Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Transl Med ; 21(1): 483, 2023 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-37468934

RESUMEN

BACKGROUND: Viral therapies developed for cancer treatment have classically prioritized direct oncolytic effects over their immune activating properties. However, recent clinical insights have challenged this longstanding prioritization and have shifted the focus to more immune-based mechanisms. Through the potential utilization of novel, inherently immune-stimulating, oncotropic viruses there is a therapeutic opportunity to improve anti-tumor outcomes through virus-mediated immune activation. PV001-DV is an attenuated strain of Dengue virus (DEN-1 #45AZ5) with a favorable clinical safety profile that also maintains the potent immune stimulatory properties characterstic of Dengue virus infection. METHODS: In this study, we utilized in vitro tumor killing and immune multiplex assays to examine the anti-tumor effects of PV001-DV as a potential novel cancer immunotherapy. RESULTS: In vitro assays demonstrated that PV001-DV possesses the ability to directly kill human melanoma cells lines as well as patient melanoma tissue ex vivo. Importantly, further work demonstrated that, when patient peripheral blood mononuclear cells (PBMCs) were exposed to PV001-DV, a substantial induction in the production of apoptotic factors and immunostimulatory cytokines was detected. When tumor cells were cultured with the resulting soluble mediators from these PBMCs, rapid cell death of melanoma and breast cancer cell lines was observed. These soluble mediators also increased dengue virus binding ligands and immune checkpoint receptor, PD-L1 expression. CONCLUSIONS: The direct in vitro tumor-killing and immune-mediated tumor cytotoxicity facilitated by PV001-DV contributes support of its upcoming clinical evaluation in patients with advanced melanoma who have failed prior therapy.


Asunto(s)
Virus del Dengue , Dengue , Melanoma , Viroterapia Oncolítica , Virus Oncolíticos , Humanos , Virus del Dengue/fisiología , Leucocitos Mononucleares , Melanoma/terapia , Células MCF-7 , Inmunidad , Muerte Celular , Viroterapia Oncolítica/métodos
2.
J Immunother Cancer ; 11(7)2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37433716

RESUMEN

BACKGROUND: Immunotherapies are becoming front-line treatments for many advanced cancers, and combinations of two or more therapies are beginning to be investigated. Based on their individual antitumor capabilities, we sought to determine whether combination oncolytic virus (OV) and radiation therapy (RT) may improve cancer outcomes. METHODS: To investigate the activity of this combination therapy, we used in vitro mouse and human cancer cell lines as well as a mouse model of skin cancer. After initial results, we further included immune checkpoint blockade, whose addition constituted a triple combination immunotherapy. RESULTS: Our findings demonstrate that OV and RT reduce tumor growth via conversion of immunologically 'cold' tumors to 'hot', via a CD8+ T cell-dependent and IL-1α-dependent mechanism that is associated with increased PD-1/PD-L1 expression, and the triple combination of OV, RT, and PD-1 checkpoint inhibition impedes tumor growth and prolongs survival. Further, we describe the response of a PD-1-refractory patient with cutaneous squamous cell carcinoma who received the triple combination of OV, RT, and immune checkpoint inhibitor (ICI), and went on to experience unexpected, prolonged control and survival. He remains off-treatment and is without evidence of progression for >44 months since study entry. CONCLUSIONS: Effective systemic antitumor immune response is rarely elicited by a single therapy. In a skin cancer mouse model, we demonstrate improved outcomes with combination OV, RT, and ICI treatment, which is associated with mechanisms involving augmented CD8+ T cell infiltration and IL-1α expression. We report tumor reduction and prolonged survival of a patient with skin cancer treated with combination OV, RT, and ICI. Overall, our data provide strong rationale for combining OV, RT, and ICI for treatment of patients with ICI-refractory skin and potentially other cancers.


Asunto(s)
Carcinoma de Células Escamosas , Inhibidores de Puntos de Control Inmunológico , Viroterapia Oncolítica , Neoplasias Cutáneas , Animales , Humanos , Masculino , Ratones , Carcinoma de Células Escamosas/terapia , Modelos Animales de Enfermedad , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunoterapia , Neoplasias Cutáneas/terapia , Línea Celular Tumoral , Terapia Combinada
3.
Int J Mol Sci ; 25(1)2023 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-38203396

RESUMEN

Breast cancer continues to have a high disease burden worldwide and presents an urgent need for novel therapeutic strategies to improve outcomes. The influenza vaccine offers a unique approach to enhance the anti-tumor immune response in patients with breast cancer. Our study explores the intratumoral use of the influenza vaccine in a triple-negative 4T1 mouse model of breast cancer. We show that the influenza vaccine attenuated tumor growth using a three-dose intratumoral regimen. More importantly, prior vaccination did not alter this improved anti-tumor response. Furthermore, we characterized the effect that the influenza vaccine has on the tumor microenvironment and the underlying mechanisms of action. We established that the vaccine facilitated favorable shifts in restructuring the tumor microenvironment. Additionally, we show that the vaccine's ability to bind sialic acid residues, which have been implicated in having oncogenic functions, emerged as a key mechanism of action. Influenza hemagglutinin demonstrated binding ability to breast cancer cells through sialic acid expression. When administered intratumorally, the influenza vaccine offers a promising therapeutic strategy for breast cancer patients by reshaping the tumor microenvironment and modestly suppressing tumor growth. Its interaction with sialic acids has implications for effective therapeutic application and future research.


Asunto(s)
Neoplasias de la Mama , Vacunas contra la Influenza , Animales , Ratones , Humanos , Femenino , Neoplasias de la Mama/terapia , Hemaglutininas , Microambiente Tumoral , Ácido N-Acetilneuramínico
4.
J Immunol Methods ; 505: 113253, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35358495

RESUMEN

Understanding the dynamics of the tumor microenvironment (TME) has become vital in discovering new targets for effective immunotherapies and enhancing current treatments. However, localization and distribution of immune cells and treatment biomolecules are poorly characterized to date. In this study, a murine Luminal B mammary adenocarcinoma model received a combinatorial treatment of fluorescently labeled anti-PD-1-Cy3 and IL-15 complex-Cy5 injected interperitoneally and intratumorally, respectively. Fluorescent labeling allowed for the visualization of the distribution of IL-15 complexes and anti-PD-1, as well as their localization to immune cells in the TME and tumor-draining lymph node. Using fluorescent microscopy and light sheet microscopy of whole-clarified tumors and draining lymph nodes, the localization of IL-15 complexes was found to be distributed around the periphery of the tumor at 4 h post injection and medially located at the center of the tumor at 24 h post injection, corresponding with high densities of CD8 cells in the tumor present at 48 h and 72 h post injection. Anti-PD-1 was distributed around the perimeter of the tumor and colocalized to IL-15 in the draining lymph nodes 24 h post injection. Colocalization of IL-15 was also established with NK cells, CD8+ T cells, and macrophages. This study develops a novel method to spatiotemporally track fluorescently labeled immunotherapeutic biomolecules in vivo, with implications for optimizing and further understanding the pharmacokinetics of clinical immunotherapies.


Asunto(s)
Neoplasias , Microambiente Tumoral , Animales , Linfocitos T CD8-positivos , Interleucina-15 , Ganglios Linfáticos , Ratones , Neoplasias/patología
5.
Vaccines (Basel) ; 9(12)2021 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-34960243

RESUMEN

Currently approximately 10 million people die each year due to cancer, and cancer is the cause of every sixth death worldwide. Tremendous efforts and progress have been made towards finding a cure for cancer. However, numerous challenges have been faced due to adverse effects of chemotherapy, radiotherapy, and alternative cancer therapies, including toxicity to non-cancerous cells, the inability of drugs to reach deep tumor tissue, and the persistent problem of increasing drug resistance in tumor cells. These challenges have increased the demand for the development of alternative approaches with greater selectivity and effectiveness against tumor cells. Cancer immunotherapy has made significant advancements towards eliminating cancer. Our understanding of cancer-directed immune responses and the mechanisms through which immune cells invade tumors have extensively helped us in the development of new therapies. Among immunotherapies, the application of bacteria and bacterial-based products has promising potential to be used as treatments that combat cancer. Bacterial targeting of tumors has been developed as a unique therapeutic option that meets the ongoing challenges of cancer treatment. In comparison with other cancer therapeutics, bacterial-based therapies have capabilities for suppressing cancer. Bacteria are known to accumulate and proliferate in the tumor microenvironment and initiate antitumor immune responses. We are currently well-informed regarding various methods by which bacteria can be manipulated by simple genetic engineering or synthetic bioengineering to induce the production of anti-cancer drugs. Further, bacterial-based cancer therapy (BBCT) can be either used as a monotherapy or in combination with other anticancer therapies for better clinical outcomes. Here, we review recent advances, current challenges, and prospects of bacteria and bacterial products in the development of BBCTs.

6.
Proc Natl Acad Sci U S A ; 117(2): 1119-1128, 2020 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-31888983

RESUMEN

Reprogramming the tumor microenvironment to increase immune-mediated responses is currently of intense interest. Patients with immune-infiltrated "hot" tumors demonstrate higher treatment response rates and improved survival. However, only the minority of tumors are hot, and a limited proportion of patients benefit from immunotherapies. Innovative approaches that make tumors hot can have immediate impact particularly if they repurpose drugs with additional cancer-unrelated benefits. The seasonal influenza vaccine is recommended for all persons over 6 mo without prohibitive contraindications, including most cancer patients. Here, we report that unadjuvanted seasonal influenza vaccination via intratumoral, but not intramuscular, injection converts "cold" tumors to hot, generates systemic CD8+ T cell-mediated antitumor immunity, and sensitizes resistant tumors to checkpoint blockade. Importantly, intratumoral vaccination also provides protection against subsequent active influenza virus lung infection. Surprisingly, a squalene-based adjuvanted vaccine maintains intratumoral regulatory B cells and fails to improve antitumor responses, even while protecting against active influenza virus lung infection. Adjuvant removal, B cell depletion, or IL-10 blockade recovers its antitumor effectiveness. Our findings propose that antipathogen vaccines may be utilized for both infection prevention and repurposing as a cancer immunotherapy.


Asunto(s)
Inmunoterapia/métodos , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/uso terapéutico , Inyecciones Intralesiones , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Animales , Linfocitos B , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Linfocitos T CD8-positivos/inmunología , Humanos , Inmunidad Celular , Subtipo H1N1 del Virus de la Influenza A/inmunología , Gripe Humana , Interleucina-10 , Pulmón/patología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos C57BL , Proteínas Represoras/genética , Estaciones del Año , Piel , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/patología , Escualeno/administración & dosificación , Microambiente Tumoral/efectos de los fármacos , Vacunación
7.
J Immunol ; 199(7): 2536-2546, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28814601

RESUMEN

IL-15 is an essential cytokine known to promote T cell survival and activate the effector function of memory phenotype CD8 T cells. Blocking IL-15 signals also significantly impacts tissue-specific effector and memory CD8 T cell formation. In this study, we demonstrate that IL-15 influences the generation of memory CD8 T cells by first promoting their accumulation into mucosal tissues and second by sustaining expression of Bcl-6 and T-bet. We show that the mechanism for this recruitment is largely dependent on mammalian target of rapamycin and its subsequent inactivation of FoxO1. Last, we show that IL-15 complexes delivered locally to mucosal tissues without reinfection is an effective strategy to enhance establishment of tissue resident memory CD8 T cells within mucosal tissues. This study provides mechanistic insight into how IL-15 controls the generation of memory CD8 T cells and influences their trafficking and ability to take up residence within peripheral tissues.


Asunto(s)
Linfocitos T CD8-positivos/fisiología , Memoria Inmunológica , Interleucina-15/fisiología , Membrana Mucosa/inmunología , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Movimiento Celular , Proteína Forkhead Box O1/metabolismo , Interleucina-15/genética , Interleucina-15/farmacología , Ratones , Ratones Endogámicos C57BL , Membrana Mucosa/citología , Membrana Mucosa/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-6/genética , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Proteínas de Dominio T Box/genética , Subgrupos de Linfocitos T/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo
8.
Cell Rep ; 17(4): 957-965, 2016 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-27760326

RESUMEN

In light of increased cancer prevalence and cancer-specific deaths in patients with infections, we investigated whether infections alter anti-tumor immune responses. We report that acute influenza infection of the lung promotes distal melanoma growth in the dermis and leads to accelerated cancer-specific host death. Furthermore, we show that during influenza infection, anti-melanoma CD8+ T cells are shunted from the tumor to the infection site, where they express high levels of the inhibitory receptor programmed cell death protein 1 (PD-1). Immunotherapy to block PD-1 reverses this loss of anti-tumor CD8+ T cells from the tumor and decreases infection-induced tumor growth. Our findings show that acute non-oncogenic infection can promote cancer growth, raising concerns regarding acute viral illness sequelae. They also suggest an unexpected role for PD-1 blockade in cancer immunotherapy and provide insight into the immune response when faced with concomitant challenges.


Asunto(s)
Melanoma/inmunología , Melanoma/patología , Oncogenes , Infecciones por Orthomyxoviridae/patología , Enfermedad Aguda , Animales , Linfocitos T CD8-positivos/inmunología , Proliferación Celular , Pulmón/patología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Receptor de Muerte Celular Programada 1/metabolismo
10.
J Immunol ; 193(5): 2067-71, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25070853

RESUMEN

Mucosal tissues are subject to frequent pathogen exposure and are major sites for transmission of infectious disease. CD8 T cells play a critical role in controlling mucosa-acquired infections even though their migration into mucosal tissues is tightly regulated. The mechanisms and signals that control the formation of tissue-resident memory CD8 T cells are poorly understood; however, one key regulator of memory CD8 T cell differentiation, mammalian target of rapamycin kinase, can be inhibited by rapamycin. We report that, despite enhancing the formation of memory CD8 T cells in secondary lymphoid tissues, rapamycin inhibits the formation of resident memory CD8 T cells in the intestinal and vaginal mucosa. The ability of rapamycin to block the formation of functional resident CD8 T cells in mucosal tissues protected mice from a model of CD8 T cell-mediated lethal intestinal autoimmunity. These findings demonstrate an opposing role for mammalian target of rapamycin in the formation of resident versus nonresident CD8 T cell immunity.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Inmunidad Mucosa/fisiología , Memoria Inmunológica/fisiología , Mucosa Intestinal/lesiones , Modelos Inmunológicos , Serina-Treonina Quinasas TOR/inmunología , Animales , Autoinmunidad/fisiología , Linfocitos T CD8-positivos/citología , Femenino , Mucosa Intestinal/citología , Ratones , Vagina/citología , Vagina/inmunología
12.
Oncoimmunology ; 2(2): e22731, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23525138

RESUMEN

The major objectives of tumor vaccination are to induce the regression of established tumors and to favor the establishment of long-lasting tumor-specific immunity, capable of protecting the host from relapse. Immunotherapeutic strategies such as the administration of tumor-associated antigenic peptides offer one means to boost preexisting antitumor CD8+ T cell immunity. Our recent work reveals that established breast tumors are rejected and tumor recurrence prevented when low-dose irradiation is combined with the adoptive transfer of Mammaglobin A epitope-specific CD8+ T cells.

13.
PLoS One ; 7(7): e41240, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22911764

RESUMEN

Adoptive T cell therapy has proven to be beneficial in a number of tumor systems by targeting the relevant tumor antigen. The tumor antigen targeted in our model is Mammaglobin-A, expressed by approximately 80% of human breast tumors. Here we evaluated the use of adoptively transferred Mammaglobin-A specific CD8 T cells in combination with low dose irradiation to induce breast tumor rejection and prevent relapse. We show Mammaglobin-A specific CD8 T cells generated by DNA vaccination with all epitopes (Mammaglobin-A2.1, A2.2, A2.4 and A2.6) and full-length DNA in vivo resulted in heterogeneous T cell populations consisting of both effector and central memory CD8 T cell subsets. Adoptive transfer of spleen cells from all Mammaglobin-A2 immunized mice into tumor-bearing SCID/beige mice induced tumor regression but this anti-tumor response was not sustained long-term. Additionally, we demonstrate that only the adoptive transfer of Mammaglobin-A2 specific CD8 T cells in combination with a single low dose of irradiation prevents tumors from recurring. More importantly we show that this single dose of irradiation results in the down regulation of the macrophage scavenger receptor 1 on dendritic cells within the tumor and reduces lipid uptake by tumor resident dendritic cells potentially enabling the dendritic cells to present tumor antigen more efficiently and aid in tumor clearance. These data reveal the potential for adoptive transfer combined with a single low dose of total body irradiation as a suitable therapy for the treatment of established breast tumors and the prevention of tumor recurrence.


Asunto(s)
Traslado Adoptivo , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/terapia , Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/inmunología , Mamoglobina A/inmunología , Irradiación Corporal Total , Animales , Neoplasias de la Mama/mortalidad , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Citotoxicidad Inmunológica , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Epítopos de Linfocito T/genética , Femenino , Humanos , Memoria Inmunológica , Interferón gamma/biosíntesis , Subunidad alfa del Receptor de Interleucina-7/metabolismo , Selectina L/metabolismo , Metabolismo de los Lípidos , Mamoglobina A/química , Ratones , Ratones SCID , Ratones Transgénicos , Receptores Depuradores de Clase A/metabolismo , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis , Vacunas de ADN/inmunología
14.
Nat Med ; 18(3): 422-8, 2012 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-22366950

RESUMEN

CD4-unhelped CD8(+) T cells are functionally defective T cells primed in the absence of CD4(+) T cell help. Given the co-stimulatory role of natural-killer group 2, member D protein (NKG2D) on CD8(+) T cells, we investigated its ability to rescue these immunologically impotent cells. We demonstrate that augmented co-stimulation through NKG2D during priming paradoxically rescues memory, but not effector, CD8(+) T cell responses. NKG2D-mediated rescue is characterized by reversal of elevated transcription factor T-box expressed in T cells (T-bet) expression and recovery of interleukin-2 and interferon-γ production and cytolytic responses. Rescue is abrogated in CD8(+) T cells lacking NKG2D. Augmented co-stimulation through NKG2D confers a high rate of survival to mice lacking CD4(+) T cells in a CD4-dependent influenza model and rescues HIV-specific CD8(+) T cell responses from CD4-deficient HIV-positive donors. These findings demonstrate that augmented co-stimulation through NKG2D is effective in rescuing CD4-unhelped CD8(+) T cells from their pathophysiological fate and may provide therapeutic benefits.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Infecciones por VIH/inmunología , Gripe Humana/inmunología , Subfamilia K de Receptores Similares a Lectina de Células NK/genética , Subfamilia K de Receptores Similares a Lectina de Células NK/inmunología , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD8-positivos/citología , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Expresión Génica , VIH-1/inmunología , Humanos , Inmunidad Celular , Gripe Humana/genética , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-2/inmunología , Interleucina-2/metabolismo , Ratones , Ratones Endogámicos C57BL , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Proteínas de Dominio T Box/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Vacunas de ADN/inmunología
15.
PLoS One ; 6(2): e16529, 2011 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-21364749

RESUMEN

Memory CD4 T cells play a vital role in protection against re-infection by pathogens as diverse as helminthes or influenza viruses. Inducible costimulator (ICOS) is highly expressed on memory CD4 T cells and has been shown to augment proliferation and survival of activated CD4 T cells. However, the role of ICOS costimulation on the development and maintenance of memory CD4 T cells remains controversial. Herein, we describe a significant defect in the number of effector memory (EM) phenotype cells in ICOS(-/-) and ICOSL(-/-) mice that becomes progressively more dramatic as the mice age. This decrease was not due to a defect in the homeostatic proliferation of EM phenotype CD4 T cells in ICOS(-/-) or ICOSL(-/-) mice. To determine whether ICOS regulated the development or survival of EM CD4 T cells, we utilized an adoptive transfer model. We found no defect in development of EM CD4 T cells, but long-term survival of ICOS(-/-) EM CD4 T cells was significantly compromised compared to wild-type cells. The defect in survival was specific to EM cells as the central memory (CM) ICOS(-/-) CD4 T cells persisted as well as wild type cells. To determine the physiological consequences of a specific defect in EM CD4 T cells, wild-type and ICOS(-/-) mice were infected with influenza virus. ICOS(-/-) mice developed significantly fewer influenza-specific EM CD4 T cells and were more susceptible to re-infection than wild-type mice. Collectively, our findings demonstrate a role for ICOS costimulation in the maintenance of EM but not CM CD4 T cells.


Asunto(s)
Antígenos de Diferenciación de Linfocitos T/fisiología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/fisiología , Memoria Inmunológica/genética , Proteínas/fisiología , Animales , Antígenos de Diferenciación de Linfocitos T/genética , Linfocitos T CD4-Positivos/metabolismo , Supervivencia Celular/genética , Citoprotección/genética , Citoprotección/inmunología , Predisposición Genética a la Enfermedad , Memoria Inmunológica/fisiología , Ligando Coestimulador de Linfocitos T Inducibles , Proteína Coestimuladora de Linfocitos T Inducibles , Virus de la Influenza A/fisiología , Interleucina-4/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/inmunología , Fenotipo , Proteínas/genética
16.
Adv Exp Med Biol ; 684: 69-78, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20795541

RESUMEN

Immunological memory is considered the hallmark of adaptive, or acquired, immunity. That ability of our immune system to recognize and respond to those pathogens we have encountered before not only typifies acquired immunity but has provided the basis for the most notable of medical interventions: vaccination. Yet, as much as we now know about this process, we are still on the cusp of fully understanding how memory T cells develop, how they are maintained and the importance of memory T-cell heterogeneity. In this review we will primarily focus on our understanding of CD8 T-cell memory generated during acute infections and how precursor frequency influences their development and functional attributes.


Asunto(s)
Memoria Inmunológica/inmunología , Células Precursoras de Linfocitos T/fisiología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/fisiología , Animales , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular , Humanos , Activación de Linfocitos/inmunología , Recuento de Linfocitos , Linfopoyesis , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Transgenes
17.
Blood ; 114(10): 2121-30, 2009 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-19617575

RESUMEN

The ability of the adaptive immune system to respond rapidly and robustly upon repeated antigen exposure is known as immunologic memory, and it is thought that acquisition of memory T-cell function is an irreversible differentiation event. In this study, we report that many phenotypic and functional characteristics of antigen-specific CD8 memory T cells are lost when they are deprived of contact with dendritic cells. Under these circumstances, memory T cells reverted from G(1) to the G(0) cell-cycle state and responded to stimulation like naive T cells, as assessed by proliferation, dependence upon costimulation, and interferon-gamma production, without losing cell surface markers associated with memory. The memory state was maintained by signaling via members of the tumor necrosis factor receptor superfamily, CD27 and 4-1BB. Foxo1, a transcription factor involved in T-cell quiescence, was reduced in memory cells, and stimulation of naive CD8 cells via CD27 caused Foxo1 to be phosphorylated and emigrate from the nucleus in a phosphatidylinositol-3 kinase-dependent manner. Consistent with these results, maintenance of G(1) in vivo was compromised in antigen-specific memory T cells in vesicular stomatitis virus-infected CD27-deficient mice. Therefore, sustaining the functional phenotype of T memory cells requires active signaling and maintenance.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Comunicación Celular/inmunología , Células Dendríticas/inmunología , Memoria Inmunológica/inmunología , Transducción de Señal/inmunología , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Transporte Activo de Núcleo Celular/genética , Transporte Activo de Núcleo Celular/inmunología , Animales , Antígenos Virales/inmunología , Comunicación Celular/genética , Núcleo Celular/inmunología , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/inmunología , Fase G1/inmunología , Memoria Inmunológica/genética , Interferón gamma/inmunología , Ratones , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/inmunología , Fosforilación/genética , Fosforilación/inmunología , Fase de Descanso del Ciclo Celular/inmunología , Transducción de Señal/genética , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Estomatitis Vesicular/inmunología , Vesiculovirus/inmunología
18.
J Immunol ; 179(1): 36-40, 2007 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-17579018

RESUMEN

Memory CD8 T cells, essential for defense against intracellular pathogens, are heterogeneous with respect to phenotype and function. Constitutively lytic effector memory cells primarily reside in nonlymphoid tissues, whereas secondary lymphoid tissues contain functionally quiescent central memory cells. However, the mechanism by which functionally distinct memory populations are maintained is unknown. In this study, we show that resting CD8 memory cells modified their functional abilities upon entry into nonlymphoid tissues, as exemplified by the induction of granzyme B and lytic activity. Contemporaneously, the costimulator CD27 was down-regulated. These findings hold important implications for memory cell lineage development and tissue-specific immunity.


Asunto(s)
Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Movimiento Celular/inmunología , Memoria Inmunológica , Traslado Adoptivo , Animales , Linfocitos T CD8-positivos/enzimología , Linfocitos T CD8-positivos/trasplante , Citotoxicidad Inmunológica , Regulación hacia Abajo/inmunología , Granzimas/biosíntesis , Inmunofenotipificación , Hígado/citología , Hígado/inmunología , Pulmón/citología , Pulmón/inmunología , Tejido Linfoide/citología , Tejido Linfoide/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/biosíntesis , Regulación hacia Arriba/inmunología
19.
Eur J Immunol ; 37(4): 925-34, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17330818

RESUMEN

The role of CD44 in T cell biology remains incompletely understood. Although studies using anti-CD44 antibodies have implicated this cell adhesion molecule in a variety of important T cell processes, few T cell defects have been reported in CD44-deficient mice. We have assessed the requirement for CD44 in T cell development and mature T cell function by analyzing mice in which CD44(-/-) and WT cells were produced simultaneously. In mixed (CD44(-/-) + CD44(+/+)) bone marrow chimeras, production of CD44(-/-) T cells was shown to be reduced compared to WT cells due to inefficient intrathymic development. In addition, mature CD44(-/-) CD8(+) T cells generated a substantially lower response than WT T cells after infection of mice with lymphocytic choriomeningitis virus, with the reduction in response apparent in both lymphoid and non-lymphoid tissues. Overall, these results demonstrate a poor capacity of CD44(-/-) T lineage cells to compete with WT cells at multiple levels, implicating CD44 in normal T cell function.


Asunto(s)
Comunicación Celular/inmunología , Diferenciación Celular/inmunología , Receptores de Hialuranos/fisiología , Subgrupos de Linfocitos T/inmunología , Linfocitos T/inmunología , Animales , Infecciones por Arenaviridae/inmunología , Comunicación Celular/genética , Diferenciación Celular/genética , Receptores de Hialuranos/biosíntesis , Receptores de Hialuranos/genética , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Subgrupos de Linfocitos T/metabolismo , Linfocitos T/clasificación , Linfocitos T/citología
20.
Infect Immun ; 75(4): 2046-62, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17220323

RESUMEN

We examined the interactions of live and lysed spirochetes with innate immune cells. THP-1 monocytoid cells were activated to comparable extents by live Borrelia burgdorferi and by B. burgdorferi and Treponema pallidum lysates but were poorly activated by live T. pallidum. Because THP-1 cells poorly internalized live spirochetes, we turned to an ex vivo peripheral blood mononuclear cell system that would more closely reflect spirochete-mononuclear phagocyte interactions that occur during actual infection. In this system, B. burgdorferi induced significantly greater monocyte activation and inflammatory cytokine production than did borrelial lysates or T. pallidum, and only B. burgdorferi elicited gamma interferon (IFN-gamma) from NK cells. B. burgdorferi was phagocytosed avidly by monocytes, while T. pallidum was not, suggesting that the enhanced response to live B. burgdorferi was due to phagocytosis of the organism. When cytochalasin D was used to block phagocytosis of live B. burgdorferi, cytokine production decreased to levels comparable to those induced by B. burgdorferi lysates, while the IFN-gamma response was abrogated altogether. In the presence of human syphilitic serum, T. pallidum was efficiently internalized and initiated responses resembling those observed with live B. burgdorferi, including the production of IFN-gamma by NK cells. Depletion of monocytes revealed that they were the primary source of inflammatory cytokines, while dendritic cells (DCs) directed IFN-gamma production from innate lymphocytes. Thus, phagocytosis of live spirochetes initiates cell activation programs in monocytes and DCs that differ qualitatively and quantitatively from those induced at the cell surface by lipoprotein-enriched lysates. The greater stimulatory capacity of B. burgdorferi versus T. pallidum appears to be explained by the successful recognition and phagocytosis of B. burgdorferi by host cells and the ability of T. pallidum to avoid detection and uptake by virtue of its denuded outer membrane rather than by differences in surface lipoprotein expression.


Asunto(s)
Borrelia burgdorferi/inmunología , Interferón gamma/biosíntesis , Linfocitos/inmunología , Monocitos/inmunología , Fagocitosis , Treponema pallidum/inmunología , Línea Celular , Células Cultivadas , Citocalasina D/farmacología , Células Dendríticas/inmunología , Citometría de Flujo , Humanos , Inmunidad Innata , Factores Inmunológicos/farmacología , Células Asesinas Naturales/inmunología , Procedimientos de Reducción del Leucocitos , Monocitos/microbiología , Suero/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...