Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Methods Mol Biol ; 2660: 23-41, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37191788

RESUMEN

RNA sequencing (RNA-seq) is a method used for the high-throughput quantification of mRNA in a biological sample. It is widely used to investigate differential gene expression between drug-resistant and sensitive cancers to identify genetic mediators of drug resistance. Here, we describe a comprehensive experimental and bioinformatic pipeline to isolate mRNA from human cell lines, prepare mRNA libraries for next-generation sequencing, and perform post-sequencing bioinformatics analyses.


Asunto(s)
Biología Computacional , Neoplasias , Humanos , Biología Computacional/métodos , ARN Mensajero/genética , Línea Celular , Análisis de Secuencia de ARN/métodos , Perfilación de la Expresión Génica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Transcriptoma , ARN/genética , Neoplasias/genética
2.
BMC Cancer ; 23(1): 138, 2023 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-36765275

RESUMEN

BACKGROUND: Rearranged during transfection (RET) tyrosine kinase signaling has been previously implicated in endocrine resistant breast cancer, however the mechanism by which this signaling cascade promotes resistance is currently not well described. We recently reported that glial cell-derived neurotrophic factor (GDNF)-RET signaling appears to promote a positive feedback loop with the transcription factor early growth response 1 (EGR1). Here we investigate the mechanism behind this feedback loop and test the hypothesis that GDNF-RET signaling forms a regulatory loop with EGR1 to upregulate cyclin D1 (CCND1) transcription, leading to cell cycle progression and tamoxifen resistance. METHODS: To gain a better understanding of the GDNF-RET-EGR1 resistance mechanism, we studied the GDNF-EGR1 positive feedback loop and the role of GDNF and EGR1 in endocrine resistance by modulating their transcription levels using CRISPR-dCAS9 in tamoxifen sensitive (TamS) and tamoxifen resistant (TamR) MCF-7 cells. Additionally, we performed kinetic studies using recombinant GDNF (rGDNF) treatment of TamS cells. Finally, we performed cell proliferation assays using rGDNF, tamoxifen (TAM), and Palbociclib treatments in TamS cells. Statistical significance for qPCR and chromatin immunoprecipitation (ChIP)-qPCR experiments were determined using a student's paired t-test and statistical significance for the cell viability assay was a one-way ANOVA. RESULTS: GDNF-RET signaling formed a positive feedback loop with EGR1 and also downregulated estrogen receptor 1 (ESR1) transcription. Upregulation of GDNF and EGR1 promoted tamoxifen resistance in TamS cells and downregulation of GDNF promoted tamoxifen sensitivity in TamR cells. Additionally, we show that rGDNF treatment activated GDNF-RET signaling in TamS cells, leading to recruitment of phospho-ELK-1 to the EGR1 promoter, upregulation of EGR1 mRNA and protein, binding of EGR1 to the GDNF and CCND1 promoters, increased GDNF protein expression, and subsequent upregulation of CCND1 mRNA levels. We also show that inhibition of cyclin D1 with Palbociclib, in the presence of rGDNF, decreases cell proliferation and resensitizes cells to TAM. CONCLUSION: Outcomes from these studies support the hypotheses that GDNF-RET signaling forms a positive feedback loop with the transcription factor EGR1, and that GDNF-RET-EGR1 signaling promotes endocrine resistance via signaling to cyclin D1. Inhibition of components of this signaling pathway could lead to therapeutic insights into the treatment of endocrine resistant breast cancer.


Asunto(s)
Factor Neurotrófico Derivado de la Línea Celular Glial , Tamoxifeno , Línea Celular Tumoral , Proliferación Celular , Ciclina D1/genética , Ciclina D1/metabolismo , Resistencia a Antineoplásicos/genética , Retroalimentación , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Cinética , ARN Mensajero , Transducción de Señal , Tamoxifeno/farmacología , Tamoxifeno/uso terapéutico , Factores de Transcripción , Humanos
3.
Cancer Drug Resist ; 5(2): 304-316, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35800369

RESUMEN

Ovarian cancer is one of the most lethal gynecologic cancers. The standard therapy for ovarian cancer has been the same for the past two decades, a combination treatment of platinum with paclitaxel. Recently, the FDA approved three new therapeutic drugs, two poly (ADP-ribose) polymerase inhibitors (olaparib and niraparib) and one vascular endothelial growth factor inhibitor (bevacizumab) as maintenance therapies for ovarian cancer. In this review, we summarize the resistance mechanisms for conventional platinum-based chemotherapy and for the newly FDA-approved drugs.

4.
Pharmacol Res ; 178: 106162, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35259479

RESUMEN

Poly (ADP-ribose) polymerase (PARP) inhibitors (PARPis) have become a mainstay of therapy in ovarian cancer and other malignancies, including BRCA-mutant breast, prostate, and pancreatic cancers. However, a growing number of patients develop resistance to PARPis, highlighting the need to further understand the mechanisms of PARPi resistance and develop effective treatment strategies. Targeting cell cycle checkpoint protein kinases, e.g., ATR, CHK1, and WEE1, which are upregulated in response to replication stress, represents one such therapeutic approach for PARPi-resistant cancers. Mechanistically, activated cell cycle checkpoints promote cell cycle arrest, replication fork stabilization, and DNA repair, demonstrating the interplay of DNA repair proteins with replication stress in the development of PARPi resistance. Inhibitors of these cell cycle checkpoints are under investigation in PARPi-resistant ovarian and other cancers. In this review, we discuss the cell cycle checkpoints and their roles beyond mere cell cycle regulation as part of the arsenal to overcome PARPi-resistant cancers. We also address the current status and recent advancements as well as limitations of cell cycle checkpoint inhibitors in clinical trials.


Asunto(s)
Antineoplásicos , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Neoplasias Ováricas , Proteínas Tirosina Quinasas , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Puntos de Control del Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Femenino , Humanos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Proteínas Tirosina Quinasas/metabolismo
5.
Cancers (Basel) ; 14(5)2022 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-35267530

RESUMEN

RecQ helicases are essential for DNA replication, recombination, DNA damage repair, and other nucleic acid metabolic pathways required for normal cell growth, survival, and genome stability. More recently, RecQ helicases have been shown to be important for replication fork stabilization, one of the major mechanisms of PARP inhibitor resistance. Cancer cells often have upregulated helicases and depend on these enzymes to repair rapid growth-promoted DNA lesions. Several studies are now evaluating the use of RecQ helicases as potential biomarkers of breast and gynecologic cancers. Furthermore, RecQ helicases have attracted interest as possible targets for cancer treatment. In this review, we discuss the characteristics of RecQ helicases and their interacting partners that may be utilized for effective treatment strategies (as cancers depend on helicases for survival). We also discuss how targeting helicase in combination with DNA repair inhibitors (i.e., PARP and ATR inhibitors) can be used as novel approaches for cancer treatment to increase sensitivity to current treatment to prevent rise of treatment resistance.

6.
Cancer Drug Resist ; 4: 620-633, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34308273

RESUMEN

The brain is protected from toxins by a tightly regulated network of specialized cells, including endothelial cells, pericytes, astrocyes, and neurons, known collectively as the blood-brain barrier (BBB). This selectively permeable barrier permits only the most crucial molecules essential for brain function to enter and employs a number of different mechanisms to prevent the entry of potentially harmful toxins and pathogens. In addition to a physical barrier comprised of endothelial cells that form tight junctions to restrict paracellular transport, there is an active protective mechanism made up of energy-dependent transporters that efflux compounds back into the bloodstream. Two of these ATP-binding cassette (ABC) transporters are highly expressed at the BBB: P-glycoprotein (P-gp, encoded by the ABCB1 gene) and ABCG2 (encoded by the ABCG2 gene). Although a number of in vitro and in vivo systems have been developed to examine the role that ABC transporters play in keeping compounds out of the brain, all have inherent advantages and disadvantages. Zebrafish (Danio rerio) have become a model of interest for studies of the BBB due to the similarities between the zebrafish and mammalian BBB systems. In this review, we discuss what is known about ABC transporters in zebrafish and what information is still needed before the zebrafish can be recommended as a model to elucidate the role of ABC transporters at the BBB.

7.
Virology ; 552: 32-42, 2021 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-33059318

RESUMEN

The HEK-293 cell line was created in 1977 by transformation of primary human embryonic kidney cells with sheared adenovirus type 5 DNA. A previous study determined that the HEK-293 cells have neuronal markers rather than kidney markers. In this study, we tested the hypothesis whether Zika virus (ZIKV), a neurotropic virus, is able to infect and replicate in the HEK-293 cells. We show that the HEK-293 cells infected with ZIKV support viral replication as shown by indirect immunofluorescence (IFA) and quantitative reverse transcriptase-PCR (qRT-PCR). We performed RNA-seq analysis on the ZIKV-infected and the control uninfected HEK-293 cells and find 659 genes that are differentially transcribed in ZIKV-infected HEK-293 cells as compared to uninfected cells. The results show that the top 10 differentially transcribed and upregulated genes are involved in antiviral and inflammatory responses. Seven upregulated genes, IFNL1, DDX58, CXCL10, ISG15, KCNJ15, IFNIH1, and IFIT2, were validated by qRT-PCR. Altogether, our findings show that ZIKV infection alters host gene expression by affecting their antiviral and inflammatory responses.


Asunto(s)
Regulación de la Expresión Génica , Inflamación/virología , Infección por el Virus Zika/metabolismo , Infección por el Virus Zika/virología , Virus Zika/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Quimiocina CXCL10/metabolismo , Citocinas/metabolismo , Proteína 58 DEAD Box/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta/métodos , Células HEK293 , Interacciones Microbiota-Huesped , Humanos , Helicasa Inducida por Interferón IFIH1/metabolismo , Interferones/metabolismo , Interleucinas/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Proteínas de Unión al ARN/metabolismo , RNA-Seq , Receptores Inmunológicos/metabolismo , Ubiquitinas/metabolismo , Virus Zika/inmunología , Infección por el Virus Zika/inmunología
8.
Transl Oncol ; 14(1): 100917, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33129114

RESUMEN

Although the first line of therapy for epithelial ovarian cancer typically consists of taxane-platinum combination therapy, many patients develop a platinum-resistant tumor within a year. Several previous studies have looked at this cross-resistance between cisplatin and anti-microtubule drugs, but their findings have been somewhat conflicting. Here, we developed cisplatin-resistant cell lines that are resistant to low and high levels of cisplatin and explored the effects of three anti-microtubule drugs (paclitaxel, vincristine, and colchicine) on the parental and cisplatin-resistant cells. We found that cells resistant to lower levels of cisplatin were no more resistant to anti-microtubule drugs than parental cells, while cells that were resistant to higher levels of cisplatin had a subpopulation of cells that were cross-resistant to anti-microtubule drugs, clarifying discrepancies within the field. We then isolated this subpopulation by applying selective pressure with anti-microtubule drugs and performed RNA sequencing and gene set enrichment analysis to identify resistance mechanisms. This subpopulation was found to express increased levels of pro-survival TNF/NFκB signaling, among other enriched pathways, suggesting that cross-resistance was due to more general survival mechanisms found in the cisplatin-selected cells.

9.
Curr Cancer Drug Targets ; 20(7): 532-544, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32329691

RESUMEN

Improved understanding of the genomic and molecular landscape of acute myeloid leukemia (AML) has resulted in a significant evolution of our understanding of AML biology and allows refined prognostication for those receiving standard combination chemotherapy induction. This dramatic increase in knowledge preceded, and was somewhat responsible for, at least some of eight new FDA drug approvals for AML. This review discusses the impact of genomics on clinical care of AML patients and highlights newly approved FDA drugs. Despite these recent clinical advances, however, the outcome for most patients diagnosed with AML remains dire. Thus, we describe here some of the challenges identified with treating AML including off-target toxicity, drug transporters, clonal heterogeneity, and adaptive resistance, and some of the most promising opportunities for improved therapy.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Antimetabolitos Antineoplásicos/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Mutación , Inhibidores de Proteínas Quinasas/uso terapéutico , Aprobación de Drogas , Genómica/métodos , Humanos , Leucemia Mieloide Aguda/diagnóstico , Medición de Riesgo , Resultado del Tratamiento
10.
J Med Chem ; 63(3): 1434-1439, 2020 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-31702923

RESUMEN

We previously reported that some, but not all, multidrug-resistant cells that overexpressed various drug-resistance transporters were collaterally sensitive to tiopronin. In recent follow-up studies, we discovered that sensitivity to tiopronin in the original study was mediated by infection of the cells by a human-specific strain of mycoplasma. These results strongly support the need to constantly monitor cells for mycoplasma infection and keep stored samples of all cells that are used for in vitro studies.


Asunto(s)
Resistencia a Múltiples Medicamentos/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Infecciones por Mycoplasma/fisiopatología , Tiopronina/farmacología , Acetilcisteína/farmacología , Línea Celular Tumoral , Resistencia a Múltiples Medicamentos/fisiología , Resistencia a Antineoplásicos/fisiología , Humanos , Mycoplasma fermentans/fisiología
11.
Mol Cell Oncol ; 6(6): e1650631, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31692823

RESUMEN

We recently identified three sub-populations of refractory acute myeloid leukemia (AML) patients with distinct intrinsic resistance mechanisms. Furthermore, we were able to risk-stratify the overall survival of the patients and identify patients who would likely benefit from alternative therapies.

12.
Proc Natl Acad Sci U S A ; 116(21): 10494-10503, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-31064876

RESUMEN

Successful clinical remission to therapy for acute myeloid leukemia (AML) is required for long-term survival to be achieved. Despite trends in improved survival due to better supportive care, up to 40% of patients will have refractory disease, which has a poorly understood biology and carries a dismal prognosis. The development of effective treatment strategies has been hindered by a general lack of knowledge about mechanisms of chemotherapy resistance. Here, through transcriptomic analysis of 154 cases of treatment-naive AML, three chemorefractory patient groups with distinct expression profiles are identified. A classifier, four key refractory gene signatures (RG4), trained based on the expression profile of the highest risk refractory patients, validated in an independent cohort (n = 131), was prognostic for overall survival (OS) and refined an established 17-gene stemness score. Refractory subpopulations have differential expression in pathways involved in cell cycle, transcription, translation, metabolism, and/or stem cell properties. Ex vivo drug sensitivity to 122 small-molecule inhibitors revealed effective group-specific targeting of pathways among these three refractory groups. Gene expression profiling by RNA sequencing had a suboptimal ability to correctly predict those individuals resistant to conventional cytotoxic induction therapy, but could risk-stratify for OS and identify subjects most likely to have superior responses to a specific alternative therapy. Such personalized therapy may be studied prospectively in clinical trials.


Asunto(s)
Resistencia a Antineoplásicos , Quimioterapia de Inducción , Leucemia Mieloide Aguda/tratamiento farmacológico , Adulto , Anciano , Antineoplásicos/uso terapéutico , Estudios de Cohortes , Femenino , Flavonoides/uso terapéutico , Perfilación de la Expresión Génica , Heterogeneidad Genética , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/mortalidad , Masculino , Persona de Mediana Edad , Proteínas Nucleares/genética , Nucleofosmina , Piperidinas/uso terapéutico , Estados Unidos/epidemiología , Adulto Joven
13.
PLoS One ; 13(4): e0194023, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29608602

RESUMEN

The RET tyrosine kinase signaling pathway is involved in the development of endocrine resistant ER+ breast cancer. However, we know little about how ER+ cells activate RET signaling and initiate an endocrine resistant phenotype. Here we show that both ER+ endocrine resistant and sensitive breast cancers have a functional RET tyrosine kinase signaling pathway, but that endocrine sensitive breast cancer cells lack RET ligands that are necessary to drive endocrine resistance. Transcription of one RET ligand, GDNF, is necessary and sufficient to confer resistance in the ER+ MCF-7 cell line. Endogenous GDNF produced by endocrine resistant cells is translated, secreted into the media, and activates RET signaling in nearby cells. In patients, RET ligand expression predicts responsiveness to endocrine therapies and correlates with survival. Collectively, our findings show that ER+ tumor cells are "poised" for RET mediated endocrine resistance, expressing all components of the RET signaling pathway, but endocrine sensitive cells lack high expression of RET ligands that are necessary to initiate the resistance phenotype.


Asunto(s)
Neoplasias de la Mama/metabolismo , Resistencia a Antineoplásicos/genética , Proteínas Proto-Oncogénicas c-ret/metabolismo , Receptores de Estrógenos/metabolismo , Transducción de Señal/fisiología , Neoplasias de la Mama/patología , Proliferación Celular/genética , Femenino , Humanos , Células MCF-7 , Proteínas Proto-Oncogénicas c-ret/genética , Receptores de Estrógenos/genética
14.
PLoS One ; 13(4): e0194522, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29614078

RESUMEN

Discovering regulatory interactions between genes that specify the behavioral properties of cells remains an important challenge. We used the dynamics of transcriptional changes resolved by PRO-seq to identify a regulatory network responsible for endocrine resistance in breast cancer. We show that GDNF leads to endocrine resistance by switching the active state in a bi-stable feedback loop between GDNF, EGR1, and the master transcription factor ERα. GDNF stimulates MAP kinase, activating the transcription factors SRF and AP-1. SRF initiates an immediate transcriptional response, activating EGR1 and suppressing ERα. Newly translated EGR1 protein activates endogenous GDNF, leading to constitutive GDNF and EGR1 up-regulation, and the sustained down-regulation of ERα. Endocrine resistant MCF-7 cells are constitutively in the GDNF-high/ ERα-low state, suggesting that the state in the bi-stable feedback loop may provide a 'memory' of endocrine resistance. Thus, we identified a regulatory network switch that contributes to drug resistance in breast cancer.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Neoplasias de la Mama/metabolismo , Resistencia a Antineoplásicos , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Receptor alfa de Estrógeno/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Transducción de Señal , Antineoplásicos Hormonales/uso terapéutico , Sitios de Unión , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Línea Celular Tumoral , ADN Polimerasa II , Resistencia a Antineoplásicos/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Elementos de Facilitación Genéticos , Receptor alfa de Estrógeno/genética , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Humanos , Células MCF-7 , Motivos de Nucleótidos , Unión Proteica , Reproducibilidad de los Resultados , Transducción de Señal/efectos de los fármacos
15.
Cell Tissue Res ; 370(2): 275-283, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28766045

RESUMEN

We previously found that transgenic mice overexpressing MMTV-FLAG-hPAD2 (PAD2OE) developed spontaneous skin lesions, with a subset of these lesions progressing to invasive squamous cell carcinoma (SCC). The goal of this report was to better understand the potential mechanisms by which PAD2 overexpression promotes skin cancer. Here, PAD2OE mice were treated with the carcinogen, 9,10-dimethyl-1,2-benzanthracene and with O-tetradecanoylphorbol-13-acetate and then scored for papilloma formation. Additionally, tumor sections were evaluated for evidence of tumor cell invasion and inflammation. We found that the total number of papillomas was significantly increased in PAD2OE mice compared to controls. Histopathologic analysis of the lesions found that in PAD2OE skin tumors progressed to invasive SCC more frequently than controls. Additionally, we found that PAD2OE lesions were highly inflamed, with a dense inflammatory cell infiltrate and an associated increase in nuclear phospho-STAT3 (signal transducer and activator of transcription 3) in the transgenic tumors. These data suggest that overexpression of the hPAD2 transgene in the epidermis increases the malignant conversion rate of benign tumors by promoting an inflammatory microenvironment.


Asunto(s)
Inflamación/genética , Papiloma/genética , Desiminasas de la Arginina Proteica/genética , Neoplasias Cutáneas/genética , Regulación hacia Arriba , 9,10-Dimetil-1,2-benzantraceno , Animales , Carcinogénesis/inducido químicamente , Carcinogénesis/genética , Carcinogénesis/patología , Carcinógenos , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Inflamación/complicaciones , Inflamación/patología , Masculino , Ratones , Ratones Transgénicos , Papiloma/inducido químicamente , Papiloma/complicaciones , Papiloma/patología , Arginina Deiminasa Proteína-Tipo 2 , Piel/efectos de los fármacos , Piel/metabolismo , Piel/patología , Neoplasias Cutáneas/inducido químicamente , Neoplasias Cutáneas/complicaciones , Neoplasias Cutáneas/patología , Acetato de Tetradecanoilforbol
16.
BMC Cancer ; 17(1): 378, 2017 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-28549415

RESUMEN

BACKGROUND: Penetration of the mammary gland basement membrane by cancer cells is a crucial first step in tumor invasion. Using a mouse model of ductal carcinoma in situ, we previously found that inhibition of peptidylarginine deiminase 2 (PAD2, aka PADI2) activity appears to maintain basement membrane integrity in xenograft tumors. The goal of this investigation was to gain insight into the mechanisms by which PAD2 mediates this process. METHODS: For our study, we modulated PAD2 activity in mammary ductal carcinoma cells by lentiviral shRNA-mediated depletion, lentiviral-mediated PAD2 overexpression, or PAD inhibition and explored the effects of these treatments on changes in cell migration and cell morphology. We also used these PAD2-modulated cells to test whether PAD2 may be required for EGF-induced cell migration. To determine how PAD2 might promote tumor cell migration in vivo, we tested the effects of PAD2 inhibition on the expression of several cell migration mediators in MCF10DCIS.com xenograft tumors. In addition, we tested the effect of PAD2 inhibition on EGF-induced ductal invasion and elongation in primary mouse mammary organoids. Lastly, using a transgenic mouse model, we investigated the effects of PAD2 overexpression on mammary gland development. RESULTS: Our results indicate that PAD2 depletion or inhibition suppresses cell migration and alters the morphology of MCF10DCIS.com cells. In addition, we found that PAD2 depletion suppresses the expression of the cytoskeletal regulatory proteins RhoA, Rac1, and Cdc42 and also promotes a mesenchymal to epithelial-like transition in tumor cells with an associated increase in the cell adhesion marker, E-cadherin. Our mammary gland organoid study found that inhibition of PAD2 activity suppresses EGF-induced ductal invasion. In vivo, we found that PAD2 overexpression causes hyperbranching in the developing mammary gland. CONCLUSION: Together, these results suggest that PAD2 plays a critical role in breast cancer cell migration. Our findings that EGF treatment increases protein citrullination and that PAD2 inhibition blocks EGF-induced cell migration suggest that PAD2 likely functions within the EGF signaling pathway to mediate cell migration.


Asunto(s)
Carcinoma Intraductal no Infiltrante/patología , Movimiento Celular/fisiología , Neoplasias Mamarias Experimentales/patología , Desiminasas de la Arginina Proteica/metabolismo , Animales , Carcinoma Intraductal no Infiltrante/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Ratones Transgénicos , Organoides
17.
J Vis Exp ; (99): e52727, 2015 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-26067809

RESUMEN

Given the inherent difficulties in investigating the mechanisms of tumor progression in vivo, cell-based assays such as the soft agar colony formation assay (hereafter called soft agar assay), which measures the ability of cells to proliferate in semi-solid matrices, remain a hallmark of cancer research. A key advantage of this technique over conventional 2D monolayer or 3D spheroid cell culture assays is the close mimicry of the 3D cellular environment to that seen in vivo. Importantly, the soft agar assay also provides an ideal tool to rigorously test the effects of novel compounds or treatment conditions on cell proliferation and migration. Additionally, this assay enables the quantitative assessment of cell transformation potential within the context of genetic perturbations. We recently identified peptidylarginine deiminase 2 (PADI2) as a potential breast cancer biomarker and therapeutic target. Here we highlight the utility of the soft agar assay for preclinical anti-cancer studies by testing the effects of the PADI inhibitor, BB-Cl-amidine (BB-CLA), on the tumorigenicity of human ductal carcinoma in situ (MCF10DCIS) cells.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Células Madre Neoplásicas/efectos de los fármacos , Agar , Neoplasias de la Mama/enzimología , Carcinogénesis/efectos de los fármacos , Carcinoma in Situ/tratamiento farmacológico , Carcinoma in Situ/enzimología , Carcinoma in Situ/patología , Carcinoma Ductal de Mama/tratamiento farmacológico , Carcinoma Ductal de Mama/enzimología , Carcinoma Ductal de Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Progresión de la Enfermedad , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Hidrolasas/antagonistas & inhibidores , Ornitina/análogos & derivados , Ornitina/farmacología , Arginina Deiminasa Proteína-Tipo 2 , Desiminasas de la Arginina Proteica , Sefarosa/química
18.
Cancer Res ; 74(21): 6306-17, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25213324

RESUMEN

Peptidylarginine deiminase 2 (PAD2/PADI2) has been implicated in various inflammatory diseases and, more recently, cancer. The goal of this study was to test the hypothesis that PAD2 promotes oncogenesis using a transgenic mouse model. We found that about 37% of transgenic mice overexpressing human FLAG-PAD2 downstream of the MMTV-LTR promoter develop spontaneous neoplastic skin lesions. Molecular and histopathologic analyses of the resulting lesions find that they contain increased levels of markers for invasion, inflammation, and epithelial-to-mesenchymal transition (EMT) and that a subset of the lesions progress to invasive squamous cell carcinoma (SCC). We then stably overexpressed FLAG-PAD2 in the human SCC cell line, A431, and found that the PAD2-overexpressing cells were more tumorigenic in vitro and also contained elevated levels of markers for inflammation and EMT. Collectively, these studies provide the first genetic evidence that PAD2 functions as an oncogene and suggest that PAD2 may promote tumor progression by enhancing inflammation within the tumor microenvironment.


Asunto(s)
Carcinoma de Células Escamosas/genética , Regulación Neoplásica de la Expresión Génica , Hidrolasas/biosíntesis , Neoplasias Cutáneas/genética , Animales , Carcinoma de Células Escamosas/patología , Transformación Celular Neoplásica , Transición Epitelial-Mesenquimal/genética , Humanos , Hidrolasas/genética , Ratones , Ratones Transgénicos , Arginina Deiminasa Proteína-Tipo 2 , Desiminasas de la Arginina Proteica , Neoplasias Cutáneas/patología , Microambiente Tumoral
19.
Bioessays ; 36(8): 736-40, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24889365

RESUMEN

Histone post-translational modifications (PTMs) alter the chromatin architecture, generating "open" and "closed" states, and these structural changes can modulate gene expression under specific cellular conditions. While methylation and acetylation are the best-characterized histone PTMs, citrullination by the protein arginine deiminases (PADs) represents another important player in this process. In addition to "fine tuning" chromatin structure at specific loci, histone citrullination can also promote rapid global chromatin decondensation during the formation of extracellular traps (ETs) in immune cells. Recent studies now show that PAD4-mediated citrullination of histone H1 at promoter elements can also promote localized chromatin decondensation in stem cells, thus regulating the pluripotent state. These observations suggest that PAD-mediated histone deimination profoundly affects chromatin structure, possibly above and beyond that of other PTMs. Additionally, these recent findings further enhance our understanding of PAD biology and the important contributions that these enzymes play in development, health, and disease.


Asunto(s)
Histonas/metabolismo , Hidrolasas/fisiología , Células Madre Pluripotentes Inducidas/enzimología , Animales , Artritis Reumatoide/enzimología , Reprogramación Celular , Ensamble y Desensamble de Cromatina , Citrulina/metabolismo , Humanos , Procesamiento Proteico-Postraduccional , Arginina Deiminasa Proteína-Tipo 4 , Desiminasas de la Arginina Proteica , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética
20.
Mol Cancer ; 13: 129, 2014 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-24886523

RESUMEN

Over three decades have passed since the first report on the expression of CA125 by ovarian tumors. Since that time our understanding of ovarian cancer biology has changed significantly to the point that these tumors are now classified based on molecular phenotype and not purely on histological attributes. However, CA125 continues to be, with the recent exception of HE4, the only clinically reliable diagnostic marker for ovarian cancer. Many large-scale clinical trials have been conducted or are underway to determine potential use of serum CA125 levels as a screening modality or to distinguish between benign and malignant pelvic masses. CA125 is a peptide epitope of a 3-5 million Da mucin, MUC16. Here we provide an in-depth review of the literature to highlight the importance of CA125 as a prognostic and diagnostic marker for ovarian cancer. We focus on the increasing body of literature describing the biological role of MUC16 in the progression and metastasis of ovarian tumors. Finally, we consider previous and on-going efforts to develop therapeutic approaches to eradicate ovarian tumors by targeting MUC16. Even though CA125 is a crucial marker for ovarian cancer, the exact structural definition of this antigen continues to be elusive. The importance of MUC16/CA125 in the diagnosis, progression and therapy of ovarian cancer warrants the need for in-depth research on the biochemistry and biology of this mucin. A renewed focus on MUC16 is likely to culminate in novel and more efficient strategies for the detection and treatment of ovarian cancer.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/genética , Antígeno Ca-125/genética , Inmunoterapia , Proteínas de la Membrana/genética , Neoplasias Ováricas/terapia , Biomarcadores de Tumor/antagonistas & inhibidores , Biomarcadores de Tumor/inmunología , Antígeno Ca-125/inmunología , Ensayos Clínicos como Asunto , Progresión de la Enfermedad , Femenino , Expresión Génica , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/patología , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/inmunología , Metástasis de la Neoplasia , Neoplasias Ováricas/diagnóstico , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Pronóstico , Proteínas/genética , Proteínas/inmunología , Proteína 2 de Dominio del Núcleo de Cuatro Disulfuros WAP
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA