Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
J Biol Chem ; 294(14): 5246-5260, 2019 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-30737284

RESUMEN

Cumulative evidence suggests that the heat shock protein 90 (Hsp90) co-chaperone UNC-45 myosin chaperone A (UNC45A) contributes to tumorigenesis and that its expression in cancer cells correlates with proliferation and metastasis of solid tumors. However, the molecular mechanism by which UNC45A regulates cancer cell proliferation remains largely unknown. Here, using siRNA-mediated gene silencing and various human cells, we report that UNC45A is essential for breast cancer cell growth, but is dispensable for normal cell proliferation. Immunofluorescence microscopy, along with gene microarray and RT-quantitative PCR analyses, revealed that UNC45A localizes to the cancer cell nucleus, where it up-regulates the transcriptional activity of the glucocorticoid receptor and thereby promotes expression of the mitotic kinase NIMA-related kinase 7 (NEK7). We observed that UNC45A-deficient cancer cells exhibit extensive pericentrosomal material disorganization, as well as defects in centrosomal separation and mitotic chromosome alignment. Consequently, these cells stalled in metaphase and cytokinesis and ultimately underwent mitotic catastrophe, phenotypes that were rescued by heterologous NEK7 expression. Our results identify a key role for the co-chaperone UNC45A in cell proliferation and provide insight into the regulatory mechanism. We propose that UNC45A represents a promising new therapeutic target to inhibit cancer cell growth in solid tumor types.


Asunto(s)
Neoplasias de la Mama/metabolismo , Carcinogénesis/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Quinasas Relacionadas con NIMA/biosíntesis , Proteínas de Neoplasias/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Carcinogénesis/genética , Carcinogénesis/patología , Femenino , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Células MCF-7 , Mitosis/genética , Quinasas Relacionadas con NIMA/genética , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Células PC-3
2.
Mol Cell ; 47(4): 570-84, 2012 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-22819326

RESUMEN

Although inactivation of the PTEN gene has been implicated in the development of resistance to the HER2 targeting antibody trastuzumab, the mechanisms mediating this resistance remain elusive. We generated trastuzumab resistant cells by knocking down PTEN expression in HER2 overexpressing breast cancer cell lines and demonstrate that development of trastuzumab resistance in these cells is mediated by activation of an IL6 inflammatory feedback loop leading to expansion of the cancer stem cell (CSC) population. Long term trastuzumab treatment generates highly enriched CSCs which display an EMT phenotype secreting over 100-fold more IL6 than parental cells. An IL6 receptor antibody interrupted this inflammatory feedback loop reducing the cancer stem cell population resulting in decreased tumor growth and metastasis in mouse xenographs. These studies demonstrate that trastuzumab resistance may be mediated by an IL6 inflammatory loop and suggest that blocking this loop may provide alternative strategy to overcome trastuzumab resistance.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Neoplasias de la Mama/metabolismo , Inflamación/metabolismo , Interleucina-6/metabolismo , Células Madre Neoplásicas/metabolismo , Receptor ErbB-2/metabolismo , Animales , Antineoplásicos/farmacología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Quimiocina CCL5/genética , Quimiocina CCL5/metabolismo , Regulación hacia Abajo/genética , Resistencia a Antineoplásicos , Femenino , Humanos , Inflamación/genética , Inflamación/patología , Interleucina-6/genética , Interleucina-8/genética , Interleucina-8/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Madre Neoplásicas/patología , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Receptor ErbB-2/genética , Trastuzumab
3.
Int J Mol Sci ; 20(9)2019 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-31085991

RESUMEN

It is now widely believed that mammary epithelial cell plasticity, an important physiological process during the stages of mammary gland development, is exploited by the malignant cells for their successful disease progression. Normal mammary epithelial cells are heterogeneous and organized in hierarchical fashion, in which the mammary stem cells (MaSC) lie at the apex with regenerative capacity as well as plasticity. Despite the fact that the majority of studies supported the existence of multipotent MaSCs giving rise to both basal and luminal lineages, others proposed lineage restricted unipotent MaSCs. Consistent with the notion, the latest research has suggested that although normal MaSC subsets mainly stay in a quiescent state, they differ in their reconstituting ability, spatial localization, and molecular and epigenetic signatures in response to physiological stimuli within the respective microenvironment during the stages of mammary gland development. In this review, we will focus on current research on the biology of normal mammary stem cells with an emphasis on properties of cellular plasticity, self-renewal and quiescence, as well as the role of the microenvironment in regulating these processes. This will include a discussion of normal breast stem cell heterogeneity, stem cell markers, and lineage tracing studies.


Asunto(s)
Células Epiteliales/citología , Glándulas Mamarias Animales/citología , Células Madre Multipotentes/citología , Células Madre/citología , Animales , Diferenciación Celular/fisiología , Femenino , Humanos
4.
Int J Cancer ; 143(9): 2177-2186, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-29752723

RESUMEN

Suppressor of cytokine signaling (SOCS) family of proteins plays critical role in the regulation of immune responses controlling JAK/STAT mediated inflammatory cytokines. Among the members, SOCS1 and SOCS3 contain a kinase inhibitory region (KIR) and SOCS3 binds to JAK/STAT/gp130 complex by inhibiting the downstream signaling and suppressing inflammatory cytokines. Loss or reduced levels of SOCS3 have been linked to cancer-associated inflammation and suppressive immunity leading to enhanced tumor growth and metastasis. In line with these reports, we previously demonstrated that proteolytic degradation of SOCS3 in triple negative breast cancer (TNBC) subtype drives the expression of inflammatory cytokines. Therefore, we postulated that SOCS3 mimetics might suppress the inflammatory cytokine production in TNBC subtype and inhibit tumor growth and metastasis. Here we designed and characterized five linear peptides derived from the N-terminal region of SOCS3 encompassing regions that interface with the JAK2/gp130 complex using the Circular Dichroism and Surface Plasmon Resonance spectroscopies. The KIRESS peptide resulted the sequence containing the most part of the hot-spots required for binding to JAK2 and was further investigated in vivo in mouse xenografts of MDA-MB-231-luci tumors as models of human TNBC subtype. Expectedly, this peptide showed a significant inhibition of primary tumor growth and pulmonary metastasis. Our studies suggest that SOCS3 peptidomimetics may possess a therapeutic potential in aggressive cancers, such as TNBC subtype, with activated inflammatory cytokines.


Asunto(s)
Biomimética , Neoplasias Pulmonares/tratamiento farmacológico , Fragmentos de Péptidos/farmacología , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Secuencia de Aminoácidos , Animales , Apoptosis , Biomarcadores de Tumor/metabolismo , Proliferación Celular , Citocinas/metabolismo , Femenino , Humanos , Janus Quinasa 2/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Fragmentos de Péptidos/química , Conformación Proteica , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Proteína 3 Supresora de la Señalización de Citocinas/química , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Mol Cancer ; 14: 6, 2015 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-25608686

RESUMEN

BACKGROUND: While angiogenesis inhibitors represent a viable cancer therapy, there is preclinical and clinical data to suggest that many tumors develop resistance to such treatments. Moreover, previous studies have revealed a complex association between autophagy and angiogenesis, and their collective influence on tumorigenesis. Autophagy has been implicated in cytoprotection and tumor promotion, and as such may represent an alternative way of targeting apoptosis-resistant cancer cells. This study explored the anti-cancer agent and boswellic acid analog BA145 as an inducer of autophagy and angiogenesis-mediated cytoprotection of tumor cells. METHODS: Flow cytometry, western blotting, and confocal microscopy were used to investigate the role of BA145 mediated autophagy. ELISA, microvessel sprouting, capillary structure formation, aortic ring and wound healing assays were performed to determine the relationship between BA145 triggered autophagy and angiogenesis. Flow cytometery, western blotting, and microscopy were employed to examine the mechanism of BA145 induced cell death and apoptosis. Live imaging and tumor volume analysis were carried out to evaluate the effect of BA145 triggered autophagy on mouse tumor xenografts. RESULTS: BA145 induced autophagy in PC-3 cancer cells and HUVECs significantly impeded its negative regulation on cell proliferation, migration, invasion and tube formation. These effects of BA145 induced autophagy were observed under both normoxic and hypoxic conditions. However, inhibition of autophagy using either pharmacological inhibitors or RNA interference enhanced the BA145 mediated death of these cells. Similar observations were noticed with sunitinib, the anti-angiogenic properties of which were significantly enhanced during combination treatments with autophagy inhibitors. In mouse tumor xenografts, co-treatment with chloroquinone and BA145 led to a considerable reduction in tumor burden and angiogenesis compared to BA145 alone. CONCLUSION: These studies reveal the essential role of BA145 triggered autophagy in the regulation of angiogenesis and cytoprotection. It also suggests that the combination of the autophagy inhibitors with chemotherapy or anti-angiogenic agents may be an effective therapeutic approach against cancer.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Triterpenos/química , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Indoles/farmacología , Pirroles/farmacología , Sunitinib
6.
Proc Natl Acad Sci U S A ; 109(8): 2784-9, 2012 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-22308314

RESUMEN

Antiangiogenic therapy has been thought to hold significant potential for the treatment of cancer. However, the efficacy of such treatments, especially in breast cancer patients, has been called into question, as recent clinical trials reveal only limited effectiveness of antiangiogenic agents in prolonging patient survival. New research using preclinical models further suggests that antiangiogenic agents actually increase invasive and metastatic properties of breast cancer cells. We demonstrate that by generating intratumoral hypoxia in human breast cancer xenografts, the antiangiogenic agents sunitinib and bevacizumab increase the population of cancer stem cells. In vitro studies revealed that hypoxia-driven stem/progenitor cell enrichment is primarily mediated by hypoxia-inducible factor 1α. We further show that the Akt/ß-catenin cancer stem cell regulatory pathway is activated in breast cancer cells under hypoxic conditions in vitro and in sunitinib-treated mouse xenografts. These studies demonstrate that hypoxia-driven cancer stem cell stimulation limits the effectiveness of antiangiogenic agents, and suggest that to improve patient outcome, these agents might have to be combined with cancer stem cell-targeting drugs.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Neoplasias de la Mama/patología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Neoplasias de la Mama/tratamiento farmacológico , Recuento de Células , Hipoxia de la Célula/efectos de los fármacos , Línea Celular Tumoral , Femenino , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Indoles/farmacología , Indoles/uso terapéutico , Ratones , Pirroles/farmacología , Pirroles/uso terapéutico , Transducción de Señal/efectos de los fármacos , Sunitinib , beta Catenina/metabolismo
7.
PLoS Genet ; 8(6): e1002751, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22685420

RESUMEN

MicroRNAs (miRNAs) play important roles in normal cellular differentiation and oncogenesis. microRNA93 (mir-93), a member of the mir106b-25 cluster, located in intron 13 of the MCM7 gene, although frequently overexpressed in human malignancies may also function as a tumor suppressor gene. Using a series of breast cancer cell lines representing different stages of differentiation and mouse xenograft models, we demonstrate that mir-93 modulates the fate of breast cancer stem cells (BCSCs) by regulating their proliferation and differentiation states. In "claudin(low)" SUM159 cells, expression of mir-93 induces Mesenchymal-Epithelial Transition (MET) associated with downregulation of TGFß signaling and downregulates multiple stem cell regulatory genes, including JAK1, STAT3, AKT3, SOX4, EZH1, and HMGA2, resulting in cancer stem cell (CSC) depletion. Enforced expression of mir-93 completely blocks tumor development in mammary fat pads and development of metastases following intracardiac injection in mouse xenografts. The effect of mir-93 on the CSC population is dependent on the cellular differentiation state, with mir-93 expression increasing the CSC population in MCF7 cells that display a more differentiated "luminal" phenotype. mir-93 also regulates the proliferation and differentiation of normal breast stem cells isolated from reduction mammoplasties. These studies demonstrate that miRNAs can regulate the states and fates of normal and malignant mammary stem cells, findings which have important biological and clinical implications.


Asunto(s)
Neoplasias de la Mama/genética , Diferenciación Celular/genética , Transformación Celular Neoplásica , MicroARNs/genética , Células Madre Neoplásicas , Animales , Neoplasias de la Mama/metabolismo , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/genética , Proteínas de Unión al ADN/genética , Transición Epitelial-Mesenquimal/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Glándulas Mamarias Humanas/metabolismo , Ratones , MicroARNs/metabolismo , Componente 7 del Complejo de Mantenimiento de Minicromosoma , Neoplasias Experimentales , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/metabolismo , Proteínas Nucleares/genética
8.
Res Sq ; 2024 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-38464167

RESUMEN

Triple negative breast cancer (TNBC) subtype is characterized with higher EMT/stemness properties and immune suppressive tumor microenvironment (TME). Women with advanced TNBC exhibit aggressive disease and have limited treatment options. Although immune suppressive TME is implicated in driving aggressive properties of basal/TNBC subtype and therapy resistance, effectively targeting it remains a challenge. Minnelide, a prodrug of triptolide currently being tested in clinical trials, has shown anti-tumorigenic activity in multiple malignancies via targeting super enhancers, Myc and anti-apoptotic pathways such as HSP70. Distinct super-enhancer landscape drives cancer stem cells (CSC) in TNBC subtype while inducing immune suppressive TME. We show that Minnelide selectively targets CSCs in human and murine TNBC cell lines compared to cell lines of luminal subtype by targeting Myc and HSP70. Minnelide in combination with cyclophosphamide significantly reduces the tumor growth and eliminates metastasis by reprogramming the tumor microenvironment and enhancing cytotoxic T cell infiltration in 4T1 tumor-bearing mice. Resection of residual tumors following the combination treatment leads to complete eradication of disseminated tumor cells as all mice are free of local and distant recurrences. All control mice showed recurrences within 3 weeks of post-resection while single Minnelide treatment delayed recurrence and one mouse was free of tumor. We provide evidence that Minnelide targets tumor intrinsic pathways and reprograms the immune suppressive microenvironment. Our studies also suggest that Minnelide in combination with cyclophosphamide may lead to durable responses in patients with basal/TNBC subtype warranting its clinical investigation.

9.
Sci Rep ; 13(1): 13790, 2023 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-37612326

RESUMEN

Heat shock protein 90 (Hsp90) and its co-chaperones promote cancer, and targeting Hsp90 holds promise for cancer treatment. Most of the efforts to harness this potential have focused on targeting the Hsp90 N-terminus ATP binding site. Although newer-generation inhibitors have shown improved efficacy in aggressive cancers, induction of the cellular heat shock response (HSR) by these inhibitors is thought to limit their clinical efficacy. Therefore, Hsp90 inhibitors with novel mechanisms of action and that do not trigger the HSR would be advantageous. Here, we investigated the mechanism by which capsaicin inhibits Hsp90. Through mutagenesis, chemical modifications, and proteomic studies, we show that capsaicin binds to the N-terminus of Hsp90 and inhibits its ATPase activity. Consequently, capsaicin and its analogs inhibit Hsp90 ATPase-dependent progesterone receptor reconstitution in vitro. Capsaicin did not induce the HSR, instead, it promoted the degradation of Hsp70 through the lysosome-autophagy pathway. Remarkably, capsaicin did not induce degradation of the constitutively expressed cognate Hsc70, indicating selectivity for Hsp70. Combined treatments of capsaicin and the Hsp90 inhibitor 17-AAG improved the anti-tumor efficacy of 17-AAG in cell culture and tridimensional tumor spheroid growth assays using breast and prostate cancer models. Consistent with this, in silico docking studies revealed that capsaicin binding to the ATP binding site of Hsp90 was distinct from classical N-terminus Hsp90 inhibitors, indicating a novel mechanism of action. Collectively, these findings support the use of capsaicin as a chemical scaffold to develop novel Hsp90 N-terminus inhibitors as well as its ability to be a potential cancer co-therapeutic.


Asunto(s)
Capsaicina , Neoplasias de la Próstata , Masculino , Humanos , Capsaicina/farmacología , Proteómica , Proteínas HSP70 de Choque Térmico , Proteínas HSP90 de Choque Térmico , Lisosomas , Adenosina Trifosfatasas , Adenosina Trifosfato
10.
iScience ; 26(12): 108308, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-38025772

RESUMEN

Low response rates and immune-related adverse events limit the remarkable impact of cancer immunotherapy. To improve clinical outcomes, preclinical studies have shown that combining immunotherapies with N-terminal Hsp90 inhibitors resulted in improved efficacy, even though induction of an extensive heat shock response (HSR) and less than optimal dosing of these inhibitors limited their clinical efficacy as monotherapies. We discovered that the natural product Enniatin A (EnnA) targets Hsp90 and destabilizes its client oncoproteins without inducing an HSR. EnnA triggers immunogenic cell death in triple-negative breast cancer (TNBC) syngeneic mouse models and exhibits superior antitumor activity compared to Hsp90 N-terminal inhibitors. EnnA reprograms the tumor microenvironment (TME) to promote CD8+ T cell-dependent antitumor immunity by reducing PD-L1 levels and activating the chemokine receptor CX3CR1 pathway. These findings provide strong evidence for transforming the immunosuppressive TME into a more tumor-hostile milieu by engaging Hsp90 with therapeutic agents involving novel mechanisms of action.

11.
Breast Cancer Res Treat ; 135(3): 913-22, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22941572

RESUMEN

Though xenografts are used extensively for drug development in breast cancer, how well xenografts reflect the breadth of primary breast tumor subtypes has not been well characterized. Moreover, few studies have compared the gene expression of xenograft tumors to the primary tumors from which they were derived. Here we investigate whether the ability of human breast tumors (n = 20) to create xenografts in immune-deficient mice is associated with breast cancer immunohistochemical (IHC) and intrinsic subtype. We also characterize how precisely the gene expression of xenografts reprises that of parent breast tumors, using hierarchical clustering and other correlation-based techniques applied to Agilent 44K gene expression data from 16 samples including four matched primary tumor-xenograft pairs. Of the breast tumors studied, 25 % (5/20) generated xenografts. Receptor and intrinsic subtype were significant predictors of xenograft success, with all (4/4) triple-negative (TN) tumors and no (0/12) HR+Her2- tumors forming xenografts (P = 0.0005). Tumor cell expression of ALDH1, a stem cell marker, trended toward successful engraftment (P = 0.14), though CDK5/6, a basal marker, did not. Though hierarchical clustering across the 500 most variable genes segregated human breast tumors from xenograft tumors, when clustering was performed over the PAM50 gene set the primary tumor-xenograft pairs clustered together, with all IHC subtypes clustered in distinct groups. Greater similarity between primary tumor-xenograft pairs relative to random pairings was confirmed by calculation of the within-pair between-pair scatter ratio (WPBPSR) distribution (P = 0.0269), though there was a shift in the xenografts toward more aggressive features including higher proliferation scores relative to the primary. Triple-negative breast tumors demonstrate superior ability to create xenografts compared to HR+ tumors, which may reflect higher proliferation or relatively stroma-independent growth of this subtype. Xenograft tumors' gene expression faithfully resembles that of their parent tumors, yet also demonstrates a shift toward more aggressive molecular features.


Asunto(s)
Neoplasias de la Mama/genética , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Familia de Aldehído Deshidrogenasa 1 , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Femenino , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Ratones , Ratones SCID , Familia de Multigenes , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Retinal-Deshidrogenasa/genética , Retinal-Deshidrogenasa/metabolismo , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
12.
PLoS Biol ; 7(6): e1000121, 2009 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-19492080

RESUMEN

Recent evidence suggests that many malignancies, including breast cancer, are driven by a cellular subcomponent that displays stem cell-like properties. The protein phosphatase and tensin homolog (PTEN) is inactivated in a wide range of human cancers, an alteration that is associated with a poor prognosis. Because PTEN has been reported to play a role in the maintenance of embryonic and tissue-specific stem cells, we investigated the role of the PTEN/Akt pathway in the regulation of normal and malignant mammary stem/progenitor cell populations. We demonstrate that activation of this pathway, via PTEN knockdown, enriches for normal and malignant human mammary stem/progenitor cells in vitro and in vivo. Knockdown of PTEN in normal human mammary epithelial cells enriches for the stem/progenitor cell compartment, generating atypical hyperplastic lesions in humanized NOD/SCID mice. Akt-driven stem/progenitor cell enrichment is mediated by activation of the Wnt/beta-catenin pathway through the phosphorylation of GSK3-beta. In contrast to chemotherapy, the Akt inhibitor perifosine is able to target the tumorigenic cell population in breast tumor xenografts. These studies demonstrate an important role for the PTEN/PI3-K/Akt/beta-catenin pathway in the regulation of normal and malignant stem/progenitor cell populations and suggest that agents that inhibit this pathway are able to effectively target tumorigenic breast cancer cells.


Asunto(s)
Mama , Fosfohidrolasa PTEN/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Células Madre/metabolismo , beta Catenina/metabolismo , Animales , Mama/citología , Mama/metabolismo , Mama/patología , Neoplasias de la Mama/patología , Femenino , Regulación de la Expresión Génica , Humanos , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Células Madre/patología , beta Catenina/genética
13.
Oncol Rep ; 45(3): 1171-1181, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33469683

RESUMEN

Myeloid­derived suppressor cells (MDSCs) are an indispensable component of the tumor microenvironment (TME). Along with the role of MDSC immunosuppression and antitumor immunity, MDSCs facilitate tumor growth, differentiation, and metastasis in several ways that are yet to be explored. Like any other cell type, MDSCs also release a tremendous number of exosomes, or nanovesicles of endosomal origin, that participate in intercellular communications by dispatching biological macromolecules. There have been no investigational studies conducted to characterize the role of MDSC­derived exosomes (MDSC exo) in modulating the TME. In this study, we isolated MDSC exo and demonstrated that they carry a significant level of proteins that play an indispensable role in tumor growth, invasion, angiogenesis, and immunomodulation. We observed a higher yield and more substantial immunosuppressive potential of exosomes isolated from MDSCs in the primary tumor area than those in the spleen or bone marrow. Our in vitro data suggest that MDSC exo are capable of hyper­activating or exhausting CD8 T­cells and induce reactive oxygen species production that elicits activation­induced cell death. We confirmed the depletion of CD8 T­cells in vivo by treating mice with MDSC exo. We also observed a reduction in pro­inflammatory M1­macrophages in the spleen of those animals. Our results indicate that the immunosuppressive and tumor­promoting functions of MDSCs are also implemented by MDSC­derived exosomes which would open up a new avenue of MDSC research and MDSC­targeted therapy.


Asunto(s)
Exosomas/metabolismo , Células Supresoras de Origen Mieloide/inmunología , Neoplasias/inmunología , Escape del Tumor , Microambiente Tumoral/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Comunicación Celular/inmunología , Línea Celular Tumoral/trasplante , Modelos Animales de Enfermedad , Humanos , Macrófagos/inmunología , Ratones , Células Supresoras de Origen Mieloide/metabolismo , Neoplasias/patología , Cultivo Primario de Células
14.
Mol Cancer Res ; 19(9): 1486-1497, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34099522

RESUMEN

DNA damage, induced by either chemical carcinogens or environmental pollutants, plays an important role in the initiation of colorectal cancer. DNA repair processes, however, are involved in both protecting against cancer formation, and also contributing to cancer development, by ensuring genomic integrity and promoting the efficient DNA repair in tumor cells, respectively. Although DNA repair pathways have been well exploited in the treatment of breast and ovarian cancers, the role of DNA repair processes and their therapeutic efficacy in colorectal cancer is yet to be appreciably explored. To understand the role of DNA repair, especially homologous recombination (HR), in chemical carcinogen-induced colorectal cancer growth, we unraveled the role of RAD51AP1 (RAD51-associated protein 1), a protein involved in HR, in genotoxic carcinogen (azoxymethane, AOM)-induced colorectal cancer. Although AOM treatment alone significantly increased RAD51AP1 expression, the combination of AOM and dextran sulfate sodium (DSS) treatment dramatically increased by several folds. RAD51AP1 expression is found in mouse colonic crypt and proliferating cells. RAD51AP1 expression is significantly increased in majority of human colorectal cancer tissues, including BRAF/KRAS mutant colorectal cancer, and associated with reduced treatment response and poor prognosis. Rad51ap1-deficient mice were protected against AOM/DSS-induced colorectal cancer. These observations were recapitulated in a genetically engineered mouse model of colorectal cancer (ApcMin /+ ). Furthermore, chemotherapy-resistant colorectal cancer is associated with increased RAD51AP1 expression. This phenomenon is associated with reduced cell proliferation and colorectal cancer stem cell (CRCSC) self-renewal. Overall, our studies provide evidence that RAD51AP1 could be a novel diagnostic marker for colorectal cancer and a potential therapeutic target for colorectal cancer prevention and treatment. IMPLICATIONS: This study provides first in vivo evidence that RAD51AP1 plays a critical role in colorectal cancer growth and drug resistance by regulating CRCSC self-renewal.


Asunto(s)
Autorrenovación de las Células , Neoplasias Colorrectales/tratamiento farmacológico , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/fisiología , Resistencia a Antineoplásicos , Fluorouracilo/farmacología , Células Madre Neoplásicas/efectos de los fármacos , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/fisiología , Animales , Antimetabolitos Antineoplásicos/farmacología , Apoptosis , Estudios de Casos y Controles , Proliferación Celular , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Proteínas de Unión al ADN/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Pronóstico , Proteínas de Unión al ARN/genética , Tasa de Supervivencia , Células Tumorales Cultivadas
15.
Breast Cancer Res Treat ; 122(3): 777-85, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19898931

RESUMEN

The cancer stem cell hypothesis asserts that malignancies arise in tissue stem and/or progenitor cells through the dysregulation or acquisition of self-renewal. In order to determine whether the dietary polyphenols, curcumin, and piperine are able to modulate the self-renewal of normal and malignant breast stem cells, we examined the effects of these compounds on mammosphere formation, expression of the breast stem cell marker aldehyde dehydrogenase (ALDH), and Wnt signaling. Mammosphere formation assays were performed after curcumin, piperine, and control treatment in unsorted normal breast epithelial cells and normal stem and early progenitor cells, selected by ALDH positivity. Wnt signaling was examined using a Topflash assay. Both curcumin and piperine inhibited mammosphere formation, serial passaging, and percent of ALDH+ cells by 50% at 5 microM and completely at 10 microM concentration in normal and malignant breast cells. There was no effect on cellular differentiation. Wnt signaling was inhibited by both curcumin and piperine by 50% at 5 microM and completely at 10 microM. Curcumin and piperine separately, and in combination, inhibit breast stem cell self-renewal but do not cause toxicity to differentiated cells. These compounds could be potential cancer preventive agents. Mammosphere formation assays may be a quantifiable biomarker to assess cancer preventive agent efficacy and Wnt signaling assessment can be a mechanistic biomarker for use in human clinical trials.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Mama/patología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Aldehído Deshidrogenasa/metabolismo , Alcaloides/administración & dosificación , Benzodioxoles/administración & dosificación , Mama/efectos de los fármacos , Mama/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Curcumina/administración & dosificación , Femenino , Humanos , Técnicas para Inmunoenzimas , Células Madre Neoplásicas/metabolismo , Piperidinas/administración & dosificación , Alcamidas Poliinsaturadas/administración & dosificación , Transducción de Señal/efectos de los fármacos , Proteínas Wnt/metabolismo
16.
Clin Cancer Res ; 15(6): 1845-7, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19276254

RESUMEN

Increasing evidence indicates that tumor-initiating (cancer stem) cells may contribute to treatment resistance and relapse, suggesting that improved clinical outcome will require effective targeting of this cell population. Recent studies suggest that the remarkable clinical efficacy of trastuzumab may relate to its ability to target cancer stem cell populations.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral/efectos de los fármacos , Receptor ErbB-2/fisiología , Receptores Notch/fisiología , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Neoplasias de la Mama/patología , Femenino , Humanos , Receptor ErbB-2/antagonistas & inhibidores , Receptor ErbB-2/genética , Transducción de Señal , Trastuzumab
17.
Biomedicines ; 8(2)2020 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-32028692

RESUMEN

Cisplatin (CP) is one of the most preferred platinum-containing antineoplastic drugs. However, even in nontoxic plasma concentrations, it may cause kidney injury. To be able to increase its effective pharmacological dose, its side effects need to be regarded. Diet restriction (DR) has been demonstrated to improve cellular survival in a number of disorders. In this context, we investigated the role of DR in CP-induced nephrotoxicity (CPN). Besides alternate DR, animals were exposed to DR for 3 days prior or after CP treatment. Here, we observed that both 3 days of DR reverses the nephrotoxic effect of CP, which was associated with improved physiological outcomes, such as serum creatine, blood-urea nitrogen and urea. These treatments significantly increased phosphorylation of survival kinases PI3K/Akt and ERK-1/2 and decreased the level of stress kinase JNK were noted. In addition, the activation level of signal transduction mediator p38 MAPK phosphorylation was higher particularly in both three-day DR groups. Next, animals were fed with carbohydrate-, protein- or fat-enriched diets in the presence of CP. Results indicated that not only fasting but also dietary content itself may play a determinant role in the severity of CPN. Our data suggest that DR is a promising approach to reduce CPN by regulating metabolism and cell signaling pathways.

18.
Cancer Res ; 80(18): 3855-3866, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32665355

RESUMEN

RAD51-associated protein 1 (RAD51AP1) plays an integral role in homologous recombination by activating RAD51 recombinase. Homologous recombination is essential for preserving genome integrity and RAD51AP1 is critical for D-loop formation, a key step in homologous recombination. Although RAD51AP1 is involved in maintaining genomic stability, recent studies have shown that RAD51AP1 expression is significantly upregulated in human cancers. However, the functional role of RAD51AP1 in tumor growth and the underlying molecular mechanism(s) by which RAD51AP1 regulates tumorigenesis have not been fully understood. Here, we use Rad51ap1-knockout mice in genetically engineered mouse models of breast cancer to unravel the role of RAD51AP1 in tumor growth and metastasis. RAD51AP1 gene transcript was increased in both luminal estrogen receptor-positive breast cancer and basal triple-negative breast cancer, which is associated with poor prognosis. Conversely, knockdown of RAD51AP1 (RADP51AP1 KD) in breast cancer cell lines reduced tumor growth. Rad51ap1-deficient mice were protected from oncogene-driven spontaneous mouse mammary tumor growth and associated lung metastasis. In vivo, limiting dilution studies provided evidence that Rad51ap1 plays a critical role in breast cancer stem cell (BCSC) self-renewal. RAD51AP1 KD improved chemotherapy and radiotherapy response by inhibiting BCSC self-renewal and associated pluripotency. Overall, our study provides genetic and biochemical evidences that RAD51AP1 is critical for tumor growth and metastasis by increasing BCSC self-renewal and may serve as a novel target for chemotherapy- and radiotherapy-resistant breast cancer. SIGNIFICANCE: This study provides in vivo evidence that RAD51AP1 plays a critical role in breast cancer growth and metastasis by regulating breast cancer stem cell self-renewal.


Asunto(s)
Neoplasias de la Mama/patología , Autorrenovación de las Células/genética , Proteínas de Unión al ADN/deficiencia , Neoplasias Mamarias Animales/patología , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/terapia , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Activación Enzimática , Femenino , Humanos , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/terapia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Madre Neoplásicas , Proteínas de Unión al ARN/genética , Recombinasa Rad51/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Regulación hacia Arriba
19.
Oncogene ; 38(28): 5749, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31197211

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

20.
Oncogene ; 38(4): 469-482, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30166590

RESUMEN

TNFα is a pleiotropic cytokine which fuels tumor cell growth, invasion, and metastasis in some malignancies, while in others it induces cytotoxic cell death. However, the molecular mechanism by which TNFα exerts its diverse effects on breast cancer subtypes remains elusive. Using in vitro assays and mouse xenografts, we show here that TNFα contributes to the aggressive properties of triple negative breast cancer (TNBC) cell lines via upregulation of TNFAIP3(A20). In a striking contrast, TNFα induces a potent cytotoxic cell death in luminal (ER+) breast cancer cell lines which fail to upregulate A20 expression. Overexpression of A20 not only protects luminal breast cancer cell lines from TNFα-induced cell death via inducing HSP70-mediated anti-apoptotic pathway but also promotes a robust EMT/CSC phenotype by activating the pStat3-mediated inflammatory signaling. Furthermore, A20 overexpression in luminal breast cancer cells induces aggressive metastatic properties in mouse xenografts via generating a permissive inflammatory microenvironment constituted by granulocytic-MDSCs. Collectively, our results reveal a mechanism by which A20 mediates pleiotropic effects of TNFα playing role in aggressive behaviors of TNBC subtype while its deficiency results in TNFα-induced apoptotic cell death in luminal breast cancer subtype.


Asunto(s)
Neoplasias de la Mama/patología , Regulación Neoplásica de la Expresión Génica , Pleiotropía Genética , Proteínas de Neoplasias/fisiología , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Animales , Apoptosis/fisiología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Transición Epitelial-Mesenquimal , Femenino , Proteínas del Choque Térmico HSP72/antagonistas & inhibidores , Proteínas del Choque Térmico HSP72/fisiología , Xenoinjertos , Humanos , Inflamación , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos NOD , Ratones SCID , Invasividad Neoplásica/genética , Proteínas de Neoplasias/genética , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Nucleósidos de Purina/farmacología , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Factor de Transcripción STAT3/fisiología , Transducción de Señal , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/biosíntesis , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda