Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
2.
Res Sq ; 2023 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-36711701

RESUMO

Chronic inflammation is a major cause of cancer worldwide. Interleukin 33 (IL-33) is a critical initiator of cancer-prone chronic inflammation; however, its induction mechanism by the environmental causes of chronic inflammation is unknown. Herein, we demonstrate that Toll-like receptor (TLR)3/4-TBK1-IRF3 pathway activation links environmental insults to IL-33 induction in the skin and pancreas. FDA-approved drug library screen identified pitavastatin as an effective IL-33 inhibitor by blocking TBK1 membrane recruitment/activation through the mevalonate pathway inhibition. Accordingly, pitavastatin prevented chronic pancreatitis and its cancer sequela in an IL-33-dependent manner. IRF3-IL-33 axis was highly active in chronic pancreatitis and its associated pancreatic cancer in humans. Interestingly, pitavastatin use correlated with a significantly reduced risk of chronic pancreatitis and pancreatic cancer in patients. Our findings demonstrate that blocking the TBK1-IRF3 signaling pathway suppresses IL-33 expression and cancer-prone chronic inflammation. Statins present a safe and effective therapeutic strategy to prevent chronic inflammation and its cancer sequela.

3.
Leukemia ; 36(2): 426-437, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34465866

RESUMO

Persistence of malignant clones is a major determinant of adverse outcome in patients with hematologic malignancies. Despite the fact that the majority of patients with acute myeloid leukemia (AML) achieve complete remission after chemotherapy, a large proportion of them relapse as a result of residual malignant cells. These persistent clones have a competitive advantage and can re-establish disease. Therefore, targeting strategies that specifically diminish cell competition of malignant cells while leaving normal cells unaffected are clearly warranted. Recently, our group identified YBX1 as a mediator of disease persistence in JAK2-mutated myeloproliferative neoplasms. The role of YBX1 in AML, however, remained so far elusive. Here, inactivation of YBX1 confirms its role as an essential driver of leukemia development and maintenance. We identify its ability to amplify the translation of oncogenic transcripts, including MYC, by recruitment to polysomal chains. Genetic inactivation of YBX1 disrupts this regulatory circuit and displaces oncogenic drivers from polysomes, with subsequent depletion of protein levels. As a consequence, leukemia cells show reduced proliferation and are out-competed in vitro and in vivo, while normal cells remain largely unaffected. Collectively, these data establish YBX1 as a specific dependency and therapeutic target in AML that is essential for oncogenic protein expression.


Assuntos
Biomarcadores Tumorais/metabolismo , Competição entre as Células , Janus Quinase 2/metabolismo , Leucemia Mieloide Aguda/patologia , Mutação , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteína 1 de Ligação a Y-Box/metabolismo , Animais , Apoptose , Biomarcadores Tumorais/genética , Proliferação de Células , Humanos , Janus Quinase 2/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Camundongos , Prognóstico , Proteínas Proto-Oncogênicas c-myc/genética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto , Proteína 1 de Ligação a Y-Box/genética
4.
Sci Adv ; 7(24)2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34117055

RESUMO

Skin cancer is one of the most common types of cancer in the United States and worldwide. Topical products are effective for treating cancerous skin lesions when surgery is not feasible. However, current topical products induce severe irritation, light-sensitivity, burning, scaling, and inflammation. Using hyaluronic acid (HA), we engineered clinically translatable polymer-drug conjugates of doxorubicin and camptothecin termed, DOxorubicin and Camptothecin Tailored at Optimal Ratios (DOCTOR) for topical treatment of skin cancers. When compared to the clinical standard, Efudex, DOCTOR exhibited high cancer-cell killing specificity with superior safety to healthy skin cells. In vivo studies confirmed its efficacy in treating cancerous lesions without irritation or systemic absorption. When tested on patient-derived primary cells and live-skin explants, DOCTOR killed the cancer with a selectivity as high as 21-fold over healthy skin tissue from the same donor. Collectively, DOCTOR provides a safe and potent option for treating skin cancer in the clinic.


Assuntos
Dermatopatias , Neoplasias Cutâneas , Administração Tópica , Camptotecina/farmacologia , Doxorrubicina/farmacologia , Humanos , Ácido Hialurônico , Neoplasias Cutâneas/tratamento farmacológico
6.
Adv Drug Deliv Rev ; 153: 18-27, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-32526451

RESUMO

The process of aging influences every bodily organ and tissue, and those with rapid epithelial cell turnover, are particularly affected. The most visible of these, however, is the skin (including the epidermis), the largest human organ that provides a barrier to external insults, structure to the body and its movements, facilitates thermoregulation, harbors immune cells, and incorporates sensory neurons (including mechanoreceptors, nociceptors, and thermoreceptors). Skin aging has traditionally been categorized into intrinsic and extrinsic, with the latter nearly exclusively restricted to "photoaging," (i.e., aging due to exposure to solar or artificial ultraviolet radiation). However, both intrinsic and extrinsic aging share similar causes, including oxidative damage, telomere shortening, and mitochondrial senescence. Also, like other malignancies, the risk of malignant and nonmalignant lesions increases with age. Herein, we review the most recent findings in skin aging and nonmelanoma skin cancer, including addition to traditional and developing therapies.


Assuntos
Antineoplásicos/uso terapêutico , Fármacos Dermatológicos/uso terapêutico , Envelhecimento da Pele/fisiologia , Neoplasias Cutâneas/fisiopatologia , Administração Cutânea , Envelhecimento/fisiologia , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Técnicas Cosméticas , Fármacos Dermatológicos/administração & dosagem , Fármacos Dermatológicos/efeitos adversos , Fármacos Dermatológicos/farmacologia , Sistemas de Liberação de Medicamentos/métodos , Epigênese Genética/fisiologia , Humanos , Pele/fisiopatologia , Envelhecimento da Pele/efeitos dos fármacos , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/prevenção & controle , Raios Ultravioleta/efeitos adversos
7.
J Invest Dermatol ; 140(6): 1266-1275.e3, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31837302

RESUMO

Epigenetic regulation has a profound influence on stem cell fate during normal development in maintenance of physiologic tissue homeostasis. Here we report diminished ten-eleven translocation (TET) methylcytosine dioxygenase expression and loss of the DNA hydroxymethylation mark 5-hydroxymethylcytosine (5-hmC) in keratinocyte stem cells and transit amplifying cells in human psoriasis and in imiquimod-induced murine psoriasis. Loss of 5-hmC was associated with dysregulated keratinocyte stem cell kinetics, resulting in accumulation of nestin and FABP5-expressing transit amplifying cells to produce classic psoriatic epidermal architecture. Moreover, 5-hmC loss was accompanied by diminished TET1 and TET2 mRNA expression. Genome-wide mapping of epidermal 5-hmC in murine psoriasis revealed loci-specific loss of 5-hmC in genes regulating stem cell homeostasis, including MBD1, RTN1, STRN4, PRKD2, AKT1, and MAPKAP2, as well as those associated with RAR and Wnt/ß-catenin signaling pathways. In vitro restoration of TET expression by ascorbic acid was accomplished in cultured human keratinocyte stem cells to show similar Ca++-induced differentiation, resulting in increased 5-hmC levels and reduced nestin expression. To our knowledge, an epigenetic deficiency in psoriasis with relevance to stem cell dysregulation has not been previously reported. This observation raises the possibility that epigenetic modifiers that impact on the TET-5-hmC pathway may be a relevant approach of heretofore unappreciated therapeutic utility.


Assuntos
Metilação de DNA , Epigênese Genética , Psoríase/genética , 5-Metilcitosina/metabolismo , Animais , Proteínas de Ligação a DNA/metabolismo , Dioxigenases , Modelos Animais de Doenças , Regulação para Baixo , Feminino , Código das Histonas/genética , Humanos , Queratinócitos/patologia , Camundongos , Oxigenases de Função Mista/metabolismo , Cultura Primária de Células , Proteínas Proto-Oncogênicas/metabolismo , Psoríase/patologia , Análise de Sequência de DNA , Células-Tronco/patologia
9.
EMBO Rep ; 20(12): e48375, 2019 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-31668005

RESUMO

Outcomes for metastatic Ewing sarcoma and osteosarcoma are dismal and have not changed for decades. Oxidative stress attenuates melanoma metastasis, and melanoma cells must reduce oxidative stress to metastasize. We explored this in sarcomas by screening for oxidative stress sensitizers, which identified the class I HDAC inhibitor MS-275 as enhancing vulnerability to reactive oxygen species (ROS) in sarcoma cells. Mechanistically, MS-275 inhibits YB-1 deacetylation, decreasing its binding to 5'-UTRs of NFE2L2 encoding the antioxidant factor NRF2, thereby reducing NFE2L2 translation and synthesis of NRF2 to increase cellular ROS. By global acetylomics, MS-275 promotes rapid acetylation of the YB-1 RNA-binding protein at lysine-81, blocking binding and translational activation of NFE2L2, as well as known YB-1 mRNA targets, HIF1A, and the stress granule nucleator, G3BP1. MS-275 dramatically reduces sarcoma metastasis in vivo, but an MS-275-resistant YB-1K81-to-alanine mutant restores metastatic capacity and NRF2, HIF1α, and G3BP1 synthesis in MS-275-treated mice. These studies describe a novel function for MS-275 through enhanced YB-1 acetylation, thus inhibiting YB-1 translational control of key cytoprotective factors and its pro-metastatic activity.


Assuntos
Antineoplásicos/uso terapêutico , Benzamidas/uso terapêutico , Neoplasias Ósseas/tratamento farmacológico , Inibidores de Histona Desacetilases/uso terapêutico , Piridinas/uso terapêutico , Sarcoma de Ewing/tratamento farmacológico , Fatores de Transcrição/metabolismo , Acetilação , Animais , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Células Cultivadas , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Fator 2 Relacionado a NF-E2/metabolismo , Metástase Neoplásica , Estresse Oxidativo , Sarcoma de Ewing/metabolismo , Sarcoma de Ewing/patologia
10.
Nature ; 561(7723): 420, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30046103

RESUMO

This Letter is being retracted owing to issues with Fig. 1d and Supplementary Fig. 31b, and the unavailability of original data for these figures that raise concerns regarding the integrity of the figures. Nature published two previous corrections related to this Letter1,2. These issues in aggregate undermine the confidence in the integrity of this study. Authors Michael Foley, Monica Schenone, Nicola J. Tolliday, Todd R. Golub, Steven A. Carr, Alykhan F. Shamji, Andrew M. Stern and Stuart L. Schreiber agree with the Retraction. Authors Lakshmi Raj, Takao Ide, Aditi U. Gurkar, Anna Mandinova and Sam W. Lee disagree with the Retraction. Author Xiaoyu Li did not respond.

11.
Nat Commun ; 9(1): 1734, 2018 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-29712925

RESUMO

The integrity of stratified epithelia depends on the ability of progenitor cells to maintain a balance between proliferation and differentiation. While much is known about the transcriptional pathways underlying progenitor cells' behavior in the epidermis, the role of posttranscriptional regulation by mRNA binding proteins-a rate-limiting step in sculpting the proteome-remains poorly understood. Here we report that the RNA binding protein YBX1 (Y-box binding protein-1) is a critical effector of progenitors' function in the epidermis. YBX1 expression is restricted to the cycling keratinocyte progenitors in vivo and its genetic ablation leads to defects in the architecture of the skin. We further demonstrate that YBX1 negatively controls epidermal progenitor senescence by regulating the translation of a senescence-associated subset of cytokine mRNAs via their 3' untranslated regions. Our study establishes YBX1 as a posttranscriptional effector required for maintenance of epidermal homeostasis.


Assuntos
Queratinócitos/metabolismo , Processamento Pós-Transcricional do RNA , Células-Tronco/metabolismo , Fatores de Transcrição/genética , Proteína 1 de Ligação a Y-Box/genética , Regiões 3' não Traduzidas , Animais , Ciclo Celular/genética , Diferenciação Celular , Proliferação de Células , Senescência Celular , Quimiocina CXCL1/genética , Quimiocina CXCL1/metabolismo , Embrião de Mamíferos , Células Epidérmicas , Epiderme/crescimento & desenvolvimento , Epiderme/metabolismo , Humanos , Interleucina-8/genética , Interleucina-8/metabolismo , Queratinócitos/citologia , Camundongos , Cultura Primária de Células , Ligação Proteica , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Células-Tronco/citologia , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/metabolismo , Proteína 1 de Ligação a Y-Box/antagonistas & inibidores , Proteína 1 de Ligação a Y-Box/metabolismo
12.
Cell Chem Biol ; 25(2): 175-184.e4, 2018 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-29249695

RESUMO

Progressive kidney diseases affect approximately 500 million people worldwide. Podocytes are terminally differentiated cells of the kidney filter, the loss of which leads to disease progression and kidney failure. To date, there are no therapies to promote podocyte survival. Drug repurposing may therefore help accelerate the development of cures in an area of tremendous unmet need. In a newly developed high-throughput screening assay of podocyte viability, we identified the BRAFV600E inhibitor GDC-0879 and the adenylate cyclase agonist forskolin as podocyte-survival-promoting compounds. GDC-0879 protects podocytes from injury through paradoxical activation of the MEK/ERK pathway. Forskolin promotes podocyte survival by attenuating protein biosynthesis. Importantly, GDC-0879 and forskolin are shown to promote podocyte survival against an array of cellular stressors. This work reveals new therapeutic targets for much needed podocyte-protective therapies and provides insights into the use of GDC-0879-like molecules for the treatment of progressive kidney diseases.


Assuntos
Indenos/farmacologia , Nefropatias/tratamento farmacológico , Podócitos/efeitos dos fármacos , Pirazóis/farmacologia , Morte Celular/efeitos dos fármacos , Colforsina/química , Colforsina/farmacologia , Humanos , Indenos/química , Nefropatias/metabolismo , Nefropatias/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Podócitos/metabolismo , Podócitos/patologia , Pirazóis/química , Transdução de Sinais/efeitos dos fármacos , Tapsigargina/antagonistas & inibidores , Tapsigargina/farmacologia
13.
Science ; 357(6356): 1156-1160, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28912244

RESUMO

Growing evidence suggests that microbes can influence the efficacy of cancer therapies. By studying colon cancer models, we found that bacteria can metabolize the chemotherapeutic drug gemcitabine (2',2'-difluorodeoxycytidine) into its inactive form, 2',2'-difluorodeoxyuridine. Metabolism was dependent on the expression of a long isoform of the bacterial enzyme cytidine deaminase (CDDL), seen primarily in Gammaproteobacteria. In a colon cancer mouse model, gemcitabine resistance was induced by intratumor Gammaproteobacteria, dependent on bacterial CDDL expression, and abrogated by cotreatment with the antibiotic ciprofloxacin. Gemcitabine is commonly used to treat pancreatic ductal adenocarcinoma (PDAC), and we hypothesized that intratumor bacteria might contribute to drug resistance of these tumors. Consistent with this possibility, we found that of the 113 human PDACs that were tested, 86 (76%) were positive for bacteria, mainly Gammaproteobacteria.


Assuntos
Antimetabólitos Antineoplásicos/uso terapêutico , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/microbiologia , Desoxicitidina/análogos & derivados , Resistencia a Medicamentos Antineoplásicos , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/microbiologia , Animais , Neoplasias do Colo/microbiologia , Desoxicitidina/uso terapêutico , Gammaproteobacteria/isolamento & purificação , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Mycoplasma hyorhinis/isolamento & purificação , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/microbiologia , Gencitabina , Neoplasias Pancreáticas
15.
Nat Rev Immunol ; 16(12): 741-750, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27667712

RESUMO

Tumour-suppressor genes are indispensable for the maintenance of genomic integrity. Recently, several of these genes, including those encoding p53, PTEN, RB1 and ARF, have been implicated in immune responses and inflammatory diseases. In particular, the p53 tumour- suppressor pathway is involved in crucial aspects of tumour immunology and in homeostatic regulation of immune responses. Other studies have identified roles for p53 in various cellular processes, including metabolism and stem cell maintenance. Here, we discuss the emerging roles of p53 and other tumour-suppressor genes in tumour immunology, as well as in additional immunological settings, such as virus infection. This relatively unexplored area could yield important insights into the homeostatic control of immune cells in health and disease and facilitate the development of more effective immunotherapies. Consequently, tumour-suppressor genes are emerging as potential guardians of immune integrity.


Assuntos
Imunidade , Imunomodulação , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Imunidade Adaptativa , Animais , Autoimunidade , Suscetibilidade a Doenças , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Sistema Imunitário/citologia , Sistema Imunitário/imunologia , Sistema Imunitário/metabolismo , Imunidade Inata , Transdução de Sinais
16.
PLoS One ; 11(9): e0162686, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27622560

RESUMO

Deficiency of mitochondrial complex I is encountered in both rare and common diseases, but we have limited therapeutic options to treat this lesion to the oxidative phosphorylation system (OXPHOS). Idebenone and menadione are redox-active molecules capable of rescuing OXPHOS activity by engaging complex I-independent pathways of entry, often referred to as "complex I bypass." In the present study, we created a cellular model of complex I deficiency by using CRISPR genome editing to knock out Ndufa9 in mouse myoblasts, and utilized this cell line to develop a high-throughput screening platform for novel complex I bypass factors. We screened a library of ~40,000 natural product extracts and performed bioassay-guided fractionation on a subset of the top scoring hits. We isolated four plant-derived 1,4-naphthoquinone complex I bypass factors with structural similarity to menadione: chimaphilin and 3-chloro-chimaphilin from Chimaphila umbellata and dehydro-α-lapachone and dehydroiso-α-lapachone from Stereospermum euphoroides. We also tested a small number of structurally related naphthoquinones from commercial sources and identified two additional compounds with complex I bypass activity: 2-methoxy-1,4-naphthoquinone and 2-methoxy-3-methyl-1,4,-naphthoquinone. The six novel complex I bypass factors reported here expand this class of molecules and will be useful as tool compounds for investigating complex I disease biology.


Assuntos
Produtos Biológicos/farmacologia , Complexo I de Transporte de Elétrons/deficiência , Complexo I de Transporte de Elétrons/metabolismo , Naftoquinonas/farmacologia , Animais , Bignoniaceae/química , Produtos Biológicos/química , Sistemas CRISPR-Cas , Linhagem Celular , Avaliação Pré-Clínica de Medicamentos , Complexo I de Transporte de Elétrons/genética , Ericaceae/química , Edição de Genes , Técnicas de Inativação de Genes , Ensaios de Triagem em Larga Escala , Camundongos , Mioblastos/efeitos dos fármacos , Mioblastos/metabolismo , Naftoquinonas/química , Fosforilação Oxidativa , Smegmamorpha/metabolismo
17.
Cell Rep ; 16(1): 28-36, 2016 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-27320923

RESUMO

The Wnt/ß-catenin signaling pathway plays a major role in tissue homeostasis, and its dysregulation can lead to various human diseases. Aberrant activation of ß-catenin is oncogenic and is a critical driver in the development and progression of human cancers. Despite the significant potential of targeting the oncogenic ß-catenin pathway for cancer therapy, the development of specific inhibitors remains insufficient. Using a T cell factor (TCF)-dependent luciferase-reporter system, we screened for small-molecule compounds that act against Wnt/ß-catenin signaling and identified MSAB (methyl 3-{[(4-methylphenyl)sulfonyl]amino}benzoate) as a selective inhibitor of Wnt/ß-catenin signaling. MSAB shows potent anti-tumor effects selectively on Wnt-dependent cancer cells in vitro and in mouse cancer models. MSAB binds to ß-catenin, promoting its degradation, and specifically downregulates Wnt/ß-catenin target genes. Our findings might represent an effective therapeutic strategy for cancers addicted to the Wnt/ß-catenin signaling pathway.


Assuntos
Benzoatos/farmacologia , Oncogenes , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Sulfonamidas/farmacologia , Via de Sinalização Wnt/efeitos dos fármacos , beta Catenina/metabolismo , meta-Aminobenzoatos/farmacologia , Animais , Benzoatos/química , Linhagem Celular Tumoral , Camundongos , Sulfonamidas/química , Ensaios Antitumorais Modelo de Xenoenxerto , meta-Aminobenzoatos/química
18.
J Invest Dermatol ; 136(3): 696-705, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26739095

RESUMO

Seborrheic keratoses (SKs) are common benign skin tumors that share many morphological features with their malignant counterpart, squamous cell carcinoma. SKs frequently have acquired oncogenic mutations in the receptor tyrosine kinase/phosphatidylinositol 3-kinase/Akt signaling cascade. We developed a reliable culture system to study SKs in vitro and screened these cells using a library of selective kinase inhibitors to evaluate effects on cell survival. These benign tumors are sensitive to inhibition by ATP-competitive Akt inhibitors, including A-443654 and GSK690693. RNA interference-mediated Akt suppression mimicked the effects of enzyme inhibition in cultured cells. Akt inhibition suppressed phosphorylation of downstream targets of Akt kinase that are critical for cell survival, including MDM2 and FOXO3a, and induced apoptosis. Cell death was also dependent on p53, mutations in which, although common in cutaneous squamous cell carcinoma, have not been identified in SKs. Intact explants of SKs were also sensitive to Akt inhibition. In addition to the obvious therapeutic implications of these findings, identifying the signaling characteristics that differentiate benign and malignant tumors may inform our understanding of the malignant state.


Assuntos
Sobrevivência Celular/genética , Transformação Celular Neoplásica/patologia , Ceratose Seborreica/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Apoptose/genética , Western Blotting , Carcinoma de Células Escamosas/patologia , Células Cultivadas , Análise Mutacional de DNA , Humanos , Imuno-Histoquímica , Ceratose Seborreica/genética , Neoplasias Cutâneas/patologia
20.
Chem Biol ; 22(9): 1206-16, 2015 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-26320861

RESUMO

TP53 is the most frequently mutated gene in human cancer, and small-molecule reactivation of mutant p53 function represents an important anticancer strategy. A cell-based, high-throughput small-molecule screen identified chetomin (CTM) as a mutant p53 R175H reactivator. CTM enabled p53 to transactivate target genes, restored MDM2 negative regulation, and selectively inhibited the growth of cancer cells harboring mutant p53 R175H in vitro and in vivo. We found that CTM binds to Hsp40 and increases the binding capacity of Hsp40 to the p53 R175H mutant protein, causing a potential conformational change to a wild-type-like p53. Thus, CTM acts as a specific reactivator of the p53 R175H mutant form through Hsp40. These results provide new insights into the mechanism of reactivation of this specific p53 mutant.


Assuntos
Antineoplásicos/farmacologia , Dissulfetos/farmacologia , Proteínas de Choque Térmico HSP40/metabolismo , Alcaloides Indólicos/farmacologia , Bibliotecas de Moléculas Pequenas/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Animais , Antineoplásicos/química , Linhagem Celular Tumoral , Dissulfetos/química , Ensaios de Seleção de Medicamentos Antitumorais , Células HCT116 , Ensaios de Triagem em Larga Escala , Humanos , Alcaloides Indólicos/química , Camundongos , Camundongos Nus , Mutação , Bibliotecas de Moléculas Pequenas/química , Proteína Supressora de Tumor p53/genética , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...