Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Nature ; 523(7561): 431-436, 2015 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-26176913

RESUMEN

Traumatic brain injury (TBI), characterized by acute neurological dysfunction, is one of the best known environmental risk factors for chronic traumatic encephalopathy and Alzheimer's disease, the defining pathologic features of which include tauopathy made of phosphorylated tau protein (P-tau). However, tauopathy has not been detected in the early stages after TBI, and how TBI leads to tauopathy is unknown. Here we find robust cis P-tau pathology after TBI in humans and mice. After TBI in mice and stress in vitro, neurons acutely produce cis P-tau, which disrupts axonal microtubule networks and mitochondrial transport, spreads to other neurons, and leads to apoptosis. This process, which we term 'cistauosis', appears long before other tauopathy. Treating TBI mice with cis antibody blocks cistauosis, prevents tauopathy development and spread, and restores many TBI-related structural and functional sequelae. Thus, cis P-tau is a major early driver of disease after TBI and leads to tauopathy in chronic traumatic encephalopathy and Alzheimer's disease. The cis antibody may be further developed to detect and treat TBI, and prevent progressive neurodegeneration after injury.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Lesiones Encefálicas/patología , Lesiones Encefálicas/prevención & control , Tauopatías/prevención & control , Proteínas tau/antagonistas & inhibidores , Proteínas tau/química , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/prevención & control , Animales , Anticuerpos Monoclonales/uso terapéutico , Afinidad de Anticuerpos , Axones/metabolismo , Axones/patología , Encéfalo/metabolismo , Encéfalo/patología , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/metabolismo , Modelos Animales de Enfermedad , Epítopos/química , Epítopos/inmunología , Femenino , Humanos , Masculino , Ratones , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/biosíntesis , Fosfoproteínas/inmunología , Fosfoproteínas/toxicidad , Estrés Fisiológico , Tauopatías/complicaciones , Tauopatías/metabolismo , Tauopatías/patología , Proteínas tau/biosíntesis , Proteínas tau/inmunología , Proteínas tau/toxicidad
2.
Mol Cell ; 46(6): 771-83, 2012 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-22608923

RESUMEN

Fbw7 is the substrate recognition component of the Skp1-Cullin-F-box (SCF)-type E3 ligase complex and a well-characterized tumor suppressor that targets numerous oncoproteins for destruction. Genomic deletion or mutation of FBW7 has been frequently found in various types of human cancers; however, little is known about the upstream signaling pathway(s) governing Fbw7 stability and cellular functions. Here we report that Fbw7 protein destruction and tumor suppressor function are negatively regulated by the prolyl isomerase Pin1. Pin1 interacts with Fbw7 in a phoshorylation-dependent manner and promotes Fbw7 self-ubiquitination and protein degradation by disrupting Fbw7 dimerization. Consequently, overexpressing Pin1 reduces Fbw7 abundance and suppresses Fbw7's ability to inhibit proliferation and transformation. By contrast, depletion of Pin1 in cancer cells leads to elevated Fbw7 expression, which subsequently reduces Mcl-1 abundance, sensitizing cancer cells to Taxol. Thus, Pin1-mediated inhibition of Fbw7 contributes to oncogenesis, and Pin1 may be a promising drug target for anticancer therapy.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteínas F-Box/genética , Regulación de la Expresión Génica , Isomerasa de Peptidilprolil/metabolismo , Ubiquitina-Proteína Ligasas/genética , Secuencia de Aminoácidos , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proteínas F-Box/antagonistas & inhibidores , Proteínas F-Box/metabolismo , Proteína 7 que Contiene Repeticiones F-Box-WD , Genes Supresores de Tumor , Humanos , Datos de Secuencia Molecular , Peptidilprolil Isomerasa de Interacción con NIMA , Isomerasa de Peptidilprolil/genética , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
3.
Mol Cell ; 42(2): 147-59, 2011 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-21497122

RESUMEN

Pin1 is a phospho-specific prolyl isomerase that regulates numerous key signaling molecules and whose deregulation contributes to disease notably cancer. However, since prolyl isomerases are often believed to be constitutively active, little is known whether and how Pin1 catalytic activity is regulated. Here, we identify death-associated protein kinase 1 (DAPK1), a known tumor suppressor, as a kinase responsible for phosphorylation of Pin1 on Ser71 in the catalytic active site. Such phosphorylation fully inactivates Pin1 catalytic activity and inhibits its nuclear location. Moreover, DAPK1 inhibits the ability of Pin1 to induce centrosome amplification and cell transformation. Finally, Pin1 pSer71 levels are positively correlated with DAPK1 levels and negatively with centrosome amplification in human breast cancer. Thus, phosphorylation of Pin1 Ser71 by DAPK1 inhibits its catalytic activity and cellular function, providing strong evidence for an essential role of the Pin1 enzymatic activity for its cellular function.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Neoplasias de la Mama/enzimología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Isomerasa de Peptidilprolil/metabolismo , Transducción de Señal , Transporte Activo de Núcleo Celular , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Quinasas Dependientes de Calcio-Calmodulina/genética , Dominio Catalítico , Ciclo Celular , Núcleo Celular/enzimología , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Centrosoma/metabolismo , Proteínas Quinasas Asociadas a Muerte Celular , Estabilidad de Enzimas , Femenino , Células HeLa , Humanos , Inmunohistoquímica , Ratones , Ratones Noqueados , Microscopía Fluorescente , Mutación , Células 3T3 NIH , Peptidilprolil Isomerasa de Interacción con NIMA , Isomerasa de Peptidilprolil/antagonistas & inhibidores , Isomerasa de Peptidilprolil/genética , Fosforilación , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas , Proteínas Recombinantes de Fusión/metabolismo , Serina , Factores de Tiempo , Análisis de Matrices Tisulares , Transfección
4.
Hum Mol Genet ; 25(12): 2498-2513, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27094130

RESUMEN

Extracellular deposition of amyloid-beta (Aß) peptide, a metabolite of sequential cleavage of amyloid precursor protein (APP), is a critical step in the pathogenesis of Alzheimer's disease (AD). While death-associated protein kinase 1 (DAPK1) is highly expressed in AD brains and its genetic variants are linked to AD risk, little is known about the impact of DAPK1 on APP metabolism and Aß generation. In this study, we demonstrated a novel effect of DAPK1 in the regulation of APP processing using cell culture and mouse models. DAPK1, but not its kinase deficient mutant (K42A), significantly increased human Aß secretion in neuronal cell culture models. Moreover, knockdown of DAPK1 expression or inhibition of DAPK1 catalytic activity significantly decreased Aß secretion. Furthermore, DAPK1, but not K42A, triggered Thr668 phosphorylation of APP, which may initiate and facilitate amyloidogenic APP processing leading to the generation of Aß. In Tg2576 APPswe-overexpressing mice, knockout of DAPK1 shifted APP processing toward non-amyloidogenic pathway and decreased Aß generation. Finally, in AD brains, elevated DAPK1 levels showed co-relation with the increase of APP phosphorylation. Combined together, these results suggest that DAPK1 promotes the phosphorylation and amyloidogenic processing of APP, and that may serve a potential therapeutic target for AD.


Asunto(s)
Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Proteínas Quinasas Asociadas a Muerte Celular/genética , Neuronas/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/biosíntesis , Animales , Encéfalo/metabolismo , Encéfalo/patología , Proteínas Quinasas Asociadas a Muerte Celular/antagonistas & inhibidores , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Ratones , Neuronas/patología , Fosforilación
5.
Neurobiol Dis ; 76: 13-23, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25576397

RESUMEN

The unique proline isomerase Pin1 is pivotal for protecting against age-dependent neurodegeneration in Alzheimer's disease (AD), with its inhibition providing a molecular link between tangle and plaque pathologies. Pin1 is oxidatively modified in human AD brains, but little is known about its regulatory mechanisms and pathological significance of such Pin1 modification. In this paper, our determination of crystal structures of oxidized Pin1 reveals a series of Pin1 oxidative modifications on Cys113 in a sequential fashion. Cys113 oxidization is further confirmed by generating antibodies specifically recognizing oxidized Cys113 of Pin1. Furthermore, Pin1 oxidation on Cys113 inactivates its catalytic activity in vitro, and Ala point substitution of Cys113 inactivates the ability of Pin1 to isomerize tau as well as to promote protein turnover of tau and APP. Moreover, redox regulation affects Pin1 subcellular localization and Pin1-mediated neuronal survival in response to hypoxia treatment. Importantly, Cys113-oxidized Pin1 is significantly increased in human AD brain comparing to age-matched controls. These results not only identify a novel Pin1 oxidation site to be the critical catalytic residue Cys113, but also provide a novel oxidative regulation mechanism for inhibiting Pin1 activity in AD. These results suggest that preventing Pin1 oxidization might help to reduce the risk of AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Isomerasa de Peptidilprolil/química , Isomerasa de Peptidilprolil/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Anticuerpos , Dominio Catalítico , Línea Celular Tumoral , Hipocampo/metabolismo , Humanos , Peptidilprolil Isomerasa de Interacción con NIMA , Oxidación-Reducción , Isomerasa de Peptidilprolil/inmunología , Proteínas tau/metabolismo
6.
Circ Res ; 112(9): 1244-52, 2013 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-23487407

RESUMEN

RATIONALE: Cardiac hypertrophy results from the complex interplay of differentially regulated cascades based on the phosphorylation status of involved signaling molecules. Although numerous critical regulatory kinases and phosphatases have been identified in the myocardium, the intracellular mechanism for temporal regulation of signaling duration and intensity remains obscure. In the nonmyocyte context, control of folding, activity, and stability of proteins is mediated by the prolyl isomerase Pin1, but the role of Pin1 in the heart is unknown. OBJECTIVE: To establish the role of Pin1 in the heart. METHODS AND RESULTS: Here, we show that either genetic deletion or cardiac overexpression of Pin1 blunts hypertrophic responses induced by transaortic constriction and consequent cardiac failure in vivo. Mechanistically, we find that Pin1 directly binds to Akt, mitogen activated protein kinase (MEK), and Raf-1 in cultured cardiomyocytes after hypertrophic stimulation. Furthermore, loss of Pin1 leads to diminished hypertrophic signaling of Akt and MEK, whereas overexpression of Pin1 increases Raf-1 phosphorylation on the autoinhibitory site Ser259, leading to reduced MEK activation. CONCLUSIONS: Collectively, these data support a role for Pin1 as a central modulator of the intensity and duration of 2 major hypertrophic signaling pathways, thereby providing a novel target for regulation and control of cardiac hypertrophy.


Asunto(s)
Cardiomegalia/enzimología , Miocitos Cardíacos/enzimología , Isomerasa de Peptidilprolil/metabolismo , Transducción de Señal , Animales , Cardiomegalia/diagnóstico por imagen , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Cardiomegalia/prevención & control , Dependovirus/genética , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Miocitos Cardíacos/patología , Peptidilprolil Isomerasa de Interacción con NIMA , Isomerasa de Peptidilprolil/deficiencia , Isomerasa de Peptidilprolil/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Ratas , Factores de Tiempo , Transducción Genética , Transfección , Ultrasonografía , Quinasas raf/metabolismo
7.
EMBO J ; 29(10): 1748-61, 2010 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-20389280

RESUMEN

Death-associated protein kinase (DAPK) was identified as a mediator of interferon (IFN)-induced cell death. How IFN controls DAPK activation remains largely unknown. Here, we identify the BTB-Kelch protein KLHL20 as a negative regulator of DAPK. KLHL20 binds DAPK and Cullin 3 (Cul3) via its Kelch-repeat domain and BTB domain, respectively. The KLHL20-Cul3-ROC1 E3 ligase complex promotes DAPK polyubiquitination, thereby inducing the proteasomal degradation of DAPK. Accordingly, depletion of KLHL20 diminishes DAPK ubiquitination and degradation. The KLHL20-mediated DAPK ubiquitination is suppressed in cells receiving IFN-alpha or IFN-gamma, which induces an enrichment/sequestration of KLHL20 in the PML nuclear bodies, thereby separating KLHL20 from DAPK. Consequently, IFN triggers the stabilization of DAPK. This mechanism of DAPK stabilization is crucial for determining IFN responsiveness of tumor cells and contributes to IFN-induced autophagy. This study identifies KLHL20-Cul3-ROC1 as an E3 ligase for DAPK ubiquitination and reveals a regulatory mechanism of DAPK, through blocking its accessibility to this E3 ligase, in IFN-induced apoptotic and autophagic death. Our findings may be relevant to the problem of IFN resistance in cancer therapy.


Asunto(s)
Proteínas Portadoras/química , Proteínas Cullin/química , Regulación de la Expresión Génica , Interferones/química , Ubiquitina/química , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Autofagia , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Proteínas Quinasas Asociadas a Muerte Celular , Células HeLa , Humanos , Interferones/metabolismo , Ratones , Modelos Biológicos , Células 3T3 NIH , Neoplasias/terapia , Fenotipo , Ubiquitina-Proteína Ligasas/metabolismo
8.
Sci Transl Med ; 13(577)2021 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-33472951

RESUMEN

Angiotensin converting enzyme 2 (ACE2) is an enzyme that belongs to the renin-angiotensin system (RAS) and antagonizes the classical angiotensin (Ang) II/angiotensin II receptor type 1 (AT1) receptor pathway. Here, we report that higher ACE2 expression correlates with better overall survival in patients with clear cell renal cell carcinoma (ccRCC). Moreover, ACE2 has inhibitory effects on tumor proliferation in ccRCC in vitro and in preclinical animal models of ccRCC. We further show that Ang-(1-7), a heptapeptide generated by ACE2, is the likely mediator of this effect. Vascular endothelial growth factor receptor-tyrosine kinase inhibitor (VEGFR-TKI) treatment of ccRCC xenografts decreased ACE2 expression, and combination treatment with VEGFR-TKI and Ang-(1-7) generated additive suppression of tumor growth and improved survival outcomes. Last, the addition of Ang-(1-7) to programmed death-ligand 1 (PD-L1) pathway inhibitor and VEGFR-TKI showed further growth suppression in an immunocompetent RCC model. Together, these results suggest that targeting the ACE2/Ang-(1-7) axis is a promising therapeutic strategy against ccRCC.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Angiotensina I , Angiotensina II , Enzima Convertidora de Angiotensina 2 , Animales , Carcinoma de Células Renales/tratamiento farmacológico , Humanos , Neoplasias Renales/tratamiento farmacológico , Fragmentos de Péptidos , Peptidil-Dipeptidasa A , Inhibidores de Proteínas Quinasas , Factor A de Crecimiento Endotelial Vascular
9.
Cancer Immunol Res ; 9(2): 156-169, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33229411

RESUMEN

Blockade of the PD1 pathway is a broadly effective cancer therapy, but additional immune-inhibitory pathways contribute to tumor immune evasion. HERV-H LTR-associating 2 (HHLA2; also known as B7H5 and B7H7) is a member of the B7 family of immunoregulatory ligands that mediates costimulatory effects through its interaction with the CD28 family member transmembrane and immunoglobulin domain containing 2 (TMIGD2). However, HHLA2 has also been known to have inhibitory effects on T cells. Here, we report that we have identified killer cell immunoglobulin-like receptor, three immunoglobulin domains and long cytoplasmic tail 3 (KIR3DL3) as an inhibitory receptor for HHLA2 in T cells and natural killer (NK) cells and have generated HHLA2 and KIR3DL3 antibodies that block the immune-inhibitory activity of HHLA2, preserving the costimulatory signal. It is known that HHLA2 is frequently expressed in several tumor types, including clear cell renal cell carcinoma (ccRCC). We found that HHLA2 expression was nonoverlapping with PDL1 expression in ccRCC, suggesting that HHLA2 mediates a mechanism of tumor immune evasion that is independent from PDL1. Blockade of both the PD1 and KIR3DL3 pathways may be a more effective way to reverse tumor immune evasion.See related Spotlight on p. 128.


Asunto(s)
Antígeno B7-H1/metabolismo , Carcinoma de Células Renales/inmunología , Inmunoglobulinas/metabolismo , Neoplasias Renales/inmunología , Receptores KIR/metabolismo , Animales , Antígeno B7-H1/inmunología , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Humanos , Inmunoglobulinas/inmunología , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Células Asesinas Naturales/inmunología , Ratones , Ratones Endogámicos BALB C , Receptores KIR/inmunología , Linfocitos T/inmunología
10.
Cancer Res ; 80(14): 3033-3045, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32193285

RESUMEN

PARP inhibitor monotherapies are effective to treat patients with breast, ovary, prostate, and pancreatic cancer with BRCA1 mutations, but not to the much more frequent BRCA wild-type cancers. Searching for strategies that would extend the use of PARP inhibitors to BRCA1-proficient tumors, we found that the stability of BRCA1 protein following ionizing radiation (IR) is maintained by postphosphorylational prolyl-isomerization adjacent to Ser1191 of BRCA1, catalyzed by prolyl-isomerase Pin1. Extinction of Pin1 decreased homologous recombination (HR) to the level of BRCA1-deficient cells. Pin1 stabilizes BRCA1 by preventing ubiquitination of Lys1037 of BRCA1. Loss of Pin1, or introduction of a BRCA1-mutant refractory to Pin1 binding, decreased the ability of BRCA1 to localize to repair foci and augmented IR-induced DNA damage. In vitro growth of HR-proficient breast, prostate, and pancreatic cancer cells were modestly repressed by olaparib or Pin1 inhibition using all-trans retinoic acid (ATRA), while combination treatment resulted in near-complete block of cell proliferation. In MDA-MB-231 xenografts and triple-negative breast cancer patient-derived xenografts, either loss of Pin1 or ATRA treatment reduced BRCA1 expression and sensitized breast tumors to olaparib. Together, our study reveals that Pin1 inhibition, with clinical widely used ATRA, acts as an effective HR disrupter that sensitizes BRCA1-proficient tumors to PARP inhibition. SIGNIFICANCE: PARP inhibitors have been limited to treat homologous recombination-deficient tumors. All-trans retinoic acid, by inhibiting Pin1 and destabilizing BRCA1, extends benefit of PARP inhibitors to patients with homologous recombination-proficient tumors.See related commentary by Cai, p. 2977.


Asunto(s)
Inhibidores de Poli(ADP-Ribosa) Polimerasas , Neoplasias de la Mama Triple Negativas , Proteína BRCA1/genética , Línea Celular Tumoral , Femenino , Humanos , Masculino , Peptidilprolil Isomerasa de Interacción con NIMA/genética , Isomerasa de Peptidilprolil , Ftalazinas/farmacología , Ftalazinas/uso terapéutico , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética
11.
Oncogenesis ; 8(3): 15, 2019 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-30796200

RESUMEN

Several therapeutic options are available for metastatic RCC, but responses are almost never complete, and resistance to therapy develops in the vast majority of patients. Consequently, novel treatments are needed to combat resistance to current therapies and to improve patient outcomes. We have applied integrated transcriptome and proteome analyses to identify cathepsin B (CTSB), a cysteine proteinase of the papain family, as one of the most highly upregulated gene products in established human RCC xenograft models of resistance to vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitors (TKI). We used established RCC models to test the significance of CTSB in the progression of renal cancer. Our evaluation of CTSB showed that stable CTSB knockdown suppressed RCC growth in vitro and in vivo. Stable over-overexpression of wild-type CTSB (CTSBwt/hi), but not of an CTSB active site mutant (CTSBN298A), rescued cell growth in CTSB knockdown cells and abolished the efficacy of VEGFR TKI treatment. Genome-wide transcriptome profiling of CTSB knockdown cells demonstrated significant effects on multiple metabolic and stem cell-related pathways, with ALDHA1A (ALDH1) as one of the most significantly downregulated genes. Importantly, survival analysis across 16 major TCGA cancers revealed that CTSB overexpression is associated with low rates of three and five year patient survival rates (P = 2.5e-08, HR = 1.4). These data strongly support a contribution of CTSB activity to RCC cell growth and tumorigenicity. They further highlight the promise of CTSB inhibition in development of novel combination therapies designed to improve efficacy of current TKI treatments of metastatic RCC.

13.
Nat Commun ; 9(1): 3069, 2018 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-30093655

RESUMEN

Arsenic trioxide (ATO) and all-trans retinoic acid (ATRA) combination safely cures fatal acute promyelocytic leukemia, but their mechanisms of action and efficacy are not fully understood. ATRA inhibits leukemia, breast, and liver cancer by targeting isomerase Pin1, a master regulator of oncogenic signaling networks. Here we show that ATO targets Pin1 and cooperates with ATRA to exert potent anticancer activity. ATO inhibits and degrades Pin1, and suppresses its oncogenic function by noncovalent binding to Pin1's active site. ATRA increases cellular ATO uptake through upregulating aquaporin-9. ATO and ATRA, at clinically safe doses, cooperatively ablate Pin1 to block numerous cancer-driving pathways and inhibit the growth of triple-negative breast cancer cells and tumor-initiating cells in cell and animal models including patient-derived orthotopic xenografts, like Pin1 knockout, which is substantiated by comprehensive protein and microRNA analyses. Thus, synergistic targeting of Pin1 by ATO and ATRA offers an attractive approach to combating breast and other cancers.


Asunto(s)
Trióxido de Arsénico/farmacología , Regulación Neoplásica de la Expresión Génica , Peptidilprolil Isomerasa de Interacción con NIMA/metabolismo , Neoplasias/metabolismo , Tretinoina/metabolismo , Animales , Antineoplásicos/farmacología , Proliferación Celular , Femenino , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Humanos , Leucemia Promielocítica Aguda/metabolismo , Espectroscopía de Resonancia Magnética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Trasplante de Neoplasias , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Proteómica , Transducción de Señal
14.
Nat Commun ; 9(1): 1603, 2018 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-29686383

RESUMEN

Interleukin 33 (IL-33) is among the earliest-released cytokines in response to allergens that orchestrate type 2 immunity. The prolyl cis-trans isomerase PIN1 is known to induce cytokines for eosinophil survival and activation by stabilizing cytokines mRNAs, but the function of PIN1 in upstream signaling pathways in asthma is unknown. Here we show that interleukin receptor associated kinase M (IRAK-M) is a PIN1 target critical for IL-33 signaling in allergic asthma. NMR analysis and docking simulations suggest that PIN1 might regulate IRAK-M conformation and function in IL-33 signaling. Upon IL-33-induced airway inflammation, PIN1 is activated for binding with and isomerization of IRAK-M, resulting in IRAK-M nuclear translocation and induction of selected proinflammatory genes in dendritic cells. Thus, the IL-33-PIN1-IRAK-M is an axis critical for dendritic cell activation, type 2 immunity and IL-33 induced airway inflammation.


Asunto(s)
Asma/inmunología , Inmunidad Celular , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Interleucina-33/inmunología , Peptidilprolil Isomerasa de Interacción con NIMA/metabolismo , Células Th2/inmunología , Adulto , Animales , Antígenos Dermatofagoides/inmunología , Asma/sangre , Asma/patología , Pruebas de Provocación Bronquial , Líquido del Lavado Bronquioalveolar/citología , Línea Celular , Modelos Animales de Enfermedad , Eosinófilos/inmunología , Femenino , Células HEK293 , Humanos , Quinasas Asociadas a Receptores de Interleucina-1/química , Quinasas Asociadas a Receptores de Interleucina-1/genética , Interleucina-33/metabolismo , Pulmón/inmunología , Pulmón/patología , Masculino , Ratones , Ratones Noqueados , Simulación del Acoplamiento Molecular , Peptidilprolil Isomerasa de Interacción con NIMA/química , Peptidilprolil Isomerasa de Interacción con NIMA/genética , Cultivo Primario de Células , Dominios Proteicos , Transducción de Señal/inmunología , Células Th2/metabolismo , Adulto Joven
15.
Cell Death Differ ; 24(2): 238-250, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28141794

RESUMEN

Death-associated protein kinase 1 (DAPK1) has been shown to have important roles in neuronal cell death in several model systems and has been implicated in multiple diseases, including Alzheimer's disease (AD). However, little is known about the molecular mechanisms by which DAPK1 signals neuronal cell death. In this study, N-myc downstream-regulated gene 2 (NDRG2) was identified as a novel substrate of DAPK1 using phospho-peptide library screening. DAPK1 interacted with NDRG2 and directly phosphorylated the Ser350 residue in vitro and in vivo. Moreover, DAPK1 overexpression increased neuronal cell death through NDRG2 phosphorylation after ceramide treatment. In contrast, inhibition of DAPK1 by overexpression of a DAPK1 kinase-deficient mutant and small hairpin RNA, or by treatment with a DAPK1 inhibitor significantly decreased neuronal cell death, and abolished NDRG2 phosphorylation in cell culture and in primary neurons. Furthermore, NDRG2-mediated cell death by DAPK1 was required for a caspase-dependent poly-ADP-ribose polymerase cleavage. In addition, DAPK1 ablation suppressed ceramide-induced cell death in mouse brain and neuronal cell death in Tg2576 APPswe-overexpressing mice. Finally, levels of phosphorylated NDRG2 Ser350 and DAPK1 were significantly increased in human AD brain samples. Thus, phosphorylation of NDRG2 on Ser350 by DAPK1 is a novel mechanism activating NDRG2 function and involved in neuronal cell death regulation in vivo.


Asunto(s)
Proteínas Quinasas Asociadas a Muerte Celular/metabolismo , Proteínas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Clorometilcetonas de Aminoácidos/farmacología , Animales , Apoptosis/efectos de los fármacos , Encéfalo/metabolismo , Caspasa 3/química , Caspasa 3/metabolismo , Células Cultivadas , Ceramidas/farmacología , Proteínas Quinasas Asociadas a Muerte Celular/antagonistas & inhibidores , Proteínas Quinasas Asociadas a Muerte Celular/genética , Células HEK293 , Células HeLa , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/citología , Neuronas/metabolismo , Fosforilación/efectos de los fármacos , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas/antagonistas & inhibidores , Proteínas/genética , Interferencia de ARN
16.
Cell Rep ; 11(1): 111-24, 2015 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-25818297

RESUMEN

Proline-directed phosphorylation is regulated by the prolyl isomerase Pin1, which plays a fundamental role in driving breast cancer stem-like cells (BCSCs). Rab2A is a small GTPase critical for vesicle trafficking. Here, we show that Pin1 increases Rab2A transcription to promote BCSC expansion and tumorigenesis in vitro and in vivo. Mechanistically, Rab2A directly interacts with and prevents dephosphorylation/inactivation of Erk1/2 by the MKP3 phosphatase, resulting in Zeb1 upregulation and ß-catenin nuclear translocation. In cancer cells, Rab2A is activated via gene amplification, mutation or Pin1 overexpression. Rab2A overexpression or mutation endows BCSC traits to primary normal human breast epithelial cells, whereas silencing Rab2A potently inhibits the expansion and tumorigenesis of freshly isolated BCSCs. Finally, Rab2A overexpression correlates with poor clinical outcome in breast cancer patients. Thus, Pin1/Rab2A/Erk drives BCSC expansion and tumorigenicity, suggesting potential drug targets.


Asunto(s)
Neoplasias de la Mama/genética , Sistema de Señalización de MAP Quinasas/genética , Isomerasa de Peptidilprolil/biosíntesis , Proteínas de Unión al GTP rab/genética , Neoplasias de la Mama/patología , Carcinogénesis/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Peptidilprolil Isomerasa de Interacción con NIMA , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Isomerasa de Peptidilprolil/genética , Activación Transcripcional/genética , Proteínas de Unión al GTP rab/biosíntesis
17.
Cell Rep ; 13(4): 812-828, 2015 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-26489457

RESUMEN

Axon guidance relies on precise translation of extracellular signal gradients into local changes in cytoskeletal dynamics, but the molecular mechanisms regulating dose-dependent responses of growth cones are still poorly understood. Here, we show that during embryonic development in growing axons, a low level of Semaphorin3A stimulation is buffered by the prolyl isomerase Pin1. We demonstrate that Pin1 stabilizes CDK5-phosphorylated CRMP2A, the major isoform of CRMP2 in distal axons. Consequently, Pin1 knockdown or knockout reduces CRMP2A levels specifically in distal axons and inhibits axon growth, which can be fully rescued by Pin1 or CRMP2A expression. Moreover, Pin1 knockdown or knockout increases sensitivity to Sema3A-induced growth cone collapse in vitro and in vivo, leading to developmental abnormalities in axon guidance. These results identify an important isoform-specific function and regulation of CRMP2A in controlling axon growth and uncover Pin1-catalyzed prolyl isomerization as a regulatory mechanism in axon guidance.


Asunto(s)
Axones/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Isomerasa de Peptidilprolil/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Línea Celular Tumoral , Femenino , Humanos , Inmunohistoquímica , Inmunoprecipitación , Masculino , Peptidilprolil Isomerasa de Interacción con NIMA , Proteínas del Tejido Nervioso/genética , Isomerasa de Peptidilprolil/genética , Fosforilación , Transducción de Señal , Pez Cebra , Proteínas de Pez Cebra/genética
18.
Nat Med ; 21(5): 457-66, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25849135

RESUMEN

A common key regulator of oncogenic signaling pathways in multiple tumor types is the unique isomerase Pin1. However, available Pin1 inhibitors lack the required specificity and potency for inhibiting Pin1 function in vivo. By using mechanism-based screening, here we find that all-trans retinoic acid (ATRA)--a therapy for acute promyelocytic leukemia (APL) that is considered the first example of targeted therapy in cancer, but whose drug target remains elusive--inhibits and degrades active Pin1 selectively in cancer cells by directly binding to the substrate phosphate- and proline-binding pockets in the Pin1 active site. ATRA-induced Pin1 ablation degrades the protein encoded by the fusion oncogene PML-RARA and treats APL in APL cell and animal models as well as in human patients. ATRA-induced Pin1 ablation also potently inhibits triple-negative breast cancer cell growth in human cells and in animal models by acting on many Pin1 substrate oncogenes and tumor suppressors. Thus, ATRA simultaneously blocks multiple Pin1-regulated cancer-driving pathways, an attractive property for treating aggressive and drug-resistant tumors.


Asunto(s)
Neoplasias de la Mama/metabolismo , Regulación Leucémica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Leucemia Promielocítica Aguda/metabolismo , Isomerasa de Peptidilprolil/genética , Tretinoina/metabolismo , Animales , Antineoplásicos/química , Neoplasias de la Mama/genética , Catálisis , Dominio Catalítico , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Femenino , Fibroblastos/metabolismo , Células HEK293 , Humanos , Leucemia Promielocítica Aguda/genética , Células MCF-7 , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Peptidilprolil Isomerasa de Interacción con NIMA , Trasplante de Neoplasias , Fosfatos/química , Fosforilación , Prolina/química , Neoplasias de la Mama Triple Negativas/metabolismo
19.
Cancer Res ; 74(13): 3603-16, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24786790

RESUMEN

Breast cancer stem-like cells (BCSC) have been implicated in tumor growth, metastasis, drug resistance, and relapse but druggable targets in appropriate subsets of this cell population have yet to be identified. Here we identify a fundamental role for the prolyl isomerase Pin1 in driving BCSC expansion, invasiveness, and tumorigenicity, defining it as a key target of miR200c, which is known to be a critical regulator in BCSC. Pin1 overexpression expanded the growth and tumorigenicity of BCSC and triggered epithelial-mesenchymal transition. Conversely, genetic or pharmacological inhibition of Pin1 reduced the abundance and self-renewal activity of BCSC. Moreover, moderate overexpression of miR200c-resistant Pin1 rescued the BCSC defect in miR200c-expressing cells. Genetic deletion of Pin1 also decreased the abundance and repopulating capability of normal mouse mammary stem cells. In human cells, freshly isolated from reduction mammoplasty tissues, Pin1 overexpression endowed BCSC traits to normal breast epithelial cells, expanding both luminal and basal/myoepithelial lineages in these cells. In contrast, Pin1 silencing in primary breast cancer cells freshly isolated from clinical samples inhibited the expansion, self-renewal activity, and tumorigenesis of BCSC in vitro and in vivo. Overall, our work demonstrated that Pin1 is a pivotal regulator acting downstream of miR200c to drive BCSC and breast tumorigenicity, highlighting a new therapeutic target to eradicate BCSC.


Asunto(s)
Neoplasias de la Mama/genética , Transformación Celular Neoplásica/genética , MicroARNs/genética , Células Madre Neoplásicas/patología , Isomerasa de Peptidilprolil/genética , Animales , Transformación Celular Neoplásica/patología , Transición Epitelial-Mesenquimal/genética , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , MicroARNs/biosíntesis , Peptidilprolil Isomerasa de Interacción con NIMA , Invasividad Neoplásica , Trasplante de Neoplasias , Isomerasa de Peptidilprolil/biosíntesis , Fenantrolinas/farmacología , Interferencia de ARN , Transducción Genética , Células Tumorales Cultivadas
20.
Cancer Res ; 73(13): 3951-62, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23633483

RESUMEN

The Pin1 prolyl isomerase regulates phosphorylation signaling by controlling protein conformation after phosphorylation, and its upregulation promotes oncogenesis via acting on numerous oncogenic molecules. SUMOylation and deSUMOylation are dynamic mechanisms regulating a spectrum of protein activities. The SUMO proteases (SENP) remove SUMO conjugate from proteins, and their expression is deregulated in cancers. However, nothing is known about the role of SUMOylation in regulating Pin1 function. Here, we show that Pin1 is SUMOylated on Lys6 in the WW domain and on Lys63 in the PPIase domain. Pin1 SUMOylation inhibits its protein activity and oncogenic function. We further identify that SENP1 binds to and deSUMOylates Pin1. Importantly, either overexpression of SENP1 or disruption of Pin1 SUMOylation promotes the ability of Pin1 to induce centrosome amplification and cell transformation. Moreover, SENP1 also increases Pin1 protein stability in cell cultures, and Pin1 levels are positively correlated with SENP1 levels in human breast cancer specimens. These results not only uncover Pin1 SUMOylation on Lys6/63 as a novel mechanism to inhibit its activity and function but also identify a critical role for SENP1-mediated deSUMOylation in promoting Pin1 function during tumorigenesis.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Endopeptidasas/metabolismo , Isomerasa de Peptidilprolil/metabolismo , Sumoilación , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Neoplasias de la Mama/enzimología , Línea Celular , Centrosoma/metabolismo , Inestabilidad Cromosómica , Ciclina D1 , Cisteína Endopeptidasas , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Peptidilprolil Isomerasa de Interacción con NIMA , Estrés Oxidativo , Isomerasa de Peptidilprolil/química , Isomerasa de Peptidilprolil/genética , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA