Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 167
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 21(8): 938-949, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32572242

RESUMEN

The central nervous system (CNS) is classically viewed as immune-privileged; however, recent advances highlight interactions between the peripheral immune system and CNS in controlling infections and tissue homeostasis. Tissue-resident memory (TRM) CD8+ T cells in the CNS are generated after brain infections, but it is unknown whether CNS infection is required to generate brain TRM cells. We show that peripheral infections generate antigen-specific CD8+ memory T cells in the brain that adopt a unique TRM signature. Upon depletion of circulating and perivascular memory T cells, this brain signature was enriched and the surveilling properties of brain TRM cells was revealed by intravital imaging. Notably, peripherally induced brain TRM cells showed evidence of rapid activation and enhanced cytokine production and mediated protection after brain infections. These data reveal that peripheral immunizations can generate brain TRM cells and will guide potential use of T cells as therapeutic strategies against CNS infections and neurological diseases.


Asunto(s)
Encéfalo/inmunología , Linfocitos T CD8-positivos/inmunología , Infecciones del Sistema Nervioso Central/inmunología , Memoria Inmunológica/inmunología , Animales , Infecciones Bacterianas/inmunología , Encéfalo/citología , Activación de Linfocitos/inmunología , Ratones , Virosis/inmunología
3.
Nat Immunol ; 18(8): 931-939, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28604718

RESUMEN

Activated CD8+ T cells differentiate into cytotoxic effector (TEFF) cells that eliminate target cells. How TEFF cell identity is established and maintained is not fully understood. We found that Runx3 deficiency limited clonal expansion and impaired upregulation of cytotoxic molecules in TEFF cells. Runx3-deficient CD8+ TEFF cells aberrantly upregulated genes characteristic of follicular helper T (TFH) cell lineage, including Bcl6, Tcf7 and Cxcr5. Mechanistically, the Runx3-CBFß transcription factor complex deployed H3K27me3 to Bcl6 and Tcf7 genes to suppress the TFH program. Ablating Tcf7 in Runx3-deficient CD8+ TEFF cells prevented the upregulation of TFH genes and ameliorated their defective induction of cytotoxic genes. As such, Runx3-mediated Tcf7 repression coordinately enforced acquisition of cytotoxic functions and protected the cytotoxic lineage integrity by preventing TFH-lineage deviation.


Asunto(s)
Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Linfopoyesis/genética , Linfocitos T Citotóxicos/citología , Linfocitos T Colaboradores-Inductores/citología , Animales , Linaje de la Célula , Ensayo de Inmunoadsorción Enzimática , Epigénesis Genética , Regulación de la Expresión Génica , Factor Nuclear 1-alfa del Hepatocito/genética , Inmunohistoquímica , Ratones , Proteínas Proto-Oncogénicas c-bcl-6/genética , Receptores CXCR5/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ARN , Regulación hacia Arriba
5.
Immunol Rev ; 316(1): 84-103, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37014087

RESUMEN

Nearly half of the world's population is at risk of malaria, a disease caused by the protozoan parasite Plasmodium, which is estimated to cause more than 240,000,000 infections and kill more than 600,000 people annually. The emergence of Plasmodia resistant to chemoprophylactic treatment highlights the urgency to develop more effective vaccines. In this regard, whole sporozoite vaccination approaches in murine models and human challenge studies have provided substantial insight into the immune correlates of protection from malaria. From these studies, CD8+ T cells have come to the forefront, being identified as critical for vaccine-mediated liver-stage immunity that can prevent the establishment of the symptomatic blood stages and subsequent transmission of infection. However, the unique biological characteristics required for CD8+ T cell protection from liver-stage malaria dictate that more work must be done to design effective vaccines. In this review, we will highlight a subset of studies that reveal basic aspects of memory CD8+ T cell-mediated protection from liver-stage malaria infection.


Asunto(s)
Vacunas contra la Malaria , Malaria , Plasmodium , Ratones , Humanos , Animales , Memoria Inmunológica , Hígado , Linfocitos T CD8-positivos
6.
Immunity ; 47(5): 835-847.e4, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29150238

RESUMEN

Immune response (Ir) genes, originally proposed by Baruj Benacerraf to explain differential antigen-specific responses in animal models, have become synonymous with the major histocompatibility complex (MHC). We discovered a non-MHC-linked Ir gene in a T cell receptor (TCR) locus that was required for CD8+ T cell responses to the Plasmodium berghei GAP5040-48 epitope in mice expressing the MHC class I allele H-2Db. GAP5040-48-specific CD8+ T cell responses emerged from a very large pool of naive Vß8.1+ precursors, which dictated susceptibility to cerebral malaria and conferred protection against recombinant Listeria monocytogenes infection. Structural analysis of a prototypical Vß8.1+ TCR-H-2Db-GAP5040-48 ternary complex revealed that germline-encoded complementarity-determining region 1ß residues present exclusively in the Vß8.1 segment mediated essential interactions with the GAP5040-48 peptide. Collectively, these findings demonstrated that Vß8.1 functioned as an Ir gene that was indispensable for immune reactivity against the malaria GAP5040-48 epitope.


Asunto(s)
Antígeno de Histocompatibilidad H-2D/genética , Plasmodium berghei/inmunología , Proteínas Protozoarias/inmunología , Receptores de Antígenos de Linfocitos T/genética , Animales , Linfocitos T CD8-positivos/inmunología , Regiones Determinantes de Complementariedad , Epítopos , Genes Codificadores de la Cadena beta de los Receptores de Linfocito T , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Fragmentos de Péptidos/inmunología
7.
J Immunol ; 212(4): 563-575, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38149923

RESUMEN

Patients infected with SARS-CoV-2 experience variable disease susceptibility, and patients with comorbidities such as sepsis are often hospitalized for COVID-19 complications. However, the extent to which initial infectious inoculum dose determines disease outcomes and whether this can be used for immunological priming in a genetically susceptible host has not been completely defined. We used an established SARS-like murine model in which responses to primary and/or secondary challenges with murine hepatitis virus type 1 (MHV-1) were analyzed. We compared the response to infection in genetically susceptible C3H/HeJ mice, genetically resistant C57BL/6J mice, and genetically diverse, variably susceptible outbred Swiss Webster mice. Although defined as genetically susceptible to MHV-1, C3H/HeJ mice displayed decreasing dose-dependent pathological changes in disease severity and lung infiltrate/edema, as well as lymphopenia. Importantly, an asymptomatic dose (500 PFU) was identified that yielded no measurable morbidity/mortality postinfection in C3H/HeJ mice. Polymicrobial sepsis induced via cecal ligation and puncture converted asymptomatic infections in C3H/HeJ and C57BL/6J mice to more pronounced disease, modeling the impact of sepsis as a comorbidity to ß-coronavirus infection. We then used low-dose infection as an immunological priming event in C3H/HeJ mice, which provided neutralizing Ab-dependent, but not circulating CD4/CD8 T cell-dependent, protection against a high-dose MHV-1 early rechallenge. Together, these data define how infection dose, immunological status, and comorbidities modulate outcomes of primary and secondary ß-coronavirus infections in hosts with variable susceptibility.


Asunto(s)
Virus de la Hepatitis Murina , Sepsis , Humanos , Ratones , Animales , Ratones Endogámicos C57BL , Ratones Endogámicos C3H , Ratones Endogámicos , Predisposición Genética a la Enfermedad
8.
Proc Natl Acad Sci U S A ; 120(27): e2302785120, 2023 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-37364124

RESUMEN

The increasing use of nuclear energy sources inevitably raises the risk of accidental or deliberate radiation exposure and associated immune dysfunction. However, the extent to which radiation exposure impacts memory CD8 T cells, potent mediators of immunity to recurring intracellular infections and malignancies, remains understudied. Using P14 CD8 T cell chimeric mice (P14 chimeras) with an lymphocytic choriomeningitis virus (LCMV) infection model, we observed that sublethal (5Gy) whole-body irradiation (WBI) induced a rapid decline in the number of naive (TN) and P14 circulating memory CD8 T cells (TCIRCM), with the former being more susceptible to radiation-induced numeric loss. While TN cell numbers rapidly recovered, as previously described, the number of P14 TCIRCM cells remained low at least 9 mo after radiation exposure. Additionally, the remaining P14 TCIRCM in irradiated hosts exhibited an inefficient transition to a central memory (CD62L+) phenotype compared to nonirradiated P14 chimeras. WBI also resulted in long-lasting T cell intrinsic deficits in memory CD8 T cells, including diminished cytokine and chemokine production along with impaired secondary expansion upon cognate Ag reencounter. Irradiated P14 chimeras displayed significantly higher bacterial burden after challenge with Listeria monocytogenes expressing the LCMV GP33-41 epitope relative to nonirradiated controls, likely due to radiation-induced numerical and functional impairments. Taken together, our findings suggest that sublethal radiation exposure caused a long-term numerical, impaired differentiation, and functional dysregulation in preexisting TCIRCM, rendering previously protected hosts susceptible to reinfection.


Asunto(s)
Coriomeningitis Linfocítica , Irradiación Corporal Total , Ratones , Animales , Recurrencia Local de Neoplasia , Linfocitos T CD8-positivos , Virus de la Coriomeningitis Linfocítica , Memoria Inmunológica , Ratones Endogámicos C57BL
9.
Proc Natl Acad Sci U S A ; 120(2): e2210181120, 2023 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-36595704

RESUMEN

Malaria, caused by Plasmodium parasites is a severe disease affecting millions of people around the world. Plasmodium undergoes obligatory development and replication in the hepatocytes, before initiating the life-threatening blood-stage of malaria. Although the natural immune responses impeding Plasmodium infection and development in the liver are key to controlling clinical malaria and transmission, those remain relatively unknown. Here we demonstrate that the DNA of Plasmodium parasites is sensed by cytosolic AIM2 (absent in melanoma 2) receptors in the infected hepatocytes, resulting in Caspase-1 activation. Remarkably, Caspase-1 was observed to undergo unconventional proteolytic processing in hepatocytes, resulting in the activation of the membrane pore-forming protein, Gasdermin D, but not inflammasome-associated proinflammatory cytokines. Nevertheless, this resulted in the elimination of Plasmodium-infected hepatocytes and the control of malaria infection in the liver. Our study uncovers a pathway of natural immunity critical for the control of malaria in the liver.


Asunto(s)
Malaria , Parásitos , Plasmodium , Animales , Humanos , Hepatocitos/metabolismo , Hígado , Malaria/parasitología , Caspasas/metabolismo , Proteínas de Unión al ADN/metabolismo
10.
PLoS Pathog ; 19(10): e1011720, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37824591

RESUMEN

Sepsis, an amplified immune response to systemic infection, is characterized by a transient cytokine storm followed by chronic immune dysfunction. Consequently, sepsis survivors are highly susceptible to newly introduced infections, suggesting sepsis can influence the function and composition of the naïve CD8 T cell pool and resulting pathogen-induced primary CD8 T cell responses. Here, we explored the extent to which sepsis induces phenotypic and functional changes within the naïve CD8 T cell pool. To interrogate this, the cecal ligation and puncture (CLP) mouse model of polymicrobial sepsis was used. In normal, non-septic mice, we show type-I interferon (IFN I)-mediated signaling plays an important role in driving the phenotypic and functional heterogeneity in the naïve CD8 T cell compartment leading to increased representation of Ly6C+ naïve CD8 T cells. In response to viral infection after sepsis resolution, naïve Ly6C+ CD8 T cells generated more primary effector and memory CD8 T cells with slower conversion to a central memory CD8 T cell phenotype (Tcm) than Ly6C- naïve CD8 T cells. Importantly, as a potent inducer of cytokine storm and IFN I production, sepsis leads to increased representation of Ly6C+ naïve CD8 T cells that maintained their heightened ability to respond (i.e., effector and memory CD8 T cell accumulation and cytokine production) to primary LCMV infection. Lastly, longitudinal analyses of peripheral blood samples obtained from septic patients revealed profound changes in CD8 T cell subset composition and frequency compared to healthy controls. Thus, sepsis has the capacity to alter the composition of naïve CD8 T cells, directly influencing primary CD8 T cell responses to newly introduced infections.


Asunto(s)
Síndrome de Liberación de Citoquinas , Sepsis , Humanos , Ratones , Animales , Linfocitos T CD8-positivos , Inmunidad Innata , Fenotipo , Ratones Endogámicos C57BL , Memoria Inmunológica
11.
Trends Immunol ; 43(12): 1018-1031, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36369103

RESUMEN

The mammalian central nervous system (CNS) contains a vibrant community of resident adaptive immune cells at homeostasis. Among these are memory CD8+ and CD4+ T cells, which reside in the CNS in the settings of health, aging, and neurological disease. These T cells commonly exhibit a tissue-resident memory (TRM) phenotype, suggesting that they are antigen-experienced and remain separate from the circulation. Despite these characterizations, T cell surveillance of the CNS has only recently been studied through the lens of TRM immunology. In this Review, we outline emerging concepts of CNS TRM generation, localization, maintenance, function, and specificity. In this way, we hope to highlight roles of CNS TRM in health and disease to inform future studies of adaptive neuroimmunity.


Asunto(s)
Linfocitos T CD8-positivos , Memoria Inmunológica , Animales , Antígenos , Homeostasis , Mamíferos
12.
Immunity ; 45(6): 1341-1354, 2016 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-27986453

RESUMEN

Differentiation of effector and memory CD8+ T cells is accompanied by extensive changes in the transcriptome and histone modifications at gene promoters; however, the enhancer repertoire and associated gene regulatory networks are poorly defined. Using histone mark chromatin immunoprecipitation coupled with deep sequencing, we mapped the enhancer and super-enhancer landscapes in antigen-specific naive, differentiated effector, and central memory CD8+ T cells during LCMV infection. Epigenomics-based annotation revealed a highly dynamic repertoire of enhancers, which were inherited, de novo activated, decommissioned and re-activated during CD8+ T cell responses. We employed a computational algorithm to pair enhancers with target gene promoters. On average, each enhancer targeted three promoters and each promoter was regulated by two enhancers. By identifying enriched transcription factor motifs in enhancers, we defined transcriptional regulatory circuitry at each CD8+ T cell response stage. These multi-dimensional datasets provide a blueprint for delineating molecular mechanisms underlying functional differentiation of CD8+ T cells.


Asunto(s)
Infecciones por Arenaviridae/inmunología , Linfocitos T CD8-positivos/inmunología , Elementos de Facilitación Genéticos/inmunología , Regulación de la Expresión Génica/inmunología , Activación de Linfocitos/inmunología , Algoritmos , Animales , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Inmunoprecipitación de Cromatina , Biología Computacional/métodos , Modelos Animales de Enfermedad , Elementos de Facilitación Genéticos/genética , Epigenómica/métodos , Redes Reguladoras de Genes , Secuenciación de Nucleótidos de Alto Rendimiento , Activación de Linfocitos/genética , Virus de la Coriomeningitis Linfocítica , Ratones , Regiones Promotoras Genéticas/genética , Regiones Promotoras Genéticas/inmunología
13.
J Immunol ; 210(8): 1025-1030, 2023 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-36912465

RESUMEN

Although tissue resident memory T cells (TRM) in the lung confer robust protection against secondary influenza infection, their in vivo production of IFN-γ is unknown. In this study, using a mouse model, we evaluated production of IFN-γ by influenza-induced TRM (defined as CD103+) that localize to the airways or lung parenchyma. Airway TRM consist of both CD11ahi and CD11alo populations, with low CD11a expression signifying prolonged airway residence. In vitro, high-dose peptide stimulation evoked IFN-γ from most CD11ahi airway and parenchymal TRM, whereas most CD11alo airway TRM did not produce IFN-γ. In vivo production of IFN-γ was clearly detectable in CD11ahi airway and parenchymal TRM but essentially absent in CD11alo airway TRM, irrespective of airway-instilled peptide concentration or influenza reinfection. The majority of IFN-γ-producing airway TRM in vivo were CD11ahi, suggesting recent airway entry. These results question the contribution of long-term CD11alo airway TRM to influenza immunity and reinforce the importance of defining TRM tissue compartment-specific contributions to protective immunity.


Asunto(s)
Gripe Humana , Humanos , Linfocitos T CD8-positivos , Células T de Memoria , Memoria Inmunológica , Pulmón , Interferón gamma , Receptores de Antígenos de Linfocitos T/metabolismo
14.
J Immunol ; 210(9): 1305-1313, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36939394

RESUMEN

Production of IFN-γ by CD4 T cells is widely theorized to control Plasmodium parasite burden during blood-stage malaria. Surprisingly, the specific and crucial mechanisms through which this highly pleiotropic cytokine acts to confer protection against malarial disease remain largely untested in vivo. Here we used a CD4 T cell-restricted Cre-Lox IFN-γ excision mouse model to test whether and how CD4 T cell-derived IFN-γ controls blood-stage malaria. Although complete absence of IFN-γ compromised control of the acute and the chronic, recrudescent blood-stage infections with P. c. chabaudi, we identified a specific, albeit modest, role for CD4 T cell-derived IFN-γ in limiting parasite burden only during the chronic stages of P. c. chabaudi malaria. CD4 T cell IFN-γ promoted IgG Ab class switching to the IgG2c isotype during P. c. chabaudi malaria in C57BL/6 mice. Unexpectedly, our data do not support gross defects in phagocytic activity in IFN-γ-deficient hosts infected with blood-stage malaria. Together, our data confirm CD4 T cell-dependent roles for IFN-γ but suggest CD4 T cell-independent roles for IFN-γ in immune responses to blood-stage malaria.


Asunto(s)
Malaria , Plasmodium chabaudi , Ratones , Animales , Linfocitos T CD4-Positivos , Ratones Endogámicos C57BL , Interferón gamma
15.
Brain ; 147(2): 566-589, 2024 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-37776513

RESUMEN

Cerebral malaria is the deadliest complication that can arise from Plasmodium infection. CD8 T-cell engagement of brain vasculature is a putative mechanism of neuropathology in cerebral malaria. To define contributions of brain endothelial cell major histocompatibility complex (MHC) class I antigen-presentation to CD8 T cells in establishing cerebral malaria pathology, we developed novel H-2Kb LoxP and H-2Db LoxP mice crossed with Cdh5-Cre mice to achieve targeted deletion of discrete class I molecules, specifically from brain endothelium. This strategy allowed us to avoid off-target effects on iron homeostasis and class I-like molecules, which are known to perturb Plasmodium infection. This is the first endothelial-specific ablation of individual class-I molecules enabling us to interrogate these molecular interactions. In these studies, we interrogated human and mouse transcriptomics data to compare antigen presentation capacity during cerebral malaria. Using the Plasmodium berghei ANKA model of experimental cerebral malaria (ECM), we observed that H-2Kb and H-2Db class I molecules regulate distinct patterns of disease onset, CD8 T-cell infiltration, targeted cell death and regional blood-brain barrier disruption. Strikingly, ablation of either molecule from brain endothelial cells resulted in reduced CD8 T-cell activation, attenuated T-cell interaction with brain vasculature, lessened targeted cell death, preserved blood-brain barrier integrity and prevention of ECM and the death of the animal. We were able to show that these events were brain-specific through the use of parabiosis and created the novel technique of dual small animal MRI to simultaneously scan conjoined parabionts during infection. These data demonstrate that interactions of CD8 T cells with discrete MHC class I molecules on brain endothelium differentially regulate development of ECM neuropathology. Therefore, targeting MHC class I interactions therapeutically may hold potential for treatment of cases of severe malaria.


Asunto(s)
Malaria Cerebral , Ratones , Humanos , Animales , Malaria Cerebral/patología , Malaria Cerebral/prevención & control , Células Endoteliales/patología , Encéfalo/patología , Barrera Hematoencefálica/patología , Linfocitos T CD8-positivos , Endotelio/patología , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad
17.
Nat Immunol ; 13(2): 188-95, 2011 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-22157630

RESUMEN

Infection of erythrocytes with Plasmodium species induces clinical malaria. Parasite-specific CD4(+) T cells correlate with lower parasite burdens and severity of human malaria and are needed to control blood-stage infection in mice. However, the characteristics of CD4(+) T cells that determine protection or parasite persistence remain unknown. Here we show that infection of humans with Plasmodium falciparum resulted in higher expression of the inhibitory receptor PD-1 associated with T cell dysfunction. In vivo blockade of the PD-1 ligand PD-L1 and the inhibitory receptor LAG-3 restored CD4(+) T cell function, amplified the number of follicular helper T cells and germinal-center B cells and plasmablasts, enhanced protective antibodies and rapidly cleared blood-stage malaria in mice. Thus, chronic malaria drives specific T cell dysfunction, and proper function can be restored by inhibitory therapies to enhance parasite control.


Asunto(s)
Antígenos CD/efectos de los fármacos , Antígeno B7-H1/antagonistas & inhibidores , Linfocitos T CD4-Positivos/efectos de los fármacos , Malaria Falciparum/tratamiento farmacológico , Plasmodium falciparum/efectos de los fármacos , Enfermedad Aguda , Animales , Antígenos CD/inmunología , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Linfocitos B/parasitología , Antígeno B7-H1/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/parasitología , Niño , Preescolar , Enfermedad Crónica , Eritrocitos/inmunología , Eritrocitos/parasitología , Femenino , Centro Germinal/efectos de los fármacos , Centro Germinal/inmunología , Centro Germinal/parasitología , Humanos , Malaria Falciparum/inmunología , Malí , Ratones , Ratones Endogámicos C57BL , Plasmodium falciparum/inmunología , Estados Unidos , Regulación hacia Arriba/efectos de los fármacos , Proteína del Gen 3 de Activación de Linfocitos
18.
J Immunol ; 207(11): 2631-2635, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34716185

RESUMEN

Radiation-attenuated sporozoite (RAS) vaccination offers hope for global malaria control through induction of protective liver-stage-specific memory CD8 T cells. Effective RAS vaccination regimens exist; however, widespread implementation remains unfeasible. A key difficulty resides in the need to administer three or more doses i.v. to achieve sufficient immunity. Strategies to reduce the number of RAS doses are therefore desirable. Here we used mice to model human immune responses to a single, suboptimal weight-normalized RAS dose administered i.v. followed by subunit vaccination to amplify liver-stage-specific memory CD8 T cells. RAS+subunit prime-boost regimens increased the numbers of liver-stage-specific memory CD8 T cells to a level greater than is present after one RAS vaccination. Both i.v. and i.m. subunit vaccine delivery induced immunity in mice, and many vaccinated mice completely cleared liver infection. These findings are particularly relevant to human vaccine development because RAS+subunit prime-boost vaccination would reduce the logistical challenges of multiple RAS-only immunizations.


Asunto(s)
Hepatopatías/inmunología , Vacunas contra la Malaria/inmunología , Malaria/inmunología , Esporozoítos/inmunología , Vacunas Atenuadas/inmunología , Vacunas de Subunidad/inmunología , Animales , Inmunización , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Vacunación
19.
J Immunol ; 206(6): 1171-1180, 2021 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-33514512

RESUMEN

The dysregulated sepsis-induced cytokine storm evoked during systemic infection consists of biphasic and interconnected pro- and anti-inflammatory responses. The contrasting inflammatory cytokine responses determine the severity of the septic event, lymphopenia, host survival, and the ensuing long-lasting immunoparalysis state. NK cells, because of their capacity to elaborate pro- (i.e., IFN-γ) and anti-inflammatory (i.e., IL-10) responses, exist at the inflection of sepsis-induced inflammatory responses. Thus, NK cell activity could be beneficial or detrimental during sepsis. In this study, we demonstrate that murine NK cells promote host survival during sepsis by limiting the scope and duration of the cytokine storm. Specifically, NK cell-derived IL-10, produced in response to IL-15, is relevant to clinical manifestations in septic patients and critical for survival during sepsis. This role of NK cells demonstrates that regulatory mechanisms of classical inflammatory cells are beneficial and critical for controlling systemic inflammation, a notion relevant for therapeutic interventions during dysregulated infection-induced inflammatory responses.


Asunto(s)
Síndrome de Liberación de Citoquinas/inmunología , Interleucina-10/metabolismo , Células Asesinas Naturales/inmunología , Sepsis/complicaciones , Animales , Síndrome de Liberación de Citoquinas/sangre , Síndrome de Liberación de Citoquinas/diagnóstico , Humanos , Interferón gamma/metabolismo , Interleucina-10/genética , Interleucina-15/metabolismo , Células Asesinas Naturales/metabolismo , Ratones , Ratones Transgénicos , Sepsis/sangre , Sepsis/diagnóstico , Sepsis/inmunología , Índice de Severidad de la Enfermedad , Transducción de Señal/inmunología
20.
J Immunol ; 207(7): 1871-1881, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34479943

RESUMEN

Sepsis reduces the number and function of memory CD8 T cells within the host, contributing to the long-lasting state of immunoparalysis. Interestingly, the relative susceptibility of memory CD8 T cell subsets to quantitative/qualitative changes differ after cecal ligation and puncture (CLP)-induced sepsis. Compared with circulatory memory CD8 T cells (TCIRCM), moderate sepsis (0-10% mortality) does not result in numerical decline of CD8 tissue-resident memory T cells (TRM), which retain their "sensing and alarm" IFN-γ-mediated effector function. To interrogate this biologically important dichotomy, vaccinia virus-immune C57BL/6 (B6) mice containing CD8 TCIRCM and skin TRM underwent moderate or severe (∼50% mortality) sepsis. Severe sepsis led to increased morbidity and mortality characterized by increased inflammation compared with moderate CLP or sham controls. Severe CLP mice also displayed increased vascular permeability in the ears. Interestingly, skin CD103+ CD8 TRM, detected by i.v. exclusion or two-photon microscopy, underwent apoptosis and subsequent numerical loss following severe sepsis, which was not observed in mice that experienced moderate CLP or sham surgeries. Consequently, severe septic mice showed diminished CD8 T cell-mediated protection to localized skin reinfection. Finally, the relationship between severity of sepsis and demise in circulatory versus tissue-embedded memory CD8 T cell populations was confirmed by examining tumor-infiltrating and nonspecific CD8 T cells in B16 melanoma tumors. Thus, sepsis can differentially affect the presence and function of Ag-specific CD8 T cells that reside inside tissues/tumors depending on the severity of the insult, a notion with direct relevance to sepsis survivors and their ability to mount protective memory CD8 T cell-dependent responses to localized Ag re-encounter.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/fisiología , Sepsis/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Circulación Sanguínea , Células Cultivadas , Progresión de la Enfermedad , Humanos , Memoria Inmunológica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Especificidad de Órganos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA