling is essential to protect mice from fatal systemic disease, but that classical T-bet-expressing Th1 cells are non-essential for primary clearance within the FRT. Exploring the protective contribution of Th1 cells versus other CD4 effector lineages could provide important information for the generation of new Chlamydia vaccines.


Asunto(s)
Infecciones por Chlamydia , Chlamydia , Infecciones del Sistema Genital , Animales , Linfocitos T CD4-Positivos , Infecciones por Chlamydia/microbiología , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Dominio T Box/genética , Células TH1 , Células Th17
9.
PLoS Pathog ; 17(10): e1010004, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34695149

RESUMEN

While Salmonella enterica is seen as an archetypal facultative intracellular bacterial pathogen where protection is mediated by CD4+ T cells, identifying circulating protective cells has proved very difficult, inhibiting steps to identify key antigen specificities. Exploiting a mouse model of vaccination, we show that the spleens of C57BL/6 mice vaccinated with live-attenuated Salmonella serovar Typhimurium (S. Typhimurium) strains carried a pool of IFN-γ+ CD4+ T cells that could adoptively transfer protection, but only transiently. Circulating Salmonella-reactive CD4+ T cells expressed the liver-homing chemokine receptor CXCR6, accumulated over time in the liver and assumed phenotypic characteristics associated with tissue-associated T cells. Liver memory CD4+ T cells showed TCR selection bias and their accumulation in the liver could be inhibited by blocking CXCL16. These data showed that the circulation of CD4+ T cells mediating immunity to Salmonella is limited to a brief window after which Salmonella-specific CD4+ T cells migrate to peripheral tissues. Our observations highlight the importance of triggering tissue-specific immunity against systemic infections.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Memoria Inmunológica/inmunología , Hígado/inmunología , Salmonelosis Animal/inmunología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Salmonella typhimurium/inmunología
10.
Proc Natl Acad Sci U S A ; 118(21)2021 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-34001624

RESUMEN

Anatomical positioning of memory lymphocytes within barrier tissues accelerates secondary immune responses and is thought to be essential for protection at mucosal surfaces. However, it remains unclear whether resident memory in the female reproductive tract (FRT) is required for Chlamydial immunity. Here, we describe efficient generation of tissue-resident memory CD4 T cells and memory lymphocyte clusters within the FRT after vaginal infection with Chlamydia Despite robust establishment of localized memory lymphocytes within the FRT, naïve mice surgically joined to immune mice, or mice with only circulating immunity following intranasal immunization, were fully capable of resisting Chlamydia infection via the vaginal route. Blocking the rapid mobilization of circulating memory CD4 T cells to the FRT inhibited this protective response. These data demonstrate that secondary protection in the FRT can occur in the complete absence of tissue-resident immune cells. The ability to confer robust protection to barrier tissues via circulating immune memory provides an unexpected opportunity for vaccine development against infections of the FRT.


Asunto(s)
Anticuerpos Antibacterianos/biosíntesis , Linfocitos T CD4-Positivos/inmunología , Infecciones por Chlamydia/prevención & control , Chlamydia muridarum/inmunología , Genitales Femeninos/inmunología , Inmunización/métodos , Administración Intranasal , Administración Intravaginal , Animales , Antígenos Bacterianos/administración & dosificación , Vacunas Bacterianas/administración & dosificación , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/microbiología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/inmunología , Infecciones por Chlamydia/inmunología , Infecciones por Chlamydia/microbiología , Chlamydia muridarum/efectos de los fármacos , Chlamydia muridarum/crecimiento & desarrollo , Chlamydia muridarum/patogenicidad , Femenino , Genitales Femeninos/efectos de los fármacos , Genitales Femeninos/microbiología , Inmunidad Mucosa/efectos de los fármacos , Memoria Inmunológica/efectos de los fármacos , Ratones , Parabiosis/métodos
11.
Nat Commun ; 12(1): 541, 2021 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-33483492

RESUMEN

CD4 T follicular helper (Tfh) cells are important for the generation of durable and specific humoral protection against viral infections. The degree to which SARS-CoV-2 infection generates Tfh cells and stimulates the germinal center (GC) response is an important question as we investigate vaccine induced immunity against COVID-19. Here, we report that SARS-CoV-2 infection in rhesus macaques, either infused with convalescent plasma, normal plasma, or receiving no infusion, resulted in transient accumulation of pro-inflammatory monocytes and proliferating Tfh cells with a Th1 profile in peripheral blood. CD4 helper cell responses skewed predominantly toward a Th1 response in blood, lung, and lymph nodes. SARS-CoV-2 Infection induced GC Tfh cells specific for the SARS-CoV-2 spike and nucleocapsid proteins, and a corresponding early appearance of antiviral serum IgG antibodies. Collectively, the data show induction of GC responses in a rhesus model of mild COVID-19.


Asunto(s)
COVID-19/inmunología , Centro Germinal/inmunología , SARS-CoV-2/inmunología , Células T Auxiliares Foliculares/inmunología , Células TH1/inmunología , Animales , Anticuerpos Antivirales/sangre , COVID-19/terapia , Línea Celular , Chlorocebus aethiops , Proteínas de la Nucleocápside de Coronavirus/inmunología , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunidad Humoral/inmunología , Inmunización Pasiva , Inmunogenicidad Vacunal/inmunología , Inmunoglobulina G/sangre , Macaca mulatta , Masculino , Fosfoproteínas/inmunología , SARS-CoV-2/aislamiento & purificación , Glicoproteína de la Espiga del Coronavirus/inmunología , Células Vero , Sueroterapia para COVID-19
12.
PLoS One ; 15(7): e0235518, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32614928

RESUMEN

Interruption of the programmed death 1 (PD-1) / programmed death ligand 1 (PD-L1) pathway is an established and effective therapeutic strategy in human oncology and holds promise for veterinary oncology. We report the generation and characterization of monoclonal antibodies specific for canine PD-1 and PD-L1. Antibodies were initially assessed for their capacity to block the binding of recombinant canine PD-1 to recombinant canine PD-L1 and then ranked based on efficiency of binding as judged by flow cytometry. Selected antibodies were capable of detecting PD-1 and PD-L1 on canine tissues by flow cytometry and Western blot. Anti-PD-L1 worked for immunocytochemistry and anti-PD-1 worked for immunohistochemistry on formalin-fixed paraffin embedded canine tissues, suggesting the usage of this antibody with archived tissues. Additionally, anti-PD-L1 (JC071) revealed significantly increased PD-L1 expression on canine monocytes after stimulation with peptidoglycan or lipopolysaccharide. Together, these antibodies display specificity for the natural canine ligand using a variety of potential diagnostic applications. Importantly, multiple PD-L1-specific antibodies amplified IFN-γ production in a canine peripheral blood mononuclear cells (PBMC) concanavlin A (Con A) stimulation assay, demonstrating functional activity.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Antígeno B7-H1/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Linfocitos T/inmunología , Animales , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Perros , Interferón gamma/metabolismo , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/metabolismo , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos BALB C , Monocitos/efectos de los fármacos , Monocitos/inmunología , Monocitos/metabolismo , Peptidoglicano/farmacología , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/metabolismo , Linfocitos T/metabolismo
13.
bioRxiv ; 2020 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-32676606

RESUMEN

CD4 T follicular helper (T fh ) cells are important for the generation of long-lasting and specific humoral protection against viral infections. The degree to which SARS-CoV-2 infection generates T fh cells and stimulates the germinal center response is an important question as we investigate vaccine options for the current pandemic. Here we report that, following infection with SARS-CoV-2, adult rhesus macaques exhibited transient accumulation of activated, proliferating T fh cells in their peripheral blood on a transitory basis. The CD4 helper cell responses were skewed predominantly toward a T h 1 response in blood, lung, and lymph nodes, reflective of the interferon-rich cytokine environment following infection. We also observed the generation of germinal center T fh cells specific for the SARS-CoV-2 spike (S) and nucleocapsid (N) proteins, and a corresponding early appearance of antiviral serum IgG antibodies but delayed or absent IgA antibodies. Our data suggest that a vaccine promoting Th1-type Tfh responses that target the S protein may lead to protective immunity.

14.
mBio ; 11(3)2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32487756

RESUMEN

The inflammatory response to Chlamydia infection is likely to be multifactorial and involve a variety of ligand-dependent and -independent recognition pathways. We previously reported the presence of NOD1/NOD2-dependent endoplasmic reticulum (ER) stress-induced inflammation during Chlamydia muridarum infection in vitro, but the relevance of this finding to an in vivo context is unclear. Here, we examined the ER stress response to in vivo Chlamydia infection. The induction of interleukin 6 (IL-6) production after systemic Chlamydia infection correlated with expression of ER stress response genes. Furthermore, when tauroursodeoxycholate (TUDCA) was used to inhibit the ER stress response, an increased bacterial burden was detected, suggesting that ER stress-driven inflammation can contribute to systemic bacterial clearance. Mice lacking both NOD1 and NOD2 or RIP2 exhibited slightly higher systemic bacterial burdens after infection with Chlamydia Overall, these data suggest a model where RIP2 and NOD1/NOD2 proteins link ER stress responses with the induction of Chlamydia-specific inflammatory responses.IMPORTANCE Understanding the initiation of the inflammatory response during Chlamydia infection is of public health importance given the impact of this disease on young women in the United States. Many young women are chronically infected with Chlamydia but are asymptomatic and therefore do not seek treatment, leaving them at risk of long-term reproductive harm due to inflammation in response to infection. Our manuscript explores the role of the endoplasmic reticulum stress response pathway initiated by an innate receptor in the development of this inflammation.


Asunto(s)
Infecciones por Chlamydia/inmunología , Estrés del Retículo Endoplásmico/genética , Inmunidad Innata , Proteína Adaptadora de Señalización NOD1/genética , Proteína Adaptadora de Señalización NOD2/genética , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/genética , Animales , Carga Bacteriana , Chlamydia muridarum , Inflamación , Ratones , Ratones Endogámicos C57BL , Transducción de Señal , Organismos Libres de Patógenos Específicos
15.
Eur J Immunol ; 50(5): 676-684, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32026472

RESUMEN

The obligate intracellular bacterium Chlamydia trachomatis causes the most prevalent bacterial sexually transmitted infection worldwide. CD4 T cells play a central role in the protective immunity against Chlamydia female reproductive tract (FRT) infection, while B cells are thought to be dispensable for resolution of primary Chlamydia infection in mouse models. We recently reported an unexpected requirement of B cells in local Chlamydia-specific CD4 T-cell priming and bacterial containment within the FRT. Here, we sought to tackle the precise effector function of B cells during Chlamydia primary infection. Using mixed bone marrow chimeras that lack B-cell-dependent Ag presentation (MHCIIB-/- ) or devoid of circulating antibodies (AID-/- × µS-/- ), we show that Chlamydia-specific CD4 T-cell expansion does not rely on Ag presentation by B cells. Importantly, we demonstrate that antibody, but not B-cell-dependent Ag presentation, is required for preventing systemic bacterial dissemination following Chlamydia FRT infection.


Asunto(s)
Anticuerpos Antibacterianos/biosíntesis , Linfocitos B/inmunología , Bacteriemia/inmunología , Células de la Médula Ósea/inmunología , Linfocitos T CD4-Positivos/inmunología , Infecciones por Chlamydia/inmunología , Chlamydia trachomatis/inmunología , Animales , Presentación de Antígeno , Linfocitos B/microbiología , Bacteriemia/microbiología , Bacteriemia/patología , Células de la Médula Ósea/microbiología , Linfocitos T CD4-Positivos/microbiología , Infecciones por Chlamydia/microbiología , Infecciones por Chlamydia/patología , Chlamydia trachomatis/crecimiento & desarrollo , Chlamydia trachomatis/patogenicidad , Modelos Animales de Enfermedad , Femenino , Inmunidad Humoral , Isotipos de Inmunoglobulinas , Ratones , Quimera por Trasplante , Vagina/inmunología , Vagina/microbiología , Irradiación Corporal Total
16.
mBio ; 11(6)2020 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-33443133

RESUMEN

The Helicobacter pylori type IV secretion system (T4SS) encoded on the cag pathogenicity island (cagPAI) secretes the CagA oncoprotein and other effectors into the gastric epithelium. During murine infection, T4SS function is lost in an immune-dependent manner, typically as a result of in-frame recombination in the middle repeat region of cagY, though single nucleotide polymorphisms (SNPs) in cagY or in other essential genes may also occur. Loss of T4SS function also occurs in gerbils, nonhuman primates, and humans, suggesting that it is biologically relevant and not simply an artifact of the murine model. Here, we sought to identify physiologically relevant conditions under which T4SS function is maintained in the murine model. We found that loss of H. pylori T4SS function in mice was blunted by systemic Salmonella coinfection and completely eliminated by dietary iron restriction. Both have epidemiologic parallels in humans, since H. pylori strains from individuals in developing countries, where iron deficiency and systemic infections are common, are also more often cagPAI+ than strains from developed countries. These results have implications for our fundamental understanding of the cagPAI and also provide experimental tools that permit the study of T4SS function in the murine model.IMPORTANCE The type IV secretion system (T4SS) is the major Helicobacter pylori virulence factor, though its function is lost during murine infection. Loss of function also occurs in gerbils and in humans, suggesting that it is biologically relevant, but the conditions under which T4SS regulation occurs are unknown. Here, we found that systemic coinfection with Salmonella and iron deprivation each promote retention of T4SS function. These results improve our understanding of the cag pathogenicity island (cagPAI) and provide experimental tools that permit the study of T4SS function in the murine model.


Asunto(s)
Antígenos Bacterianos/genética , Proteínas Bacterianas/genética , Islas Genómicas , Infecciones por Helicobacter/microbiología , Helicobacter pylori/genética , Sistemas de Secreción Tipo IV/genética , Animales , Coinfección/microbiología , Femenino , Mucosa Gástrica , Helicobacter pylori/metabolismo , Helicobacter pylori/patogenicidad , Hierro/metabolismo , Ratones , Ratones Endogámicos C57BL , Salmonelosis Animal/sangre , Salmonelosis Animal/microbiología , Sistemas de Secreción Tipo IV/metabolismo , Factores de Virulencia
17.
Infect Immun ; 87(11)2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31427450

RESUMEN

Salmonella infection can cause gastroenteritis in healthy individuals or a serious, systemic infection in immunocompromised patients and has a global impact. CD4 Th1 cells represent the main lymphocyte population that participates in bacterial clearance during both primary and secondary infections in mice of the H-2b haplotype. Previous studies have used congenic mice to examine the function of major histocompatibility complex (MHC) molecules in elimination of this pathogen from the host. In this study, we further characterized the ability of H-2b, H-2k, and H-2u molecules to influence adaptive immunity to Salmonella in MHC congenic mice. By depleting different cell populations during infection, we unexpectedly found that CD8 T cells, in addition to CD4 T cells, play a major role in accelerated clearance of bacteria from H-2k congenic hosts. Our data suggest that CD8 T cells accelerate clearance in some MHC congenic mouse strains and could therefore represent an unexpected contributor to the protective efficacy of Salmonella vaccines outside the typical studies in C57BL/6 mice.


Asunto(s)
Linfocitos T CD8-positivos/fisiología , Salmonelosis Animal/inmunología , Salmonelosis Animal/microbiología , Salmonella typhimurium/inmunología , Animales , Linfocitos T CD4-Positivos/metabolismo , Infecciones por Chlamydia/inmunología , Infecciones por Chlamydia/microbiología , Chlamydia muridarum , Haplotipos , Interferón gamma , Complejo Mayor de Histocompatibilidad/genética , Ratones
18.
Vet Comp Oncol ; 17(3): 242-252, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30684301

RESUMEN

Our lack of understanding of the immune microenvironment in canine osteosarcoma (cOSA) has limited the identification of potential immunotherapeutic targets. In particular, our ability to utilize readily available tissue from a dog's primary tumour to predict the type and extent of immune response in their pulmonary metastatic lesions is unknown. We, therefore, collected 21 matched pairs of primary tumours and pulmonary metastatic lesions from dogs with OSA and performed immunohistochemistry to quantify T-lymphocyte (CD3), FOXP3+ cell, B-lymphocyte (Pax-5), and CD204+ macrophage infiltration. We found that T-lymphocytes and FOXP3+ infiltrates in primary tumours positively correlated with that of metastatic lesions (ρ = 0.512, P = 0.038 and ρ = 0.698, P = 0.007, respectively), while a strong trend existed for CD204+ infiltrates (ρ = 0.404, P = 0.087). We also observed T- and B-lymphocytes, and CD204+ macrophages to be significantly higher in a dog's pulmonary metastasis compared to their primary tumour (P = 0.018, P = 0.018, P = 0.016, respectively), while FOXP3+ cells were only significantly higher in metastases when all primary tumour and metastasis lesions were compared without pairing (P = 0.036). Together, these findings suggest that the metastatic immune microenvironment may be influenced by that of the primary cOSA, and that primary tumour immune biomarkers could potentially be applied to predict immunotherapeutic responses in gross metastatic disease. We, therefore, provide a rationale for the treatment of cOSA pulmonary metastases with immunotherapeutics that enhance the anti-tumour activity of these immune cells, particularly in dogs with moderate to high immune cell infiltration in their primary tumours.


Asunto(s)
Neoplasias Óseas/veterinaria , Enfermedades de los Perros/patología , Regulación Neoplásica de la Expresión Génica/fisiología , Osteosarcoma/veterinaria , Linfocitos T/metabolismo , Animales , Neoplasias Óseas/patología , Perros , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Inmunohistoquímica , Osteosarcoma/patología , Factor de Transcripción PAX5/genética , Factor de Transcripción PAX5/metabolismo , Receptores Depuradores de Clase A/genética , Receptores Depuradores de Clase A/metabolismo
19.
Vet Comp Oncol ; 17(1): 49-60, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30156029

RESUMEN

Immunotherapeutic strategies have shown promise for the treatment of canine osteosarcoma (cOSA). Very little is known about the immune microenvironment within cOSA, however, limiting our ability to identify potential immune targets and biomarkers of therapeutic response. We therefore prospectively assessed the disease-free interval (DFI) and overall survival time (ST) of 30 dogs with cOSA treated with amputation and six doses of adjuvant carboplatin. We then quantified lymphocytic (CD3+, FOXP3+) and macrophage (CD204+) infiltrates within the primary tumours of this cohort using immunohistochemistry, and evaluated their association with outcome. Overall, the median DFI and ST were 392 and 455 days, respectively. The median number of CD3+ and FOXP3+ infiltrates were 45.8 cells/mm2 (4.6-607.6 cells/mm2 ) and 8.5 mm2 (0-163.1 cells/mm2 ), respectively. The median area of CD204+ macrophages was 4.7% (1.3%-23.3%), and dogs with tumours containing greater than 4.7% CD204+ macrophages experienced a significantly longer DFI (P = 0.016). Interestingly, a significantly lower percentage of CD204+ macrophages was detected in cOSA arising from the proximal humerus compared to other appendicular bone locations (P = 0.016). Lymphocytic infiltrates did not appear to correlate with outcome in cOSA. Overall, our findings suggest that macrophages may play a role in inhibiting cOSA progression, as has been suggested in human osteosarcoma.


Asunto(s)
Neoplasias Óseas/veterinaria , Enfermedades de los Perros/patología , Linfocitos/patología , Macrófagos/patología , Osteosarcoma/veterinaria , Animales , Neoplasias Óseas/patología , Perros , Femenino , Humanos , Masculino , Osteosarcoma/patología
20.
Proc Natl Acad Sci U S A ; 115(41): 10416-10421, 2018 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-30254173

RESUMEN

While CD4 Th1 cells are required for resistance to intramacrophage infections, adoptive transfer of Th1 cells is insufficient to protect against Salmonella infection. Using an epitope-tagged vaccine strain of Salmonella, we found that effective protection correlated with expanded Salmonella-specific memory CD4 T cells in circulation and nonlymphoid tissues. However, naive mice that previously shared a blood supply with vaccinated partners lacked T cell memory with characteristics of tissue residence and did not acquire robust protective immunity. Using a YFP-IFN-γ reporter system, we identified Th1 cells in the liver of immunized mice that displayed markers of tissue residence, including P2X7, ARTC2, LFA-1, and CD101. Adoptive transfer of liver memory cells after ARTC2 blockade increased protection against highly virulent bacteria. Taken together, these data demonstrate that noncirculating memory Th1 cells are a vital component of immunity to Salmonella infection and should be the focus of vaccine strategies.


Asunto(s)
Memoria Inmunológica/inmunología , Hígado/inmunología , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Linfocitos T/inmunología , Células TH1/inmunología , Animales , Células Cultivadas , Femenino , Inmunización , Hígado/microbiología , Ratones , Ratones Endogámicos C57BL , Infecciones por Salmonella/microbiología , Infecciones por Salmonella/prevención & control , Linfocitos T/microbiología , Células TH1/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...