Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cancer Res Commun ; 3(8): 1486-1500, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37559947

RESUMEN

While the role of prostaglandin E2 (PGE2) in promoting malignant progression is well established, how to optimally block the activity of PGE2 signaling remains to be demonstrated. Clinical trials with prostaglandin pathway targeted agents have shown activity but without sufficient significance or dose-limiting toxicities that have prevented approval. PGE2 signals through four receptors (EP1-4) to modulate tumor progression. EP2 and EP4 signaling exacerbates tumor pathology and is immunosuppressive through potentiating cAMP production. EP1 and EP3 signaling has the opposite effect through increasing IP3 and decreasing cAMP. Using available small-molecule antagonists of single EP receptors, the cyclooxygenase-2 (COX-2) inhibitor celecoxib, or a novel dual EP2/EP4 antagonist generated in this investigation, we tested which approach to block PGE2 signaling optimally restored immunologic activity in mouse and human immune cells and antitumor activity in syngeneic, spontaneous, and xenograft tumor models. We found that dual antagonism of EP2 and EP4 together significantly enhanced the activation of PGE2-suppressed mouse and human monocytes and CD8+ T cells in vitro as compared with single EP antagonists. CD8+ T-cell activation was dampened by single EP1 and EP3 antagonists. Dual EP2/EP4 PGE2 receptor antagonists increased tumor microenvironment lymphocyte infiltration and significantly reduced disease burden in multiple tumor models, including in the adenomatous polyposis coli (APC)min+/- spontaneous colorectal tumor model, compared with celecoxib. These results support a hypothesis that redundancy of EP2 and EP4 receptor signaling necessitates a therapeutic strategy of dual blockade of EP2 and EP4. Here we describe TPST-1495, a first-in-class orally available small-molecule dual EP2/EP4 antagonist. Significance: Prostaglandin (PGE2) drives tumor progression but the pathway has not been effectively drugged. We demonstrate significantly enhanced immunologic potency and antitumor activity through blockade of EP2 and EP4 PGE2 receptor signaling together with a single molecule.


Asunto(s)
Neoplasias , Prostaglandinas , Humanos , Animales , Ratones , Dinoprostona/metabolismo , Subtipo EP2 de Receptores de Prostaglandina E/metabolismo , Celecoxib/farmacología , Linfocitos T CD8-positivos/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Inhibidores de la Ciclooxigenasa 2 , Microambiente Tumoral
2.
J Immunol ; 200(12): 4012-4023, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29703862

RESUMEN

Maintenance of the regulatory T (Treg) cell pool is essential for peripheral tolerance and prevention of autoimmunity. Integrins, heterodimeric transmembrane proteins consisting of α and ß subunits that mediate cell-to-cell and cell-to-extracellular matrix interactions, play an important role in facilitating Treg cell contact-mediated suppression. In this article, we show that integrin activation plays an essential, previously unappreciated role in maintaining murine Treg cell function. Treg cell-specific loss of talin, a ß integrin-binding protein, or expression of talin(L325R), a mutant that selectively abrogates integrin activation, resulted in lethal systemic autoimmunity. This dysfunction could be attributed, in part, to a global dysregulation of the Treg cell transcriptome. Activation of integrin α4ß1 led to increased suppressive capacity of the Treg cell pool, suggesting that modulating integrin activation on Treg cells may be a useful therapeutic strategy for autoimmune and inflammatory disorders. Taken together, these results reveal a critical role for integrin-mediated signals in controlling peripheral tolerance by virtue of maintaining Treg cell function.


Asunto(s)
Integrinas/inmunología , Tolerancia Periférica/inmunología , Linfocitos T Reguladores/inmunología , Animales , Autoinmunidad/inmunología , Inflamación/inmunología , Ratones , Talina/inmunología , Transcriptoma/inmunología
3.
J Clin Invest ; 127(10): 3609-3623, 2017 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-28846070

RESUMEN

During an immune response, CD8+ T lymphocytes can undergo asymmetric division, giving rise to daughter cells that exhibit distinct tendencies to adopt terminal effector and memory cell fates. Here we show that "pre-effector" and "pre-memory" cells resulting from the first CD8+ T cell division in vivo exhibited low and high rates of endogenous proteasome activity, respectively. Pharmacologic reduction of proteasome activity in CD8+ T cells early during differentiation resulted in acquisition of terminal effector cell characteristics, whereas enhancement of proteasome activity conferred attributes of memory lymphocytes. Transcriptomic and proteomic analyses revealed that modulating proteasome activity in CD8+ T cells affected cellular metabolism. These metabolic changes were mediated, in part, through differential expression of Myc, a transcription factor that controls glycolysis and metabolic reprogramming. Taken together, these results demonstrate that proteasome activity is an important regulator of CD8+ T cell fate and raise the possibility that increasing proteasome activity may be a useful therapeutic strategy to enhance the generation of memory lymphocytes.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/inmunología , División Celular/inmunología , Glucólisis/inmunología , Memoria Inmunológica , Complejo de la Endopetidasa Proteasomal/inmunología , Animales , Linfocitos T CD8-positivos/metabolismo , Ratones , Ratones Mutantes , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Proto-Oncogénicas c-myc/inmunología , Proteínas Proto-Oncogénicas c-myc/metabolismo
4.
J Immunol ; 198(12): 4639-4651, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28515282

RESUMEN

Talin, a cytoskeletal protein essential in mediating integrin activation, has been previously shown to be involved in the regulation of T cell proliferation and function. In this study, we describe a role for talin in maintaining the homeostasis and survival of the regulatory T (Treg) cell pool. T cell-specific deletion of talin in Tln1fl/flCd4Cre mice resulted in spontaneous lymphocyte activation, primarily due to numerical and functional deficiencies of Treg cells in the periphery. Peripheral talin-deficient Treg cells were unable to maintain high expression of IL-2Rα, resulting in impaired IL-2 signaling and ultimately leading to increased apoptosis through downregulation of prosurvival proteins Bcl-2 and Mcl-1. The requirement for talin in maintaining high IL-2Rα expression by Treg cells was due, in part, to integrin LFA-1-mediated interactions between Treg cells and dendritic cells. Collectively, our data suggest a critical role for talin in Treg cell-mediated maintenance of immune homeostasis.


Asunto(s)
Homeostasis , Activación de Linfocitos , Transducción de Señal , Linfocitos T Reguladores/inmunología , Talina/metabolismo , Animales , Apoptosis , Células Dendríticas/inmunología , Genes bcl-2 , Interleucina-2/inmunología , Interleucina-2/metabolismo , Subunidad alfa del Receptor de Interleucina-2/genética , Subunidad alfa del Receptor de Interleucina-2/inmunología , Antígeno-1 Asociado a Función de Linfocito/inmunología , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Ratones , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Linfocitos T Reguladores/fisiología , Talina/deficiencia , Talina/inmunología
5.
Nat Immunol ; 18(4): 422-432, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28218746

RESUMEN

During microbial infection, responding CD8+ T lymphocytes differentiate into heterogeneous subsets that together provide immediate and durable protection. To elucidate the dynamic transcriptional changes that underlie this process, we applied a single-cell RNA-sequencing approach and analyzed individual CD8+ T lymphocytes sequentially throughout the course of a viral infection in vivo. Our analyses revealed a striking transcriptional divergence among cells that had undergone their first division and identified previously unknown molecular determinants that controlled the fate specification of CD8+ T lymphocytes. Our findings suggest a model for the differentiation of terminal effector cells initiated by an early burst of transcriptional activity and subsequently refined by epigenetic silencing of transcripts associated with memory lymphocytes, which highlights the power and necessity of single-cell approaches.


Asunto(s)
Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/genética , Epigénesis Genética , Transcripción Genética , Animales , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/inmunología , Análisis por Conglomerados , Biología Computacional/métodos , Perfilación de la Expresión Génica , Silenciador del Gen , Heterogeneidad Genética , Histonas/metabolismo , Memoria Inmunológica/genética , Memoria Inmunológica/inmunología , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Transcriptoma
6.
J Immunol ; 196(10): 4237-45, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-27029586

RESUMEN

The microtubule-associated protein lissencephaly 1 (Lis1) is a key regulator of cell division during stem cell renewal and differentiation. In this study, we examined the role of Lis1 in T lymphocyte homeostasis and fate diversification in response to microbial infection. T cell-specific deletion of Lis1 resulted in depletion of the peripheral CD4(+) and CD8(+) T lymphocyte pool owing to a loss of homeostatic, cytokine-induced proliferation. In contrast, cognate Ag-triggered proliferation was much less affected, enabling Lis1-deficient CD8(+) T cells to differentiate into terminal effector cells in response to microbial infection. Strikingly, however, the specification of Lis1-deficient long-lived memory CD8(+) T lymphocytes was impaired due, in part, to an apparent failure to differentiate appropriately to IL-15. Taken together, these findings suggest that Lis1 plays an important role in T cell homeostasis and the generation of memory T lymphocytes.


Asunto(s)
1-Alquil-2-acetilglicerofosfocolina Esterasa/inmunología , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/inmunología , Memoria Inmunológica , Proteínas Asociadas a Microtúbulos/inmunología , 1-Alquil-2-acetilglicerofosfocolina Esterasa/genética , Animales , Linfocitos T CD4-Positivos/inmunología , División Celular , Homeostasis/inmunología , Inmunofenotipificación , Interleucina-15/inmunología , Interleucina-7/inmunología , Listeria monocytogenes , Listeriosis/inmunología , Activación de Linfocitos , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/genética , Transducción de Señal , Timo/inmunología
7.
Oncotarget ; 7(16): 21527-41, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-26930717

RESUMEN

The epithelial-mesenchymal transition (EMT) endows carcinoma cells with phenotypic plasticity that can facilitate the formation of cancer stem cells (CSCs) and contribute to the metastatic cascade. While there is substantial support for the role of EMT in driving cancer cell dissemination, less is known about the intracellular molecular mechanisms that govern formation of CSCs via EMT. Here we show that ß2 and ß5 proteasome subunit activity is downregulated during EMT in immortalized human mammary epithelial cells. Moreover, selective proteasome inhibition enabled mammary epithelial cells to acquire certain morphologic and functional characteristics reminiscent of cancer stem cells, including CD44 expression, self-renewal, and tumor formation. Transcriptomic analyses suggested that proteasome-inhibited cells share gene expression signatures with cells that have undergone EMT, in part, through modulation of the TGF-ß signaling pathway. These findings suggest that selective downregulation of proteasome activity in mammary epithelial cells can initiate the EMT program and acquisition of a cancer stem cell-like phenotype. As proteasome inhibitors become increasingly used in cancer treatment, our findings highlight a potential risk of these therapeutic strategies and suggest a possible mechanism by which carcinoma cells may escape from proteasome inhibitor-based therapy.


Asunto(s)
Regulación hacia Abajo , Transición Epitelial-Mesenquimal , Células Madre Neoplásicas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Animales , Biocatálisis/efectos de los fármacos , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Carcinogénesis/genética , Carcinogénesis/metabolismo , Carcinogénesis/patología , Línea Celular , Línea Celular Transformada , Femenino , Perfilación de la Expresión Génica/métodos , Humanos , Estimación de Kaplan-Meier , Ratones Desnudos , Inhibidores de Proteasoma/farmacología , Trasplante Heterólogo
8.
Nat Immunol ; 17(3): 286-96, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26829767

RESUMEN

SHARPIN forms a linear-ubiquitin-chain-assembly complex that promotes signaling via the transcription factor NF-κB. SHARPIN deficiency leads to progressive multi-organ inflammation and immune system malfunction, but how SHARPIN regulates T cell responses is unclear. Here we found that SHARPIN deficiency resulted in a substantial reduction in the number of and defective function of regulatory T cells (Treg cells). Transfer of SHARPIN-sufficient Treg cells into SHARPIN-deficient mice considerably alleviated their systemic inflammation. SHARPIN-deficient T cells displayed enhanced proximal signaling via the T cell antigen receptor (TCR) without an effect on the activation of NF-κB. SHARPIN conjugated with Lys63 (K63)-linked ubiquitin chains, which led to inhibition of the association of TCRζ with the signaling kinase Zap70; this affected the generation of Treg cells. Our study therefore identifies a role for SHARPIN in TCR signaling whereby it maintains immunological homeostasis and tolerance by regulating Treg cells.


Asunto(s)
Proteínas Portadoras/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Animales , Proteínas Portadoras/genética , Colitis/inmunología , Citocinas/inmunología , Femenino , Citometría de Flujo , Humanos , Tolerancia Inmunológica/inmunología , Immunoblotting , Inmunoprecipitación , Técnicas In Vitro , Inflamación , Péptidos y Proteínas de Señalización Intracelular , Células Jurkat , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , FN-kappa B/inmunología , Transducción de Señal , Ubiquitinación , Proteína Tirosina Quinasa ZAP-70/inmunología
9.
Sci Rep ; 6: 19182, 2016 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-26765121

RESUMEN

Naïve CD8(+) T lymphocytes responding to microbial pathogens give rise to effector T cells that provide acute defense and memory T cells that provide long-lived immunity. Upon activation, CD8(+) T lymphocytes can undergo asymmetric division, unequally distributing factors to the nascent daughter cells that influence their eventual fate towards the effector or memory lineages. Individual loss of either atypical protein kinase C (aPKC) isoform, PKCζ or PKCλ/ι, partially impairs asymmetric divisions and increases CD8(+) T lymphocyte differentiation toward a long-lived effector fate at the expense of memory T cell formation. Here, we show that deletion of both aPKC isoforms resulted in a deficit in asymmetric divisions, increasing the proportion of daughter cells that inherit high amounts of effector fate-associated molecules, IL-2Rα, T-bet, IFNγR, and interferon regulatory factor 4 (IRF4). However, unlike CD8(+) T cells deficient in only one aPKC isoform, complete loss of aPKC unexpectedly increased CD8(+) T cell differentiation toward a short-lived, terminal effector fate, as evidenced by increased rates of apoptosis and decreased expression of Eomes and Bcl2 early during the immune response. Together, these results provide evidence for an important role for asymmetric division in CD8(+) T lymphocyte fate specification by regulating the balance between effector and memory precursors at the initiation of the adaptive immune response.


Asunto(s)
División Celular Asimétrica , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/fisiología , Diferenciación Celular , Proteína Quinasa C/metabolismo , Inmunidad Adaptativa , Animales , Técnicas de Inactivación de Genes , Memoria Inmunológica , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones , Proteína Quinasa C/deficiencia , Proteína Quinasa C/genética , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/fisiología
10.
Nat Commun ; 6: 8369, 2015 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-26458291

RESUMEN

Tamoxifen is a selective oestrogen receptor modulator widely used for the treatment of breast cancer. In addition to its activity as an oestrogen receptor agonist/antagonist, tamoxifen also modulates sphingolipid biosynthesis, which has been shown to play an important role in the regulation of neutrophil activity. Here, we find that tamoxifen stimulation enhances several pro-inflammatory pathways in human neutrophils, including chemotaxis, phagocytosis and neutrophil extracellular trap (NET) formation. The enhancement of NET production occurs via a ceramide/PKCζ-mediated pathway, and treatment with synthetic ceramide is sufficient to promote NET formation. Pretreatment of human neutrophils with tamoxifen boosts neutrophil bactericidal capacity against a variety of pathogens in vitro and enhances clearance of the leading human pathogen methicillin-resistant Staphylococcus aureus in vivo. Our results suggest that tamoxifen, and the lipid signalling pathways it modulates, merit further exploration as targets for boosting host innate immune function.


Asunto(s)
Ceramidas/metabolismo , Trampas Extracelulares/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Tamoxifeno/farmacología , Animales , Femenino , Voluntarios Sanos , Humanos , Inmunidad Innata/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina , Ratones , Neutrófilos/metabolismo , Proteína Quinasa C/metabolismo
11.
J Clin Invest ; 123(12): 5165-78, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24270422

RESUMEN

Mammalian target of rapamycin (mTOR) plays a crucial role in the control of T cell fate determination; however, the precise regulatory mechanism of the mTOR pathway is not fully understood. We found that T cell-specific deletion of the gene encoding tuberous sclerosis 1 (TSC1), an upstream negative regulator of mTOR, resulted in augmented Th1 and Th17 differentiation and led to severe intestinal inflammation in a colitis model. Conditional Tsc1 deletion in Tregs impaired their suppressive activity and expression of the Treg marker Foxp3 and resulted in increased IL-17 production under inflammatory conditions. A fate-mapping study revealed that Tsc1-null Tregs that lost Foxp3 expression gained a stronger effector-like phenotype compared with Tsc1-/- Foxp3+ Tregs. Elevated IL-17 production in Tsc1-/- Treg cells was reversed by in vivo knockdown of the mTOR target S6K1. Moreover, IL-17 production was enhanced by Treg-specific double deletion of Tsc1 and Foxo3a. Collectively, these studies suggest that TSC1 acts as an important checkpoint for maintaining immune homeostasis by regulating cell fate determination.


Asunto(s)
Colitis/inmunología , Linfocitos T Reguladores/inmunología , Células TH1/inmunología , Células Th17/inmunología , Proteínas Supresoras de Tumor/fisiología , Traslado Adoptivo , Animales , Diferenciación Celular , Células Cultivadas , Colitis/genética , Citocinas/metabolismo , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/biosíntesis , Factores de Transcripción Forkhead/deficiencia , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/fisiología , Técnicas de Silenciamiento del Gen , Homeostasis , Tolerancia Inmunológica , Inmunidad Mucosa , Interleucina-17/biosíntesis , Interleucina-17/genética , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Quimera por Radiación , Proteínas Quinasas S6 Ribosómicas 90-kDa/genética , Proteínas Quinasas S6 Ribosómicas 90-kDa/fisiología , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...