Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
Mater Today Bio ; 25: 100975, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38322662

RESUMO

Diabetic wound healing is delayed due to persistent inflammation, and macrophage-immunomodulating biomaterials can control the inflammatory phase and shorten the healing time. In this study, acellular embryoid bodies (aEBs) were prepared and mixed with thermosensitive hydroxybutyl chitosan (HBC) hydrogels to produce aEB/HBC composite hydrogels. The aEB/HBC composite hydrogels exhibited reversible temperature-sensitive phase transition behavior and a hybrid porous network. In vitro analysis showed that the aEB/HBC composite hydrogels exhibited better antimicrobial activity than the PBS control, aEBs or HBC hydrogels and promoted M0 to M2 polarization but not M1 to M2 macrophage repolarization in culture. The in vivo results showed that the aEB/HBC composite hydrogels accelerated cutaneous wound closure, re-epithelialization, ingrowth of new blood vessels, and collagen deposition and reduced the scar width during wound healing in diabetic mice over time. Macrophage phenotype analysis showed that the aEB/HBC composite hydrogels induce M2 macrophage reactions continually, upregulate M2-related mRNA and protein expression and downregulate M1-related mRNA and protein expression. Therefore, the aEB/HBC composite hydrogels have excellent antimicrobial activity, promote M2 macrophage polarization and accelerate the functional and structural healing of diabetic cutaneous wounds.

2.
Cancer Res ; 83(20): 3442-3461, 2023 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-37470810

RESUMO

Although external beam radiotherapy (xRT) is commonly used to treat central nervous system (CNS) tumors in patients of all ages, young children treated with xRT frequently experience life-altering and dose-limiting neurocognitive impairment (NI) while adults do not. The lack of understanding of mechanisms responsible for these differences has impeded the development of neuroprotective treatments. Using a newly developed mouse model of xRT-induced NI, we found that neurocognitive function is impaired by ionizing radiation in a dose- and age-dependent manner, with the youngest animals being most affected. Histologic analysis revealed xRT-driven neuronal degeneration and cell death in neurogenic brain regions in young animals but not adults. BH3 profiling showed that neural stem and progenitor cells, neurons, and astrocytes in young mice are highly primed for apoptosis, rendering them hypersensitive to genotoxic damage. Analysis of single-cell RNA sequencing data revealed that neural cell vulnerability stems from heightened expression of proapoptotic genes including BAX, which is associated with developmental and mitogenic signaling by MYC. xRT induced apoptosis in primed neural cells by triggering a p53- and PUMA-initiated, proapoptotic feedback loop requiring cleavage of BID and culminating in BAX oligomerization and caspase activation. Notably, loss of BAX protected against apoptosis induced by proapoptotic signaling in vitro and prevented xRT-induced apoptosis in neural cells in vivo as well as neurocognitive sequelae. On the basis of these findings, preventing xRT-induced apoptosis specifically in immature neural cells by blocking BAX, BIM, or BID via direct or upstream mechanisms is expected to ameliorate NI in pediatric patients with CNS tumor. SIGNIFICANCE: Age- and differentiation-dependent apoptotic priming plays a pivotal role in driving radiotherapy-induced neurocognitive impairment and can be targeted for neuroprotection in pediatric patients.


Assuntos
Proteínas Reguladoras de Apoptose , Apoptose , Animais , Criança , Pré-Escolar , Humanos , Camundongos , Apoptose/fisiologia , Proteínas Reguladoras de Apoptose/metabolismo , Proteína X Associada a bcl-2/metabolismo , Morte Celular , Transdução de Sinais , Proteína Supressora de Tumor p53/genética
3.
Front Immunol ; 14: 1099799, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36936950

RESUMO

Introduction: Macrophages play an important role in the innate immunity. While macrophage inflammation is necessary for biological defense, it must be appropriately controlled. Extracellular vesicles (EVs) are small vesicles released from all types of cells and play a central role in intercellular communication. Skeletal muscle has been suggested to release anti-inflammatory factors, but the effect of myotube-derived EVs on macrophages is unknown. As an anti-inflammatory mechanism of macrophages, the immune responsive gene 1 (IRG1)-itaconate pathway is essential. In this study, we show that skeletal muscle-derived EVs suppress macrophage inflammatory responses, upregulating the IRG1-itaconate pathway. Methods: C2C12 myoblasts were differentiated into myotubes and EVs were extracted by ultracentrifugation. Skeletal myotube-derived EVs were administered to mouse bone marrow-derived macrophages, then lipopolysaccharide (LPS) stimulation was performed and inflammatory cytokine expression was measured by RT-qPCR. Metabolite abundance in macrophages after addition of EVs was measured by CE/MS, and IRG1 expression was measured by RT-PCR. Furthermore, RNA-seq analysis was performed on macrophages after EV treatment. Results: EVs attenuated the expression of LPS-induced pro-inflammatory factors in macrophages. Itaconate abundance and IRG1 expression were significantly increased in the EV-treated group. RNA-seq analysis revealed activation of the PI3K-Akt and JAK-STAT pathways in macrophages after EV treatment. The most abundant miRNA in myotube EVs was miR-206-3p, followed by miR-378a-3p, miR-30d-5p, and miR-21a-5p. Discussion: Skeletal myotube EVs are supposed to increase the production of itaconate via upregulation of IRG1 expression and exhibited an anti-inflammatory effect in macrophages. This anti-inflammatory effect was suggested to involve the PI3K-Akt and JAK-STAT pathways. The miRNA profiles within EVs implied that miR-206-3p, miR-378a-3p, miR-30d-5p, and miR-21a-5p may be responsible for the anti-inflammatory effects of the EVs. In summary, in this study we showed that myotube-derived EVs prevent macrophage inflammatory responses by activating the IRG1-itaconate pathway.


Assuntos
Vesículas Extracelulares , MicroRNAs , Animais , Camundongos , Lipopolissacarídeos/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Macrófagos , MicroRNAs/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Vesículas Extracelulares/metabolismo
4.
World J Surg Oncol ; 21(1): 78, 2023 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-36872314

RESUMO

BACKGROUND: The purpose of our study is to investigate the expression level and prognostic value of serum α-hydroxybutyrate dehydrogenase (α-HBDH) in lung cancer (LC) patients. METHOD: LC patients treated in the Department of Oncology, Shaanxi Provincial Cancer Hospital from January 2014 to December 2016 were included in this study, all of whom underwent serological detection of α-HBDH prior to admission, and were enrolled in follow-up 5-year survival. Comparing the differences between high group and normal groups based on α-HBDH and LDH expression via clinicopathological parameters and laboratory data. Univariate and multivariate regression and overall survival (OS) were analyzed to explore whether elevated α-HBDH was an independent risk factor for LC, compared to LDH. RESULTS: Multivariate regression analysis showed that age (P = 0.018), liver metastasis (P = 0.011), α-HBDH (P = 0.015), and neutrophil-to-lymphocyte ratio (NLR) (P = 0.031) were independent prognostic factors affecting OS in LC patients. The overall diagnostic efficacy of α-HBDH (AUC = 0.887) was higher than that of LDH (AUC = 0.709) in the ROC curve. The sensitivity was significantly higher of α-HBDH (sensitivity: 76.06%, specificity: 94.87%) compared with LDH (sensitivity: 49.30%, specificity: 94.87%). The median of OS was more significant in the high-α-HBDH group (6.4 months) than in the normal-α-HBDH group (12.7 months) (P = 0.023). The median of OS was significant in the high-LDH (> 245 U/L) group at 5.8 months and 12.0 months in the normal-LDH (≤ 245 U/L) group (P = 0.068). CONCLUSIONS: Elevated expression of α-HBDH may indicate a poor prognosis of LC patients. It has a higher sensitivity than LDH and can be used as a potential early biomarker and an independent risk factor predicting the prognosis of LC survival.


Assuntos
Neoplasias Hepáticas , Neoplasias Pulmonares , Humanos , Prognóstico , Institutos de Câncer , Hospitalização
5.
Int J Radiat Biol ; 99(6): 934-940, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36357962

RESUMO

PURPOSE: Unlike treatment with high doses of radiation that causes considerable DNA damage resulting in injury and p53 activation, exposure of cells or whole animals to low doses of radiation (LDR, ∼10cGy) can induce a protective radio adaptive response. Despite ample information about the contribution of the p53 pathway to high doses of radiation-induced effects, our understanding of the role of p53 in LDR-induced response remains incomplete. This review provides a brief summary of the p53 response to LDR exposure focusing on metabolic regulation. CONCLUSION: Consistent with growing evidence indicating a critical role of metabolic pathways in the modulation of stress responses, the radio adaptive response was mediated by the LDR-induced metabolic switch from oxidative phosphorylation to glycolysis. Remarkably, this metabolic reprogramming depends on p53 downregulation, suggesting a previously unrecognized p53-mediated metabolic response. Of note is that the LDR-induced p53 response is temporary but may become persistent if LDR exposure is recurrent and protracted. While further investigation is necessary, the model where LDR induces p53 downregulation concurrent with anabolic reprogramming may offer novel mechanistic insight into the radio adaptive response. It suggests a model in which LDR exposure is protective when transient or intermittent but may become detrimental when chronic because prolonged p53 downregulation would lead to inactivation of this critical tumor suppressor, causing a loss of p53-dependent DNA damage checkpoint, genomic instability, dysregulated metabolism, and thus increased cancer risk.


Assuntos
Radiação Ionizante , Proteína Supressora de Tumor p53 , Animais , Proteína Supressora de Tumor p53/metabolismo , Dano ao DNA , Relação Dose-Resposta à Radiação
6.
Circ Cardiovasc Imaging ; 15(7): e014116, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35861979

RESUMO

BACKGROUND: Remnant cholesterol (RC) contributes to residual risk of atherosclerotic cardiovascular disease, but population-based evidence on the prospective relationship between RC and coronary artery calcium (CAC) progression is rare. METHODS: We pooled data obtained from 6544 atherosclerotic cardiovascular disease-free individuals from the CARDIA study (Coronary Artery Risk Development in Young Adults; n=2635) and MESA (Multi-Ethnic Study of Atherosclerosis; n=3909), with a mean±SD age of 47.2±19.8 years; 3019 (46.1%) were men who completed computed tomography of CAC at baseline. RC was measured as total cholesterol minus HDL (high-density lipoprotein) cholesterol minus calculated LDL (low-density lipoprotein) cholesterol (LDL-C) estimated by using the Martin/Hopkins equation. Adjusted Cox models were used to assess the relationships between RC levels and CAC progression. We also performed discordance analyses examining the risk of CAC progression in RC versus LDL-C discordant/concordant groups using median cut points and clinically relevant LDL-C targets. RESULTS: During a median follow-up of 8.6 years, 2778 (42.5%) participants had CAC progression. After multivariable adjustment for demographics and cardiovascular risk factors, a 1-mg/dL increase in RC levels was associated with a 1.3% higher risk of CAC progression (hazard ratio, 1.013 [95% CI, 1.008-1.017]). Results were similar when we categorized individuals by RC quartiles. Furthermore, the discordant high RC/low LDL-C group had a significantly higher risk of CAC progression than the concordant low RC/LDL-C group regarding their medians (hazard ratio, 1.195 [95% CI, 1.063-1.343]) or when setting the clinical LDL-C cut points at 100 and 130 but not 70 mg/dL. The association remained robust across a series of sensitivity analyses. CONCLUSIONS: Elevated RC levels were associated with an increased risk of CAC progression independent of traditional cardiovascular risk factors, even in individuals with optimal LDL-C levels. REGISTRATION: URL: https://www. CLINICALTRIALS: gov; Unique identifiers: NCT00005130 (CARDIA) and NCT00005487 (MESA).


Assuntos
Aterosclerose , Doença da Artéria Coronariana , Adulto , Idoso , Cálcio , Colesterol , HDL-Colesterol , LDL-Colesterol , Doença da Artéria Coronariana/diagnóstico por imagem , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Fatores de Risco
7.
Front Cell Dev Biol ; 9: 775312, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34888311

RESUMO

Apart from mutations in the p53 gene, p53 functions can be alternatively compromised by a decrease in nuclear p53 protein levels or activities. In accordance, enhanced p53 protein turnover due to elevated expression of the critical p53 E3 ligase MDM2 or MDM2/MDMX is found in many human cancers. Likewise, the HPV viral E6 protein-mediated p53 degradation critically contributes to the tumorigenesis of cervical cancer. In addition, growth-promoting signaling-induced cell proliferation is accompanied by p53 downregulation. Animal studies have also shown that loss of p53 is essential for oncogenes to drive malignant transformation. The close association between p53 downregulation and carcinogenesis implicates a critical role of basally expressed p53. In accordance, available evidence indicates that a reduced level of basal p53 is usually associated with disruption of homeostasis, suggesting a homeostatic function mediated by basal p53. However, basally expressed p53 under non-stress conditions is maintained at a relatively low abundance with little transcriptional activity, raising the question of how basal p53 could protect homeostasis. In this review, we summarize the findings pertinent to basal p53-mediated activities in the hope of developing a model in which basally expressed p53 functions as a barrier to anabolic metabolism to preserve homeostasis. Future investigation is necessary to characterize basal p53 functionally and to obtain an improved understanding of p53 homeostatic function, which would offer novel insight into the role of p53 in tumor suppression.

8.
Ultrasonics ; 110: 106243, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32961400

RESUMO

Skeletal muscle is an important secretory organ in mammals, producing myriad chemical mediators ("myokines") with distinct biological action in different tissues, including anti-inflammatory activity. Extracellular vesicles (EVs) have recently been identified as a mode of myokine transport from muscle, facilitating such anti-inflammatory activity. In this report, we have demonstrated that high-intensity ultrasound (US) strongly induces EV secretion from cultured myotubes without a reduction in cell viability. High-intensity US of 3.0 W/cm2 with 20% duty cycle increased the number of EVs by 2-fold compared to control at 6 h. This effect was specific to EVs in the 100-150 nm size range. Thus, high-intensity US is a novel modality for inducing myocellular EV release and may hold therapeutic value.


Assuntos
Vesículas Extracelulares/metabolismo , Tratamento por Ondas de Choque Extracorpóreas/métodos , Fibras Musculares Esqueléticas/metabolismo , Animais , Células Cultivadas , Camundongos
9.
J Mol Cell Biol ; 12(9): 713-722, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32706867

RESUMO

A role of tumor-suppressive activity of p53 in the tumor microenvironment (TME) has been implicated but remains fairly understudied. To address this knowledge gap, we leveraged our MdmxS314A mice as recipients to investigate how implanted tumor cells incapacitate host p53 creating a conducive TME for tumor progression. We found that tumor cell-associated stress induced p53 downregulation in peritumor cells via an MDMX-Ser314 phosphorylation-dependent manner. As a result, an immunosuppressive TME was developed, as reflected by diminished immune cell infiltration into tumors and compromised macrophage M1 polarization. Remarkably, ablation of MDMX-Ser314 phosphorylation attenuated p53 decline in peritumor cells, which was associated with mitigation of immunosuppression and significant tumor growth delay. Our data collectively uncover a novel role of p53 in regulating the tumor immune microenvironment, suggesting that p53 restoration in the TME can be exploited as a potential strategy of anticancer therapy.


Assuntos
Regulação para Baixo , Terapia de Imunossupressão , Proteínas Proto-Oncogênicas/metabolismo , Microambiente Tumoral/imunologia , Proteína Supressora de Tumor p53/metabolismo , Animais , Linhagem Celular Tumoral , Polaridade Celular , Técnicas de Introdução de Genes , Humanos , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Fosforilação , Fosfosserina/metabolismo
10.
Prostate ; 80(12): 986-992, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32557725

RESUMO

BACKGROUND: Focal therapies for prostate cancer (PC) can reduce adverse events and do not lead to androgen-independent progression. Ultrasound could be used for cancer treatments if the repetition frequency is fitted to the purpose. We investigated the possible therapeutic effect of ultrasound irradiation on PC cells. MATERIALS AND METHODS: We irradiated two PC cell lines, androgen-dependent LNCaP and -independent PC-3 with ultrasound (3.0 W/cm2 , 3 MHz, irradiation time rate: 20%) for 2 minutes for 1 day or 3 consecutive days at a repetition frequency of 1, 10, or 100 Hz in vitro. Cell proliferation and apoptosis were determined after irradiation. RESULTS: Cell proliferation of PC-3 was significantly inhibited after 1 day (P < .0001) and 3 days (P < .0001) of 10 Hz ultrasound irradiation, and that of LNCaP after 1 day (P < .0001) and 3 days (P < .0001) of irradiation. LNCaP was more sensitive to ultrasound at both lower and higher cell density but PC-3 was only sensitive at a lower cell density (P < .01). Irradiation with 10 Hz ultrasound-induced significantly more PC-3 apoptotic cells than control (1 day, P = .0137; 3 days, P = .0386) rather than irradiation with 1 Hz. Apoptosis via caspase-3 was induced at 10 Hz in 1-day (P < .05) irradiation in both cell lines. CONCLUSIONS: Ultrasound irradiation with even 1 day of 10 Hz significantly inhibited cell proliferation in both LNCaP and PC-3, especially by the remarkable induction of apoptosis in vitro. Our study indicated that ultrasound irradiation can be a therapeutic option for PC and further studies in vivo will be undertaken.


Assuntos
Neoplasias da Próstata/radioterapia , Terapia por Ultrassom/métodos , Apoptose/efeitos da radiação , Linhagem Celular Tumoral , Proliferação de Células/efeitos da radiação , Sobrevivência Celular/efeitos da radiação , Humanos , Masculino , Células PC-3 , Neoplasias da Próstata/patologia , Neoplasias de Próstata Resistentes à Castração/patologia , Neoplasias de Próstata Resistentes à Castração/radioterapia
11.
Cell Death Differ ; 27(5): 1618-1632, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31659281

RESUMO

Apart from its well-known prodeath activity, p53 is also implicated in promoting cell survival. How p53 can mediate such seemingly opposing effects is largely unclear. We report here a novel mechanism in which p53-mediated proapoptosis is switched to antiapoptosis via its interaction with a p53 isoform, Δ133p53. We show that the expression of Δ133p53 is induced by mild or a moderate level of stress via an HIF1-dependent mechanism. Increased Δ133p53 levels contribute to the adaptive response by shifting the p53 binding at the Bcl2 promoter from suppressive responsive elements (RE) to activating REs, resulting in induction of Bcl2. In accordance with this mode of action, pretreatment of mice with mild stress induces Δ133p53 and Bcl2, which is associated with protection of animals from toxicity caused by high doses of DNA damage agents. Collectively, our work uncovers a novel functional interplay between p53 and Δ133p53 determining cell fate; survival or death in response to stress.


Assuntos
Mutação/genética , Estresse Fisiológico , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Animais , Apoptose , Sequência de Bases , Sítios de Ligação , Linhagem Celular , Relação Dose-Resposta à Radiação , Glicólise , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas/genética , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Regulação para Cima
12.
Transl Cancer Res ; 9(3): 1993-1997, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35117546

RESUMO

Via regulation of cellular stress responses, p53 contributes to the maintenance of homeostasis. Contrary to its well-established pro-death function, p53 is also implicated in promoting cell survival by mediating the adaptive stress response. Emerging data reveal that the adaptive stress response is coupled with p53 decline that is a prerequisite for the induction of pro-survival pathways augmenting cell fitness. However, if the adaptive stress responses persist or become chronic, the sustained p53 downregulation would result in a permanent loss of p53 function and p53-dependent homeostasis. The available information suggests a model in which cells respond to different levels of stress by governing the activity and abundance of p53 that, in turn, determines the cell fate dependent on not only the intensity but also the duration of stress.

13.
Cell Rep ; 29(11): 3693-3707.e5, 2019 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-31825845

RESUMO

When the core body temperature is higher than 40°C, life is threatened due to heatstroke. Tumor repressor p53 is required for heat-induced apoptosis at hyperthermia conditions (>41°C). However, its role in sub-heatstroke conditions (≤40°C) remains unclear. Here, we reveal that both zebrafish and human p53 promote survival at 40°C, the heatstroke threshold temperature, by preventing a hyperreactive heat shock response (HSR). At 40°C, both Hsf1 and Hsp90 are activated. Hsf1 upregulates the expression of Hsc70 to trigger Hsc70-mediated protein degradation, whereas Hsp90 stabilizes p53 to repress the expression of Hsf1 and Hsc70, which prevents excessive HSR to maintain cell homeostasis. Under hyperthermia conditions, ATM is activated to phosphorylate p53 at S37, which increases BAX expression to induce apoptosis. Furthermore, growth of p53-deficient tumor xenografts, but not that of their p53+/+ counterparts, was inhibited by 40°C treatment. Our findings may provide a strategy for individualized therapy for p53-deficient cancers.


Assuntos
Apoptose , Golpe de Calor/metabolismo , Resposta ao Choque Térmico , Proteína Supressora de Tumor p53/metabolismo , Animais , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Células HCT116 , Proteínas de Choque Térmico HSC70/metabolismo , Fatores de Transcrição de Choque Térmico/metabolismo , Golpe de Calor/genética , Células Hep G2 , Humanos , Células MCF-7 , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Estabilidade Proteica , Proteína Supressora de Tumor p53/genética , Peixe-Zebra , Proteína X Associada a bcl-2/metabolismo
14.
J Mol Cell Biol ; 10(4): 273-284, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-30265334

RESUMO

ZBTB7A, a member of the POZ/BTB and Krüppel (POK) family of transcription factors, has been shown to have a context-dependent role in cancer development and progression. The role of ZBTB7A in estrogen receptor alpha (ERα)-positive breast cancer is largely unknown. Approximately 70% of breast cancers are classified as ERα-positive. ERα carries out the biological effects of estrogen and its expression level dictates response to endocrine therapies and prognosis for breast cancer patients. In this study, we find that ZBTB7A transcriptionally regulates ERα expression in ERα-positive breast cancer cell lines by binding to the ESR1 promoter leading to increased transcription of ERα. Inhibition of ZBTB7A in ERα-positive cells results in decreased estrogen responsiveness as demonstrated by diminished estrogen-response element-driven luciferase reporter activity, induction of estrogen target genes, and estrogen-stimulated growth. We also report that ERα potentiates ZBTB7A expression via a post-translational mechanism, suggesting the presence of a positive feedback loop between ZBTB7A and ERα, conferring sensitivity to estrogen in breast cancer. Clinically, we find that ZBTB7A and ERα are often co-expressed in breast cancers and that high ZBTB7A expression correlates with improved overall and relapse-free survival for breast cancer patients. Importantly, high ZBTB7A expression predicts a more favorable outcome for patients treated with endocrine therapies. Together, these findings demonstrate that ZBTB7A contributes to the transcriptional program maintaining ERα expression and potentially an endocrine therapy-responsive phenotype in breast cancer.


Assuntos
Neoplasias da Mama/genética , Proteínas de Ligação a DNA/genética , Receptor alfa de Estrogênio/genética , Regulação Neoplásica da Expressão Gênica , Fatores de Transcrição/genética , Animais , Mama/metabolismo , Mama/patologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Regiões Promotoras Genéticas , Ativação Transcricional
15.
Asia Pac J Clin Nutr ; 27(3): 655-661, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29737814

RESUMO

BACKGROUND AND OBJECTIVES: Previous studies have obtained conflicting findings regarding the possible associations between glycemic load (GL) indices and diabetes. In the present study, we examined cross-sectional associations between several GL indices, including the total dietary GL, the energy-adjusted GL, and the prevalence of abnormal glucose metabolism, including prediabetes and diabetes. METHODS AND STUDY DESIGN: This study was conducted in Guangzhou, China from July 2011 to December 2011. It included 2,022 participants (602 men and 1,420 women), between 45 and 75 years of age. The prevalence of abnormal glucose metabolism was compared across the quartiles of GL indices to discover any potential linear correlations. Stratified analysis was conducted according to the body mass index (BMI) and waist circumference (WC) measurements. RESULTS: Energy-adjusted GL was positively associated with the prevalence of diabetes and the multivariable-adjusted estimate of the OR comparing the highest versus the lowest quartile was 2.50 (95% CI, 1.49-4.19). For the stratified analysis by sex, BMI or WC, similar associations were observed. For the overweight and obese (BMI ≥24.0 kg/m2) or centrally obese (WC ≥85 cm for men or ≥80 cm for women) participants, compared to participants in the lowest quartile of energy-adjusted GL, those in the highest quartile showed an increased risk of abnormal glucose metabolism. The OR estimates were 2.25 (95% CI: 1.45-3.52) and 1.54 (95% CI: 1.06-2.25), respectively. CONCLUSIONS: High dietary energy-adjusted GL is associated with the prevalence of diabetes as well as abnormal glucose metabolism among middle-aged and elderly adults.


Assuntos
Carga Glicêmica , Estado Pré-Diabético/epidemiologia , Estado Pré-Diabético/etiologia , Idoso , Glicemia , Peso Corporal , China/epidemiologia , Estudos Transversais , Diabetes Mellitus/epidemiologia , Dieta , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
16.
Proc Natl Acad Sci U S A ; 115(13): 3452-3457, 2018 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-29540569

RESUMO

Renewable tissues exhibit heightened sensitivity to DNA damage, which is thought to result from a high level of p53. However, cell proliferation in renewable tissues requires p53 down-regulation, creating an apparent discrepancy between the p53 level and elevated sensitivity to DNA damage. Using a combination of genetic mouse models and pharmacologic inhibitors, we demonstrate that it is p53-regulated MDM2 that functions together with MDMX to regulate DNA damage sensitivity by targeting EZH2 (enhancer of zeste homolog 2) for ubiquitination/degradation. As a methyltransferase, EZH2 promotes H3K27me3, and therefore chromatin compaction, to determine sensitivity to DNA damage. We demonstrate that genetic and pharmacologic interference of the association between MDM2 and MDMX stabilizes EZH2, resulting in protection of renewable tissues from radio-/chemotherapy-induced acute injury. In cells with p53 mutation, there are diminished MDM2 levels, and thus accumulation of EZH2, underpinning the resistant phenotype. Our work uncovers an epigenetic mechanism behind tissue sensitivity to DNA damage, carrying important translation implications.


Assuntos
Cromatina/metabolismo , Dano ao DNA , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Epigênese Genética , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas Proto-Oncogênicas/genética , Proteína Supressora de Tumor p53/genética , Animais , Apoptose , Cromatina/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Camundongos , Camundongos Transgênicos , Ligação Proteica , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Ubiquitinação
17.
Oncogene ; 37(16): 2150-2164, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29371679

RESUMO

Although ΔNp63 is known to promote cancer cell proliferation, the underlying mechanism behind its oncogenic function remains elusive. We report here a functional interplay between ΔNp63 and Δ133p53. These two proteins are co-overexpressed in a subset of human cancers and cooperate to promote cell proliferation. Mechanistically, Δ133p53 binds to ΔNp63 and utilizes its transactivation domain to upregulate GLUT1, GLUT4, and PGM expression driving glycolysis. While increased glycolysis provides cancer cells with anabolic metabolism critical for proliferation and survival, it can be harnessed for selective cancer cell killing. Indeed, we show that tumors overexpressing both ΔNp63 and Δ133p53 exhibit heightened sensitivity to vitamin C that accumulate to a lethal level due to accelerated uptake via overexpressed GLUT1. These observations offer a new therapeutic avenue that could be exploited for clinical applications.


Assuntos
Proliferação de Células/genética , Glicólise/genética , Neoplasias/patologia , Fatores de Transcrição/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Células A549 , Animais , Metabolismo dos Carboidratos/genética , Células Cultivadas , Códon sem Sentido , Feminino , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Células MCF-7 , Camundongos , Camundongos Nus , Proteínas Mutantes/genética , Proteínas Mutantes/fisiologia , Neoplasias/genética , Neoplasias/metabolismo , Isoformas de Proteínas/fisiologia , Fatores de Transcrição/genética , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor/genética
18.
Cell Stress ; 2(7): 176-180, 2018 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-31225484

RESUMO

Epigenetic regulation of gene expression in cells is a complex and dynamic process that remains incompletely understood. The architecture of the chromatin itself and its level of condensation can greatly impact the expression of genes as well as the sensitivity of the DNA to damage. The compact nature of heterochromatin typically results in gene silencing and resistance to DNA-damaging agents, while less compact euchromatin results in gene expression and increased sensitivity to injury. There are diverse ways in which the chromatin structure, and therefore the sensitivity of cells to damage, can be regulated, including post-translational modifications to both the histones within the chromatin and the DNA itself. These modifications are tightly controlled and correspond to various factors such as metabolism and cell cycle. When these processes are dysregulated, as in cancer cells, the chromatin structure is also altered, ultimately changing the gene expression profile as well as the susceptibility of cells to DNA-damaging agents commonly used for cancer treatments. Recent studies have shown that manipulating the various players involved in regulating post-translational modifications to chromatin and exploiting differences in metabolism may prove to be effective methods for modifying cancer and normal cell sensitivity to damaging agents. In this review we discuss various ways of regulating chromatin structure and how these changes can influence cellular sensitivity to damage as well as the implications of these relationships for improving the efficacy and safety of cancer treatments.

19.
J Mol Cell Biol ; 9(2): 154-165, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-27927748

RESUMO

Deregulation of the tyrosine kinase signalling is often associated with tumour progression and drug resistance, but its underlying mechanisms are only partly understood. In this study, we investigated the effects of the receptor tyrosine kinase AXL on the stability of the MDMX-MDM2 heterocomplex and the activity of p53 in melanoma cells. Our data demonstrated that AXL overexpression or activation through growth arrest-specific 6 (Gas6) ligand stimulation increases MDMX and MDM2 protein levels and decreases p53 activity. Upon activation, AXL stabilizes MDMX through a post-translational modification that involves phosphorylation of MDMX on the phosphosite Ser314, leading to increased affinity between MDMX and MDM2 and favouring MDMX nuclear translocation. Ser314 phosphorylation can also protect MDMX from MDM2-mediated degradation, leading to stabilization of the MDMX-MDM2 complex. We identified CDK4/6 and p38 MAPK as the two kinases mediating AXL-induced modulation of the MDMX-MDM2 complex, and demonstrated that suppression of AXL, either through siRNA silencing or pharmacological inhibition, increases expression levels of p53 target genes P21, MDM2, and PUMA, improves p53 pathway response to chemotherapy, and sensitizes cells to both Cisplatin and Vemurafenib. Our findings offer an insight into a novel signalling axis linking AXL to p53 and provide a potentially druggable pathway to restore p53 function in melanoma.


Assuntos
Melanoma/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Cisplatino/farmacologia , Quinases Ciclina-Dependentes/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Indóis/farmacologia , Melanoma/patologia , Fosforilação/efeitos dos fármacos , Fosfosserina/metabolismo , Estabilidade Proteica/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Sulfonamidas/farmacologia , Transcrição Gênica/efeitos dos fármacos , Proteína Supressora de Tumor p53/genética , Vemurafenib , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Receptor Tirosina Quinase Axl
20.
J Biol Chem ; 291(50): 25937-25949, 2016 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-27777309

RESUMO

Deregulated receptor tyrosine kinase (RTK) signaling is frequently associated with tumorigenesis and therapy resistance, but its underlying mechanisms still need to be elucidated. In this study, we have shown that the RTK human epidermal growth factor receptor 4 (Her4, also known as Erbb4) can inhibit the tumor suppressor p53 by regulating MDMX-mouse double minute 2 homolog (MDM2) complex stability. Upon activation by either overexpression of a constitutively active vector or ligand binding (Neuregulin-1), Her4 was able to stabilize the MDMX-MDM2 complex, resulting in suppression of p53 transcriptional activity, as shown by p53-responsive element-driven luciferase assay and mRNA levels of p53 target genes. Using a phospho-proteomics approach, we functionally identified a novel Her4-induced posttranslational modification on MDMX at Ser-314, a putative phosphorylation site for the CDK4/6 kinase. Remarkably, inhibition of Ser-314 phosphorylation either with Ser-to-Ala substitution or with a specific inhibitor of CDK4/6 kinase blocked Her4-induced stabilization of MDMX-MDM2 and rescued p53 activity. Our study offers insights into the mechanisms of deregulated RTK-induced carcinogenesis and provides the basis for the use of inhibitors targeting RTK-mediated signals for p53 restoration.


Assuntos
Proteínas Nucleares/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptor ErbB-4/metabolismo , Transcrição Gênica , Proteína Supressora de Tumor p53/metabolismo , Animais , Proteínas de Ciclo Celular , Quinase 4 Dependente de Ciclina/genética , Quinase 4 Dependente de Ciclina/metabolismo , Quinase 6 Dependente de Ciclina/genética , Quinase 6 Dependente de Ciclina/metabolismo , Células HEK293 , Humanos , Células MCF-7 , Camundongos , Fosforilação , Proteínas Proto-Oncogênicas c-mdm2/genética , Receptor ErbB-4/genética , Proteína Supressora de Tumor p53/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...