Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Invest Dermatol ; 142(8): 2159-2172.e9, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35090950

RESUMEN

TNF-α, a proinflammatory cytokine, is a crucial mediator of psoriasis pathogenesis. TNF-α functions by activating TNFR1 and TNFR2. Anti-TNF drugs that neutralize TNF-α, thus blocking the activation of TNFR1 and TNFR2, have been proven highly therapeutic in psoriatic diseases. TNF-α also plays an important role in host defense; thus, anti-TNF therapy can cause potentially serious adverse effects, including opportunistic infections and latent tuberculosis reactivation. These adverse effects are attributed to TNFR1 inactivation. Therefore, understanding the relative contributions of TNFR1 and TNFR2 has clinical implications in mitigating psoriasis versus global TNF-α blockade. We found a significant reduction in psoriasis lesions as measured by epidermal hyperplasia, characteristic gross skin lesion, and IL-23 or IL-17A levels in Tnfr2-knockout but not in Tnfr1-knockout mice in the imiquimod psoriasis model. Furthermore, imiquimod-mediated increase in the myeloid dendritic cells, TNF/inducible nitric oxide synthase‒producing dendritic cells, and IL-23 expression in the draining lymph nodes were dependent on TNFR2 but not on TNFR1. Together, our results support that psoriatic inflammation is not dependent on TNFR1 activity but is driven by a TNFR2-dependent IL-23/IL-17 pathway activation. Thus, targeting the TNFR2 pathway may emerge as a potential next-generation therapeutic approach for psoriatic diseases.


Asunto(s)
Psoriasis , Receptores Tipo II del Factor de Necrosis Tumoral , Animales , Células Dendríticas/metabolismo , Imiquimod , Inflamación/patología , Interleucina-17 , Interleucina-23 , Ganglios Linfáticos/patología , Ratones , Ratones Endogámicos C57BL , Psoriasis/tratamiento farmacológico , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Inhibidores del Factor de Necrosis Tumoral , Factor de Necrosis Tumoral alfa/metabolismo
2.
Hum Mol Genet ; 30(22): 2135-2148, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34218272

RESUMEN

Germline mutation of PTEN is causally observed in Cowden syndrome (CS) and is one of the most common, penetrant risk genes for autism spectrum disorder (ASD). However, the majority of individuals who present with CS-like clinical features are PTEN-mutation negative. Reassessment of PTEN promoter regulation may help explain abnormal PTEN dosage, as only the minimal promoter and coding regions are currently included in diagnostic PTEN mutation analysis. Therefore, we reanalyzed the architecture of the PTEN promoter using next-generation sequencing datasets. Specifically, run-on sequencing assays identified two additional transcription start regions (TSRs) at -2053 and -1906 basepairs from the canonical start of PTEN, thus extending the PTEN 5'UTR and redefining the PTEN promoter. We show that these novel upstream TSRs are active in cancer cell lines, human cancer and normal tissue. Furthermore, these TSRs can produce novel PTEN transcripts due to the introduction of new splice donors at -2041, -1826 and -1355, which may allow for splicing out of the PTEN 5'UTR or the first and second exon in upstream-initiated transcripts. Combining ENCODE ChIP-seq and pertinent literature, we also compile and analyze all transcription factors (TFs) binding at the redefined PTEN locus. Enrichment analyses suggest that TFs bind specifically to the upstream TSRs may be implicated in inflammatory processes. Altogether, these data redefine the architecture of the PTEN promoter, an important step toward a comprehensive model of PTEN transcription regulation, a basis for future investigations into the new promoters' role in disease pathogenesis.


Asunto(s)
Fosfohidrolasa PTEN/genética , Regiones Promotoras Genéticas , Transcripción Genética , Biología Computacional/métodos , Regulación de la Expresión Génica , Ontología de Genes , Humanos , Sitios de Carácter Cuantitativo , Sitio de Iniciación de la Transcripción
3.
Transl Psychiatry ; 11(1): 348, 2021 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-34091589

RESUMEN

Genetic studies in ASD have mostly focused on the proband, with no clear understanding of parental genetic contributions to fetal neurodevelopment. Among parental etiological factors, perinatal maternal inflammation secondary to autoimmunity, infections, and toxins is associated with ASD. However, the inherent impact of maternal genetics on in-utero inflammation and fetal neurodevelopment in the absence of strong external inflammatory exposures is not known. We used the PtenWT/m3m4 mouse model for ASD to demonstrate the impact of maternal genetics on the penetrance of ASD-like phenotypes in the offspring. PtenWT/m3m4 (Momm3m4) or PtenWT/WT (MomWT) females, their offspring, and placental interface were analyzed for inflammatory markers, gene expression, and cellular phenotypes at E17.5. Postnatal behavior was tested by comparing pups from Momm3m4 vs. MomWT. Mothers of the PtenWT/m3m4 genotype (Momm3m4) showed inadequate induction of IL-10 mediated immunosuppression during pregnancy. Low IL-10 in the mother was directly correlated with decreased complement expression in the fetal liver. Fetuses from Momm3m4 had increased breakdown of the blood-brain-barrier, neuronal loss, and lack of glial cell maturation during in-utero stages. This impact of maternal genotype translated to a postnatal increase in the risk of newborn mortality, visible macrocephaly and ASD-like repetitive and social behaviors. Depending on maternal genotype, non-predisposed (wildtype) offspring showed ASD-like phenotypes, and phenotypic penetrance was decreased in predisposed pups from MomWT. Our study introduces the concept that maternal genetics alone, without any added external inflammatory insults, can modulate fetal neurodevelopment and ASD-related phenotypes in the offspring via alteration of IL-10 mediated materno-fetal immunosuppression.


Asunto(s)
Trastorno del Espectro Autista , Tolerancia Inmunológica , Animales , Femenino , Humanos , Recién Nacido , Ratones , Embarazo , Trastorno del Espectro Autista/genética , Feto , Fenotipo , Placenta
4.
Mol Autism ; 12(1): 5, 2021 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-33509259

RESUMEN

BACKGROUND: PTEN is a well-established risk gene for autism spectrum disorder (ASD). Yet, little is known about how PTEN mutations and associated molecular processes influence neurobehavioral function in mutation carriers with (PTEN-ASD) and without ASD (PTEN no-ASD). The primary aim of the present study was to examine group differences in peripheral blood-derived PTEN pathway protein levels between PTEN-ASD, PTEN no-ASD, and idiopathic macrocephalic ASD patients (macro-ASD). Secondarily, associations between protein levels and neurobehavioral functions were examined in the full cohort. METHODS: Patients were recruited at four tertiary medical centers. Peripheral blood-derived protein levels from canonical PTEN pathways (PI3K/AKT and MAPK/ERK) were analyzed using Western blot analyses blinded to genotype and ASD status. Neurobehavioral measures included standardized assessments of global cognitive ability and multiple neurobehavioral domains. Analysis of variance models examined group differences in demographic, neurobehavioral, and protein measures. Bivariate correlations, structural models, and statistical learning procedures estimated associations between molecular and neurobehavioral variables. To complement patient data, Western blots for downstream proteins were generated to evaluate canonical PTEN pathways in the PTEN-m3m4 mouse model. RESULTS: Participants included 61 patients (25 PTEN-ASD, 16 PTEN no-ASD, and 20 macro-ASD). Decreased PTEN and S6 were observed in both PTEN mutation groups. Reductions in MnSOD and increases in P-S6 were observed in ASD groups. Elevated neural P-AKT/AKT and P-S6/S6 from PTEN murine models parallel our patient observations. Patient PTEN and AKT levels were independently associated with global cognitive ability, and p27 expression was associated with frontal sub-cortical functions. As a group, molecular measures added significant predictive value to several neurobehavioral domains over and above PTEN mutation status. LIMITATIONS: Sample sizes were small, precluding within-group analyses. Protein and neurobehavioral data were limited to a single evaluation. A small number of patients were excluded with invalid protein data, and cognitively impaired patients had missing data on some assessments. CONCLUSIONS: Several canonical PTEN pathway molecules appear to influence the presence of ASD and modify neurobehavioral function in PTEN mutation patients. Protein assays of the PTEN pathway may be useful for predicting neurobehavioral outcomes in PTEN patients. Future longitudinal analyses are needed to replicate these findings and evaluate within-group relationships between protein and neurobehavioral measures. TRIAL REGISTRATION: ClinicalTrials.gov Identifier NCT02461446.


Asunto(s)
Trastorno del Espectro Autista/diagnóstico , Trastorno del Espectro Autista/etiología , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal , Heterocigoto , Fosfohidrolasa PTEN/genética , Adolescente , Alelos , Animales , Trastorno del Espectro Autista/psicología , Biomarcadores , Niño , Preescolar , Modelos Animales de Enfermedad , Femenino , Genotipo , Humanos , Masculino , Pruebas Neuropsicológicas , Fosfohidrolasa PTEN/metabolismo , Fenotipo , Transducción de Señal , Adulto Joven
5.
Mol Psychiatry ; 26(5): 1458-1471, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-32055008

RESUMEN

Germline mutations in PTEN account for ~10% of cases of autism spectrum disorder (ASD) with coincident macrocephaly. To explore the importance of nuclear PTEN in the development of ASD and macrocephaly, we previously generated a mouse model with predominantly cytoplasmic localization of Pten (Ptenm3m4/m3m4).Cytoplasmic predominant Pten localization results in a phenotype of extreme macrocephaly and autistic-like traits. Transcriptomic analysis of the Ptenm3m4/m3m4 cortex found upregulated gene pathways related to myeloid cell activation, myeloid cell migration, and phagocytosis. These transcriptomic findings were used to direct in vitro assays on Pten wild-type and Ptenm3m4/m3m4 microglia. We found increased Iba1 and C1q expression with enhanced phagocytic capacity in Ptenm3m4/m3m4 microglia, indicating microglial activation. Moreover, through a series of neuron-microglia co-culture experiments, we found Ptenm3m4/m3m4 microglia are more efficient at synaptic pruning compared with wild-type controls. In addition, we found evidence for neuron-microglia cross-talk, where Ptenm3m4/m3m4 neurons elicit enhanced pruning from innately activated microglia. Subsequent in vivo studies validated our in vitro findings. We observed a concurrent decline in the expression of Pten and synaptic markers in the Ptenm3m4/m3m4 cortex. At ~3 weeks of age, with a 50% drop in Pten expression compared with wild-type levels, we observed enhanced activation of microglia in the Ptenm3m4/m3m4 brain. Collectively, our data provide evidence that dysregulated Pten in microglia has an etiological role in microglial activation, phagocytosis, and synaptic pruning, creating avenues for future studies on the importance of PTEN in maintaining microglia homeostasis.


Asunto(s)
Trastorno del Espectro Autista , Trastorno Autístico , Fosfohidrolasa PTEN/genética , Animales , Trastorno del Espectro Autista/genética , Trastorno Autístico/genética , Modelos Animales de Enfermedad , Ratones , Microglía , Plasticidad Neuronal , Fenotipo
6.
Hum Mol Genet ; 29(14): 2353-2364, 2020 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-32588888

RESUMEN

Individuals with germline mutations in the gene encoding phosphatase and tensin homolog on chromosome ten (PTEN) are diagnosed with PTEN hamartoma tumor syndrome (PHTS) and are at high risk for developing breast, thyroid and other cancers and/or autoimmunity or neurodevelopmental issues including autism spectrum disorders. Although well recognized as a tumor suppressor, involvement of PTEN mutations in mediating such a diverse range of phenotypes indicates a more central involvement for PTEN in immunity than previously recognized. To address this, sequencing of the T-cell receptor variable-region ß-chain was performed on peripheral blood from PHTS patients. Based on patient findings, we performed mechanistic studies in two Pten knock-in murine models, distinct from each other in cell compartment-specific predominance of Pten. We found that PTEN mutations in humans and mice are associated with a skewed T- and B-cell gene repertoire, characterized by increased prevalence of high-frequency clones. Immunological characterization showed that Pten mutants have increased B-cell proliferation and a proclivity towards increased T-cell reactivity upon Toll-like-receptor stimulation. Furthermore, decreases in nuclear but not cytoplasmic Pten levels associated with a reduction in expression of the autoimmune regulator (Aire), a critical mediator of central immune tolerance. Mechanistically, we show that nuclear PTEN most likely regulates Aire expression via its emerging role in splicing regulation. We conclude that germline disruption of PTEN, both in human and mouse, results in compromised central immune tolerance processes that may significantly impact individual stress responses and therefore predisposition to autoimmunity and cancer.


Asunto(s)
Síndrome de Hamartoma Múltiple/genética , Fosfohidrolasa PTEN/genética , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Factores de Transcripción/genética , Animales , Linfocitos B/inmunología , Linfocitos B/patología , Proliferación Celular/genética , Modelos Animales de Enfermedad , Femenino , Técnicas de Sustitución del Gen , Mutación de Línea Germinal/genética , Síndrome de Hamartoma Múltiple/sangre , Síndrome de Hamartoma Múltiple/inmunología , Síndrome de Hamartoma Múltiple/patología , Humanos , Tolerancia Inmunológica/genética , Masculino , Ratones , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Linfocitos T/inmunología , Linfocitos T/patología , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología , Proteína AIRE
7.
Mol Autism ; 11(1): 43, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32487265

RESUMEN

BACKGROUND: PTEN, a syndromic autism spectrum disorder (ASD) risk gene, is mutated in approximately 10% of macrocephalic ASD cases. Despite the described genetic association between PTEN and ASD and ensuing studies, we continue to have a limited understanding of how PTEN disruption drives ASD pathogenesis and maintenance. METHODS: We derived neural stem cells (NSCs) from the dentate gyrus (DG) of Ptenm3m4 mice, a model that recapitulates PTEN-ASD phenotypes. We subsequently characterized the expression of stemness factors, proliferation, and differentiation of neurons and glia in Ptenm3m4 NSCs using immunofluorescent and immunoblotting approaches. We also measured Creb phosphorylation by Western blot analysis and expression of Creb-regulated genes with qRT-PCR. RESULTS: The m3m4 mutation decreases Pten localization to the nucleus and its global expression over time. Ptenm3m4 NSCs exhibit persistent stemness characteristics associated with increased proliferation and a resistance to neuronal maturation during differentiation. Given the increased proliferation of Ptenm3m4 NSCs, a significant increase in the population of immature neurons relative to mature neurons occurs, an approximately tenfold decrease in the ratio between the homozygous mutant and wildtype. There is an opposite pattern of differentiation in some Ptenm3m4 glia, specifically an increase in astrocytes. These aberrant differentiation patterns associate with changes in Creb activation in Ptenm3m4/m3m4 NSCs. We specifically observed loss of Creb phosphorylation at S133 in Ptenm3m4/m3m4 NSCs and a subsequent decrease in expression of Creb-regulated genes important to neuronal function (i.e., Bdnf). Interestingly, Bdnf treatment is able to partially rescue the stunted neuronal maturation phenotype in Ptenm3m4/m3m4 NSCs. CONCLUSIONS: Constitutional disruption of Pten nuclear localization with subsequent global decrease in Pten expression generates abnormal patterns of differentiation, a stunting of neuronal maturation. The propensity of Pten disruption to restrain neurons to a more progenitor-like state may be an important feature contributing to PTEN-ASD pathogenesis.


Asunto(s)
Diferenciación Celular/genética , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis/genética , Neuronas/citología , Neuronas/metabolismo , Fosfohidrolasa PTEN/genética , Animales , Trastorno del Espectro Autista/etiología , Trastorno del Espectro Autista/metabolismo , Biomarcadores , Núcleo Celular , Proliferación Celular , Autorrenovación de las Células , Células Cultivadas , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados , Mutación , Neuroglía/citología , Neuroglía/metabolismo , Fosfohidrolasa PTEN/metabolismo
8.
J Biomol Struct Dyn ; 37(7): 1766-1782, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29663862

RESUMEN

Individuals with germline mutations in the tumor suppressor gene phosphatase and tensin homolog (PTEN), irrespective of clinical presentation, are diagnosed with PTEN hamartoma tumor syndrome (PHTS). PHTS confers a high risk of breast, thyroid, and other cancers or autism spectrum disorder (ASD) with macrocephaly. It remains unclear why mutations in one gene can lead to seemingly disparate phenotypes. Thus, we sought to identify differences in ASD vs. cancer-associated germline PTEN missense mutations by investigating putative structural effects induced by each mutation. We utilized a theoretical computational approach combining in silico structural analysis and molecular dynamics (MD) to interrogate 17 selected mutations from our patient population: six mutations were observed in patients with ASD (only), six mutations in patients with PHTS-associated cancer (only), four mutations shared across both phenotypes, and one mutation with both ASD and cancer. We demonstrate structural stability changes where all six cancer-associated mutations showed a global decrease in structural stability and increased dynamics across the domain interface with a proclivity to unfold, mediating a closed (inactive) active site. In contrast, five of the six ASD-associated mutations showed localized destabilization that contribute to the partial opening of the active site. Our results lend insight into distinctive structural effects of germline PTEN mutations associated with PTEN-ASD vs. those associated with PTEN-cancer, potentially aiding in identification of the shared and separate molecular features that contribute to autism or cancer, thus, providing a deeper understanding of genotype-phenotype relationships for germline PTEN mutations.


Asunto(s)
Trastorno del Espectro Autista/genética , Mutación de Línea Germinal , Simulación de Dinámica Molecular , Neoplasias/genética , Fosfohidrolasa PTEN/química , Fosfohidrolasa PTEN/genética , Conformación Proteica , Alelos , Sustitución de Aminoácidos , Sitios de Unión , Dominio Catalítico , Predisposición Genética a la Enfermedad , Humanos , Mutación Missense , Unión Proteica , Estabilidad Proteica , Relación Estructura-Actividad
9.
Oncotarget ; 8(20): 32536-32549, 2017 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-28430646

RESUMEN

Immunotherapy has historically been successful in highly antigenic tumors but has shown limited therapeutic efficacy in non-antigenic tumors such as breast cancers. Our previous studies in autoimmunity have demonstrated that increased antigen load within a tissue enhances immune reactivity against it. We therefore hypothesized that enhancing expression of target proteins on breast tumors can increase efficacy of targeted immunotherapy. We hypothesized that antagonism of the estrogen receptor (ER) can increase expression of targets that are hormonally regulated and facilitate enhanced tumor recognition by targeted immunotherapy. We used a lactation protein α-Lactalbumin, a known immunotherapeutic target on breast tumors, as our model target antigen. Enhancement of target protein expression in human and murine breast tumors was tested in vitro and in vivo by ER antagonism using clinically established ER modulators, Tamoxifen and Fulvestrant. We show that antagonism of the ER can induce a 2-3 fold increase in expression of target proteins on tumors leaving the normal breast tissue unaffected. Tumor progression studies in 4T1 tumor-bearing mice show that efficacy of adoptively transferred cell based targeted immunotherapy was enhanced by target antigen amplification resulting in significantly higher tumor inhibition. However, in spite of increased target expression, anti-tumor efficacy of direct immunization was not enhanced probably due to other limiting factors involved in the immune priming process. Our study provides a novel combinatorial clinical strategy for enhancing efficacy of immunotherapy not only on breast tumors but potentially also for other hormonally driven tumors such as those of the prostate, testis and ovary.


Asunto(s)
Receptores de Estrógenos/antagonistas & inhibidores , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inmunoterapia/métodos , Ratones , Ratones Endogámicos BALB C
10.
Hum Mol Genet ; 26(7): 1365-1375, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28164237

RESUMEN

Thyroid cancer is a major component cancer of Cowden syndrome (CS), a disorder typically associated with germline mutations in PTEN. Germline variants in succinate dehydrogenase genes (SDHx) co-occurring with PTEN germline mutations confer a 2-fold increased prevalence (OR 2.7) of thyroid cancer compared to PTEN-associated CS but 50% decreased prevalence (OR 0.54) of thyroid cancer compared to SDHx-associated CS. We have previously shown that CS-associated SDHD variants G12S and H50R induce PTEN oxidation and nuclear accumulation in thyroid cancer. Our current study shows that SDHD-G12S and -H50R variants cause down-regulation of autophagy, demonstrating a role for SDHD in autophagy-associated pathogenesis of differentiated thyroid cancer. These findings could explain the increased prevalence of thyroid cancer in CS patients with SDHx germline mutations compared to those with PTEN mutations alone. Importantly, we demonstrate the dependence of this process on functional wild-type PTEN with reversal of decreased autophagy after PTEN knockdown. The latter could explain the clinically observed decrease in thyroid cancer prevalence in patients with co-existent PTEN mutations and SDHx variants. We also show that SDHD-G12S/H50R promotes mono-ubiquitination of PTEN, causing its translocation into the nucleus, upregulation of AKT and consequent phosphorylation of FOXO3a. Furthermore, SDHD-G12S/H50R-mediated increase in acetylation of FOXO3a further enhances AKT-associated phosphorylation of FOXO3a. This combination of phosphorylation and acetylation of FOXO3a results in its nuclear export for degradation and consequent down-regulation of FOXO3a-target autophagy-related gene (ATG) expression. Overall, our study reveals a novel mechanism of crosstalk amongst SDHD, PTEN and autophagy pathways and their potential roles in thyroid carcinogenesis.


Asunto(s)
Carcinogénesis/genética , Síndrome de Hamartoma Múltiple/genética , Fosfohidrolasa PTEN/genética , Succinato Deshidrogenasa/genética , Neoplasias de la Tiroides/genética , Autofagia/genética , Homólogo de la Proteína 1 Relacionada con la Autofagia/genética , Línea Celular Tumoral , Proteína Forkhead Box O3/biosíntesis , Proteína Forkhead Box O3/genética , Regulación Neoplásica de la Expresión Génica , Mutación de Línea Germinal/genética , Síndrome de Hamartoma Múltiple/patología , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Proteína Oncogénica v-akt/genética , Transducción de Señal/genética , Neoplasias de la Tiroides/patología
11.
Cancers (Basel) ; 8(6)2016 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-27322324

RESUMEN

We have proposed that safe and effective protection against the development of adult onset cancers may be achieved by vaccination against tissue-specific self-proteins that are "retired" from expression at immunogenic levels in normal tissues as we age, but are overexpressed in emerging tumors. α-Lactalbumin is an example of a "retired" self-protein because its expression in normal tissues is confined exclusively to the breast during late pregnancy and lactation, but is also expressed in the vast majority of human triple negative breast cancers (TNBC)-the most aggressive and lethal form of breast cancer and the predominant form that occurs in women at high genetic risk including those with mutated BRCA1 genes. In anticipation of upcoming clinical trials, here we provide preclinical data indicating that α-lactalbumin has the potential as a vaccine target for inducing safe and effective primary immunoprevention as well as immunotherapy against TNBC.

12.
J Immunol Res ; 2015: 630287, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26618181

RESUMEN

Anti-Müllerian hormone receptor, type II (AMHR2), is a differentiation protein expressed in 90% of primary epithelial ovarian carcinomas (EOCs), the most deadly gynecologic malignancy. We propose that AMHR2 may serve as a useful target for vaccination against EOC. To this end, we generated the recombinant 399-amino acid cytoplasmic domain of mouse AMHR2 (AMHR2-CD) and tested its efficacy as a vaccine target in inhibiting growth of the ID8 transplantable EOC cell line in C57BL/6 mice and in preventing growth of autochthonous EOCs that occur spontaneously in transgenic mice. We found that AMHR2-CD immunization of C57BL/6 females induced a prominent antigen-specific proinflammatory CD4+ T cell response that resulted in a mild transient autoimmune oophoritis that resolved rapidly with no detectable lingering adverse effects on ovarian function. AMHR2-CD vaccination significantly inhibited ID8 tumor growth when administered either prophylactically or therapeutically, and protection against EOC growth was passively transferred into naive recipients with AMHR2-CD-primed CD4+ T cells but not with primed B cells. In addition, prophylactic AMHR2-CD vaccination of TgMISIIR-TAg transgenic mice significantly inhibited growth of autochthonous EOCs and provided a 41.7% increase in mean overall survival. We conclude that AMHR2-CD vaccination provides effective immunotherapy of EOC with relatively benign autoimmune complications.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Vacunas contra el Cáncer/administración & dosificación , Carcinoma/inmunología , Ooforitis/prevención & control , Neoplasias Ováricas/inmunología , Receptores de Péptidos/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteínas Recombinantes/administración & dosificación , Traslado Adoptivo , Animales , Linfocitos T CD4-Positivos/trasplante , Vacunas contra el Cáncer/efectos adversos , Carcinoma/genética , Carcinoma/prevención & control , Procesos de Crecimiento Celular , Línea Celular Tumoral , Células Cultivadas , Células Epiteliales/fisiología , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Terapia Molecular Dirigida , Trasplante de Neoplasias , Ooforitis/etiología , Neoplasias Ováricas/genética , Neoplasias Ováricas/prevención & control , Ingeniería de Proteínas , Estructura Terciaria de Proteína/genética , Receptores de Péptidos/genética , Receptores de Factores de Crecimiento Transformadores beta/genética
13.
J Neuroimmunol ; 279: 71-4, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25670003

RESUMEN

Multiple sclerosis (MS) is widely viewed as a prototypic human autoimmune disease involving proinflammatory T cells that induce lesions in the central nervous system (CNS) in response to myelin self proteins. Although the impact of sex hormones on MS is well recognized, the converse effects of autoimmunity on sex hormones are still unclear. The current study was designed to assess the impact of CNS autoimmunity on female reproductive physiology. In order to identify subtle hormonal disturbances as a result of autoimmunity, we analyzed the estrous cycle in SJL/J mice after active induction of experimental autoimmune encephalomyelitis (EAE), an animal model with substantial similarities to MS. Here we show that CNS autoimmunity significantly shortens the murine estrous cycle. This shortening of the estrous cycle is characterized by a dramatic decrease in the length of the metestrus-diestrus luteal phase partially offset by a highly significant but less dramatic elongation of the proestrus-estrus follicular phase of the uterine cycle. Thus, our study provides experimental evidence for a direct causal link between CNS autoimmunity and disruption of the homeostatic balance of the uterine cycle often observed in women with MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/fisiopatología , Ciclo Estral/fisiología , Homeostasis/fisiología , Animales , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Ciclo Estral/efectos de los fármacos , Ciclo Estral/inmunología , Femenino , Homeostasis/efectos de los fármacos , Homeostasis/inmunología , Humanos , Ratones , Ratones Endogámicos C57BL , Proteína Proteolipídica de la Mielina/toxicidad , Fragmentos de Péptidos/toxicidad
14.
Exp Eye Res ; 121: 35-40, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24582892

RESUMEN

Myofibroblasts, the primary cells associated with corneal stromal haze (opacity), can be derived from both cornea-derived and bone marrow-derived precursor cells. In the present study, the role of TGFß or PDGF blockage on bone marrow-derived myofibroblast development was investigated using a green fluorescent protein (GFP) chimeric bone marrow mouse model and plasmid vectors that blocked TGFß or PDGF signaling. At the peak of corneal haze one month after irregular phototherapeutic keratectomy the central stroma had significantly less alpha-smooth muscle actin (α-SMA)-positive cells derived from GFP+ bone marrow-derived cells or GFP- keratocyte/corneal fibroblast-derived cells when corneas were treated with the TGFß blocking vector pGFPC1.TGFRBKDEL or the PDGF blocking vector pCMV.PDGFRB.23KDEL compared with the corresponding empty vector treated or untreated control groups. In individual animals, 30-60% of myofibroblasts were derived from bone marrow-derived precursor cells and 40-70% of myofibroblasts were derived from keratocyte-derived precursor cells. TGFß and PDGF regulate corneal myofibroblast development from bone marrow-derived precursor cells and keratocyte/corneal fibroblast-derived precursor cells.


Asunto(s)
Células de la Médula Ósea/citología , Queratocitos de la Córnea/citología , Miofibroblastos/citología , Proteínas Proto-Oncogénicas c-sis/fisiología , Transducción de Señal/efectos de los fármacos , Células Madre/citología , Factor de Crecimiento Transformador beta/fisiología , Actinas/metabolismo , Animales , Células de la Médula Ósea/metabolismo , Queratocitos de la Córnea/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Vectores Genéticos/farmacología , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Ratones Endogámicos C57BL , Miofibroblastos/metabolismo , Queratectomía Fotorrefractiva , Plásmidos , Proteínas Proto-Oncogénicas c-sis/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/antagonistas & inhibidores
15.
Int J Cancer ; 134(7): 1695-705, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24105638

RESUMEN

Sunitinib, a protein tyrosine kinase inhibitor is the frontline therapy for renal and gastrointestinal cancers. We hypothesized that by virtue of its well documented tumor apoptosis and immune adjuvant properties, combination of Sunitinib with anti-tumor immunotherapeutics will provide synergistic inhibition of tumor growth. Our study was designed to evaluate the impact of Sunitinib on immunotherapy mediated anti-tumor immune responses and evaluate its efficacy as a combinatorial therapy with tumor targeted immunotherapeutic vaccination. Mice immunized with recombinant α-lactalbumin, a lactation protein expressed on majority of breast tumors were treated with 1 mg of Sunitinib for seven consecutive days beginning (1) concurrently, on the day of α-lactalbumin immunization or (2) sequentially, on day 9 after immunization. Ten-day lymph nodes or 21 day spleens were tested by ELISPOT assays and flow cytometry to evaluate responsiveness to α-lactalbumin immunization in presence of Sunitinib and distribution of cells involved in T cell antigen priming and proliferation in different lymphoid compartments. In addition, therapeutic efficacy of the α-lactalbumin/ Sunitinib combination was evaluated by monitoring tumor growth in the 4T1 transplanted tumor model. Our studies reveal that concurrent administration of Sunitinib with active vaccination against a targeted tumor antigen inhibits priming to the immunogen due to a drastic decrease in CD11b+CD11c+ antigen presenting cells, leading to failure of vaccination. However, sequential delivery of Sunitinib timed to avoid the priming phase of vaccination results in the desired vaccination mediated boost in immune responses.


Asunto(s)
Vacunas contra el Cáncer/farmacología , Indoles/farmacología , Lactalbúmina/inmunología , Pirroles/farmacología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Adyuvantes Inmunológicos/farmacología , Animales , Células Presentadoras de Antígenos/efectos de los fármacos , Células Presentadoras de Antígenos/inmunología , Vacunas contra el Cáncer/inmunología , Proliferación Celular/efectos de los fármacos , Progresión de la Enfermedad , Femenino , Inmunoterapia/métodos , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos BALB C , Bazo/efectos de los fármacos , Bazo/inmunología , Sunitinib , Vacunación/métodos
16.
Exp Eye Res ; 116: 366-70, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24140502

RESUMEN

GFP-chimeric mice are important tools to study the role of bone marrow-derived cells in eye physiology. A method is described to generate GFP-chimeric mice using whole-body, sub-lethal radiation (600 rad) of wild-type C57BL/6 recipients followed by tail vein injection of bone marrow cells derived from GFP+ (GFP-transgenic C57/BL/6-Tg(UBC-GFP)30 Scha/J) mice. This method yields stable GFP+ chimeras with greater than 95% chimerism (range 95-99%), achieved within one month of bone marrow transfer confirmed by microscopy and fluorescence-assisted cell sorting (FACS) analysis, with lower mortality after irradiation than prior methods. To demonstrate the efficacy of GFP+ bone marrow chimeric mice, the role of circulating GFP+ bone marrow-derived cells in myofibroblast generation after irregular photo-therapeutic keratectomy (PTK) was analyzed. Many SMA+ myofibroblasts that were generated at one month after PTK were derived from GFP+ bone marrow-derived cells. The GFP+ bone marrow chimeric mouse provides an excellent model for studying the role of bone marrow-derived cells in corneal wound healing, glaucoma surgery, optic nerve head pathology and retinal pathophysiology and wound healing.


Asunto(s)
Células de la Médula Ósea/citología , Trasplante de Médula Ósea/métodos , Lesiones de la Cornea , Lesiones Oculares/cirugía , Cicatrización de Heridas , Animales , Córnea/patología , Córnea/cirugía , Modelos Animales de Enfermedad , Lesiones Oculares/metabolismo , Lesiones Oculares/patología , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
17.
J Neuroimmunol ; 259(1-2): 37-46, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23601904

RESUMEN

This study was designed to understand the impact of self-antigen load on manifestation of organ specific autoimmunity. Using a transgenic mouse model characterized by CNS hypermyelination, we show that larger myelin content results in greater severity of experimental autoimmune encephalomyelitis attributable to an increased number of microglia within the hypermyelinated brain. We conclude that a larger self-antigen load affects an increase in number of tissue resident antigen presenting cells (APCs) most likely due to compensatory antigen clearance mechanisms thereby enhancing the probability of productive T cell-APC interactions in an antigen abundant environment and results in enhanced severity of autoimmune disease.


Asunto(s)
Presentación de Antígeno/inmunología , Autoantígenos/inmunología , Sistema Nervioso Central/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Microglía/inmunología , Vaina de Mielina/inmunología , Animales , Sistema Nervioso Central/patología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/patología , Femenino , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos , Ratones Transgénicos , Microglía/patología , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Vaina de Mielina/patología , Neuroinmunomodulación/inmunología , Índice de Severidad de la Enfermedad , Linfocitos T/inmunología , Linfocitos T/patología
18.
Am J Pathol ; 181(3): 775-84, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22901749

RESUMEN

Mastitis is a substantial clinical problem in lactating women that may result in severe pain and abrupt termination of breastfeeding, thereby predisposing infants to long-term health risks. Many cases of mastitis involve no known infectious agent and may fundamentally be due to autoimmune-mediated inflammation of the breast. Herein, we develop a murine model of autoimmune mastitis and provide a detailed characterization of its resulting phenotype of breast failure and lactation insufficiency. To generate breast-specific autoimmunity, we immunized SWXJ mice with recombinant mouse α-lactalbumin, a lactation-dependent, breast-specific differentiation protein critical for production of lactose. Mice immunized with α-lactalbumin showed extensive T-cell-mediated inflammation in lactating normal breast parenchyma but none in nonlactating normal breast parenchyma. This targeted autoimmune attack resulted in breast failure characterized by lactation insufficiency and decreased ability to nurture offspring. Although immunization with α-lactalbumin had no effect on fertility and birth numbers, pups nursed by α-lactalbumin-immunized mice showed significantly disrupted growth often accompanied by kwashiorkor-like nutritional abnormalities, including alopecia, liver toxicity, and runting. This experimental model of autoimmune breast failure has useful applications for prophylactic breast cancer vaccination and for addressing inflammatory complications during breastfeeding. In addition, this model is suited for investigating nutritionally based "failure-to-thrive" issues, particularly regarding the long-term implications of postnatal nutritional deprivation.


Asunto(s)
Enfermedades Autoinmunes/patología , Lactancia/fisiología , Glándulas Mamarias Animales/inmunología , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/prevención & control , Vacunación , Animales , Animales Recién Nacidos , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/fisiopatología , Complejo CD3/metabolismo , Vacunas contra el Cáncer/inmunología , Movimiento Celular/inmunología , Reactividad Cruzada/inmunología , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Inmunidad/inmunología , Inmunización Pasiva , Inflamación/complicaciones , Inflamación/inmunología , Inflamación/patología , Lactalbúmina/inmunología , Lactancia/genética , Neoplasias Mamarias Experimentales/patología , Ratones , Fenómenos Fisiológicos de la Nutrición , Fenotipo , Linfocitos T/inmunología
19.
Gynecol Oncol ; 124(1): 98-104, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22004903

RESUMEN

OBJECTIVES: An immune response sufficient to induce organ failure may provide protection and therapy against tumors derived from the targeted organ particularly when removal or ablation of the organ is part of the standard therapy and does not threaten survival. We have previously shown that a targeted immune response directed against the ovarian-specific protein, inhibin-α, causes ovarian failure. Here we determined whether inhibin-α autoimmunity is effective in both prevention and treatment of ovarian tumors. METHODS: A transgene consisting of the SV40 large tumor transformation antigen under the regulation of an anti-Mullerian hormone promoter (AMH-SV40Tag) was transferred by backcrossing for 12 generations to SJL/J mice producing SJL.AMH-SV40Tag (H-2(s)) females that develop a high incidence of autochthonous granulosa cell tumors. We determined whether immunization of SJL.AMH-SV40Tag female mice with the IA(s)-restricted p215-234 peptide of mouse inhibin-α was capable of preventing and treating these ovarian tumors. RESULTS: The growth of autochthonous ovarian granulosa cell tumors in SJL.AMH-SV40Tag transgenic mice was significantly inhibited in mice immunized with Inα 215-234. In addition, significant inhibition of tumor growth occurred when mice with established ovarian granulosa cell tumors were therapeutically vaccinated with Inα 215-234. CONCLUSIONS: Our results indicate that induction of ovarian-specific autoimmunity may serve as an effective way to prevent the emergence of autochthonous ovarian tumors and control the growth of established ovarian malignancies.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/terapia , Vacunas contra el Cáncer/farmacología , Inhibinas/inmunología , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/terapia , Fragmentos de Péptidos/inmunología , Secuencia de Aminoácidos , Animales , Enfermedades Autoinmunes/patología , Enfermedades Autoinmunes/prevención & control , Autoinmunidad/inmunología , Vacunas contra el Cáncer/inmunología , Procesos de Crecimiento Celular/efectos de los fármacos , Procesos de Crecimiento Celular/inmunología , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Datos de Secuencia Molecular , Neoplasias Ováricas/patología , Neoplasias Ováricas/prevención & control
20.
Ann Med ; 43(5): 356-65, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21651440

RESUMEN

Despite the monumental success of childhood prophylactic vaccination, there is no similar program designed to provide protection as we age against adult onset diseases like breast cancer. Instead, the predominant focus of current cancer vaccine strategy is to vaccinate after the tumors become established. This strategy has at best provided incremental improvement in overall survival. We propose the development of an adult vaccination program modeled on the childhood program that provides protection against diseases we confront as we enter our middle age. Since most cases of adult cancers are not associated with definitive etiopathogenic viruses, we propose extending our selection of vaccine targets to tissue-specific self proteins that are over-expressed in developing tumors but are no longer expressed in normal tissues ('retired or former self'), are expressed in normal tissues under readily avoidable conditions ('conditional self'), or are incapable of targeting any clinically significant autoimmune complications ('irrelevant self'). By extending prophylactic vaccination to such "functional non-self" targets, prophylactic vaccination against adult onset diseases like breast cancer may occur safely in the absence of any autoimmune inflammatory complications and may potentially reduce disease incidence in a manner that mimics the impact of childhood vaccination on diseases like measles and polio.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Proteínas de Neoplasias/inmunología , Neoplasias/prevención & control , Adulto , Animales , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/prevención & control , Femenino , Humanos , Programas de Inmunización/métodos , Neoplasias/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...