Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
bioRxiv ; 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38746413

RESUMEN

The phosphoinositide-3 kinase (PI3K), a heterodimeric enzyme, plays a pivotal role in cellular metabolism and survival. Its deregulation is associated with major human diseases, particularly cancer. The p85 regulatory subunit of PI3K binds to the catalytic p110 subunit via its C-terminal domains, stabilising it in an inhibited state. Certain Src homology 3 (SH3) domains can activate p110 by binding to the proline-rich (PR) 1 motif located at the N-terminus of p85. However, the mechanism by which this N-terminal interaction activates the C-terminally bound p110 remains elusive. Moreover, the intrinsically poor ligand selectivity of SH3 domains raises the question of how they can control PI3K. Combining structural, biophysical, and functional methods, we demonstrate that the answers to both these unknown issues are linked: PI3K-activating SH3 domains engage in additional "tertiary" interactions with the C-terminal domains of p85, thereby relieving their inhibition of p110. SH3 domains lacking these tertiary interactions may still bind to p85 but cannot activate PI3K. Thus, p85 uses a functional selection mechanism that precludes nonspecific activation rather than nonspecific binding. This separation of binding and activation may provide a general mechanism for how biological activities can be controlled by promiscuous protein-protein interaction domains.

2.
Sci Adv ; 9(47): eadj0123, 2023 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-37992178

RESUMEN

Transcriptional dysregulation is a recurring pathogenic hallmark and an emerging therapeutic vulnerability in ovarian cancer. Here, we demonstrated that ovarian cancer exhibited a unique dependency on the regulatory machinery of transcriptional termination, particularly, cleavage and polyadenylation specificity factor (CPSF) complex. Genetic abrogation of multiple CPSF subunits substantially hampered neoplastic cell viability, and we presented evidence that their indispensable roles converged on the endonuclease CPSF3. Mechanistically, CPSF perturbation resulted in lengthened 3'-untranslated regions, diminished intronic polyadenylation and widespread transcriptional readthrough, and consequently suppressed oncogenic pathways. Furthermore, we reported the development of specific CPSF3 inhibitors building upon the benzoxaborole scaffold, which exerted potent antitumor activity. Notably, CPSF3 blockade effectively exacerbated genomic instability by down-regulating DNA damage repair genes and thus acted in synergy with poly(adenosine 5'-diphosphate-ribose) polymerase inhibition. These findings establish CPSF3-dependent transcriptional termination as an exploitable driving mechanism of ovarian cancer and provide a promising class of boron-containing compounds for targeting transcription-addicted human malignancies.


Asunto(s)
Recurrencia Local de Neoplasia , Neoplasias Ováricas , Femenino , Humanos , Factor de Especificidad de Desdoblamiento y Poliadenilación/genética , Factor de Especificidad de Desdoblamiento y Poliadenilación/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética
3.
Lancet Reg Health West Pac ; 36: 100775, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37547050

RESUMEN

Background: The integration of next-generation sequencing (NGS) comprehensive gene profiling (CGP) into clinical practice is playing an increasingly important role in oncology. Therefore, the HKU-HKSH Multi-disciplinary Molecular Tumour Board (MTB) was established to advance precision oncology in Hong Kong. A multicenter retrospective study investigated the feasibility of the HKU-HKSH MTB in determining genome-guided therapy for treatment-refractory solid cancers in Hong Kong. Methods: Patients who were presented at the HKU-HKSH MTB between August 2018 and June 2022 were included in this study. The primary study endpoints were the proportion of patients who receive MTB-guided therapy based on genomic analysis and overall survival (OS). Secondary endpoints included the proportion of patients with actionable genomic alterations, objective response rate (ORR), and disease control rate (DCR). The Kaplan-Meier method was used in the survival analyses, and hazard ratios were calculated using univariate Cox regression. Findings: 122 patients were reviewed at the HKU-HKSH MTB, and 63% (n = 77) adopted treatment per the MTB recommendations. These patients achieved a significantly longer median OS than those who did not receive MTB-guided therapy (12.7 months vs. 5.2 months, P = 0.0073). Their ORR and DCR were 29% and 65%, respectively. Interpretation: Our study demonstrated that among patients with heavily pre-treated advanced solid cancers, MTB-guided treatment could positively impact survival outcomes, thus illustrating the applicability of NGS CGPs in real-world clinical practice. Funding: The study was supported by the Li Shu Pui Medical Foundation. Dr Aya El Helali was supported by the Li Shu Pui Medical Foundation Fellowship grant from the Li Shu Pui Medical Foundation. Funders had no role in study design, data collection, data analysis, interpretation, or writing of the report.

5.
Genes Dis ; 10(1): 199-211, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37013053

RESUMEN

The insulin-like growth factor (IGF) axis plays important roles in cancer development and metastasis. The type 1 IGF receptor (IGF-1R) is a key member in the IGF axis and has long been recognized for its oncogenic role in multiple cancer lineages. Here we review the occurrence of IGF-1R aberrations and activation mechanisms in cancers, which justify the development of anti-IGF-1R therapies. We describe the therapeutic agents available for IGF-1R inhibition, with focuses on the recent or ongoing pre-clinical and clinical studies. These include antisense oligonucleotide, tyrosine kinase inhibitors and monoclonal antibodies which may be conjugated with cytotoxic drug. Remarkably, simultaneous targeting of IGF-1R and several other oncogenic vulnerabilities has shown early promise, highlighting the potential benefits of combination therapy. Further, we discuss the challenges in targeting IGF-1R so far and new concepts to improve therapeutic efficacy such as blockage of the nuclear translocation of IGF-1R.

6.
Br J Cancer ; 128(11): 2054-2062, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36997661

RESUMEN

BACKGROUND: Ovarian clear cell carcinoma (OCCC) is a challenging disease due to its intrinsic chemoresistance. Immunotherapy is an emerging treatment option but currently impeded by insufficient understanding of OCCC immunophenotypes and their molecular determinants. METHODS: Whole-genome sequencing on 23 pathologically confirmed patients was employed to depict the genomic profile of primary OCCCs. APOBEC3B expression and digital pathology-based Immunoscore were assessed by performing immunohistochemistry and correlated with clinical outcomes. RESULTS: An APOBEC-positive (APOBEC+) subtype was identified based on the characteristic mutational signature and prevalent kataegis events. APOBEC + OCCC displayed favourable prognosis across one internal and two external patient cohorts. The improved outcome was ascribable to increased lymphocytic infiltration. Similar phenomena of APOBEC3B expression and T-cell accumulation were observed in endometriotic tissues, suggesting that APOBEC-induced mutagenesis and immunogenicity could occur early during OCCC pathogenesis. Corroborating these results, a case report was presented for an APOBEC + patient demonstrating inflamed tumour microenvironment and clinical response to immune checkpoint blockade. CONCLUSIONS: Our findings implicate APOBEC3B as a novel mechanism of OCCC stratification with prognostic value and as a potential predictive biomarker that may inform immunotherapeutic opportunities.


Asunto(s)
Adenocarcinoma de Células Claras , Carcinoma , Neoplasias Ováricas , Femenino , Humanos , Neoplasias Ováricas/genética , Pronóstico , Mutación , Linfocitos T/patología , Adenocarcinoma de Células Claras/genética , Microambiente Tumoral , Citidina Desaminasa , Antígenos de Histocompatibilidad Menor
7.
Cell Rep ; 41(11): 111821, 2022 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-36516775

RESUMEN

Recurrent deletion of 16q12.2 is observed in luminal breast cancer, yet the causal genomic alterations in this region are largely unknown. In this study, we identify that loss of AKTIP, which is located on 16q12.2, drives tumorigenesis of estrogen receptor alpha (ERα)-positive, but not ERα-negative, breast cancer cells and is associated with poor prognosis of patients with ERα-positive breast cancer. Intriguingly, AKTIP-depleted tumors have increased ERα protein level and activity. Cullin-associated and neddylation-dissociated protein 1 (CAND1), which regulates the cullin-RING E3 ubiquitin ligases, protects ERα from cullin 2-dependent proteasomal degradation. Apart from ERα signaling, AKTIP loss triggers JAK2-STAT3 activation, which provides an alternative survival signal when ERα is inhibited. AKTIP-depleted MCF7 cells and ERα-positive patient-derived organoids are more resistant to ERα antagonists. Importantly, the resistance can be overcome by co-inhibition of JAK2/STAT3. Together, our results highlight the subtype-specific functional consequences of AKTIP loss and provide a mechanistic explanation for the enriched AKTIP copy-number loss in ERα-positive breast cancer.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Neoplasias de la Mama/patología , Proteínas Cullin/metabolismo , Regulación Neoplásica de la Expresión Génica , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Células MCF-7 , Carcinogénesis/genética , Transformación Celular Neoplásica/genética , Resistencia a Antineoplásicos/genética , Línea Celular Tumoral , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo
8.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34507989

RESUMEN

The phosphoinositide 3-kinase regulatory subunit p85α is a key regulator of kinase signaling and is frequently mutated in cancers. In the present study, we showed that in addition to weakening the inhibitory interaction between p85α and p110α, a group of driver mutations in the p85α N-terminal SH2 domain activated EGFR, HER2, HER3, c-Met, and IGF-1R in a p110α-independent manner. Cancer cells expressing these mutations exhibited the activation of p110α and the AKT pathway. Interestingly, the activation of EGFR, HER2, and c-Met was attributed to the ability of driver mutations to inhibit HER3 ubiquitination and degradation. The resulting increase in HER3 protein levels promoted its heterodimerization with EGFR, HER2, and c-Met, as well as the allosteric activation of these dimerized partners; however, HER3 silencing abolished this transactivation. Accordingly, inhibitors of either AKT or the HER family reduced the oncogenicity of driver mutations. The combination of these inhibitors resulted in marked synergy. Taken together, our findings provide mechanistic insights and suggest therapeutic strategies targeting a class of recurrent p85α mutations.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Fosfatidilinositol 3-Quinasa Clase Ia/metabolismo , Dominio Catalítico/genética , Línea Celular Tumoral , Fosfatidilinositol 3-Quinasa Clase I/genética , Fosfatidilinositol 3-Quinasa Clase Ia/genética , Fosfatidilinositol 3-Quinasa Clase Ia/fisiología , Células HCT116 , Humanos , Mutación , Neoplasias/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Dominios Proteicos/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor ErbB-3/metabolismo , Transducción de Señal , Dominios Homologos src
9.
Neoplasia ; 23(7): 718-730, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34144267

RESUMEN

EGFR signaling promotes ovarian cancer tumorigenesis, and high EGFR expression correlates with poor prognosis. However, EGFR inhibitors alone have demonstrated limited clinical benefit for ovarian cancer patients, owing partly to tumor resistance and the lack of predictive biomarkers. Cotargeting EGFR and the PI3K pathway has been previously shown to yield synergistic antitumor effects in ovarian cancer. Therefore, we reasoned that PI3K may affect cellular response to EGFR inhibition. In this study, we revealed PI3K isoform-specific effects on the sensitivity of ovarian cancer cells to the EGFR inhibitor erlotinib. Gene silencing of PIK3CA (p110α) and PIK3CB (p110ß) rendered cells more susceptible to erlotinib. In contrast, low expression of PIK3R2 (p85ß) was associated with erlotinib resistance. Depletion of PIK3R2, but not PIK3CA or PIK3CB, led to increased DNA damage and reduced level of the nonhomologous end joining DNA repair protein BRD4. Intriguingly, these defects in DNA repair were reversed upon erlotinib treatment, which caused activation and nuclear import of p38 MAPK to promote DNA repair with increased protein levels of 53BP1 and BRD4 and foci formation of 53BP1. Remarkably, inhibition of p38 MAPK or BRD4 re-sensitized PIK3R2-depleted cells to erlotinib. Collectively, these data suggest that p38 MAPK activation and the subsequent DNA repair serve as a resistance mechanism to EGFR inhibitor. Combined inhibition of EGFR and p38 MAPK or DNA repair may maximize the therapeutic potential of EGFR inhibitor in ovarian cancer.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase Ia/genética , Reparación del ADN/efectos de los fármacos , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Línea Celular Tumoral , Fosfatidilinositol 3-Quinasa Clase Ia/metabolismo , Variaciones en el Número de Copia de ADN , Manejo de la Enfermedad , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Resistencia a Antineoplásicos/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Modelos Biológicos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Inhibidores de Proteínas Quinasas/uso terapéutico , Transducción de Señal
10.
Sci Total Environ ; 762: 143159, 2021 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-33131852

RESUMEN

BACKGROUND: With the unprecedented urbanization light pollution has emerged as a ubiquitous problem, and there has been accumulating evidence on the links between exposure to light at night (LAN) and breast cancer risk. We conducted a systematic review and meta-analysis of published studies on the associations between LAN exposure and breast cancer risk. METHODS: We included all observational human studies wherein the exposure variable was LAN measured in indoor and outdoor environments, and the outcome was breast cancer. We employed summary relative risks (SRR) for breast cancer by comparing highest versus lowest categories of LAN exposure within a random-effects model. The National Toxicology Program's (NTP) Office of Health Assessment and Translation (OHAT) risk of bias rating tool was adopted to assess the risk of bias in individual studies and the Grading of Recommendations Assessment, Development and Evaluation (GRADE) guideline was employed to assess confidence in the body of evidence. RESULTS: A total 14 studies comprising four cohorts (13,155 cases among 372,802 exposed subjects), nine case-control and one case-referent studies of female subjects (39,462 cases and 20,739 controls) across seven countries and published between 2001 and 20 were included for review. Participants in the highest LAN exposure category were associated with higher risk of breast cancer in reference to those in the lowest (SRR: 1.12; 95% CI: 1.06-1.18; I2 = 39% for outdoor LAN, and SRR: 1.13; 95%CI: 1.05-1.21; I2 = 19% for indoor LAN). Pooled evidence identified relatively pronounced association of outdoor LAN exposure and breast cancer among women with estrogen receptor positive (ER+) tumor (SRR: 1.21; 95% CI: 1.04-1.40) and premenopausal status (SRR: 1.21; 95% CI: 1.06-1.37). The final rate of confidence in the body of evidence generated was graded as 'moderate' based on GRADE guideline. DISCUSSION: LAN exposure was consistently associated with higher breast cancer risk corroborating NTP's recommendations which anticipates excessive LAN as human carcinogen.


Asunto(s)
Neoplasias de la Mama , Neoplasias de la Mama/epidemiología , Neoplasias de la Mama/etiología , Estudios de Casos y Controles , Femenino , Humanos , Luz , Medición de Riesgo
13.
J Exp Clin Cancer Res ; 39(1): 262, 2020 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-33243298

RESUMEN

BACKGROUND: Recent genomic analyses revealed that druggable molecule targets were only detectable in approximately 6% of patients with nasopharyngeal carcinoma (NPC). However, a dependency on dysregulated CDK4/6-cyclinD1 pathway signaling is an essential event in the pathogenesis of NPC. In this study, we aimed to evaluate the therapeutic efficacy of a specific CDK4/6 inhibitor, palbociclib, and its compatibility with other chemotherapeutic drugs for the treatment of NPC by using newly established xenograft models and cell lines derived from primary, recurrent, and metastatic NPC. METHODS: We evaluated the efficacies of palbociclib monotherapy and concurrent treatment with palbociclib and cisplatin or suberanilohydroxamic acid (SAHA) in NPC cell lines and xenograft models. RNA sequencing was then used to profile the drug response-related pathways. Palbociclib-resistant NPC cell lines were established to determine the potential use of cisplatin as a second-line treatment after the development of palbociclib resistance. We further examined the efficacy of palbociclib treatment against cisplatin-resistant NPC cells. RESULTS: In NPC cells, palbociclib monotherapy was confirmed to induce cell cycle arrest in the G1 phase in vitro. Palbociclib monotherapy also had significant inhibitory effects in all six tested NPC tumor models in vivo, as indicated by substantial reductions in the total tumor volumes and in Ki-67 proliferation marker expression. In NPC cells, concurrent palbociclib treatment mitigated the cytotoxic effect of cisplatin in vitro. Notably, concurrent treatment with palbociclib and SAHA synergistically promoted NPC cell death both in vitro and in vivo. This combination also further inhibited tumor growth by inducing autophagy-associated cell death. NPC cell lines with induced palbociclib or cisplatin resistance remained sensitive to treatment with cisplatin or palbociclib, respectively. CONCLUSIONS: Our study findings provide essential support for the use of palbociclib as an alternative therapy for NPC and increase awareness of the effective timing of palbociclib administration with other chemotherapeutic drugs. Our results provide a foundation for the design of first-in-human clinical trials of palbociclib regimens in patients with NPC.


Asunto(s)
Antineoplásicos/uso terapéutico , Genómica/métodos , Carcinoma Nasofaríngeo/tratamiento farmacológico , Piperazinas/uso terapéutico , Piridinas/uso terapéutico , Animales , Antineoplásicos/farmacología , Humanos , Masculino , Ratones , Piperazinas/farmacología , Piridinas/farmacología , Transfección
14.
Mol Cell Oncol ; 7(5): 1780900, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32944639

RESUMEN

The phosphatidylinositol 3-kinase (PI3K), which is composed of the p85 regulatory and p110 catalytic subunits, is known to be downstream of the receptor tyrosine kinase (RTK). Our recent findings revealed that p85ß increases the protein level of AXL (an RTK) to activate p110, suggesting bidirectional regulation between PI3K and RTK.

15.
Cell Rep ; 32(6): 108020, 2020 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-32783942

RESUMEN

We present a CRISPR-based multi-gene knockout screening system and toolkits for extensible assembly of barcoded high-order combinatorial guide RNA libraries en masse. We apply this system for systematically identifying not only pairwise but also three-way synergistic therapeutic target combinations and successfully validate double- and triple-combination regimens for suppression of cancer cell growth and protection against Parkinson's disease-associated toxicity. This system overcomes the practical challenges of experimenting on a large number of high-order genetic and drug combinations and can be applied to uncover the rare synergistic interactions between druggable targets.


Asunto(s)
Sistemas CRISPR-Cas , Combinación de Medicamentos , Sistemas de Liberación de Medicamentos/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Animales , Antineoplásicos/farmacología , Drosophila melanogaster , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Ratones , Neoplasias/tratamiento farmacológico , Enfermedad de Parkinson/tratamiento farmacológico , ARN Guía de Kinetoplastida
16.
Curr Cancer Drug Targets ; 20(8): 573-585, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32392113

RESUMEN

Ovarian cancer remains the leading cause of gynecologic cancer-related deaths among women worldwide. The dismal survival rate is partially due to recurrence after standardized debulking surgery and first-line chemotherapy. In recent years, targeted therapies, including antiangiogenic agents or poly (ADP-ribose) polymerase inhibitors, represent breakthroughs in the treatment of ovarian cancer. As more therapeutic agents become available supplemented by a deeper understanding of ovarian cancer biology, a range of combination treatment approaches are being actively investigated to further improve the clinical outcomes of the disease. These combinations, which involve DNA-damaging agents, targeted therapies of signaling pathways and immunotherapies, simultaneously target multiple cancer pathways or hallmarks to induce additive or synergistic antitumor activities. Here we review the preclinical data and ongoing clinical trials for developing effective combination therapies in treating ovarian cancer. These emerging therapeutic modalities may reshape the treatment landscape of the disease.


Asunto(s)
Antineoplásicos/uso terapéutico , Inmunoterapia/métodos , Neoplasias Ováricas/terapia , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Animales , Terapia Combinada , Femenino , Humanos , Neoplasias Ováricas/patología
17.
Nat Commun ; 11(1): 2291, 2020 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-32385243

RESUMEN

PIK3R2 encodes the p85ß regulatory subunit of phosphatidylinositol 3-kinase and is frequently amplified in cancers. The signaling mechanism and therapeutic implication of p85ß are poorly understood. Here we report that p85ß upregulates the protein level of the receptor tyrosine kinase AXL to induce oncogenic signaling in ovarian cancer. p85ß activates p110 activity and AKT-independent PDK1/SGK3 signaling to promote tumorigenic phenotypes, which are all abolished upon inhibition of AXL. At the molecular level, p85ß alters the phosphorylation of TRIM2 (an E3 ligase) and optineurin (an autophagy receptor), which mediate the selective regulation of AXL by p85ß, thereby disrupting the autophagic degradation of the AXL protein. Therapeutically, p85ß expression renders ovarian cancer cells vulnerable to inhibitors of AXL, p110, or PDK1. Conversely, p85ß-depleted cells are less sensitive to these inhibitors. Together, our findings provide a rationale for pharmacological blockade of the AXL signaling axis in PIK3R2-amplified ovarian cancer.


Asunto(s)
Autofagia , Carcinogénesis/metabolismo , Fosfatidilinositol 3-Quinasa Clase Ia/metabolismo , Proteolisis , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Supervivencia sin Enfermedad , Activación Enzimática , Femenino , Ontología de Genes , Humanos , Lisosomas/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Proteínas Nucleares , Neoplasias Ováricas/patología , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ubiquitinación , Regulación hacia Arriba/genética , Tirosina Quinasa del Receptor Axl
18.
FEBS J ; 287(1): 122-144, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31569299

RESUMEN

The functional consequences of cancer-associated missense mutations are unclear for the majority of proteins. We have previously demonstrated that the activity of SOX and Pit-Oct-Unc (POU) family factors during pluripotency reprogramming can be switched and enhanced with rationally placed point mutations. Here, we interrogated cancer mutation databases and identified recurrently mutated positions at critical structural interfaces of the DNA-binding domains of paralogous SOX and POU family transcription factors. Using the conversion of mouse embryonic fibroblasts to induced pluripotent stem cells as functional readout, we identified several gain-of-function mutations that enhance pluripotency reprogramming by SOX2 and OCT4. Wild-type SOX17 cannot support reprogramming but the recurrent missense mutation SOX17-V118M is capable of inducing pluripotency. Furthermore, SOX17-V118M promotes oncogenic transformation, enhances thermostability and elevates cellular protein levels of SOX17. We conclude that the mutational profile of SOX and POU family factors in cancer can guide the design of high-performance reprogramming factors. Furthermore, we propose cellular reprogramming as a suitable assay to study the functional impact of cancer-associated mutations.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Pluripotentes Inducidas/citología , Mutación Missense , Neoplasias/patología , Factor 3 de Transcripción de Unión a Octámeros/genética , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXF/genética , Animales , Diferenciación Celular , Células Cultivadas , Reprogramación Celular , Células Madre Embrionarias/metabolismo , Perfilación de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Neoplasias/genética , Neoplasias/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Factores de Transcripción SOXB1/metabolismo , Factores de Transcripción SOXF/metabolismo
19.
Nat Commun ; 10(1): 716, 2019 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-30755611

RESUMEN

Copy number loss of PIK3R1 (p85α) most commonly occurs in ovarian cancer among all cancer types. Here we report that ovarian cancer cells manifest a spectrum of tumorigenic phenotypes upon knockdown of PIK3R1. PIK3R1 loss activates AKT and p110-independent JAK2/STAT3 signaling through inducing changes in the phosphorylation of the docking protein Gab2, thereby relieving the negative inhibition on AKT and promoting the assembly of JAK2/STAT3 signalosome, respectively. Additional mechanisms leading to AKT activation include enhanced p110α kinase activity and a decrease in PTEN level. PIK3R1 loss renders ovarian cancer cells vulnerable to inhibition of AKT or JAK2/STAT3. The combination of AKT and STAT3 inhibitors significantly increases the anti-tumor effect compared to single-agent treatments. Together, our findings provide a rationale for mechanism-based therapeutic approach that targets tumors with loss of PIK3R1.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neoplasias Ováricas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor de Transcripción STAT3/metabolismo , Apoptosis/fisiología , Ciclo Celular/fisiología , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Supervivencia Celular/fisiología , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Fosfatidilinositol 3-Quinasa Clase Ia , Femenino , Humanos , Janus Quinasa 2/metabolismo , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/patología , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/deficiencia , Fosforilación , Transducción de Señal
20.
J Am Pharm Assoc (2003) ; 59(1): 23-29.e1, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30348509

RESUMEN

OBJECTIVES: The primary objective of this study was to identify characteristics of pharmacists that contribute to their success. DESIGN: A working definition of success in pharmacy practice was derived from a scoping literature review and is based on the premise that successful pharmacists practice to full scope within the context of their practice setting. Semistructured individual interviews were conducted with selected pharmacists. Potential candidates were nominated by leading pharmacists in the field with the use of our prespecified definition of success. Lists from the nominators were compared, and pharmacists who appeared on more than 1 list were invited to participate. The interview tool was developed with the use of previous research on success in health care professions. SETTING AND PARTICIPANTS: Participants were 10 practicing pharmacists in a variety of locations (5 urban/5 rural) and practice settings (5 hospital/4 community/1 ambulatory care). OUTCOME MEASURES: Themes related to successful pharmacists practicing to full scope. RESULTS: Pharmacists meeting our definition of success were engaged in assessment and care planning, other expanded scope activities, and interpersonal activities and collaboration. The 10 interviewed pharmacists described motivation, critical thinking, emotional intelligence, core competencies, and work-life balance as significant contributors to their success. CONCLUSION: Several characteristics were identified as potentially related to success. These characteristics may be useful in pharmacists identifying areas for personal growth and development.


Asunto(s)
Características Humanas , Farmacéuticos/psicología , Rol Profesional/psicología , Femenino , Humanos , Masculino , Investigación Cualitativa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...