Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cells ; 12(2)2023 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-36672221

RESUMEN

The serine/threonine protein kinase calcium/calmodulin-dependent protein kinase kinase 2 (CAMKK2) plays critical roles in a range of biological processes. Despite its importance, only a handful of inhibitors of CAMKK2 have been disclosed. Having a selective small molecule tool to interrogate this kinase will help demonstrate that CAMKK2 inhibition can be therapeutically beneficial. Herein, we disclose SGC-CAMKK2-1, a selective chemical probe that targets CAMKK2.

2.
Cancer Immunol Res ; 11(1): 109-122, 2023 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-36301267

RESUMEN

Calcium/calmodulin-dependent protein kinase kinase 2 (CaMKK2) is a key regulator of energy homeostasis in several cell types. Expression of this enzyme in tumor cells promotes proliferation and migration, and expression in tumor-associated immune cells facilitates M2 macrophage polarization and the development of myeloid-derived suppressor cells. Thus, there has been interest in developing CaMKK2 inhibitors as potential anticancer therapeutics. One impediment to clinical development of these agents is that the roles of CaMKK2 in other cellular compartments within the tumor immune microenvironment remain to be established. We report herein that CaMKK2 is expressed at low basal levels in natural killer (NK) cells but is upregulated in tumor-infiltrating NK cells where it suppresses apoptosis and promotes proliferation. NK cell-intrinsic deletion of CaMKK2 increased metastatic progression in several murine models, establishing a critical role for this enzyme in NK cell-mediated antitumor immunity. Ablation of the CaMKK2 protein, but not inhibition of its kinase activity, resulted in decreased NK-cell survival. These results indicate an important scaffolding function for CaMKK2 in NK cells and suggest that competitive CaMKK2 inhibitors and ligand-directed degraders (LDD) are likely to have distinct therapeutic utilities. Finally, we determined that intracellular lactic acid is a key driver of CaMKK2 expression, suggesting that upregulated expression of this enzyme is an adaptive mechanism by which tumor-infiltrating NK cells mitigate the deleterious effects of a lactic acid-rich tumor microenvironment. The findings of this study should inform strategies to manipulate the CaMKK2-signaling axis as a therapeutic approach in cancer.


Asunto(s)
Neoplasias , Humanos , Ratones , Animales , Neoplasias/metabolismo , Transducción de Señal , Fosforilación , Apoptosis , Macrófagos , Microambiente Tumoral , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo
3.
Nat Commun ; 13(1): 6483, 2022 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-36309495

RESUMEN

Glioblastoma (GBM) is notorious for its immunosuppressive tumor microenvironment (TME) and is refractory to immune checkpoint blockade (ICB). Here, we identify calmodulin-dependent kinase kinase 2 (CaMKK2) as a driver of ICB resistance. CaMKK2 is highly expressed in pro-tumor cells and is associated with worsened survival in patients with GBM. Host CaMKK2, specifically, reduces survival and promotes ICB resistance. Multimodal profiling of the TME reveals that CaMKK2 is associated with several ICB resistance-associated immune phenotypes. CaMKK2 promotes exhaustion in CD8+ T cells and reduces the expansion of effector CD4+ T cells, additionally limiting their tumor penetrance. CaMKK2 also maintains myeloid cells in a disease-associated microglia-like phenotype. Lastly, neuronal CaMKK2 is required for maintaining the ICB resistance-associated myeloid phenotype, is deleterious to survival, and promotes ICB resistance. Our findings reveal CaMKK2 as a contributor to ICB resistance and identify neurons as a driver of immunotherapeutic resistance in GBM.


Asunto(s)
Glioblastoma , Humanos , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Linfocitos T CD8-positivos , Microambiente Tumoral , Terapia de Inmunosupresión , Neuronas/patología , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/genética
4.
Stem Cell Rev Rep ; 18(7): 2513-2521, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35262902

RESUMEN

Little is known about a regulatory role of CaMKK2 for hematopoietic stem (HSC) and progenitor (HPC) cell function. To assess this, we used Camkk2-/- and wild type (WT) control mouse bone marrow (BM) cells. BM cells were collected/processed and compared under hypoxia (3% oxygen; physioxia) vs. ambient air (~21% oxygen). Subjecting cells collected to ambient air, even for a few minutes, causes a stress that we termed Extra Physiological Shock/Stress (EPHOSS) that causes differentiation of HSCs and HPCs. We consider physioxia collection/processing a more relevant way to assess HSC/HPC numbers and function, as the cells remain in an oxygen tension closer physiologic conditions. Camkk2-/- cells collected/processed at 3% oxygen had positive and negative effects respectively on HSCs (by engraftment using competitive transplantation with congenic donor and competitor cells and lethally irradiated congenic recipient mice), and HPCs (by colony forming assays of CFU-GM, BFU-E, and CFU-GEMM) compared to WT cells processed in ambient air. Thus, with cells collected/processed under physioxia, and therefore never exposed and naïve to ambient air conditions, CaMKK2 not only appears to act as an HSC to HPC differentiation fate determinant, but as we found for other intracellular mediators, the Camkk-/- mouse BM cells were relatively resistant to effects of EPHOSS. This information is of potential use for modulation of WT BM HSCs and HPCs for future clinical advantage.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Oxígeno , Animales , Células de la Médula Ósea , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/genética , Ratones , Ratones Noqueados , Oxígeno/farmacología
5.
Front Immunol ; 12: 754083, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34712241

RESUMEN

Myeloid-derived suppressor cells (MDSCs) are a hetero geneous group of cells, which can suppress the immune response, promote tumor progression and impair the efficacy of immunotherapies. Consequently, the pharmacological targeting of MDSC is emerging as a new immunotherapeutic strategy to stimulate the natural anti-tumor immune response and potentiate the efficacy of immunotherapies. Herein, we leveraged genetically modified models and a small molecule inhibitor to validate Calcium-Calmodulin Kinase Kinase 2 (CaMKK2) as a druggable target to control MDSC accumulation in tumor-bearing mice. The results indicated that deletion of CaMKK2 in the host attenuated the growth of engrafted tumor cells, and this phenomenon was associated with increased antitumor T cell response and decreased accumulation of MDSC. The adoptive transfer of MDSC was sufficient to restore the ability of the tumor to grow in Camkk2-/- mice, confirming the key role of MDSC in the mechanism of tumor rejection. In vitro studies indicated that blocking of CaMKK2 is sufficient to impair the yield of MDSC. Surprisingly, MDSC generated from Camkk2-/- bone marrow cells also showed a higher ability to terminally differentiate toward more immunogenic cell types (e.g inflammatory macrophages and dendritic cells) compared to wild type (WT). Higher intracellular levels of reactive oxygen species (ROS) accumulated in Camkk2-/- MDSC, increasing their susceptibility to apoptosis and promoting their terminal differentiation toward more mature myeloid cells. Mechanistic studies indicated that AMP-activated protein kinase (AMPK), which is a known CaMKK2 proximal target controlling the oxidative stress response, fine-tunes ROS accumulation in MDSC. Accordingly, failure to activate the CaMKK2-AMPK axis can account for the elevated ROS levels in Camkk2-/- MDSC. These results highlight CaMKK2 as an important regulator of the MDSC lifecycle, identifying this kinase as a new druggable target to restrain MDSC expansion and enhance the efficacy of anti-tumor immunotherapy.


Asunto(s)
Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/fisiología , Células Supresoras de Origen Mieloide/enzimología , Proteínas de Neoplasias/fisiología , Proteínas Quinasas Activadas por AMP/fisiología , Traslado Adoptivo , Animales , Apoptosis , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/deficiencia , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/genética , Femenino , Depleción Linfocítica , Linfoma/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mitocondrias/metabolismo , Células Supresoras de Origen Mieloide/fisiología , Células Supresoras de Origen Mieloide/trasplante , Mielopoyesis , Especies Reactivas de Oxígeno , Microambiente Tumoral
6.
Front Immunol ; 11: 217, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32117323

RESUMEN

With this article, the authors aim to honor the memory of Serafino Zappacosta, who had been their mentor during the early years of their career in science. The authors discuss how the combination of Serafino Zappacosta's extraordinary commitment to teaching and passion for science created a fostering educational environment that led to the creation of the "Ruggero Ceppellini Advanced School of Immunology." The review also illustrates how the research on the MHC and the inspirational scientific context in the Zappacosta's laboratory influenced the authors' early scientific interests, and subsequent professional work as immunologists.


Asunto(s)
Alergia e Inmunología/historia , Linfocitos T/inmunología , Alergia e Inmunología/educación , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Inmunidad Celular , Complejo Mayor de Histocompatibilidad , Masculino , Mentores , Investigación
7.
Int J Nanomedicine ; 14: 3403-3411, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31190799

RESUMEN

Aim: To quantitatively and sensitively investigate the biodistribution of immune cells after systemic administration. Methods: Immune cells were loaded with plasmonic gold nanostars (GNS) tracking probes. Inductively coupled plasma mass spectrometry (ICP-MS) was used for quantitative gold mass measurement and two-photon photoluminescence (TPL) was used for high-resolution sensitive optical imaging. Results: GNS nanoparticles were loaded successfully into immune cells without negative effect on cellular vitality. Liver and spleen were identified to be the major organs for macrophage cells uptake after systematic administration. A small amount of macrophage cells were detected in the tumor site in our murine lymphoma animal model. Conclusion: GNS has great potential as a biocompatible marker for quantitative tracking and high-resolution imaging of immune cells at the cellular level.


Asunto(s)
Oro/química , Linfocitos/metabolismo , Nanopartículas del Metal/química , Animales , Línea Celular Tumoral , Supervivencia Celular , Macrófagos/metabolismo , Nanopartículas del Metal/administración & dosificación , Nanopartículas del Metal/ultraestructura , Ratones Endogámicos C57BL , Distribución Tisular
8.
Nat Commun ; 10(1): 2450, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-31164648

RESUMEN

Tumor-associated myeloid cells regulate tumor growth and metastasis, and their accumulation is a negative prognostic factor for breast cancer. Here we find calcium/calmodulin-dependent kinase kinase (CaMKK2) to be highly expressed within intratumoral myeloid cells in mouse models of breast cancer, and demonstrate that its inhibition within myeloid cells suppresses tumor growth by increasing intratumoral accumulation of effector CD8+ T cells and immune-stimulatory myeloid subsets. Tumor-associated macrophages (TAMs) isolated from Camkk2-/- mice expressed higher levels of chemokines involved in the recruitment of effector T cells compared to WT. Similarly, in vitro generated Camkk2-/- macrophages recruit more T cells, and have a reduced capability to suppress T cell proliferation, compared to WT. Treatment with CaMKK2 inhibitors blocks tumor growth in a CD8+ T cell-dependent manner, and facilitates a favorable reprogramming of the immune cell microenvironment. These data, credential CaMKK2 as a myeloid-selective checkpoint, the inhibition of which may have utility in the immunotherapy of breast cancer.


Asunto(s)
Neoplasias de la Mama/inmunología , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/inmunología , Carcinoma/inmunología , Neoplasias Mamarias Animales/inmunología , Células Mieloides/inmunología , Escape del Tumor/inmunología , Microambiente Tumoral/inmunología , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Linfocitos T CD8-positivos/inmunología , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/antagonistas & inhibidores , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/genética , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Carcinoma/genética , Carcinoma/metabolismo , Proliferación Celular , Quimiocinas/inmunología , Femenino , Humanos , Inmunohistoquímica , Técnicas In Vitro , Macrófagos/inmunología , Macrófagos/metabolismo , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Células Mieloides/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/metabolismo , Escape del Tumor/genética
9.
Cell Death Dis ; 8(10): e3076, 2017 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-28981105

RESUMEN

Hematopoietic stem and progenitor cells (HSPCs) are predominantly quiescent in adults, but proliferate in response to bone marrow (BM) injury. Here, we show that deletion of Ca2+/calmodulin (CaM)-dependent protein kinase kinase 2 (CaMKK2) promotes HSPC regeneration and hematopoietic recovery following radiation injury. Using Camkk2-enhanced green fluorescent protein (EGFP) reporter mice, we found that Camkk2 expression is developmentally regulated in HSPC. Deletion of Camkk2 in HSPC results in a significant downregulation of genes affiliated with the quiescent signature. Accordingly, HSPC from Camkk2 null mice have a high proliferative capability when stimulated in vitro in the presence of BM-derived endothelial cells. In addition, Camkk2 null mice are more resistant to radiation injury and show accelerated hematopoietic recovery, enhanced HSPC regeneration and ultimately a prolonged survival following sublethal or lethal total body irradiation. Mechanistically, we propose that CaMKK2 regulates the HSPC response to hematopoietic damage by coupling radiation signaling to activation of the anti-proliferative AMP-activated protein kinase. Finally, we demonstrated that systemic administration of the small molecule CaMKK2 inhibitor, STO-609, to irradiated mice enhanced HSPC recovery and improved survival. These findings identify CaMKK2 as an important regulator of HSPC regeneration and demonstrate CaMKK2 inhibition is a novel approach to promoting hematopoietic recovery after BM injury.


Asunto(s)
Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/genética , Calmodulina/genética , Células Madre Hematopoyéticas/metabolismo , Traumatismos por Radiación/tratamiento farmacológico , Animales , Bencimidazoles/administración & dosificación , Calcio/metabolismo , Proteínas Fluorescentes Verdes/genética , Ratones , Ratones Noqueados , Naftalimidas/administración & dosificación , Traumatismos por Radiación/genética , Traumatismos por Radiación/patología , Regeneración/genética , Transducción de Señal/efectos de los fármacos , Irradiación Corporal Total
10.
Int J Cardiol ; 224: 206-212, 2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-27657475

RESUMEN

BACKGROUND: Naive T lymphocytes recirculate through the body, traveling from secondary lymphoid organs through tissues and via lymphatic vessels and peripheral blood into other secondary lymphoid organs and into the bone marrow. In these tissues, lymphocytes are exposed to the chemokine CXCL12 which is abundantly produced in bone marrow and in lymph nodes by stromal cells. CXCL12 is known to drive lymphocytes chemotaxis and, in cells types such as stem cells, an antiapopototic effect has been described. METHODS: Here we analyzed the effect of CXCL12 exposure on naïve CD4+ T lymphocytes purified from peripheral blood by immunomagnetic negative isolation and cultured in a nutrient poor medium. We also studied, mainly by western blot analysis, the signaling pathways involved in CXCL12 action on naïve CD4+ T lymphocytes. RESULTS: We found that CXCL12-exposed cells survived longer than untreated ones and this prolonged lifespan was specific for resting naïve lymphocytes, while in vitro activated lymphoblasts died rapidly despite CXCL12 treatment. We demonstrated that the increased percentage of living cells observed upon CXCL12 administration was not due to induction of proliferation but to a prosurvival effect of this chemokine. Moreover, our data suggest that this prosurvival effect on naïve CD4+ T lymphocytes might likely be mediated by PKA-dependent CREB activation and consequent increased expression of the antiapoptotic factors Bcl2 and BclXl. CONCLUSIONS: This newly reported activity of CXCL12 might contribute to the maintenance of the naïve T lymphocytes pool in vivo, which is needed to ensure a proper immune response to new antigens.


Asunto(s)
Linfocitos T CD4-Positivos/fisiología , Supervivencia Celular/fisiología , Quimiocina CXCL12/metabolismo , Proteína de Unión a CREB/metabolismo , Células Cultivadas , Humanos , Proteínas Quinasas/metabolismo , Transducción de Señal/fisiología , Regulación hacia Arriba , Proteína bcl-X/metabolismo
11.
Genes Dev ; 28(9): 995-1004, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24788518

RESUMEN

Accidental or deliberate ionizing radiation exposure can be fatal due to widespread hematopoietic destruction. However, little is known about either the course of injury or the molecular pathways that regulate the subsequent regenerative response. Here we show that the Wnt signaling pathway is critically important for regeneration after radiation-induced injury. Using Wnt reporter mice, we show that radiation triggers activation of Wnt signaling in hematopoietic stem and progenitor cells. ß-Catenin-deficient mice, which lack the ability to activate canonical Wnt signaling, exhibited impaired hematopoietic stem cell regeneration and bone marrow recovery after radiation. We found that, as part of the mechanism, hematopoietic stem cells lacking ß-catenin fail to suppress the generation of reactive oxygen species and cannot resolve DNA double-strand breaks after radiation. Consistent with the impaired response to radiation, ß-catenin-deficient mice are also unable to recover effectively after chemotherapy. Collectively, these data indicate that regenerative responses to distinct hematopoietic injuries share a genetic dependence on ß-catenin and raise the possibility that modulation of Wnt signaling may be a path to improving bone marrow recovery after damage.


Asunto(s)
Células Madre Hematopoyéticas/fisiología , Estrés Oxidativo/genética , Regeneración/genética , beta Catenina/genética , beta Catenina/metabolismo , Animales , Antineoplásicos/farmacología , Médula Ósea/efectos de los fármacos , Médula Ósea/efectos de la radiación , Roturas del ADN de Doble Cadena/efectos de la radiación , Fluorouracilo/farmacología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de la radiación , Estimación de Kaplan-Meier , Ratones , Estrés Oxidativo/efectos de la radiación , Traumatismos por Radiación/genética , Especies Reactivas de Oxígeno/metabolismo , Regeneración/efectos de los fármacos , Regeneración/efectos de la radiación , Transducción de Señal , Vía de Señalización Wnt/efectos de la radiación
12.
J Bone Miner Res ; 28(7): 1599-610, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23408651

RESUMEN

Bone remodeling, a physiological process characterized by bone formation by osteoblasts (OBs) and resorption of preexisting bone matrix by osteoclasts (OCs), is vital for the maintenance of healthy bone tissue in adult humans. Imbalances in this vital process result in pathological conditions including osteoporosis. Owing to its initial asymptomatic nature, osteoporosis is often detected only after the patient has sustained significant bone loss or a fracture. Hence, anabolic therapeutics that stimulate bone accrual is in high clinical demand. Here we identify Ca²âº/calmodulin (CaM)-dependent protein kinase kinase 2 (CaMKK2) as a potential target for such therapeutics because its inhibition enhances OB differentiation and bone growth and suppresses OC differentiation. Mice null for CaMKK2 possess higher trabecular bone mass in their long bones, along with significantly more OBs and fewer multinuclear OCs. In vitro, although Camkk2⁻/⁻ mesenchymal stem cells (MSCs) yield significantly higher numbers of OBs, bone marrow cells from Camkk2⁻/⁻ mice produce fewer multinuclear OCs. Acute inhibition of CaMKK2 by its selective, cell-permeable pharmacological inhibitor STO-609 also results in increased OB and diminished OC formation. Further, we find phospho-protein kinase A (PKA) and Ser¹³³ phosphorylated form of cyclic adenosine monophosphate (cAMP) response element binding protein (pCREB) to be markedly elevated in OB progenitors deficient in CaMKK2. On the other hand, genetic ablation of CaMKK2 or its pharmacological inhibition in OC progenitors results in reduced pCREB as well as significantly reduced levels of its transcriptional target, nuclear factor of activated T cells, cytoplasmic (NFATc1). Moreover, in vivo administration of STO-609 results in increased OBs and diminished OCs, conferring significant protection from ovariectomy (OVX)-induced osteoporosis in adult mice. Overall, our findings reveal a novel function for CaMKK2 in bone remodeling and highlight the potential for its therapeutic inhibition as a valuable bone anabolic strategy that also inhibits OC differentiation in the treatment of osteoporosis.


Asunto(s)
Remodelación Ósea/fisiología , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Diferenciación Celular/fisiología , Osteoblastos/enzimología , Osteoclastos/enzimología , Adulto , Animales , Bencimidazoles/farmacología , Células de la Médula Ósea/enzimología , Remodelación Ósea/efectos de los fármacos , Proteína de Unión a CREB/genética , Proteína de Unión a CREB/metabolismo , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/antagonistas & inhibidores , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/genética , Diferenciación Celular/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Femenino , Humanos , Células Madre Mesenquimatosas/enzimología , Ratones , Ratones Noqueados , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Naftalimidas/farmacología , Osteoblastos/citología , Osteoclastos/citología , Osteoporosis/tratamiento farmacológico , Osteoporosis/enzimología , Osteoporosis/genética , Ovariectomía
13.
Trends Mol Med ; 19(2): 83-8, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23332598

RESUMEN

The androgen receptor (AR) is pivotal in the biology of sex hormone-regulated malignancies, with prostate cancer (PC) the most affected tumor. AR signals control the growth, survival, and migration of cancer cells, and they regulate the activation of macrophages, a cell type pivotal to the tumor ecosystem. Intriguingly, CaMKK2 has recently been identified as both an important AR-regulated gene in the context of PC and as a critical regulator of macrophage activation. By contrast, CaMKK2 is barely detectable in normal prostate or immune cells that mediate the response against tumorigenesis. These novel findings suggest that CaMKK2 resides at a critical molecular node that shapes the cancer ecosystem, and identifies this kinase as a novel therapeutic target for sex hormone-regulated cancers.


Asunto(s)
Andrógenos/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Humanos , Activación de Macrófagos , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Transducción de Señal
14.
PLoS One ; 8(1): e54156, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23326591

RESUMEN

The Toll-like receptors represent a largely evolutionarily conserved pathogen recognition machinery responsible for recognition of bacterial, fungal, protozoan, and viral pathogen associated microbial patterns and initiation of inflammatory response. Structurally the Toll-like receptors are comprised of an extracellular leucine rich repeat domain and a cytoplasmic Toll/Interleukin 1 receptor domain. Recognition takes place in the extracellular domain where as the cytoplasmic domain triggers a complex signal network required to sustain appropriate immune response. Signal transduction is regulated by the recruitment of different intracellular adaptors. The Toll-like receptors can be grouped depending on the usage of the adaptor, MyD88, into MyD88-dependent and MyD88 independent subsets. Herein, we present a unique phylogenetic analysis of domain regions of these receptors and their cognate signaling adaptor molecules. Although previously unclear from the phylogeny of full length receptors, these analyses indicate a separate evolutionary origin for the MyD88-dependent and MyD88-independent signaling pathway and provide evidence of a common ancestor for the vertebrate and invertebrate orthologs of the adaptor molecule MyD88. Together these observations suggest a very ancient origin of the MyD88-dependent pathway Additionally we show that early duplications gave rise to several adaptor molecule families. In some cases there is also strong pattern of parallel duplication between adaptor molecules and their corresponding TLR. Our results further support the hypothesis that phylogeny of specific domains involved in signaling pathway can shed light on key processes that link innate to adaptive immune response.


Asunto(s)
Evolución Molecular , Factor 88 de Diferenciación Mieloide , Filogenia , Receptores Toll-Like , Adaptación Fisiológica/genética , Adaptación Fisiológica/inmunología , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Duplicación de Gen , Inmunidad Innata/genética , Inmunidad Innata/fisiología , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/metabolismo , Transducción de Señal/genética , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
15.
J Biol Chem ; 287(38): 31658-65, 2012 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-22778263

RESUMEN

Many cellular Ca(2+)-dependent signaling cascades utilize calmodulin (CaM) as the intracellular Ca(2+) receptor. Ca(2+)/CaM binds and activates a plethora of enzymes, including CaM kinases (CaMKs). CaMKK2 is one of the most versatile of the CaMKs and will phosphorylate and activate CaMKI, CaMKIV, and AMP-activated protein kinase. Cell expression of CaMKK2 is limited, yet CaMKK2 is involved in regulating many important physiological and pathophysiological processes, including energy balance, adiposity, glucose homeostasis, hematopoiesis, inflammation, and cancer. Here, we explore known functions of CaMKK2 and discuss its potential as a target for therapeutic intervention.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Regulación de la Expresión Génica , Tejido Adiposo/enzimología , Adiposidad , Animales , Femenino , Glucosa/metabolismo , Homeostasis , Humanos , Inflamación/metabolismo , Hígado/enzimología , Macrófagos/metabolismo , Masculino , Ratones , Neuronas/metabolismo , Fosforilación , Neoplasias de la Próstata/enzimología , Ratas , Transducción de Señal , Distribución Tisular
16.
J Biol Chem ; 287(14): 11579-91, 2012 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-22334678

RESUMEN

Calcium/calmodulin-dependent kinase kinase 2 (CaMKK2) plays a key role in regulating food intake and energy expenditure at least in part by its actions in hypothalamic neurons. Previously, we showed that loss of CaMKK2 protected mice from high-fat diet (HFD)-induced obesity and glucose intolerance. However, although pair feeding HFD to WT mice to match food consumption of CAMKK2-null mice slowed weight gain, it failed to protect from glucose intolerance. Here we show that relative to WT mice, HFD-fed CaMKK2-null mice are protected from inflammation in adipose and remain glucose-tolerant. Moreover, loss of CaMKK2 also protected mice from endotoxin shock and fulminant hepatitis. We explored the expression of CaMKK2 in immune cells and found it to be restricted to those of the monocyte/macrophage lineage. CaMKK2-null macrophages exhibited a remarkable deficiency to spread, phagocytose bacteria, and synthesize cytokines in response to the Toll-like receptor 4 (TLR4) agonist lipopolysaccharide (LPS). Mechanistically, loss of CaMKK2 uncoupled the TLR4 cascade from activation of protein tyrosine kinase 2 (PYK2; also known as PTK2B). Our findings uncover an important function for CaMKK2 in mediating mechanisms that control the amplitude of macrophage inflammatory responses to excess nutrients or pathogen derivatives.


Asunto(s)
Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Macrófagos/metabolismo , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/deficiencia , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/genética , Adhesión Celular/efectos de los fármacos , Quimiocinas/biosíntesis , Dieta Alta en Grasa/efectos adversos , Activación Enzimática/efectos de los fármacos , Quinasa 2 de Adhesión Focal/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Técnicas de Inactivación de Genes , Intolerancia a la Glucosa/etiología , Intolerancia a la Glucosa/prevención & control , Hepatitis/etiología , Hepatitis/prevención & control , Humanos , Inflamación/enzimología , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Lipopolisacáridos/farmacología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Choque Séptico/prevención & control , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/metabolismo
17.
PLoS One ; 7(1): e29808, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22238658

RESUMEN

BACKGROUND: Pin1 has previously been described to regulate cells that participate in both innate and adaptive immunity. Thus far, however, no role for Pin1 has been described in modulating conventional dendritic cells, innate antigen presenting cells that potently activate naïve T cells, thereby bridging innate and adaptive immune responses. METHODOLOGY/PRINCIPAL FINDINGS: When challenged with LPS, Pin1-null mice failed to accumulate spleen conventional dendritic cells (cDC). Analysis of steady-state spleen DC populations revealed that Pin1-null mice had fewer CD8+ cDC. This defect was recapitulated by culturing Pin1-null bone marrow with the DC-instructive cytokine Flt3 Ligand. Additionally, injection of Flt3 Ligand for 9 days failed to induce robust expansion of CD8+ cDC in Pin1-null mice. Upon infection with Listeria monocytogenes, Pin1-null mice were defective in stimulating proliferation of adoptively transferred WT CD8+ T cells, suggesting that decreases in Pin1 null CD8+ cDC may affect T cell responses to infection in vivo. Finally, upon analyzing expression of proteins involved in DC development, elevated expression of PU.1 was detected in Pin1-null cells, which resulted from an increase in PU.1 protein half-life. CONCLUSIONS/SIGNIFICANCE: We have identified a novel role for Pin1 as a modulator of CD8+ cDC development. Consistent with reduced numbers of CD8+ cDC in Pin1-null mice, we find that the absence of Pin1 impairs CD8+ T cell proliferation in response to infection with Listeria monocytogenes. These data suggest that, via regulation of CD8+ cDC production, Pin1 may serve as an important modulator of adaptive immunity.


Asunto(s)
Antígenos CD8/metabolismo , Diferenciación Celular/genética , Células Dendríticas/fisiología , Isomerasa de Peptidilprolil/fisiología , Inmunidad Adaptativa/efectos de los fármacos , Inmunidad Adaptativa/genética , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/fisiología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Lipopolisacáridos/farmacología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Peptidilprolil Isomerasa de Interacción con NIMA , Isomerasa de Peptidilprolil/genética , Bazo/citología , Bazo/efectos de los fármacos , Bazo/metabolismo , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Células Madre/fisiología
19.
J Leukoc Biol ; 90(5): 897-909, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21816924

RESUMEN

Granulocytes serve a critical function in host organisms by recognizing and destroying invading microbes, as well as propagating and maintaining inflammation at sites of infection. However, the molecular pathways underpinning the development of granulocytes are poorly understood. Here, we identify a role for CaMKK2 in the restriction of granulocytic fate commitment and differentiation of myeloid progenitor cells. Following BMT, engraftment by Camkk2(-/-) donor cells resulted in the increased production of mature granulocytes in the BM and peripheral blood. Similarly, Camkk2(-/-) mice possessed elevated numbers of CMP cells and exhibited an accelerated granulopoietic phenotype in the BM. Camkk2(-/-) myeloid progenitors expressed increased levels of C/EBPα and PU.1 and preferentially differentiated into Gr1(+)Mac1(+) granulocytes and CFU-G in vitro. During normal granulopoiesis in vivo or G-CSF-induced differentiation of 32D myeloblast cells in vitro, CaMKK2 mRNA and protein were decreased as a function of time and were undetectable in mature granulocytes. Expression of ectopic CaMKK2 in Camkk2(-/-) CMPs was sufficient to rescue aberrant granulocyte differentiation and when overexpressed in 32D cells, was also sufficient to impede granulocyte differentiation in a kinase activity-dependent manner. Collectively, our results reveal a novel role for CaMKK2 as an inhibitor of granulocytic fate commitment and differentiation in early myeloid progenitors.


Asunto(s)
Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/fisiología , Linaje de la Célula , Granulocitos , Células Progenitoras Mieloides , Animales , Trasplante de Médula Ósea , Diferenciación Celular , Línea Celular , Microambiente Celular , Técnicas de Cocultivo , Granulocitos/citología , Granulocitos/inmunología , Granulocitos/metabolismo , Antígenos Comunes de Leucocito , Ratones , Células Progenitoras Mieloides/citología , Células Progenitoras Mieloides/inmunología , Células Progenitoras Mieloides/metabolismo , Células del Estroma , Irradiación Corporal Total
20.
Nat Immunol ; 12(8): 786-95, 2011 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-21743478

RESUMEN

Here we have used a systems biology approach to study innate and adaptive responses to vaccination against influenza in humans during three consecutive influenza seasons. We studied healthy adults vaccinated with trivalent inactivated influenza vaccine (TIV) or live attenuated influenza vaccine (LAIV). TIV induced higher antibody titers and more plasmablasts than LAIV did. In subjects vaccinated with TIV, early molecular signatures correlated with and could be used to accurately predict later antibody titers in two independent trials. Notably, expression of the kinase CaMKIV at day 3 was inversely correlated with later antibody titers. Vaccination of CaMKIV-deficient mice with TIV induced enhanced antigen-specific antibody titers, which demonstrated an unappreciated role for CaMKIV in the regulation of antibody responses. Thus, systems approaches can be used to predict immunogenicity and provide new mechanistic insights about vaccines.


Asunto(s)
Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/inmunología , Gripe Humana/inmunología , Gripe Humana/prevención & control , Orthomyxoviridae/inmunología , Inmunidad Adaptativa/inmunología , Adolescente , Adulto , Animales , Anticuerpos Antivirales/sangre , Perfilación de la Expresión Génica , Pruebas de Inhibición de Hemaglutinación , Humanos , Inmunidad Innata/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Estaciones del Año , Biología de Sistemas/métodos , Vacunación/métodos , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/inmunología , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...