Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Ther ; 31(7): 2132-2153, 2023 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-37194236

RESUMEN

To leverage complementary mechanisms for cancer cell removal, we developed a novel cell engineering and therapeutic strategy co-opting phagocytic clearance and antigen presentation activity into T cells. We engineered a chimeric engulfment receptor (CER)-1236, which combines the extracellular domain of TIM-4, a phagocytic receptor recognizing the "eat me" signal phosphatidylserine, with intracellular signaling domains (TLR2/TIR, CD28, and CD3ζ) to enhance both TIM-4-mediated phagocytosis and T cell cytotoxic function. CER-1236 T cells demonstrate target-dependent phagocytic function and induce transcriptional signatures of key regulators responsible for phagocytic recognition and uptake, along with cytotoxic mediators. Pre-clinical models of mantle cell lymphoma (MCL) and EGFR mutation-positive non-small cell lung cancer (NSCLC) demonstrate collaborative innate-adaptive anti-tumor immune responses both in vitro and in vivo. Treatment with BTK (MCL) and EGFR (NSCLC) inhibitors increased target ligand, conditionally driving CER-1236 function to augment anti-tumor responses. We also show that activated CER-1236 T cells exhibit superior cross-presentation ability compared with conventional T cells, triggering E7-specific TCR T responses in an HLA class I- and TLR-2-dependent manner, thereby overcoming the limited antigen presentation capacity of conventional T cells. Therefore, CER-1236 T cells have the potential to achieve tumor control by eliciting both direct cytotoxic effects and indirect-mediated cross-priming.


Asunto(s)
Antineoplásicos , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Adulto , Linfocitos T , Reactividad Cruzada , Fosfatidilserinas , Antígenos de Neoplasias , Receptores ErbB , Inmunoterapia Adoptiva , Receptores de Antígenos de Linfocitos T/genética
2.
Surgeon ; 19(5): e245-e255, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33423920

RESUMEN

BACKGROUND: There is currently no validated patient-reported outcome measure (PROM) that is specific to nipple-areola complex (NAC) reconstruction. This paper evaluates all patient-reported outcomes for NAC reconstruction in the literature. METHODS: Systematic literature searches of The Cochrane Central Register of Controlled Trials, MEDLINE and World Health Organization International Clinical Trials Registry Platform were conducted to identify all primary studies with patient-reported outcomes for NAC reconstruction. The primary outcome measures were patient satisfaction rates for appearance and symmetry of NAC reconstruction. RESULTS: Fifty-nine papers were included in this review. Reported patient satisfaction was generally high, with the pooled average satisfaction rate for appearance being 81.9% and symmetry 80.3%. 89.5% of respondents would do it again and 94.8% would recommend it to others. There is no standardised or validated PROM specific to NAC reconstruction and this contributes to a lack of conclusive findings from studies in this area. CONCLUSION: There is a need for a validated PROM that is specific to NAC reconstruction, in order to serve as a standardised outcome assessment to guide further research and improve patient care.


Asunto(s)
Neoplasias de la Mama , Mamoplastia , Neoplasias de la Mama/cirugía , Femenino , Humanos , Pezones/cirugía , Medición de Resultados Informados por el Paciente , Satisfacción del Paciente , Estudios Retrospectivos
3.
J Infect Dis ; 219(7): 1162-1171, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30371803

RESUMEN

BACKGROUND: A challenge to the design of improved therapeutic agents and prevention strategies for neuroinvasive infection and associated disease is the lack of known natural immune correlates of protection. A relevant model to study such correlates is offered by the Collaborative Cross (CC), a panel of recombinant inbred mouse strains that exhibit a range of disease manifestations upon infection. METHODS: We performed an extensive screen of CC-F1 lines infected with West Nile virus (WNV), including comprehensive immunophenotyping, to identify groups of lines that exhibited viral neuroinvasion or neuroinvasion with disease and lines that remained free of WNV neuroinvasion and disease. RESULTS: Our data reveal that protection from neuroinvasion and disease is multifactorial and that several immune outcomes can contribute. Immune correlates identified include decreased suppressive activity of regulatory T cells at steady state, which correlates with peripheral restriction of the virus. Further, a rapid contraction of WNV-specific CD8+ T cells in the brain correlated with protection from disease. CONCLUSIONS: These immune correlates of protection illustrate additional networks and pathways of the WNV immune response that cannot be observed in the C57BL/6 mouse model. Additionally, correlates of protection exhibited before infection, at baseline, provide insight into phenotypic differences in the human population that may predict clinical outcomes upon infection.


Asunto(s)
Ratones de Colaboración Cruzada/inmunología , Enfermedades del Sistema Nervioso/inmunología , Fiebre del Nilo Occidental/inmunología , Virus del Nilo Occidental/inmunología , 2',5'-Oligoadenilato Sintetasa/genética , Inmunidad Adaptativa , Animales , Encéfalo/inmunología , Encéfalo/patología , Relación CD4-CD8 , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Ratones de Colaboración Cruzada/genética , Modelos Animales de Enfermedad , Heterocigoto , Inmunidad Innata , Inmunofenotipificación , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedades del Sistema Nervioso/microbiología , Polimorfismo Genético , Bazo/inmunología , Bazo/patología , Linfocitos T Reguladores/inmunología , Fiebre del Nilo Occidental/complicaciones , Fiebre del Nilo Occidental/genética
4.
J Cell Biochem ; 120(10): 18320-18331, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31211444

RESUMEN

Alzheimer's disease (AD) is an irreversible, progressive brain disorder responsible for memory loss leading to the inability to carry out the simplest tasks. AD is one of the leading causes of death in the United States. As yet there are no effective medications to treat this debilitating disease. In recent years, a human gene called bridging integrator 1 (BIN1) has emerged as one of the most important genes in affecting the incidence of sporadic AD. Bin1 can directly bind to Tau and mediates late onset AD risk by modulating Tau pathology. Recently our group found Bin1 antibody could exert drug-like properties in an animal model of ulcerative colitis. We hypothesized that the Bin1 monoclonal antibody (mAb) could be used in the treatment of AD by lowering the levels of Tau in cell culture and animal models. Cell culture studies confirmed that the Bin1 mAb (99D) could lower the levels of phosphorylated Tau (pTau). Multiple mechanisms aided by endosomal proteins and Fc gamma receptors are involved in the uptake of Bin1 mAb into cells. In Tau expressing cell culture, the Bin1 mAb induces the proteasome machinery leading to ubiquitination of molecules thereby preventing cell stress. In vivo studies demonstrated that treatment of P301S mice expressing Tau with the Bin1 mAb survived longer than the untreated mice. Our data confirm that Bin1 mAb lowers the levels of pTau and could be a drug candidate in the treatment of AD.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Enfermedad de Alzheimer/metabolismo , Anticuerpos Monoclonales/farmacología , Proteínas Nucleares/inmunología , Proteínas Supresoras de Tumor/inmunología , Proteínas tau/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Animales , Anticuerpos Monoclonales/inmunología , Células CACO-2 , Modelos Animales de Enfermedad , Endosomas/metabolismo , Células HEK293 , Humanos , Ratones Transgénicos , Mutación , Fosforilación , Complejo de la Endopetidasa Proteasomal/metabolismo , Receptores de IgG/metabolismo , Análisis de Supervivencia , Ubiquitinación/efectos de los fármacos , Proteínas tau/antagonistas & inhibidores , Proteínas tau/genética
5.
J Cell Biochem ; 120(3): 4225-4237, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30269357

RESUMEN

Patients afflicted with ulcerative colitis (UC) are at increased risk of colorectal cancer. While its causes are not fully understood, UC is associated with defects in colonic epithelial barriers that sustain inflammation of the colon mucosa caused by recruitment of lymphocytes and neutrophils into the lamina propria. Based on genetic evidence that attenuation of the bridging integrator 1 (Bin1) gene can limit UC pathogenicity in animals, we have explored Bin1 targeting as a therapeutic option. Early feasibility studies in the dextran sodium sulfate mouse model of experimental colitis showed that administration of a cell-penetrating Bin1 monoclonal antibody (Bin1 mAb 99D) could prevent lesion formation in the colon mucosa in part by preventing rupture of lymphoid follicles. In vivo administration of Bin1 mAb altered tight junction protein expression and cecal barrier function. Strikingly, electrophysiology studies in organ cultures showed that Bin1 mAb could elevate resistance and lower 14 C-mannitol leakage across the cecal mucosa, consistent with a direct strengthening of colonic barrier function. Transcriptomic analyses of colitis tissues highlighted altered expression of genes involved in circadian rhythm, lipid metabolism, and inflammation, with a correction of the alterations by Bin1 mAb treatment to patterns characteristic of normal tissues. Overall, our results suggest that Bin1 mAb protects against UC by directly improving colonic epithelial barrier function to limit gene expression and cytokine programs associated with colonic inflammation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Colitis Ulcerosa/terapia , Inmunoterapia/métodos , Mucosa Intestinal/metabolismo , Proteínas del Tejido Nervioso/inmunología , Sustancias Protectoras/uso terapéutico , Uniones Estrechas/metabolismo , Proteínas Supresoras de Tumor/inmunología , Animales , Células CACO-2 , Colitis Ulcerosa/inducido químicamente , Citocinas/metabolismo , Sulfato de Dextran/farmacología , Modelos Animales de Enfermedad , Expresión Génica/efectos de los fármacos , Humanos , Mucosa Intestinal/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/efectos de los fármacos , Proteínas de Uniones Estrechas/metabolismo , Uniones Estrechas/efectos de los fármacos
6.
PLoS Pathog ; 12(11): e1005996, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27806117

RESUMEN

Infection with West Nile virus (WNV) leads to a range of disease outcomes, including chronic infection, though lack of a robust mouse model of chronic WNV infection has precluded identification of the immune events contributing to persistent infection. Using the Collaborative Cross, a population of recombinant inbred mouse strains with high levels of standing genetic variation, we have identified a mouse model of persistent WNV disease, with persistence of viral loads within the brain. Compared to lines exhibiting no disease or marked disease, the F1 cross CC(032x013)F1 displays a strong immunoregulatory signature upon infection that correlates with restraint of the WNV-directed cytolytic response. We hypothesize that this regulatory T cell response sufficiently restrains the immune response such that a chronic infection can be maintained in the CNS. Use of this new mouse model of chronic neuroinvasive virus will be critical in developing improved strategies to prevent prolonged disease in humans.


Asunto(s)
Linfocitos T Reguladores/inmunología , Fiebre del Nilo Occidental/inmunología , Animales , Enfermedad Crónica , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Humanos , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , Virus del Nilo Occidental/inmunología
7.
Dig Dis Sci ; 61(2): 423-32, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26195312

RESUMEN

BACKGROUND: Ulcerative colitis (UC) is associated with defects in colonic epithelial barriers as well as inflammation of the colon mucosa resulting from the recruitment of lymphocytes and neutrophils in the lamina propria. Patients afflicted with UC are at increased risk of colorectal cancer. Currently, UC management employs general anti-inflammatory strategies associated with a variety of side effects, including heightened risks of infection, in patients where the therapy is variably effective. Thus, second generation drugs that can more effectively and selectively limit UC are desired. AIM: Building on genetic evidence that attenuation of the Bin1 (Bridging integrator 1) gene can limit UC pathogenicity in the mouse, we pursued Bin1 targeting as a therapeutic option. METHODS: Mice were injected with a single dose of Bin1 mAb followed by oral administration of 3 % DSS in water for 7 days. RESULTS: In this study, we offer preclinical proof of concept for a monoclonal antibody (mAb) targeting the Bin1 protein that blunts UC pathogenicity in a mouse model of experimental colitis. Administration of Bin1 mAb reduced colitis morbidity in mice; whereas unprotected mice is characterized by severe lesions throughout the mucosa, rupture of the lymphoid follicle, high-level neutrophil and lymphocyte infiltration into the mucosal and submucosal areas, and loss of surface crypts. In vitro studies in human Caco-2 cells showed that Bin1 antibody altered the expression of tight junction proteins and improved barrier function. CONCLUSIONS: Our results suggest that a therapy based on Bin1 monoclonal antibody supporting mucosal barrier function and protecting integrity of the lymphoid follicle could offer a novel strategy to treat UC and possibly limit risks of colorectal cancer.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Anticuerpos Monoclonales/uso terapéutico , Colitis/terapia , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas Nucleares/metabolismo , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Línea Celular , Relación Dosis-Respuesta a Droga , Humanos , Inmunoglobulina G , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Ratones , Proteínas del Tejido Nervioso/inmunología , Proteínas Nucleares/genética , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/inmunología
8.
J Virol ; 88(24): 13990-4001, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25253340

RESUMEN

UNLABELLED: Pattern recognition receptors (PRR) sense certain molecular patterns uniquely expressed by pathogens. Retinoic-acid-inducible gene I (RIG-I) is a cytosolic PRR that senses viral nucleic acids and induces innate immune activation and secretion of type I interferons (IFNs). Here, using influenza vaccine antigens, we investigated the consequences of activating the RIG-I pathway for antigen-specific adaptive immune responses. We found that mice immunized with influenza vaccine antigens coadministered with 5'ppp-double-stranded RNA (dsRNA), a RIG-I ligand, developed robust levels of hemagglutination-inhibiting antibodies, enhanced germinal center reaction, and T follicular helper cell responses. In addition, RIG-I activation enhanced antibody affinity maturation and plasma cell responses in the draining lymph nodes, spleen, and bone marrow and conferred protective immunity against virus challenge. Importantly, activation of the RIG-I pathway was able to reduce the antigen requirement by 10- to 100-fold in inducing optimal influenza-specific cellular and humoral responses, including protective immunity. The effects induced by 5'ppp-dsRNA were significantly dependent on type I IFN and IPS-1 (an adapter protein downstream of the RIG-I pathway) signaling but were independent of the MyD88- and TLR3-mediated pathways. Our results show that activation of the RIG-I-like receptor pathway programs the innate immunity to achieve qualitatively and quantitatively enhanced protective cellular adaptive immune responses even at low antigen doses, and this indicates the potential utility of RIG-I ligands as molecular adjuvants for viral vaccines. IMPORTANCE: The recently discovered RNA helicase family of RIG-I-like receptors (RLRs) is a critical component of host defense mechanisms responsible for detecting viruses and triggering innate antiviral cytokines that help control viral replication and dissemination. In this study, we show that the RLR pathway can be effectively exploited to enhance adaptive immunity and protective immune memory against viral infection. Our results show that activation of the RIG-I pathway along with influenza vaccination programs the innate immunity to induce qualitatively and quantitatively superior protective adaptive immunity against pandemic influenza viruses. More importantly, RIG-I activation at the time of vaccination allows induction of robust adaptive responses even at low vaccine antigen doses. These results highlight the potential utility of exploiting the RIG-I pathway to enhance viral-vaccine-specific immunity and have broader implications for designing better vaccines in general.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , ARN Helicasas DEAD-box/metabolismo , Centro Germinal/inmunología , Vacunas contra la Influenza/inmunología , ARN Bicatenario/administración & dosificación , Transducción de Señal , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Anticuerpos Antivirales/sangre , Proliferación Celular , Proteína 58 DEAD Box , Modelos Animales de Enfermedad , Pruebas de Inhibición de Hemaglutinación , Vacunas contra la Influenza/administración & dosificación , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Linfocitos T Colaboradores-Inductores/fisiología , Vacunación/métodos
10.
J Cell Biochem ; 115(2): 391-6, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24123235

RESUMEN

Indoleamine 2,3-dioxygenase-1 (IDO1) catabolizes the essential amino acid tryptophan, acting as a modifier of inflammation and immune tolerance. Recent work has implicated IDO1 in many human diseases, including in cancer, chronic infection, autoimmune disorders, and neurodegenerative disease, stimulating a major surge in preclinical and clinical studies of its pathogenic functions. In the mouse, IDO1 is expressed widely but in situ detection of the enzyme in murine tissues has been unreliable due to the lack of specific antibodies that do not also react with tissues from animals that are genetically deficient in IDO1. Such probes are crucial to establish cellular mechanisms since IDO1 appears to act in different cell types depending on disease context, but reliable probes have been elusive in the field. In this report, we address this issue with the development of IDO1 monoclonal antibody 4B7 which specifically recognizes the murine enzyme in tissue sections, offering a reliable tool for immunohistology in preclinical disease models.


Asunto(s)
Tolerancia Inmunológica/genética , Indolamina-Pirrol 2,3,-Dioxigenasa/aislamiento & purificación , Inflamación/genética , Animales , Anticuerpos Monoclonales/inmunología , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Indolamina-Pirrol 2,3,-Dioxigenasa/inmunología , Inflamación/enzimología , Ratones , Distribución Tisular , Triptófano/metabolismo
11.
Cancer Immunol Immunother ; 63(7): 721-35, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24711084

RESUMEN

Genetic and pharmacological studies of indoleamine 2,3-dioxygenase (IDO) have established this tryptophan catabolic enzyme as a central driver of malignant development and progression. IDO acts in tumor, stromal and immune cells to support pathogenic inflammatory processes that engender immune tolerance to tumor antigens. The multifaceted effects of IDO activation in cancer include the suppression of T and NK cells, the generation and activation of T regulatory cells and myeloid-derived suppressor cells, and the promotion of tumor angiogenesis. Mechanistic investigations have defined the aryl hydrocarbon receptor, the master metabolic regulator mTORC1 and the stress kinase Gcn2 as key effector signaling elements for IDO, which also exerts a non-catalytic role in TGF-ß signaling. Small-molecule inhibitors of IDO exhibit anticancer activity and cooperate with immunotherapy, radiotherapy or chemotherapy to trigger rapid regression of aggressive tumors otherwise resistant to treatment. Notably, the dramatic antitumor activity of certain targeted therapeutics such as imatinib (Gleevec) in gastrointestinal stromal tumors has been traced in part to IDO downregulation. Further, antitumor responses to immune checkpoint inhibitors can be heightened safely by a clinical lead inhibitor of the IDO pathway that relieves IDO-mediated suppression of mTORC1 in T cells. In this personal perspective on IDO as a nodal mediator of pathogenic inflammation and immune escape in cancer, we provide a conceptual foundation for the clinical development of IDO inhibitors as a novel class of immunomodulators with broad application in the treatment of advanced human cancer.


Asunto(s)
Indolamina-Pirrol 2,3,-Dioxigenasa/inmunología , Inflamación/inmunología , Neoplasias/enzimología , Neoplasias/inmunología , Escape del Tumor , Animales , Humanos , Tolerancia Inmunológica , Inmunoterapia , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Inflamación/enzimología , Neoplasias/patología , Transducción de Señal
12.
BMC Pregnancy Childbirth ; 14: 270, 2014 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-25117856

RESUMEN

BACKGROUND: After the launch of Janani Suraksha Yojana, a conditional cash transfer scheme in India, the proportion of women giving birth in institutions has rapidly increased. However, there are important gaps in quality of childbirth services during institutional deliveries. The aim of this intervention was to improve the quality of childbirth services in selected high caseload public health facilities of 10 districts of Rajasthan. This intervention titled "Parijaat" was designed by Action Research & Training for Health, in partnership with the state government and United Nations Population Fund. METHODS: The intervention was carried out in 44 public health facilities in 10 districts of Rajasthan, India. These included district hospitals (9), community health centres (32) and primary health centres (3). The main intervention was orientation training of doctors and program managers and regular visits to facilities involving assessment, feedback, training and action. The adherence to evidence based practices before, during and after this intervention were measured using structured checklists and scoring sheets. Main outcome measures included changes in practices during labour, delivery or immediate postpartum period. RESULTS: Use of several unnecessary or harmful practices reduced significantly. Most importantly, proportion of facilities using routine augmentation of labour reduced (p = 0), episiotomy for primigravidas (p = 0.0003), fundal pressure (p = 0.0003), and routine suction of newborns (0 = 0.0005). Among the beneficial practices, use of oxytocin after delivery increased (p = 0.0001) and the practice of listening foetal heart sounds during labour (p = 0.0001). Some practices did not show any improvements, such as dorsal position for delivery, use of partograph, and hand-washing. CONCLUSIONS: An intervention based on repeated facility visits combined with actions at the level of decision makers can lead to substantial improvements in quality of childbirth practices at health facilities.


Asunto(s)
Centros Comunitarios de Salud , Parto Obstétrico/normas , Adhesión a Directriz , Hospitales de Distrito , Atención Perinatal/normas , Atención Primaria de Salud , Mejoramiento de la Calidad , Procedimientos Innecesarios/estadística & datos numéricos , Competencia Clínica , Educación Médica Continua , Medicina Basada en la Evidencia , Retroalimentación , Femenino , Humanos , India , Evaluación de Procesos y Resultados en Atención de Salud , Parto , Guías de Práctica Clínica como Asunto
13.
Infect Immun ; 81(11): 4171-81, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23980113

RESUMEN

Vaccination with a single dose of genetically attenuated malaria parasites can induce sterile protection against sporozoite challenge in the rodent Plasmodium yoelii model. Protection is dependent on CD8(+) T cells, involves perforin and gamma interferon (IFN-γ), and is correlated with the expansion of effector memory CD8(+) T cells in the liver. Here, we have further characterized vaccine-induced changes in the CD8(+) T cell phenotype and demonstrated significant upregulation of CD11c on CD3(+) CD8b(+) T cells in the liver, spleen, and peripheral blood. CD11c(+) CD8(+) T cells are predominantly CD11a(hi) CD44(hi) CD62L(-), indicative of antigen-experienced effector cells. Following in vitro restimulation with malaria-infected hepatocytes, CD11c(+) CD8(+) T cells expressed inflammatory cytokines and cytotoxicity markers, including IFN-γ, tumor necrosis factor alpha (TNF-α), interleukin-2 (IL-2), perforin, and CD107a. CD11c(-) CD8(+) T cells, on the other hand, expressed negligible amounts of all inflammatory cytokines and cytotoxicity markers tested, indicating that CD11c marks multifunctional effector CD8(+) T cells. Coculture of CD11c(+), but not CD11c(-), CD8(+) T cells with sporozoite-infected primary hepatocytes significantly inhibited liver-stage parasite development. Tetramer staining for the immunodominant circumsporozoite protein (CSP)-specific CD8(+) T cell epitope demonstrated that approximately two-thirds of CSP-specific cells expressed CD11c at the peak of the CD11c(+) CD8(+) T cell response, but CD11c expression was lost as the CD8(+) T cells entered the memory phase. Further analyses showed that CD11c(+) CD8(+) T cells are primarily KLRG1(+) CD127(-) terminal effectors, whereas all KLRG1(-) CD127(+) memory precursor effector cells are CD11c(-) CD8(+) T cells. Together, these results suggest that CD11c marks a subset of highly inflammatory, short-lived, antigen-specific effector cells, which may play an important role in eliminating infected hepatocytes.


Asunto(s)
Antígeno CD11c/biosíntesis , Linfocitos T CD8-positivos/inmunología , Vacunas contra la Malaria/inmunología , Plasmodium yoelii/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Sangre/inmunología , Linfocitos T CD8-positivos/química , Femenino , Inmunofenotipificación , Hígado/inmunología , Vacunas contra la Malaria/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Bazo/inmunología , Subgrupos de Linfocitos T/química , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología
14.
Front Pharmacol ; 14: 1183932, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37521457

RESUMEN

Longer lifespan produces risks of age-associated neurodegenerative disorders such as Alzheimer's disease (AD), which is characterized by declines in memory and cognitive function. The pathogenic causes of AD are thought to reflect a progressive aggregation in the brain of amyloid plaques composed of beta-amyloid (Aß) peptides and neurofibrillary tangles composed of phosphorylated tau protein. Recently, long-standing investigations of the Aß disease hypothesis gained support via a passive immunotherapy targeting soluble Aß protein. Tau-targeting approaches using antibodies are also being pursued as a therapeutic approach to AD. In genome-wide association studies, the disease modifier gene Bin1 has been identified as a top risk factor for late-onset AD in human populations, with recent studies suggesting that Bin1 binds tau and influences its extracellular deposition. Interestingly, before AD emerges in the brain, tau levels rise in the colon, where Bin1-a modifier of tissue barrier function and inflammation-acts to promote inflammatory bowel disease (IBD). This connection is provocative given clinical evidence of gut-brain communication in age-associated neurodegenerative disorders, including AD. In this review, we discuss a Bin1-targeting passive immunotherapy developed in our laboratory to treat IBD that may offer a strategy to indirectly reduce tau deposition and limit AD onset or progression.

15.
Cancer J ; 29(1): 34-37, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36693156

RESUMEN

ABSTRACT: Vaccine strategies for cancer differ from infectious disease in focusing mainly on clearing rather than preventing disease. Here we survey general vaccine strategies and combination therapy concepts being investigated for cancer treatment, with a focus on tumor antigens rather than cancer-inducing viruses or microorganisms. Many tumor antigens are "altered-self" and tend to arouse weaker immune responses than "foreign" antigens expressed by infectious agents. Further, unlike an infectious disease patient, a cancer patient's immune system is damaged, suppressed, or senescent and mainly tolerant of their disease. Thus, vaccine efficacy in a cancer patient will rely upon adjuvant or combination treatments that correct the inflammatory tumor microenvironment and degrade tumoral immunosuppression that dominates patient immunity. This brief overview is aimed at new researchers in cancer immunology seeking an overview of vaccine concepts to eradicate malignancy by provoking a selective immune attack.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias , Humanos , Vacunas contra el Cáncer/uso terapéutico , Neoplasias/terapia , Antígenos de Neoplasias , Microambiente Tumoral
16.
Sci Rep ; 13(1): 11802, 2023 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-37479833

RESUMEN

Ulcerative colitis (UC) is an idiopathic disease of the large intestine linked to high fat-high protein diets, a dysbiotic microbiome, and a metabolome linked to diet and/or aberrant circadian rhythms associated with poor sleeping patterns. Understanding diet-affected factors that negatively influence colonic health may offer new insights into how to prevent UC and enhance the efficacy of UC immunotherapy. In this preclinical study, we found that standard or high fiber diets in mice positively influenced their colonic health, whereas a high fat-high protein diet negatively influenced colonic health, consistent with clinical findings. Animals fed a high fat/high protein diet experienced obesity and a reduced colon length, illustrating a phenotype we suggest calling peinosis [hunger-like-condition; Greek, peina: hunger; osis: condition], as marked by a lack of nutrient energy remaining in fecal pellets. Notably, a high fat/high protein diet also led to signs of muscle weakness that could not be explained fully by weight gain. In contrast, mice on a high fiber diet ranked highest compared to other diets in terms of colon length and lack of muscle weakness. That said, mice on a high fiber diet were more prone to UC and toxic responses to immunotherapy, consistent with clinical observations. Recent studies have suggested that a standard diet may be needed to support the efficacy of immunotherapeutic drugs used to prevent and treat UC. Here we observed that protection against UC by Bin1 mAb, a passive UC immunotherapy that acts by coordinately enforcing intestinal barrier function, protecting enteric neurons, and normalizing the microbiome, was associated with increased colonic levels of healthful short-chain fatty acids (SCFA), particularly butyric acid and propionic acid, which help enforce intestinal barrier function. This work offers a preclinical platform to investigate how diet affects UC immunotherapy and the potential of dietary SCFA supplements to enhance it. Further, it suggests that the beneficial effects of passive immunotherapy by Bin1 mAb in UC treatment may be mediated to some extent by promoting increased levels of healthful SCFA.


Asunto(s)
Colitis Ulcerosa , Animales , Ratones , Colitis Ulcerosa/terapia , Inmunoterapia , Dieta Alta en Grasa/efectos adversos , Proteínas Adaptadoras Transductoras de Señales , Ácido Butírico , Proteínas del Tejido Nervioso , Proteínas Supresoras de Tumor
17.
PLoS Pathog ; 6(2): e1000757, 2010 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-20140199

RESUMEN

The innate immune response is essential for controlling West Nile virus (WNV) infection but how this response is propagated and regulates adaptive immunity in vivo are not defined. Herein, we show that IPS-1, the central adaptor protein to RIG-I-like receptor (RLR) signaling, is essential for triggering of innate immunity and for effective development and regulation of adaptive immunity against pathogenic WNV. IPS-1(-/-) mice exhibited increased susceptibility to WNV infection marked by enhanced viral replication and dissemination with early viral entry into the CNS. Infection of cultured bone-marrow (BM) derived dendritic cells (DCs), macrophages (Macs), and primary cortical neurons showed that the IPS-1-dependent RLR signaling was essential for triggering IFN defenses and controlling virus replication in these key target cells of infection. Intriguingly, infected IPS-1(-/-) mice displayed uncontrolled inflammation that included elevated systemic type I IFN, proinflammatory cytokine and chemokine responses, increased numbers of inflammatory DCs, enhanced humoral responses marked by complete loss of virus neutralization activity, and increased numbers of virus-specific CD8+ T cells and non-specific immune cell proliferation in the periphery and in the CNS. This uncontrolled inflammatory response was associated with a lack of regulatory T cell expansion that normally occurs during acute WNV infection. Thus, the enhanced inflammatory response in the absence of IPS-1 was coupled with a failure to protect against WNV infection. Our data define an innate/adaptive immune interface mediated through IPS-1-dependent RLR signaling that regulates the quantity, quality, and balance of the immune response to WNV infection.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Transducción de Señal/inmunología , Fiebre del Nilo Occidental/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Anticuerpos Antivirales/sangre , Western Blotting , Encéfalo/inmunología , Encéfalo/virología , Separación Celular , Citocinas/sangre , Citocinas/inmunología , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/inmunología , ARN Helicasas DEAD-box/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Expresión Génica , Inmunidad Celular/inmunología , Inflamación/inmunología , Ratones , Ratones Noqueados , Neuronas/inmunología , Neuronas/virología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Fiebre del Nilo Occidental/genética , Fiebre del Nilo Occidental/metabolismo , Virus del Nilo Occidental/inmunología
18.
Indian J Exp Biol ; 50(11): 765-70, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23305026

RESUMEN

Nervous system lesions are characterized by the loss of neuronal numbers and types. The neurotrophic factor levels in an injured tissue reflect their potential for regeneration. This hypothesis was investigated in olfactory bulb (OB), where olfactory tract was surgically transected disrupting neuronal migration and turnover. The effects were followed with quantification of mitral cells and three neurotrophic factors mRNA levels for 6 weeks. The neuronal numbers decreased by 3rd- and 4th-week in transected OBs followed by their restoration, comparable with that of controls at 5th- and 6th-week. The endogenous levels of three neurotrophic factors - (brain derived neurotrophic factor, insulin growth factor-1 and fibroblast growth factor-2) using qPCR showed increase at 2nd-week by 136-, 8- and 2-fold respectively. Also, there was a significant increase in specific neurotrophic factors at 5th-week and 6th-weeks. The results propose a temporal link between deployment of neurotrophic factors and the plausible restorative events for mitral cell numbers in OB.


Asunto(s)
Degeneración Nerviosa/patología , Factores de Crecimiento Nervioso/biosíntesis , Plasticidad Neuronal , Neuronas/patología , Bulbo Olfatorio/patología , Animales , Animales Recién Nacidos , Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Recuento de Células , Factor 2 de Crecimiento de Fibroblastos/biosíntesis , Factor I del Crecimiento Similar a la Insulina/biosíntesis , Degeneración Nerviosa/metabolismo , Neuronas/metabolismo , Bulbo Olfatorio/crecimiento & desarrollo , Bulbo Olfatorio/metabolismo , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Tiempo , Regulación hacia Arriba
19.
Methods Mol Biol ; 2410: 93-108, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34914043

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the disease COVID-19 that has decimated the health and economy of our planet. The virus causes the disease not only in people but also in companion and wild animals. As yet we do not know why the virus is highly successful in causing the pandemic within 3 months of its first report. Lack of a voice on how to handle the pandemic impacted the management of the disease globally. Publication of the importance of masks and social distancing in preprint servers reduced the spread of the disease and deaths associated with it. Very few countries have invested in science and research and development and that has impacted the development of therapies for the pandemic. Though vaccination against COVID-19 started in December 2020, slower rate of immunizations has resulted in rapid spread of the mutant strains of SARS-CoV-2. Lack of transparency and accountability coupled with anergic leadership was responsible for the high incidence of disease and death associated with the COVID-19 pandemic.


Asunto(s)
Vacunas contra la COVID-19 , COVID-19 , Pandemias , Animales , COVID-19/epidemiología , Humanos , Máscaras , Pandemias/prevención & control , Distanciamiento Físico , SARS-CoV-2
20.
Methods Mol Biol ; 2410: 265-272, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34914051

RESUMEN

COVID-19 caused by SARS-CoV-2, an RNA coronavirus has impacted the health and economy of all the countries. The virus has wide host adaptability and causes severe diseases in humans and animals. The major structural proteins of SARS-CoV-2 include spike (S), envelop (E), membrane (M), and nucleocapsid (N). The current vaccines are based on the S protein. The emergence of variants of SARS-CoV-2 has renewed interest in the use of additional structural proteins for the development of diagnostics and vaccines. Knowledge of B cell epitopes and MHC-I binding regions of the structural proteins of SARS-CoV-2 is essential in the development of effective diagnostics and therapies. This chapter provides information on the epitopes of the structural proteins of SARS-CoV-2.


Asunto(s)
Proteínas de la Envoltura de Coronavirus/inmunología , Epítopos de Linfocito B , Epítopos de Linfocito T , Glicoproteína de la Espiga del Coronavirus/inmunología , Proteínas de la Matriz Viral/inmunología , Animales , COVID-19 , Vacunas contra la COVID-19 , Humanos , SARS-CoV-2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA