Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
BMC Cancer ; 22(1): 1073, 2022 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-36258187

RESUMEN

BACKGROUND: Paclitaxel (PTX), a first-line therapy for triple negative breast cancers (TNBC) induces anti-tumor activity by microtubule stabilization and inhibition of cell division. Its dose-limiting toxicity and short half-life, however, pose clinical challenges underscoring the need for strategies that increase its efficiency. RAD6, a E2 ubiquitin conjugating enzyme, is associated with centrosomes at all phases of cell cycle. Constitutive overexpression of the RAD6B homolog in normal breast cells induces centrosome amplification and multipolar spindle formation, indicating its importance in centrosome regulation. METHODS: TNBC centrosome numbers were scored by pericentrin immunostaining. PTX sensitivities and interactions with SMI#9, a RAD6-selective small molecule inhibitor, on TNBC cell survival were analyzed by MTT and colony forming assays and an isogenic MDA-MB-468 TNBC model of PTX resistance. The molecular mechanisms underlying PTX and SMI#9 induced cytotoxicity were determined by flow cytometry, immunoblot analysis of cyclin B1 and microtubule associated protein TAU, and dual immunofluorescence staining of TAU and α-tubulin. RESULTS: Our data show aberrant centrosome numbers and that PTX sensitivities are not correlated with TNBC BRCA1 status. Combining PTX with SMI#9 synergistically enhances PTX sensitivities of BRCA1 wild-type and mutant TNBC cells. Whereas SMI#9/PTX combination treatment increased cyclin B1 levels in MDA-MB-468 cells, it induced cyclin B1 loss in HCC1937 cells with accumulation of reproductively dead giant cells, a characteristic of mitotic catastrophe. Cell cycle analysis revealed drug-induced accumulation of tetraploid cells in S and G2/M phases, and robust increases in cells with 4 N DNA content in HCC1937 cells. TAU overexpression is associated with reduced PTX efficacy. Among the six TAU isoforms, both SMI#9 and PTX downregulated 1N3R TAU in MDA-MB-468 and HCC1937 cells, suggesting a common mechanism of 1N3R regulation. Dual TAU and α-tubulin immunostaining showed that SMI#9 induces monopolar mitotic spindles. Using the isogenic model of PTX resistance, we show that SMI#9 treatment restores PTX sensitivity. CONCLUSIONS: These data support a common mechanism of microtubule regulation by SMI#9 and PTX and suggest that combining PTX with RAD6 inhibitor may be beneficial for increasing TNBC sensitivities to PTX and alleviating toxicity. This study demonstrates a new role for RAD6 in regulating microtubule dynamics.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Humanos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Ciclina B1/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Línea Celular Tumoral , Tubulina (Proteína) , Centrosoma/metabolismo , Proteínas Asociadas a Microtúbulos , ADN/uso terapéutico
2.
Oncotarget ; 13: 534-550, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35309869

RESUMEN

The accepted notion of dNTP transport following cytoplasmic biosynthesis is 'facilitated diffusion'; however, whether this alone is sufficient for moving dNTPs for DNA synthesis remains an open question. The data presented here show that the MYH9 gene encoded heavy chain of non-muscle myosin IIA binds dNTPs potentially serving as a 'reservoir'. Pull-down assays showed that MYH9 present in the cytoplasmic, mitochondrial and nuclear compartments bind to DNA and this interaction is inhibited by dNTPs and 2-deoxyribose-5-phosphate (dRP) suggesting that MYH9-DNA binding is mediated via pentose sugar recognition. Direct dNTP-MYH9 binding was demonstrated by ELISA and a novel PCR-based method, which showed that all dNTPs bind to MYH9 with varying efficiencies. Cellular thermal shift assays showed that MYH9 thermal stability is enhanced by dNTPs. MYH9 siRNA transfection or treatment with myosin II selective inhibitors ML7 or blebbistatin decreased cell proliferation compared to controls. EdU labeling and cell cycle analysis by flow cytometry confirmed MYH9 siRNA and myosin II inhibitors decreased progression to S-phase with accumulation of cells in G0/G1 phase. Taken together, our data suggest a novel role for MYH9 in dNTP binding and DNA synthesis.


Asunto(s)
Cadenas Pesadas de Miosina , Miosina Tipo IIA no Muscular , Proteínas del Citoesqueleto , ADN/genética , Desoxirribosa , Humanos , Proteínas Motoras Moleculares/genética , Proteínas Motoras Moleculares/metabolismo , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Miosina Tipo II , Miosina Tipo IIA no Muscular/genética , Miosina Tipo IIA no Muscular/metabolismo , Pentosas , Fosfatos , ARN Interferente Pequeño , Azúcares
3.
Am J Pathol ; 191(2): 368-384, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33181138

RESUMEN

Canonical Wnt signaling is critical for melanocyte lineage commitment and melanoma development. RAD6B, a ubiquitin-conjugating enzyme critical for translesion DNA synthesis, potentiates ß-catenin stability/activity by inducing proteasome-insensitive polyubiquitination. RAD6B expression is induced by ß-catenin, triggering a positive feedback loop between the two proteins. RAD6B function in melanoma development/progression was investigated by targeting RAD6B using CrispR/Cas9 or an RAD6-selective small-molecule inhibitor #9 (SMI#9). SMI#9 treatment inhibited melanoma cell proliferation but not normal melanocytes. RAD6B knockout or inhibition in metastatic melanoma cells downregulated ß-catenin, ß-catenin-regulated microphthalmia-associated transcription factor (MITF), sex-determining region Y-box 10, vimentin proteins, and MITF-regulated melan A. RAD6B knockout or inhibition decreased migration/invasion, tumor growth, and lung metastasis. RNA-sequencing and stem cell pathway real-time RT-PCR analysis revealed profound reductions in WNT1 expressions in RAD6B knockout M14 cells compared with control. Expression levels of ß-catenin-regulated genes VIM, MITF-M, melan A, and TYRP1 (a tyrosinase family member critical for melanin biosynthesis) were reduced in RAD6B knockout cells. Pathway analysis identified gene networks regulating stem cell pluripotency, Wnt signaling, melanocyte development, pigmentation signaling, and protein ubiquitination, besides DNA damage response signaling, as being impacted by RAD6B gene disruption. These data reveal an important and early role for RAD6B in melanoma development besides its bonafide translesion DNA synthesis function, and suggest that targeting RAD6B may provide a novel strategy to treat melanomas with dysregulated canonical Wnt signaling.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/fisiología , Melanoma/metabolismo , Melanoma/patología , Enzimas Ubiquitina-Conjugadoras/metabolismo , Vía de Señalización Wnt/fisiología , Animales , Línea Celular , Xenoinjertos , Humanos , Ratones , Ratones Desnudos , Fenotipo
4.
Cells ; 8(11)2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31683936

RESUMEN

Rad6B, a principal component of the translesion synthesis pathway, and activator of canonical Wnt signaling, plays an essential role in cutaneous melanoma development and progression. As Rad6 is encoded by two genes, namely, UBE2A (RAD6A) and UBE2B (RAD6B), in humans, we compared their expressions in melanomas and normal melanocytes. While both genes are weakly expressed in normal melanocytes, Rad6B is more robustly expressed in melanoma lines and patient-derived metastatic melanomas than RAD6A. The characterization of RAD6B transcripts revealed coexpression of various splice variants representing truncated or modified functional versions of wild-type RAD6B in melanomas, but not in normal melanocytes. Notably, two RAD6B isoforms with intact catalytic domains, RAD6BΔexon4 and RAD6Bintron5ins, were identified. We confirmed that RAD6BΔexon4 and RAD6Bintron5ins variants are expressed as 14 and 15 kDa proteins, respectively, with functional in vivo ubiquitin conjugating activity. Whole exome sequence analysis of 30 patient-derived melanomas showed RAD6B variants coexpressed with wild-type RAD6B in all samples analyzed, and RAD6Bintron5ins variants were found in half the cases. These variants constitute the majority of the RAD6B transcriptome in contrast to RAD6A, which was predominantly wild-type. The expression of functional RAD6B variants only in melanomas reveals RAD6B's molecular heterogeneity and its association with melanoma pathogenesis.


Asunto(s)
Melanoma/genética , Neoplasias Cutáneas/genética , Enzimas Ubiquitina-Conjugadoras/genética , Adulto , Anciano , Anciano de 80 o más Años , Empalme Alternativo , Línea Celular , Reparación del ADN , Replicación del ADN , Femenino , Humanos , Masculino , Melanoma/diagnóstico , Melanoma/metabolismo , Persona de Mediana Edad , Neoplasias Cutáneas/diagnóstico , Neoplasias Cutáneas/metabolismo , Transcripción Genética , Transcriptoma , Enzimas Ubiquitina-Conjugadoras/metabolismo , Secuenciación del Exoma/métodos , Vía de Señalización Wnt , beta Catenina/metabolismo , Melanoma Cutáneo Maligno
5.
Oncotarget ; 9(81): 35286, 2018 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-30443298

RESUMEN

[This corrects the article DOI: 10.18632/oncotarget.1795.].

6.
Nanomedicine ; 12(3): 745-757, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26563438

RESUMEN

We recently developed a small molecule inhibitor SMI#9 for Rad6, a protein overexpressed in aggressive breast cancers and involved in DNA damage tolerance. SMI#9 induces cytotoxicity in cancerous cells but spares normal breast cells; however, its therapeutic efficacy is limited by poor solubility. Here we chemically modified SMI#9 to enable its conjugation and hydrolysis from gold nanoparticle (GNP). SMI#9-GNP and parent SMI#9 activities were compared in mesenchymal and basal triple negative breast cancer (TNBC) subtype cells. Whereas SMI#9 is cytotoxic to all TNBC cells, SMI#9-GNP is endocytosed and cytotoxic only in mesenchymal TNBC cells. SMI#9-GNP endocytosis in basal TNBCs is compromised by aggregation. However, when combined with cisplatin, SMI#9-GNP is imported and synergistically increases cisplatin sensitivity. Like SMI#9, SMI#9-GNP spares normal breast cells. The released SMI#9 is active and induces cell death via mitochondrial dysfunction and PARP-1 stabilization/hyperactivation. This work signifies the development of a nanotechnology-based Rad6-targeting therapy for TNBCs. FROM THE CLINICAL EDITOR: Protein Rad6 is overexpressed in breast cancer cells and its blockade may provide a new treatment against 3N breast cancer. The authors conjugated a small molecule inhibitor SMI#9 for Rad6 to gold nanoparticles in this study and showed that this new formulation specifically targeted chemo-resistant breast cancer cells and highlighted the importance of nanotechnology in drug carrier development.


Asunto(s)
Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/farmacología , Oro/química , Nanopartículas del Metal/química , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Enzimas Ubiquitina-Conjugadoras/antagonistas & inhibidores , Mama/efectos de los fármacos , Mama/metabolismo , Mama/patología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Inhibidores Enzimáticos/química , Femenino , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/patología , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Enzimas Ubiquitina-Conjugadoras/metabolismo
7.
Mutat Res Rev Mutat Res ; 763: 258-66, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25795124

RESUMEN

Bifunctional alkylating and platinum based drugs are chemotherapeutic agents used to treat cancer. These agents induce DNA adducts via formation of intrastrand or interstrand (ICL) DNA crosslinks, and DNA lesions of the ICL type are particularly toxic as they block DNA replication and/or DNA transcription. However, the therapeutic efficacies of these drugs are frequently limited due to the cancer cell's enhanced ability to repair and tolerate these toxic DNA lesions. This ability to tolerate and survive the DNA damage is accomplished by a set of specialized low fidelity DNA polymerases called translesion synthesis (TLS) polymerases since high fidelity DNA polymerases are unable to replicate the damaged DNA template. TLS is a crucial initial step in ICL repair as it synthesizes DNA across the lesion thus preparing the damaged DNA template for repair by the homologous recombination (HR) pathway and Fanconi anemia (FA) network, processes critical for ICL repair. Here we review the molecular features and functional roles of TLS polymerases, discuss the collaborative interactions and cross-regulation of the TLS DNA damage tolerance pathway, the FA network and the BRCA-dependent HRR pathway, and the impact of TLS hyperactivation on development of chemoresistance. Finally, since TLS hyperactivation results from overexpression of Rad6/Rad18 ubiquitinating enzymes (fundamental components of the TLS pathway), increased PCNA ubiquitination, and/or increased recruitment of TLS polymerases, the potential benefits of selectively targeting critical components of the TLS pathway for enhancing anti-cancer therapeutic efficacy and curtailing chemotherapy-induced mutagenesis are also discussed.


Asunto(s)
Reparación del ADN , ADN Polimerasa Dirigida por ADN/metabolismo , ADN/biosíntesis , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Neoplasias/genética , Antineoplásicos/efectos adversos , Daño del ADN , Resistencia a Antineoplásicos , Humanos , Mutágenos/efectos adversos , Neoplasias/tratamiento farmacológico , Reparación del ADN por Recombinación , Transducción de Señal
8.
J Skin Cancer ; 2014: 439205, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24891954

RESUMEN

We have previously demonstrated that Rad6 and ß -catenin enhance each other's expression through a positive feedback loop to promote breast cancer development/progression. While ß -catenin has been implicated in melanoma pathogenesis, Rad6 function has not been investigated. Here, we examined the relationship between Rad6 and ß -catenin in melanoma development and progression. Eighty-eight cutaneous tumors, 30 nevi, 29 primary melanoma, and 29 metastatic melanomas, were immunostained with anti- ß -catenin and anti-Rad6 antibodies. Strong expression of Rad6 was observed in only 27% of nevi as compared to 100% of primary and 96% of metastatic melanomas. ß -Catenin was strongly expressed in 97% of primary and 93% of metastatic melanomas, and unlike Rad6, in 93% of nevi. None of the tumors expressed nuclear ß -catenin. ß -Catenin was exclusively localized on the cell membrane of 55% of primary, 62% of metastatic melanomas, and only 10% of nevi. Cytoplasmic ß -catenin was detected in 90% of nevi, 17% of primary, and 8% of metastatic melanoma, whereas 28% of primary and 30% of metastatic melanomas exhibited ß -catenin at both locations. These data suggest that melanoma development and progression are associated with Rad6 upregulation and membranous redistribution of ß -catenin and that ß -catenin and Rad6 play independent roles in melanoma development.

9.
Transl Oncol ; 2014 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-24831578

RESUMEN

Melanoma is the leading cause of death from skin cancer in industrialized countries. Several melanoma-related biomarkers and signaling pathways have been identified; however, their relevance to melanoma development/progression or to clinical outcome remains to be established. Aberrant activation of Wnt/ß-catenin pathway is implicated in various cancers including melanoma. We have previously demonstrated Rad6, an ubiquitin-conjugating enzyme, as an important mediator of ß-catenin stability in breast cancer cells. Similar to breast cancer, ß-catenin-activating mutations are rare in melanomas, and since ß-catenin signaling is implicated in melanoma, we examined the relationship between ß-catenin levels/activity and expression of ß-catenin transcriptional targets Rad6 and microphthalmia-associated transcription factor-M (Mitf-M) in melanoma cell models, and expression of Rad6, ß-catenin, and Melan-A in nevi and cutaneous melanoma tissue specimens. Our data show that Rad6 is only weakly expressed in normal human melanocytes but is overexpressed in melanoma lines. Unlike Mitf-M, Rad6 overexpression in melanoma lines is positively associated with high molecular weight ß-catenin protein levels and ß-catenin transcriptional activity. Double-immunofluorescence staining of Rad6 and Melan-A in melanoma tissue microarray showed that histological diagnosis of melanoma is significantly associated with Rad6/Melan-A dual positivity in the melanoma group compared to the nevi group (P=.0029). In contrast to strong ß-catenin expression in normal and tumor areas of superficial spreading malignant melanoma (SSMM), Rad6 expression is undetectable in normal areas and Rad6 expression increases coincide with increased Melan-A in the transformed regions of SSMM. These data suggest a role for Rad6 in melanoma pathogenesis and that Rad6 expression status may serve as an early marker for melanoma development.

10.
Oncotarget ; 5(4): 1101-10, 2014 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-24658355

RESUMEN

Increased activation of ERK signaling has been reported in breast cancer models of acquired tamoxifen resistance. Here, we examined the expression of Mitogen-Activated Protein Kinase Phosphatases (MKPs) 1 and 2 following tamoxifen treatment and the effects of MKP-1/MKP-2 overexpression on tamoxifen sensitivity. Treatment of MCF7 breast cancer cells with tamoxifen increased MKP-2, but not MKP-1, protein levels. Overexpression of MKP-1 or MKP-2 inhibited estrogen-induced MCF7 cell proliferation compared to vector controls. MCF7-MKP-2 cells displayed significantly increased sensitivity to tamoxifen as compared to vector control or MCF7-MKP-1 cells. MKP-1 or MKP-2 overexpression eliminated ERK1/2 phosphorylation, suggesting that decreases in estrogen-induced proliferation of MKP-1 and MKP-2 overexpressing cells are due to ERK1/2 dephosphorylation. JNK1/2 activation was not detectable in any of these cells. These data suggest that tamoxifen-induced death of these cells is not dependent upon JNK signaling, but rather that ERK is the major MAPK driving their proliferation. MCF7-TAMR cells express higher levels of MKP-2 mRNA and protein than MCF7 cells. MKP-2 and phospho-ERK1/2 proteins are constitutively expressed in MCF7-TAMR cells, and activated JNK1/2 is not detectable. These data suggest that MKP-2 rather than MKP-1 is tamoxifen-regulated and that the elevated expression of MKP-2 in MCF7-TAMR cells potentially functions to restore tamoxifen sensitivity.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/enzimología , Fosfatasa 1 de Especificidad Dual/metabolismo , Fosfatasas de Especificidad Dual/metabolismo , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/metabolismo , Tamoxifeno/farmacología , Fosfatasas de Especificidad Dual/genética , Femenino , Humanos , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/genética
11.
Bioorg Med Chem Lett ; 23(24): 6886-9, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24153206

RESUMEN

Series of substituted 4,6-diamino-1,3,5-triazine-2-carbohydrazides and -carboxamides have been synthesised, based on molecular modelling of candidate structures related to the previously reported Rad6B-inhibitory diamino-triazinylmethyl benzoate anticancer agents TZ8 and TZ9. Synthesis of the target compounds was readily accomplished in two steps from aryl biguanides via reaction of phenylhydrazine or benzylamines with key 4-amino-6-(arylamino)-1,3,5-triazine-2-carboxylate intermediates. These new triazine derivatives were tested for in vitro anticancer activity against the Rad6B expressing human breast cancer cell lines MDA-MB-231 and MCF-7. Active compounds, such as the triazinyl-carbohydrazides 3a-e, were found to exhibit low micromolar IC50 values particularly in the Rad6B-overexpressing MDA-MB-231 cell line.


Asunto(s)
Amidas/síntesis química , Amidas/farmacología , Diseño de Fármacos , Hidrazinas/síntesis química , Hidrazinas/farmacología , Triazinas/química , Amidas/química , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Sitios de Unión , Dominio Catalítico , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Hidrazinas/química , Células MCF-7 , Simulación del Acoplamiento Molecular , Relación Estructura-Actividad , Enzimas Ubiquitina-Conjugadoras/antagonistas & inhibidores , Enzimas Ubiquitina-Conjugadoras/metabolismo
12.
Oncotarget ; 4(2): 231-41, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23548208

RESUMEN

Basal breast cancer comprises ~15% of invasive ductal breast cancers, and presents as high-grade lesions with aggressive clinical behavior. Basal breast carcinomas express p63 and cytokeratin 5 (CK5) antigens characteristic of the myoepithelial lineage, and typically lack Her2/neu and hormone receptor expression. However, there is limited data about the precursor lesions from which they emerge. Here we wished to determine whether comedo-ductal carcinoma in situ (comedo- DCIS), a high-risk in situ breast lesion, serve as precursors for basal-like breast cancer. To determine this link, p63, CK5, Her2/neu, epidermal growth factor receptor (EGFR), estrogen receptor (ER) and progesterone receptor (PgR) expression were analyzed by immunohistochemistry in 17 clinical comedo- and 12 noncomedo-DCIS cases, and in tumors derived from unfractionated and CK5-overexpressing subpopulation (MCF10DCIS.com-CK5(high)) of MCF10DCIS.com cells, a model representative of clinical comedo-DCIS. p63 and Her2/neu coexpression was analyzed by immunofluorescence double labeling. A novel p63/CK5/Her2/neu expressing subpopulation of cells that are ER-/PgR-/EGFR- were identified in the myoepithelial and luminal areas of clinical comedo-DCIS and tumors derived from unfractionated MCF10DCIS.com and MCF10DCIS.com-CK5(high) cells. These data suggest that p63 and Her2/neu expressors may share a common precursor intermediate. P63, but not Her2/neu, expression was significantly associated (P = 0.038) with microinvasion/recurrence of clinical comedo-DCIS, and simultaneous expression of p63 and Her2/neu was marginally associated (P = 0.067) with comedo-DCIS. These data suggest that p63/Her2/neu expressing precursor intermediate in comedo-DCIS may provide a cellular basis for emergence of p63+/Her2/neu- or p63+/Her2/neu+ basal-like breast cancer, and that p63/Her2/neu coexpression may serve as biomarkers for identification of this subgroup of basal-like breast cancers.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Carcinoma Intraductal no Infiltrante/metabolismo , Carcinoma Intraductal no Infiltrante/patología , Proteínas de la Membrana/biosíntesis , Receptor ErbB-2/biosíntesis , Animales , Neoplasias de la Mama/genética , Carcinoma Intraductal no Infiltrante/genética , Femenino , Xenoinjertos , Humanos , Inmunohistoquímica , Proteínas de la Membrana/genética , Ratones , Ratones Desnudos , Pronóstico , Receptor ErbB-2/genética
13.
Cancer Biol Ther ; 14(5): 417-27, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23380593

RESUMEN

Ocimum genus (a.k.a holy basil or tulsi) is a dietary herb used for its multiple beneficial pharmacologic properties including anti-cancer activity. Here we show that crude extract of Ocimum gratissimum (OG) and its hydrophobic and hydrophilic fractions (HB and HL) differentially inhibit breast cancer cell chemotaxis and chemoinvasion in vitro and retard tumor growth and temporal progression of MCF10ADCIS.com xenografts, a model of human breast comedo-ductal carcinoma in situ (comedo-DCIS). OG-induced inhibition of tumor growth was associated with decreases in basement membrane disintegration, angiogenesis and MMP-2 and MMP-9 activities as confirmed by in situ gelatin zymography and cleavage of galectin-3. There was also decrease in MMP-2 and MMP-9 activities in the conditioned media of OG-treated MCF10AT1 and MCF10AT1-EIII8 premalignant human breast cancer cells as compared with control. The MMP-2 and MMP-9 inhibitory activities of OG were verified in vitro using gelatin, a synthetic fluorogenic peptide and recombinant galectin-3 as MMP substrates. Mice fed on OG-supplemented drinking water showed no adverse effects compared with control. These data suggest that OG is non-toxic and that the anti-cancer therapeutic activity of OG may in part be contributed by its MMP inhibitory activity.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Ocimum/química , Extractos Vegetales/farmacología , Animales , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/tratamiento farmacológico , Carcinoma Ductal de Mama/enzimología , Carcinoma Ductal de Mama/patología , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Humanos , Inmunohistoquímica , Inhibidores de la Metaloproteinasa de la Matriz/química , Ratones , Ratones Desnudos , Distribución Aleatoria , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Mol Cancer Ther ; 12(4): 373-83, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23339190

RESUMEN

Protein ubiquitination is important for cell signaling, DNA repair, and proteasomal degradation, and it is not surprising that alterations in ubiquitination occur frequently in cancer. Ubiquitin-conjugating enzymes (E2) mediate ubiquitination by selective interactions with ubiquitin-activating (E1) and ubiquitin ligase (E3) enzymes, and thus selective E2 small molecule inhibitor (SMI) will provide specificity unattainable with proteasome inhibitors. Here we describe synthesis and functional characterization of the first SMIs of human E2 Rad6B, a fundamental component of translesion synthesis DNA repair. A pharmacophore model for consensus E2 ubiquitin-binding sites was generated for virtual screening to identify E2 inhibitor candidates. Twelve triazine (TZ) analogs screened in silico by molecular docking to the Rad6B X-ray structure were verified by their effect on Rad6B ubiquitination of histone H2A. TZs #8 and 9 docked to the Rad6B catalytic site with highest complementarity. TZs #1, 2, 8, and 9 inhibited Rad6B-ubiquitin thioester formation and subsequent ubiquitin transfer to histone H2A. SMI #9 inhibition of Rad6 was selective as BCA2 ubiquitination by E2 UbcH5 was unaffected by SMI #9. SMI #9 more potently inhibited proliferation, colony formation, and migration than SMI #8, and induced MDA-MB-231 breast cancer cell G2-M arrest and apoptosis. Ubiquitination assays using Rad6 immunoprecipitated from SMI #8- or 9-treated cells confirmed inhibition of endogenous Rad6 activity. Consistent with our previous data showing Rad6B-mediated polyubiquitination stabilizes ß-catenin, MDA-MB-231 treatment with SMIs #8 or 9 decreased ß-catenin protein levels. Together these results describe identification of the first Rad6 SMIs.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Enzimas Ubiquitina-Conjugadoras/antagonistas & inhibidores , Secuencia de Bases , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Secuencia Conservada , Diseño de Fármacos , Inhibidores Enzimáticos/química , Humanos , Conformación Molecular , Simulación del Acoplamiento Molecular , Datos de Secuencia Molecular , Triazinas/química , Triazinas/farmacología , Enzimas Ubiquitina-Conjugadoras/química , Enzimas Ubiquitina-Conjugadoras/metabolismo , beta Catenina/metabolismo
15.
Biochim Biophys Acta ; 1823(10): 1686-96, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22705350

RESUMEN

The ubiquitin conjugating enzyme Rad6B is overexpressed in breast cancer and induces ß-catenin transcriptional activation and stabilization via K63-linked polyubiquitination. Here we identify ß-catenin and Rad6B interacting regions, identify potential Rad6B ubiquitination sites in ß-catenin, and characterize their breast cancer tissue expression. ß-catenin and Rad6B colocalize in breast carcinoma and coimmunoprecipitate from MDA-MB-231 cells. Pull-down assays using GST-ß-catenin and His-Rad6B deletion mutants identified amino acids 131-181 and 50-116, respectively, as necessary for their interaction. Ubiquitination assays using ß-catenin deletion mutants mapped Rad6B-induced ubiquitination within ß-catenin 181-422 encompassing Armadillo repeats 2-7. Lysine to arginine mutations within repeats 5-7 identified K394 as the major Rad6B ubiquitination site in vitro and in vivo, and confirmed by Rad6B ubiquitination of a ß-catenin peptide encompassing K394. Ubiquitination of wild type- but not K394R-ß-catenin was decreased by Rad6B silencing. Compared to wild type-, K312R-, K335R-, K345R-, or K354R-ß-catenin, K394R mutation caused ~50% drop in TOP/Flash activity in Wnt-silent MCF-7 cells. Consistent with these data, expression of Rad6B, itself a ß-catenin/TCF transcriptional target, was also reduced in K394R-ß-catenin transfected cells. Steady-state K394R-ß-catenin levels are decreased compared to wild type-ß-catenin. The decreased expression is not due to proteasomal degradation as treatment with MG132 failed to rescue its levels. Lymph node-positive breast carcinomas express higher levels of Rad6 protein and Rad6 activity, and K63-linked ubiquitinated ß-catenin than reduction mammoplasties. These data suggest that K394 is a novel site of ß-catenin ubiquitination that may be important for the stability and activity of ß-catenin in breast cancer.


Asunto(s)
Lisina/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitinación , beta Catenina/metabolismo , Proteínas del Dominio Armadillo/química , Proteínas del Dominio Armadillo/genética , Proteínas del Dominio Armadillo/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Dominio Catalítico , Línea Celular Tumoral , Femenino , Silenciador del Gen , Humanos , Mutación/genética , Invasividad Neoplásica , Unión Proteica , Transcripción Genética , Ubiquitina/metabolismo , Enzimas Ubiquitina-Conjugadoras/genética , Regulación hacia Arriba/genética , beta Catenina/química , beta Catenina/genética
16.
Curr Cancer Drug Targets ; 11(5): 613-23, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21486215

RESUMEN

The success of current treatment strategies is limited by the development of therapy resistance as evidenced by recurrence of the primary tumor or distant metastasis. Eradication of primary and metastatic disease requires interventions at both the cancer cell and tumor microenvironment levels. In this review, we will discuss mechanisms that are intrinsic to cancer cells, and those that are mediated by the tumor microenvironment as contributors to drug resistance. Mechanisms contributing to multidrug resistance phenotype and the challenges facing molecular targeted therapy are discussed. The DNA damage tolerance pathway confers tolerance to a variety of structurally and functionally unrelated drugs. A rationale for targeting the DNA damage tolerance pathway as a novel tool for overcoming drug resistance is discussed. We have also addressed the need for employing clinically relevant model systems for performing drug sensitivity evaluations. These model systems must take into account the three-dimensional organization and in vivo relationship of tumor with its microenvironment. Such integrative efforts would not only yield a more global understanding of the tumor- and microenvironment-derived mechanisms involved in emergence of drug resistance but would also provide novel therapeutic targets that will disrupt the interactions between the tumor cells and its microenvironment.


Asunto(s)
Antineoplásicos/uso terapéutico , Resistencia a Antineoplásicos , Neoplasias/tratamiento farmacológico , Animales , Resistencia a Múltiples Medicamentos , Matriz Extracelular/metabolismo , Humanos , Terapia Molecular Dirigida , Células del Estroma/metabolismo
17.
Med Oncol ; 27(2): 466-73, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19466589

RESUMEN

Neoadjuvant chemotherapy is a standard therapy for patients with locally advanced breast cancer (LABC) and is increasingly used for early stage operable breast cancer. Not all patients benefit from it, and reliable markers for predicting response are needed. The cytotoxic effects of chemotherapy are mediated by induction of DNA damage in tumor cells. There is evidence that resistance to chemotherapy is related to enhanced repair of DNA lesions. The postreplication DNA repair (PRR) or translesion synthesis backup DNA repair pathway is critical for cell viability, conferring tolerance to DNA damaging drugs, and maintenance of genomic integrity. However, despite its importance in conferring tolerance to a variety of DNA damaging drugs including cytotoxic chemotherapy, the involvement of this backup repair pathway in chemotherapy response has not been studied. The Rad6B protein is a fundamental component of PRR. We have shown previously that the ability of breast cells to tolerate chemotherapeutic drugs correlates with Rad6B expression levels and PRR capacity. To determine whether Rad6B expression/distribution can be used singly or in combination with p53, Mdr-1/PgP, PCNA or beta-catenin as predictors of response to neoadjuvant chemotherapy, we analyzed posttreatment samples from 20 patients with LABC in a retrospective study. Only preferential Rad6B nuclear localization was associated with response to neoadjuvant chemotherapy. Nuclear exclusion with cytoplasmic overexpression of Rad6B was observed in some patients who failed to respond, but the association with response is not statistically significant. This is the first study to report that the postreplication DNA repair protein Rad6B could be used as an independent marker for determining response to neoadjuvant chemotherapy. This is an exploratory study and larger studies utilizing interim evaluations of Rad6B expression, its subcellular localization and repair activity are required to confirm its utility as a predictor of chemotherapeutic response.


Asunto(s)
Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Quimioterapia Adyuvante , Reparación del ADN/genética , Replicación del ADN/genética , Terapia Neoadyuvante , Enzimas Ubiquitina-Conjugadoras/biosíntesis , Enzimas Ubiquitina-Conjugadoras/genética , Adulto , Anciano , Neoplasias de la Mama/tratamiento farmacológico , Reparación del ADN/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , Femenino , Humanos , Persona de Mediana Edad
18.
J Control Release ; 141(2): 137-44, 2010 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-19751777

RESUMEN

Tumor drug resistance significantly limits the success of chemotherapy in the clinic. Tumor cells utilize multiple mechanisms to prevent the accumulation of anticancer drugs at their intracellular site of action. In this study, we investigated the anticancer efficacy of doxorubicin in combination with photodynamic therapy using methylene blue in a drug-resistant mouse tumor model. Surfactant-polymer hybrid nanoparticles formulated using an anionic surfactant, Aerosol-OT (AOT), and a naturally occurring polysaccharide polymer, sodium alginate, were used for synchronized delivery of the two drugs. Balb/c mice bearing syngeneic JC tumors (mammary adenocarcinoma) were used as a drug-resistant tumor model. Nanoparticle-mediated combination therapy significantly inhibited tumor growth and improved animal survival. Nanoparticle-mediated combination treatment resulted in enhanced tumor accumulation of both doxorubicin and methylene blue, significant inhibition of tumor cell proliferation, and increased induction of apoptosis. These data suggest that nanoparticle-mediated combination chemotherapy and photodynamic therapy using doxorubicin and methylene blue has significant therapeutic potential against drug-resistant tumors.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antibióticos Antineoplásicos/farmacología , Doxorrubicina/farmacología , Portadores de Fármacos , Resistencia a Antineoplásicos , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Azul de Metileno/farmacología , Nanopartículas , Fotoquimioterapia , Adenocarcinoma/irrigación sanguínea , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Alginatos/química , Animales , Antibióticos Antineoplásicos/administración & dosificación , Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/metabolismo , Apoptosis/efectos de los fármacos , Transporte Biológico , Proliferación Celular/efectos de los fármacos , Química Farmacéutica , Ácido Dioctil Sulfosuccínico/química , Doxorrubicina/administración & dosificación , Doxorrubicina/química , Doxorrubicina/metabolismo , Composición de Medicamentos , Femenino , Ácido Glucurónico/química , Ácidos Hexurónicos/química , Neoplasias Mamarias Experimentales/irrigación sanguínea , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Azul de Metileno/administración & dosificación , Azul de Metileno/química , Azul de Metileno/metabolismo , Ratones , Ratones Endogámicos BALB C , Microvasos/efectos de los fármacos , Microvasos/patología , Factores de Tiempo
19.
J Cell Physiol ; 219(2): 288-300, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19115235

RESUMEN

Cancer progression represents an evolutionary process where overall genome level changes reflect system instability and serve as a driving force for evolving new systems. To illustrate this principle it must be demonstrated that karyotypic heterogeneity (population diversity) directly contributes to tumorigenicity. Five well characterized in vitro tumor progression models representing various types of cancers were selected for such an analysis. The tumorigenicity of each model has been linked to different molecular pathways, and there is no common molecular mechanism shared among them. According to our hypothesis that genome level heterogeneity is a key to cancer evolution, we expect to reveal that the common link of tumorigenicity between these diverse models is elevated genome diversity. Spectral karyotyping (SKY) was used to compare the degree of karyotypic heterogeneity displayed in various sublines of these five models. The cell population diversity was determined by scoring type and frequencies of clonal and non-clonal chromosome aberrations (CCAs and NCCAs). The tumorigenicity of these models has been separately analyzed. As expected, the highest level of NCCAs was detected coupled with the strongest tumorigenicity among all models analyzed. The karyotypic heterogeneity of both benign hyperplastic lesions and premalignant dysplastic tissues were further analyzed to support this conclusion. This common link between elevated NCCAs and increased tumorigenicity suggests an evolutionary causative relationship between system instability, population diversity, and cancer evolution. This study reconciles the difference between evolutionary and molecular mechanisms of cancer and suggests that NCCAs can serve as a biomarker to monitor the probability of cancer progression.


Asunto(s)
Evolución Biológica , Susceptibilidad a Enfermedades , Variación Genética , Genoma Humano , Neoplasias/genética , Animales , Pruebas de Carcinogenicidad , Línea Celular , Aberraciones Cromosómicas , Femenino , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Cariotipificación , Ratones , Ratones Desnudos , Ratones Transgénicos , Trasplante de Neoplasias , Humo/efectos adversos , Nicotiana/efectos adversos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
20.
Cancer Biol Ther ; 7(11): 1774-82, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18787417

RESUMEN

Comedo-DCIS is a histologic subtype of preinvasive breast neoplasia that is characterized by prominent apoptotic cell death and has greater malignant potential than other DCIS subtypes. We investigated the mechanisms of apoptosis in comedo-DCIS and its role in conversion of comedo-DCIS to invasive cancer. Clinical comedo-DCIS excisions and the MCF10DCIS.com human breast cancer model which produces lesions resembling comedo-DCIS were analyzed. Apoptotic luminal and myoepithelial cells were identified by TUNEL and reactivity to cleaved PARP antibody and cell death assessed by Western blotting, Mitocapture and immunohistochemical assays. MCF10DCIS.com cells undergo spontaneous apoptosis in vitro, both in monolayers and multicellular spheroids; it is associated with increased mitochondrial membrane permeability, increase in Bax/Bcl-2 ratio and occurs via caspase-9-dependent p53-independent pathway. This suggests that apoptosis is stromal-independent and that the cells are programmed to undergo apoptosis. Immunostaining with cleaved PARP antibody showed that myoepithelial apoptosis occurs before lesions progress to comedo-DCIS in both clinical comedo-DCIS and in vivo MCF10DCIS.com lesions. Intense staining for MMP-2, MMP-3, MMP-9 and MMP-11 was observed in the stroma and epithelia of solid DCIS lesions prior to conversion to comedo-DCIS in clinical and MCF10DCIS.com lesions. Gelatin zymography showed higher MMP-2 levels in lysates and conditioned media of MCF10DCIS. com cells undergoing apoptosis. These data suggest that signals arising from the outside (microenvironmental) and inside (internal genetic alterations) of the duct act in concert to trigger apoptosis of myoepithelial and luminal epithelial cells. Our findings implicate spontaneous apoptosis in both the etiology and progression of comedo-DCIS. It is possible that spontaneous apoptosis facilitates elimination of cells thus permitting expansion and malignant transformation of cancer cells that are resistant to spontaneous apoptosis.


Asunto(s)
Apoptosis , Neoplasias de la Mama/patología , Carcinoma Intraductal no Infiltrante/patología , Animales , Línea Celular Tumoral , Epitelio/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica/métodos , Cinética , Mamografía/métodos , Ratones , Poli(ADP-Ribosa) Polimerasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...