Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Nature ; 592(7854): 457-462, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33731934

RESUMEN

In metazoans, specific tasks are relegated to dedicated organs that are established early in development, occupy discrete locations and typically remain fixed in size. The adult immune system arises from a centralized haematopoietic niche that maintains self-renewing potential1,2, and-upon maturation-becomes distributed throughout the body to monitor environmental perturbations, regulate tissue homeostasis and mediate organism-wide defence. Here we examine how immunity is integrated within adult mouse tissues, and address issues of durability, expansibility and contributions to organ cellularity. Focusing on antiviral T cell immunity, we observed durable maintenance of resident memory T cells up to 450 days after infection. Once established, resident T cells did not require the T cell receptor for survival or retention of a poised, effector-like state. Although resident memory indefinitely dominated most mucosal organs, surgical separation of parabiotic mice revealed a tissue-resident provenance for blood-borne effector memory T cells, and circulating memory slowly made substantial contributions to tissue immunity in some organs. After serial immunizations or cohousing with pet-shop mice, we found that in most tissues, tissue pliancy (the capacity of tissues to vary their proportion of immune cells) enables the accretion of tissue-resident memory, without axiomatic erosion of pre-existing antiviral T cell immunity. Extending these findings, we demonstrate that tissue residence and organ pliancy are generalizable aspects that underlie homeostasis of innate and adaptive immunity. The immune system grows commensurate with microbial experience, reaching up to 25% of visceral organ cellularity. Regardless of the location, many populations of white blood cells adopted a tissue-residency program within nonlymphoid organs. Thus, residence-rather than renewal or recirculation-typifies nonlymphoid immune surveillance, and organs serve as pliant storage reservoirs that can accommodate continuous expansion of the cellular immune system throughout life. Although haematopoiesis restores some elements of the immune system, nonlymphoid organs sustain an accrual of durable tissue-autonomous cellular immunity that results in progressive decentralization of organismal immune homeostasis.


Asunto(s)
Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Microambiente Celular , Homeostasis , Memoria Inmunológica , Vigilancia Inmunológica , Inmunidad Adaptativa , Animales , Femenino , Inmunidad Innata , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T/inmunología
2.
J Immunol ; 212(11): 1621-1625, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38619284

RESUMEN

Humans experience frequent respiratory infections. Immunology and vaccinology studies in mice are typically performed in naive specific pathogen-free animals responding to their very first respiratory challenge. We found that the first respiratory infection induces lifelong enlargement of the lung-draining mediastinal lymph nodes (medLNs). Furthermore, infection-experienced medLNs supported better naive T cell surveillance and effector responses to new unrelated infections that exhibited more biased accumulation and memory establishment within the lung. Moreover, we observed that weight loss induced by influenza infection was substantially reduced in mice that had recovered from a previous unrelated respiratory viral challenge. These data show that the lack of infectious history and corresponding medLN hypoplasia in specific pathogen-free mice alter their immune response to lung infections. Preclinical vaccination and immunology studies should consider the previous infectious experience of the model organism.


Asunto(s)
Pulmón , Ganglios Linfáticos , Infecciones por Orthomyxoviridae , Animales , Ratones , Ganglios Linfáticos/inmunología , Infecciones por Orthomyxoviridae/inmunología , Pulmón/inmunología , Pulmón/virología , Pulmón/patología , Ratones Endogámicos C57BL , Organismos Libres de Patógenos Específicos , Linfocitos T/inmunología , Memoria Inmunológica/inmunología , Mediastino , Infecciones del Sistema Respiratorio/inmunología
3.
J Immunol ; 206(5): 931-935, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33441437

RESUMEN

The magnitude of SARS-CoV-2-specific T cell responses correlates inversely with human disease severity, suggesting T cell involvement in primary control. Whereas many COVID-19 vaccines focus on establishing humoral immunity to viral spike protein, vaccine-elicited T cell immunity may bolster durable protection or cross-reactivity with viral variants. To better enable mechanistic and vaccination studies in mice, we identified a dominant CD8 T cell SARS-CoV-2 nucleoprotein epitope. Infection of human ACE2 transgenic mice with SARS-CoV-2 elicited robust responses to H2-Db/N219-227, and 40% of HLA-A*02+ COVID-19 PBMC samples isolated from hospitalized patients responded to this peptide in culture. In mice, i.m. prime-boost nucleoprotein vaccination with heterologous vectors favored systemic CD8 T cell responses, whereas intranasal boosting favored respiratory immunity. In contrast, a single i.v. immunization with recombinant adenovirus established robust CD8 T cell memory both systemically and in the respiratory mucosa.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Vacunas contra la COVID-19/inmunología , COVID-19/inmunología , COVID-19/prevención & control , Epítopos de Linfocito T/inmunología , SARS-CoV-2/inmunología , Vacunación/métodos , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , COVID-19/virología , Células Cultivadas , Proteínas de la Nucleocápside de Coronavirus/inmunología , Modelos Animales de Enfermedad , Femenino , Vectores Genéticos/inmunología , Antígeno HLA-A2/inmunología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
4.
J Immunol ; 207(2): 376-379, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34193597

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the COVID-19 pandemic. Neutralizing Abs target the receptor binding domain of the spike (S) protein, a focus of successful vaccine efforts. Concerns have arisen that S-specific vaccine immunity may fail to neutralize emerging variants. We show that vaccination with a human adenovirus type 5 vector expressing the SARS-CoV-2 nucleocapsid (N) protein can establish protective immunity, defined by reduced weight loss and viral load, in both Syrian hamsters and K18-hACE2 mice. Challenge of vaccinated mice was associated with rapid N-specific T cell recall responses in the respiratory mucosa. This study supports the rationale for including additional viral Ags in SARS-CoV-2 vaccines, even if they are not a target of neutralizing Abs, to broaden epitope coverage and immune effector mechanisms.


Asunto(s)
Anticuerpos Antivirales/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra la COVID-19/inmunología , COVID-19/prevención & control , Proteínas de la Nucleocápside de Coronavirus/inmunología , SARS-CoV-2/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , COVID-19/inmunología , Línea Celular , Chlorocebus aethiops , Cricetinae , Femenino , Memoria Inmunológica/inmunología , Recuento de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Fosfoproteínas/inmunología , Vacunación , Células Vero
5.
J Immunol ; 197(7): 2635-45, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27566821

RESUMEN

Phenotypically and functionally diverse regulatory T (Tr) cell subsets populate lymphoid and nonlymphoid tissues, where their maintenance and function are governed by unique homeostatic signals. Whereas Tr cells resident in nonlymphoid tissues depend on continual TCR signaling for their survival and function, phenotypically naive Tr cells occupying secondary lymphoid organs are largely supported by paracrine IL-2 signaling. Crucially, the absence of either of these distinct Tr cell subsets results in pathogenic autoimmunity, underscoring their nonredundant roles in the preservation of self-tolerance. However, the cellular and molecular factors precipitating IL-2 release and subsequent maintenance of secondary lymphoid organ-resident Tr cells are still poorly understood. In this study, we report that IL-2-dependent Tr cells in the spleen compete for a limiting supply of paracrine IL-2 generated by autoreactive CD4(+) T cells in response to MHC class II-restricted autoantigen activation by 33D1(+)CD11b(int) dendritic cells. Manipulating this cellular circuit culminating in IL-2 production could have clinical benefits in settings in which diminished Tr cell abundance is desired.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/inmunología , Interleucina-2/inmunología , Bazo/inmunología , Linfocitos T Reguladores/inmunología , Animales , Técnicas de Cocultivo , Interleucina-2/sangre , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Bazo/citología
6.
Immunol Rev ; 259(1): 40-59, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24712458

RESUMEN

Regulatory T (Treg) cells play a vital role in the prevention of autoimmunity and the maintenance of self-tolerance, but these cells also have an active role in inhibiting immune responses during viral, bacterial, and parasitic infections. Although excessive Treg activity can lead to immunodeficiency, chronic infection, and cancer, too little Treg activity results in autoimmunity and immunopathology and impairs the quality of pathogen-specific responses. Recent studies have helped define the homeostatic mechanisms that support the diverse pool of peripheral Treg cells under steady-state conditions and delineate how the abundance and function of Treg cells changes during inflammation. These findings are highly relevant for developing effective strategies to manipulate Treg cell activity to promote allograft tolerance and treat autoimmunity, chronic infection, and cancer.


Asunto(s)
Homeostasis/inmunología , Linfocitos T Reguladores/inmunología , Animales , Autoinmunidad/genética , Autoinmunidad/inmunología , Enfermedades Transmisibles/genética , Enfermedades Transmisibles/inmunología , Enfermedades Transmisibles/metabolismo , Regulación de la Expresión Génica , Homeostasis/efectos de los fármacos , Homeostasis/genética , Humanos , Inmunomodulación , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Interleucina-2/metabolismo , Interleucina-2/farmacología , Tejido Linfoide/citología , Tejido Linfoide/inmunología , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/metabolismo , Transcripción Genética
7.
J Immunol ; 189(9): 4574-81, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23002442

RESUMEN

NETosis (neutrophil extracellular trap [NET] generation), a programmed death pathway initiated in mature neutrophils by pathogens and inflammatory mediators, can be a protective process that sequesters microbes and prevents spread of infection, but it can also be a pathological process that causes inflammation and serious tissue injury. Little is known about the regulatory mechanism. Previously, we demonstrated that serpinb1-deficient mice are highly susceptible to pulmonary bacterial and viral infections due to inflammation and tissue injury associated with increased neutrophilic death. In this study, we used in vitro and in vivo approaches to investigate whether SerpinB1 regulates NETosis. We found that serpinb1-deficient bone marrow and lung neutrophils are hypersusceptible to NETosis induced by multiple mediators in both an NADPH-dependent and -independent manner, indicating a deeply rooted regulatory role in NETosis. This role is further supported by increased nuclear expansion (representing chromatin decondensation) of PMA-treated serpinb1-deficient neutrophils compared with wild-type, by migration of SerpinB1 from the cytoplasm to the nucleus of human neutrophils that is coincident with or preceding early conversion of lobulated (segmented) nuclei to delobulated (spherical) morphology, as well as by the finding that exogenous human recombinant SerpinB1 abrogates NET production. NETosis of serpinb1-deficient neutrophils is also increased in vivo during Pseudomonas aeruginosa lung infection. The findings identify a previously unrecognized regulatory mechanism involving SerpinB1 that restricts the production of NETs.


Asunto(s)
Líquido Extracelular/inmunología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Serpinas/fisiología , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Muerte Celular/inmunología , Núcleo Celular/inmunología , Núcleo Celular/metabolismo , Núcleo Celular/patología , Células Cultivadas , Citoplasma/inmunología , Citoplasma/metabolismo , Citoplasma/patología , Líquido Extracelular/metabolismo , Femenino , Predisposición Genética a la Enfermedad , Humanos , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/patología , Neumonía Bacteriana/genética , Neumonía Bacteriana/inmunología , Neumonía Bacteriana/metabolismo , Transporte de Proteínas/genética , Transporte de Proteínas/inmunología , Infecciones por Pseudomonas/genética , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/metabolismo
8.
J Exp Med ; 220(7)2023 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-37097449

RESUMEN

The oral mucosa is a frontline for microbial exposure and juxtaposes several unique tissues and mechanical structures. Based on parabiotic surgery of mice receiving systemic viral infections or co-housing with microbially diverse pet shop mice, we report that the oral mucosa harbors CD8+ CD103+ resident memory T cells (TRM), which locally survey tissues without recirculating. Oral antigen re-encounter during the effector phase of immune responses potentiated TRM establishment within tongue, gums, palate, and cheek. Upon reactivation, oral TRM triggered changes in somatosensory and innate immune gene expression. We developed in vivo methods for depleting CD103+ TRM while sparing CD103neg TRM and recirculating cells. This revealed that CD103+ TRM were responsible for inducing local gene expression changes. Oral TRM putatively protected against local viral infection. This study provides methods for generating, assessing, and in vivo depleting oral TRM, documents their distribution throughout the oral mucosa, and provides evidence that TRM confer protection and trigger responses in oral physiology and innate immunity.


Asunto(s)
Linfocitos T CD8-positivos , Células T de Memoria , Animales , Ratones , Antígenos/metabolismo , Memoria Inmunológica , Mucosa Bucal
9.
Am J Respir Cell Mol Biol ; 47(6): 792-9, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23024061

RESUMEN

Previously, we described the protective role of the neutrophil serine protease inhibitor serpinB1 in preventing early mortality of Pseudomonas aeruginosa lung infection by fostering bacterial clearance and limiting inflammatory cytokines and proteolytic damage. Surfactant protein D (SP-D), which maintains the antiinflammatory pulmonary environment and mediates bacterial removal, was degraded in infected serpinB1-deficient mice. Based on the hypothesis that increased SP-D would rescue or mitigate the pathological effects of serpinB1 deletion, we generated two serpinB1(-/-) lines overexpressing lung-specific rat SP-D and inoculated the mice with P. aeruginosa. Contrary to predictions, bacterial counts in the lungs of SP-D(low)serpinB1(-/-) and SP-D(high) serpinB1(-/-) mice were 4 logs higher than wild-type and not different from serpinB1(-/-) mice. SP-D overexpression also failed to mitigate inflammation (TNF-α), lung injury (free protein, albumin), or excess neutrophil death (free myeloperoxidase, elastase). These pathological markers were higher for infected SP-D(high)serpinB1(-/-) mice than for serpinB1(-/-) mice, although the differences were not significant after controlling for multiple comparisons. The failure of transgenic SP-D to rescue antibacterial defense of serpinB1-deficient mice occurred despite 5-fold or 20-fold increased expression levels, largely normal structure, and dose-dependent bacteria-aggregating activity. SP-D of infected wild-type mice was intact in 43-kD monomers by reducing SDS-PAGE. By contrast, proteolytic fragments of 35, 17, and 8 kD were found in infected SP-D(low)serpinB1(-/-), SP-D(high) serpinB1(-/-) mice, and serpinB1(-/-) mice. Thus, although therapies to increase lung concentration of SP-D may have beneficial applications, the findings suggest that therapy with SP-D may not be beneficial for lung inflammation or infection if the underlying clinical condition includes excess proteolysis.


Asunto(s)
Proteína D Asociada a Surfactante Pulmonar/metabolismo , Serpinas/genética , Animales , Líquido del Lavado Bronquioalveolar , Catepsina G/metabolismo , Femenino , Lesión Pulmonar/inmunología , Lesión Pulmonar/metabolismo , Lesión Pulmonar/microbiología , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Mieloblastina/metabolismo , Neutrófilos/enzimología , Elastasa Pancreática/metabolismo , Neumonía Bacteriana/inmunología , Neumonía Bacteriana/metabolismo , Neumonía Bacteriana/microbiología , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/metabolismo , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/inmunología , Proteína D Asociada a Surfactante Pulmonar/genética , Serpinas/deficiencia
10.
Sci Immunol ; 7(78): eadd3075, 2022 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-36459542

RESUMEN

Respiratory tract resident memory T cells (TRM), typically generated by local vaccination or infection, can accelerate control of pulmonary infections that evade neutralizing antibody. It is unknown whether mRNA vaccination establishes respiratory TRM. We generated a self-amplifying mRNA vaccine encoding the influenza A virus nucleoprotein that is encapsulated in modified dendron-based nanoparticles. Here, we report how routes of immunization in mice, including contralateral versus ipsilateral intramuscular boosts, or intravenous and intranasal routes, influenced influenza-specific cell-mediated and humoral immunity. Parabiotic surgeries revealed that intramuscular immunization was sufficient to establish CD8 TRM in the lung and draining lymph nodes. Contralateral, compared with ipsilateral, intramuscular boosting broadened the distribution of lymph node TRM and T follicular helper cells but slightly diminished resulting levels of serum antibody. Intranasal mRNA delivery established modest circulating CD8 and CD4 T cell memory but augmented distribution to the respiratory mucosa. Combining intramuscular immunizations with an intranasal mRNA boost achieved high levels of both circulating T cell memory and lung TRM. Thus, routes of mRNA vaccination influence humoral and cell-mediated immunity, and intramuscular prime-boosting establishes lung TRM that can be further expanded by an additional intranasal immunization.


Asunto(s)
Linfocitos T CD4-Positivos , Vacunación , Animales , Ratones , ARN Mensajero , Anticuerpos Neutralizantes , Linfocitos T CD8-positivos , Vacunas de ARNm
11.
Cell Host Microbe ; 29(12): 1815-1827.e6, 2021 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-34731647

RESUMEN

Laboratory mice comprise an expeditious model for preclinical vaccine testing; however, vaccine immunogenicity in these models often inadequately translates to humans. Reconstituting physiologic microbial experience to specific pathogen-free (SPF) mice induces durable immunological changes that better recapitulate human immunity. We examined whether mice with diverse microbial experience better model human responses post vaccination. We co-housed laboratory mice with pet-store mice, which have varied microbial exposures, and then assessed immune responses to influenza vaccines. Human transcriptional responses to influenza vaccination are better recapitulated in co-housed mice. Although SPF and co-housed mice were comparably susceptible to acute influenza infection, vaccine-induced humoral responses were dampened in co-housed mice, resulting in poor control upon challenge. Additionally, protective heterosubtypic T cell immunity was compromised in co-housed mice. Because SPF mice exaggerated humoral and T cell protection upon influenza vaccination, reconstituting microbial experience in laboratory mice through co-housing may better inform preclinical vaccine testing.


Asunto(s)
Inmunogenicidad Vacunal , Vacunas contra la Influenza/inmunología , Animales , Femenino , Humanos , Inmunidad Humoral , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Vacunación
12.
bioRxiv ; 2021 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-33948591

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the COVID-19 pandemic. Neutralizing antibodies target the receptor binding domain of the spike (S) protein, a focus of successful vaccine efforts. Concerns have arisen that S-specific vaccine immunity may fail to neutralize emerging variants. We show that vaccination with HAd5 expressing the nucleocapsid (N) protein can establish protective immunity, defined by reduced weight loss and viral load, in both Syrian hamsters and k18-hACE2 mice. Challenge of vaccinated mice was associated with rapid N-specific T cell recall responses in the respiratory mucosa. This study supports the rationale for including additional viral antigens, even if they are not a target of neutralizing antibodies, to broaden epitope coverage and immune effector mechanisms.

13.
Curr Opin Immunol ; 63: 35-42, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32018169

RESUMEN

Advances in the field of T cell memory, including the discovery of tissue residency, continue to add to the list of defined T cell subsets. Here, we briefly review the role of resident memory T cells (TRM) in protective immunity, and propose that they exhibit developmental and migrational plasticity. We discuss T cell classification, the concept of cell type versus 'subset', and the difficulty of inferring developmental relationships between cells occupying malleable differentiation states. We propose that popular subsetting strategies do not perfectly define boundaries of developmental potential. We integrate TRM into a 'terrace' model that classifies memory T cells along a continuous axis of decreasing developmental potential. This model also segregates cells on the basis of migration properties, although different migration properties are viewed as parallel differentiation states that may be permissive to change.


Asunto(s)
Memoria Inmunológica/inmunología , Modelos Inmunológicos , Linfocitos T/inmunología , Animales , Diferenciación Celular/inmunología , Humanos
14.
J Exp Med ; 217(8)2020 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-32568362

RESUMEN

Numerous observations indicate that resident memory T cells (TRM) undergo unusually rapid attrition within the lung. Here we demonstrate that contraction of lung CD8+ T cell responses after influenza infection is contemporized with egress of CD69+/CD103+ CD8+ T cells to the draining mediastinal LN via the lymphatic vessels, which we term retrograde migration. Cells within the draining LN retained canonical markers of lung TRM, including CD103 and CD69, lacked Ly6C expression (also a feature of lung TRM), maintained granzyme B expression, and did not equilibrate among immunized parabiotic mice. Investigations of bystander infection or removal of the TCR from established memory cells revealed that the induction of the TRM phenotype was dependent on antigen recognition; however, maintenance was independent. Thus, local lung infection induces CD8+ T cells with a TRM phenotype that nevertheless undergo retrograde migration, yet remain durably committed to the residency program within the draining LN, where they provide longer-lived regional memory while chronicling previous upstream antigen experiences.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Movimiento Celular/inmunología , Memoria Inmunológica , Virus de la Influenza A/inmunología , Pulmón/inmunología , Ganglios Linfáticos/inmunología , Infecciones por Orthomyxoviridae/inmunología , Animales , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/inmunología , Antígenos Virales/inmunología , Linfocitos T CD8-positivos/patología , Movimiento Celular/genética , Femenino , Pulmón/patología , Pulmón/virología , Ganglios Linfáticos/patología , Masculino , Ratones , Ratones Transgénicos , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/patología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología
15.
Nat Commun ; 10(1): 567, 2019 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-30718505

RESUMEN

The immunosuppressive tumor microenvironment limits the success of current immunotherapies. The host retains memory T cells specific for previous infections throughout the entire body that are capable of executing potent and immediate immunostimulatory functions. Here we show that virus-specific memory T cells extend their surveillance to mouse and human tumors. Reactivating these antiviral T cells can arrest growth of checkpoint blockade-resistant and poorly immunogenic tumors in mice after injecting adjuvant-free non-replicating viral peptides into tumors. Peptide mimics a viral reinfection event to memory CD8+ T cells, triggering antigen presentation and cytotoxic pathways within the tumor, activating dendritic cells and natural killer cells, and recruiting the adaptive immune system. Viral peptide treatment of ex vivo human tumors recapitulates immune activation gene expression profiles observed in mice. Lastly, peptide therapy renders resistant mouse tumors susceptible to PD-L1 blockade. Thus, re-stimulating known antiviral immunity may provide a unique therapeutic approach for cancer immunotherapy.


Asunto(s)
Inmunoterapia/métodos , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/virología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Niño , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Persona de Mediana Edad , Neoplasias/inmunología , Neoplasias/terapia , Linfocitos T Citotóxicos/metabolismo , Microambiente Tumoral/inmunología , Microambiente Tumoral/fisiología , Adulto Joven
16.
Cell Res ; 27(7): 847-848, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28361896

RESUMEN

The consumption of exogenous free fatty acids by tissue-resident memory T (Trm) cells is critical for their long-term survival and antiviral function, and appears to be a conserved feature of Trm cells in both mouse and man, a recent paper published in Nature demonstrates.


Asunto(s)
Linfocitos T CD8-positivos , Memoria Inmunológica , Transporte Biológico , Lípidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA