Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cancers (Basel) ; 13(19)2021 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-34638504

RESUMEN

Deciphering the mechanisms that drive transdifferentiation to neuroendocrine prostate cancer (NEPC) is crucial to identifying novel therapeutic strategies against this lethal and aggressive subtype of advanced prostate cancer (PCa). Further, the role played by exosomal microRNAs (miRs) in mediating signaling mechanisms that propagate the NEPC phenotype remains largely elusive. The unbiased differential miR expression profiling of human PCa cells genetically modulated for TBX2 expression led to the identification of miR-200c-3p. Our findings have unraveled the TBX2/miR-200c-3p/SOX2/N-MYC signaling axis in NEPC transdifferentiation. Mechanistically, we found that: (1) TBX2 binds to the promoter and represses the expression of miR-200c-3p, a miR reported to be lost in castrate resistant prostate cancer (CRPC), and (2) the repression of miR-200c-3p results in the increased expression of its targets SOX2 and N-MYC. In addition, the rescue of mir-200c-3p in the context of TBX2 blockade revealed that miR-200c-3p is the critical intermediary effector in TBX2 regulation of SOX2 and N-MYC. Further, our studies show that in addition to the intracellular mode, TBX2/miR-200c-3p/SOX2/N-MYC signaling can promote NEPC transdifferentiation via exosome-mediated intercellular mechanism, an increasingly recognized and key mode of propagation of the NEPC phenotype.

2.
Nanotheranostics ; 5(1): 57-72, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33391975

RESUMEN

Successful visualization of prostate cancer (PCa) tumor margins during surgery remains a major challenge. The visualization of these tumors during surgery via near infrared fluorescence (NIRF) imaging would greatly enhance surgical resection, minimizing tumor recurrence and improving outcome. Furthermore, chemotherapy is typically administered to patients after surgery to treat any missed tumor tissue around the surgical area, minimizing metastasis and increasing patient survival. For these reasons, a theranostics fluorescent nanoparticle could be developed to assist in the visualization of PCa tumor margins, while also delivering chemotherapeutic drug after surgery. Methods: Ferumoxytol (FMX) conjugated to the fluorescent dye and PCa targeting agent, heptamethine carbocyanine (HMC), yielded the HMC-FMX nanoprobe that was tested in vitro with various PCa cell lines and in vivo with both subcutaneous and orthotopic PCa mouse models. Visualization of these tumors via NIRF imaging after administration of HMC-FMX was performed. In addition, delivery of chemotherapeutic drug and their effect on tumor growth was also assessed. Results: HMC-FMX internalized into PCa cells, labeling these cells and PCa tumors in mice with near infrared fluorescence, facilitating tumor margin visualization. HMC-FMX was also able to deliver drugs to these tumors, reducing cell migration and slowing down tumor growth. Conclusion: HMC-FMX specifically targeted PCa tumors in mice allowing for the visualization of tumor margins by NIRF imaging. Furthermore, delivery of anticancer drugs by HMC-FMX effectively reduced prostate tumor growth and reduced cell migration in vitro. Thus, HMC-FMX can potentially translate into the clinic as a nanotheranostics agent for the intraoperative visualization of PCa tumor margins, and post-operative treatment of tumors with HMC-FMX loaded with anticancer drugs.


Asunto(s)
Nanopartículas , Neoplasias de la Próstata/patología , Humanos , Cuidados Intraoperatorios , Masculino , Neoplasias de la Próstata/cirugía
3.
Oncogene ; 39(7): 1543-1556, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31685946

RESUMEN

Periodontal diseases can lead to chronic inflammation affecting the integrity of the tooth supporting tissues. Recently, a striking association has been made between periodontal diseases and primary cancers in the absence of a mechanistic understanding. Here we address the effect of periodontal inflammation (PI) on tumor progression, metastasis, and possible underlining mechanisms. We show that an experimental model of PI in mice can promote lymph node (LN) micrometastasis, as well as head and neck metastasis of 4T1 breast cancer cells, both in early and late stages of cancer progression. The cervical LNs had a greater tumor burden and infiltration of MDSC and M2 macrophages compared with LNs at other sites. Pyroptosis and the resultant IL-1ß production were detected in patients with PI, mirrored in mouse models. Anakinra, IL-1 receptor antagonist, limited metastasis, and MDSC recruitment at early stages of tumor progression, but failed to reverse established metastatic tumors. PI and the resulting production of IL-1ß was found to promote CCL5, CXCL12, CCL2, and CXCL5 expression. These chemokines recruit MDSC and macrophages, finally enabling the generation of a premetastatic niche in the inflammatory site. These findings support the idea that periodontal inflammation promotes metastasis of breast cancer by recruiting MDSC in part by pyroptosis-induced IL-1ß generation and downstream CCL2, CCL5, and CXCL5 signaling in the early steps of metastasis. These studies define the role for IL-1ß in the metastatic progression of breast cancer and highlight the need to control PI, a pervasive inflammatory condition in older patients.


Asunto(s)
Neoplasias de la Mama/complicaciones , Neoplasias de la Mama/patología , Células Mieloides/patología , Enfermedades Periodontales/complicaciones , Animales , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Interleucina-1beta/metabolismo , Ratones , Metástasis de la Neoplasia , Piroptosis
4.
Cancers (Basel) ; 11(10)2019 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-31547070

RESUMEN

Our understanding of neuroendocrine prostate cancer (NEPC) has assumed a new perspective in light of the recent advances in research. Although classical NEPC is rarely seen in the clinic, focal neuroendocrine trans-differentiation of prostate adenocarcinoma occurs in about 30% of advanced prostate cancer (PCa) cases, and represents a therapeutic challenge. Even though our knowledge of the mechanisms that mediate neuroendocrine differentiation (NED) is still evolving, the role of androgen deprivation therapy (ADT) as a key driver of this phenomenon is increasingly becoming evident. In this review, we discuss the molecular, cellular, and therapeutic mediators of NED, and emphasize the role of the tumor microenvironment (TME) in orchestrating the phenotype. Understanding the role of the TME in mediating NED could provide us with valuable insights into the plasticity associated with the phenotype, and reveal potential therapeutic targets against this aggressive form of PCa.

5.
Nanotheranostics ; 3(2): 196-211, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31183314

RESUMEN

Purpose: A successful cancer surgery requires the complete removal of cancerous tissue, while also sparing as much healthy, non-cancerous tissue as possible. To achieve this, an accurate identification of tumor boundaries during surgery is critical, but intra-operative tumor visualization remains challenging. Fluorescence imaging is a promising method to improve tumor detection and delineate tumor boundaries during surgery, but the lack of stable, long-circulating, clinically-translatable fluorescent probes that can identify tumors with high signal-to-noise ratios and low background fluorescence signals have prevented its widespread application. Methods: We screened the optical properties of several fluorescent dyes before and after nanoprobe encapsulation, and then identified nanoprobes with quenched fluorescence that were re-activated upon dye release. The physical and biological properties of these nanoprobes leading to fluorescence activation were investigated in vitro. Further, the cancer imaging properties of both free dyes and nanoprobe-encapsulated dyes were compared in vivo. Results: A novel fluorescent nanoprobe was prepared by combining two FDA-approved agents commonly used in the clinic: Feraheme (FH) and indocyanine green (ICG). The resulting FH-entrapped ICG nanoprobe [FH(ICG)] displayed quenched fluorescence compared to other nanoprobes, and this quenched fluorescence was re-activated in acidic tumor microenvironment conditions (pH 6.8) and upon uptake into cancer cells. Finally, in vivo studies in a prostate cancer mouse model demonstrated that FH(ICG) treatments enhance long-term fluorescence signals in tumors compared to ICG treatments, allowing for fluorescence-guided tumor identification using clinically relevant fluorescence cameras. Conclusions: FH(ICG) nanoprobes were identified as fluorescent nanoprobes with beneficial fluorescence activation properties compared to other FH-entrapped dyes. The activatable nature of this nanoprobe allows for a low background fluorescence signal and high signal-to-noise ratio within a long-circulating nanoagent, which allows for long-term fluorescence signals from tumors that enabled their fluorescence-guided detection. This activatable nanoprobe offers tremendous potential as a clinically translatable image-guided cancer therapy modality that can be prepared in a clinical setting.


Asunto(s)
Colorantes Fluorescentes , Nanoestructuras , Neoplasias Experimentales , Imagen Óptica , Neoplasias de la Próstata , Animales , Colorantes Fluorescentes/química , Colorantes Fluorescentes/farmacología , Humanos , Masculino , Ratones , Ratones Desnudos , Ratones SCID , Nanoestructuras/química , Nanoestructuras/uso terapéutico , Neoplasias Experimentales/diagnóstico por imagen , Neoplasias Experimentales/metabolismo , Células PC-3 , Neoplasias de la Próstata/diagnóstico por imagen , Neoplasias de la Próstata/metabolismo
6.
Nanotheranostics ; 3(1): 66-88, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30662824

RESUMEN

Biological interactions between tumor-associated macrophages (TAMs), cancer cells and other cells within the tumor microenvironment contribute to tumorigenesis, tumor growth, metastasis and therapeutic resistance. TAMs can remodel the tumor microenvironment to reduce growth barriers such as the dense extracellular matrix and shift tumors towards an immunosuppressive microenvironment that protects cancer cells from targeted immune responses. Nanoparticles can interrupt these biological interactions within tumors by altering TAM phenotypes through a process called polarization. Macrophage polarization within tumors can shift TAMs from a growth-promoting phenotype towards a cancer cell-killing phenotype that predicts treatment efficacy. Because many types of nanoparticles have been shown to preferentially accumulate within macrophages following systemic administration, there is considerable interest in identifying nanoparticle effects on TAM polarization, evaluating nanoparticle-induced TAM polarization effects on cancer treatment using drug-loaded nanoparticles and identifying beneficial types of nanoparticles for effective cancer treatment. In this review, the macrophage polarization effects of nanoparticles will be described based on their primary chemical composition. Because of their strong macrophage-polarizing and antitumor effects compared to other types of nanoparticles, the effects of iron oxide nanoparticles on macrophages will be discussed in detail. By comparing the macrophage polarization effects of various nanoparticle treatments reported in the literature, this review aims to both elucidate nanoparticle material effects on macrophage polarization and to provide insight into engineering nanoparticles with more beneficial immunological responses for cancer treatment.


Asunto(s)
Polaridad Celular , Macrófagos , Nanopartículas/uso terapéutico , Neoplasias , Escape del Tumor/efectos de los fármacos , Microambiente Tumoral , Animales , Polaridad Celular/efectos de los fármacos , Polaridad Celular/inmunología , Humanos , Macrófagos/inmunología , Macrófagos/patología , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Neoplasias/patología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
7.
Oncogene ; 38(5): 716-730, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30177832

RESUMEN

Heterogeneous prostatic carcinoma-associated fibroblasts (CAF) contribute to tumor progression and resistance to androgen signaling deprivation therapy (ADT). CAF subjected to extended passaging, compared to low passage CAF, were found to lose tumor expansion potential and heterogeneity. Cell surface endoglin (CD105), known to be expressed on proliferative endothelia and mesenchymal stem cells, was diminished in high passage CAF. RNA-sequencing revealed SFRP1 to be distinctly expressed by tumor-inductive CAF, which was further demonstrated to occur in a CD105-dependent manner. Moreover, ADT resulted in further expansion of the CD105+ fibroblastic population and downstream SFRP1 in 3-dimensional cultures and patient-derived xenograft tissues. In patients, CD105+ fibroblasts were found to circumscribe epithelia with neuroendocrine differentiation. CAF-derived SFRP1, driven by CD105 signaling, was necessary and sufficient to induce prostate cancer neuroendocrine differentiation in a paracrine manner. A partially humanized CD105 neutralizing antibody, TRC105, inhibited fibroblastic SFRP1 expression and epithelial neuroendocrine differentiation. In a novel synthetic lethality paradigm, we found that simultaneously targeting the epithelia and its microenvironment with ADT and TRC105, respectively, reduced castrate-resistant tumor progression, in a model where either ADT or TRC105 alone had little effect.


Asunto(s)
Diferenciación Celular , Endoglina/metabolismo , Fibroblastos/metabolismo , Proteínas de Neoplasias/metabolismo , Células Neuroendocrinas/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Transducción de Señal , Línea Celular Tumoral , Endoglina/genética , Fibroblastos/patología , Humanos , Masculino , Proteínas de Neoplasias/genética , Células Neuroendocrinas/patología , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/patología
8.
Cancer Med ; 7(9): 4639-4649, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30094958

RESUMEN

BACKGROUND: Recruited myeloid cells are known to promote cancer initiation, malignant progression, metastasis, and resistance to therapy in the tumor niche. We tested the hypothesis that circulating blood monocytes from advanced prostate cancer (PCa) patients exhibit a protumor phenotype and directly influence the tumor microenvironment in response to tumor-derived signals. METHODS: Blood monocytes from advanced and stable PCa patients were cultured, and the conditioned media (CM) were collected and analyzed using standard invasion and wound closure assays to measure effects on invasion and motility of PCa tumor cells. We then identified the proteome profile of these monocytes using proteome array and ELISA. RESULTS: Conditioned media from circulating monocytes in patients with metastatic prostate cancer (PCa-M) increased invasion of epithelial PCa cells in vitro. Proteome Profiler Analysis revealed that monocyte-derived CM from metastatic castration-resistant (mCRPC) patients presented high levels of chitinase-3-like 1 (CHI3L1, YKL-40) when compared to patients with stable disease (PCa-N) and healthy control individuals (HC). The only described receptor for CHI3L1, interleukin-13 receptor α2 (IL-13Rα2), was significantly up-regulated in the human metastatic PCa cell line, ARCaPM . Accordingly, we observed that the activation of IL-13Rα2 from PCa-M CM increased the invasiveness of ARCaPM cells while siRNA directed against this receptor significantly reduced invasiveness of these cells in the presence of CM from PCa-M patients. CONCLUSIONS: Thus, we show that circulating monocytes from metastatic PCa patients exert a tumor-promoting role via the secretion of CHI3L1, and CHI3L1 demands further exploration as a possible therapeutic target in advanced PCa.


Asunto(s)
Comunicación Celular , Movimiento Celular , Células Epiteliales/metabolismo , Monocitos/metabolismo , Neoplasias de la Próstata/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Proteína 1 Similar a Quitinasa-3/metabolismo , Medios de Cultivo Condicionados/farmacología , Humanos , Interleucina-1beta/metabolismo , Masculino , Neoplasias de la Próstata/patología
9.
J Clin Invest ; 128(10): 4472-4484, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30047926

RESUMEN

Prostate cancer is an androgen-dependent disease subject to interactions between the tumor epithelium and its microenvironment. Here, we found that epigenetic changes in prostatic cancer-associated fibroblasts (CAF) initiated a cascade of stromal-epithelial interactions. This facilitated lethal prostate cancer growth and development of resistance to androgen signaling deprivation therapy (ADT). We identified a Ras inhibitor, RASAL3, as epigenetically silenced in human prostatic CAF, leading to oncogenic Ras activity driving macropinocytosis-mediated glutamine synthesis. Interestingly, ADT further promoted RASAL3 epigenetic silencing and glutamine secretion by prostatic fibroblasts. In an orthotopic xenograft model, subsequent inhibition of macropinocytosis and glutamine transport resulted in antitumor effects. Stromal glutamine served as a source of energy through anaplerosis and as a mediator of neuroendocrine differentiation for prostate adenocarcinoma. Antagonizing the uptake of glutamine restored sensitivity to ADT in a castration-resistant xenograft model. In validating these findings, we found that prostate cancer patients on ADT with therapeutic resistance had elevated blood glutamine levels compared with those with therapeutically responsive disease (odds ratio = 7.451, P = 0.02). Identification of epigenetic regulation of Ras activity in prostatic CAF revealed RASAL3 as a sensor for metabolic and neuroendocrine reprogramming in prostate cancer patients failing ADT.


Asunto(s)
Silenciador del Gen , Glutamina/metabolismo , Proteínas de Neoplasias/metabolismo , Tumores Neuroendocrinos/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Activadoras de ras GTPasa/metabolismo , Animales , Fibroblastos/metabolismo , Fibroblastos/patología , Glutamina/genética , Humanos , Masculino , Ratones , Proteínas de Neoplasias/genética , Tumores Neuroendocrinos/genética , Tumores Neuroendocrinos/patología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proteínas Activadoras de ras GTPasa/genética
10.
Oncotarget ; 8(50): 87891-87902, 2017 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-29152128

RESUMEN

The tumor microenvironment (TME) is increasingly recognized as the arbiter of metastatic progression and drug resistance in advanced prostate cancer (PCa). Cabozantinib is a potent tyrosine kinase inhibitor (TKI) with reported biological activity in the PCa epithelia, but failed to provide an overall survival benefit in phase 3 clinical trials. However, the promising biologic efficacy of the drug in early trials warranted a better understanding of the mechanism of action, with the goal of improving patient selection for TKI-based therapy such as cabozantinib. We found a 100-fold lower cabozantinib IC50 in macrophages, PCa associated fibroblasts, and bone marrow fibroblasts compared to PCa epithelia. In PCa mouse models, pre-treatment with cabozantinib potentiated osseous and visceral tumor engraftment, suggesting a pro-tumorigenic host response to the drug. We further found that the host effects of cabozantinib impacted bone turnover, but not necessarily tumor expansion. Cabozantinib affected M1 macrophage polarization in mice. Analogously, circulating monocytes from PCa patients treated with cabozantinib, demonstrated a striking correlation of monocyte reprograming with therapeutic bone responsivity, to support patient selection at early stages of treatment. Thus, a re-evaluation of TKI-based therapeutic strategies in PCa can be considered for suitable patient populations based on TME responses.

11.
Cancer Res ; 77(6): 1331-1344, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28108510

RESUMEN

Identification of factors that mediate visceral and bone metastatic spread and subsequent bone remodeling events is highly relevant to successful therapeutic intervention in advanced human prostate cancer. TBX2, a T-box family transcription factor that negatively regulates cell-cycle inhibitor p21, plays critical roles during embryonic development, and recent studies have highlighted its role in cancer. Here, we report that TBX2 is overexpressed in human prostate cancer specimens and bone metastases from xenograft mouse models of human prostate cancer. Blocking endogenous TBX2 expression in PC3 and ARCaPM prostate cancer cell models using a dominant-negative construct resulted in decreased tumor cell proliferation, colony formation, and invasion in vitro Blocking endogenous TBX2 in human prostate cancer mouse xenografts decreased invasion and abrogation of bone and soft tissue metastasis. Furthermore, blocking endogenous TBX2 in prostate cancer cells dramatically reduced bone-colonizing capability through reduced tumor cell growth and bone remodeling in an intratibial mouse model. TBX2 acted in trans by promoting transcription of the canonical WNT (WNT3A) promoter. Genetically rescuing WNT3A levels in prostate cancer cells with endogenously blocked TBX2 partially restored the TBX2-induced prostate cancer metastatic capability in mice. Conversely, WNT3A-neutralizing antibodies or WNT antagonist SFRP-2 blocked TBX2-induced invasion. Our findings highlight TBX2 as a novel therapeutic target upstream of WNT3A, where WNT3A antagonists could be novel agents for the treatment of metastasis and for skeletal complications in prostate cancer patients. Cancer Res; 77(6); 1331-44. ©2017 AACR.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Biomarcadores de Tumor/metabolismo , Neoplasias Óseas/prevención & control , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias de la Próstata/prevención & control , Proteínas de Dominio T Box/antagonistas & inhibidores , Proteína Wnt3A/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Biomarcadores de Tumor/genética , Neoplasias Óseas/metabolismo , Neoplasias Óseas/secundario , Proliferación Celular/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones SCID , Terapia Molecular Dirigida , Clasificación del Tumor , Pronóstico , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Transducción de Señal/efectos de los fármacos , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Células Tumorales Cultivadas , Proteína Wnt3A/genética , Proteína Wnt3A/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Sci Rep ; 6: 39257, 2016 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-27995963

RESUMEN

Hemorrhagic cystitis is an inflammatory and ulcerative bladder condition associated with systemic chemotherapeutics, like cyclophosphomide. Earlier, we reported reactive oxygen species resulting from cyclophosphamide metabolite, acrolein, causes global methylation followed by silencing of DNA damage repair genes. Ogg1 (8-oxoguanine DNA glycosylase) is one such silenced base excision repair enzyme that can restore DNA integrity. The accumulation of DNA damage results in subsequent inflammation associated with pyroptotic death of bladder smooth muscle cells. We hypothesized that reversing inflammasome-induced imprinting in the bladder smooth muscle could prevent the inflammatory phenotype. Elevated recruitment of Dnmt1 and Dnmt3b to the Ogg1 promoter in acrolein treated bladder muscle cells was validated by the pattern of CpG methylation revealed by bisulfite sequencing. Knockout of Ogg1 in detrusor cells resulted in accumulation of reactive oxygen mediated 8-Oxo-dG and spontaneous pyroptotic signaling. Histone deacetylase (HDAC) inhibitor, suberoylanilide hydroxamic acid (SAHA), restored Ogg1 expression in cells treated with acrolein and mice treated with cyclophosphamide superior to the standard of care, mesna or nicotinamide-induced DNA demethylation. SAHA restored cyclophosphamide-induced bladder pathology to that of untreated control mice. The observed epigenetic imprinting induced by inflammation suggests a new therapeutic target for the treatment of hemorrhagic cystitis.


Asunto(s)
Ciclofosfamida/toxicidad , Cistitis/etiología , ADN Glicosilasas/metabolismo , Reparación del ADN/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Acroleína/toxicidad , Animales , Células Cultivadas , Cistitis/inducido químicamente , Cistitis/metabolismo , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Daño del ADN/efectos de los fármacos , ADN Glicosilasas/deficiencia , ADN Glicosilasas/genética , Regulación hacia Abajo/efectos de los fármacos , Femenino , Mesna/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Niacinamida/farmacología , Especies Reactivas de Oxígeno/metabolismo , Vejiga Urinaria/efectos de los fármacos , Vejiga Urinaria/metabolismo , Vejiga Urinaria/patología , Vorinostat , ADN Metiltransferasa 3B
13.
Scand J Work Environ Health ; 40(6): 631-8, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25205073

RESUMEN

OBJECTIVES: According to the "stress-as-offense-to-self" perspective, work tasks that are considered unnecessary or unreasonable - so-called "illegitimate work tasks" - are likely to elicit stress-reactions. Previous studies, mostly cross-sectional, have shown that illegitimate tasks are associated with increased self-reported stress, cortisol, and counterproductive work behavior. In this article, we examine the prospective association between unnecessary work tasks, one type of illegitimate work tasks, and mental health among Danish human service workers. Further, we explore whether this association is modified by sex, age, occupational position, and baseline mental health status. METHODS: The data were obtained from self-administered questionnaires from 1351 Danish human service workers in three waves of data-collection during 1999-2005. We measured unnecessary work tasks by a single item, and assessed mental health using the 5-item mental health inventory from the Short form 36 questionnaire. We analyzed data using multi-level modeling, adjusting for potential confounding by sex, age, cohabitation, occupational position, and baseline mental health. RESULTS: Unnecessary work tasks were prospectively associated with a decreased level of mental health. This association was stronger for employees with poor baseline mental health and tended to be more pronounced among older employees. Among participants with poor baseline mental health, the association was explained by neither psychological demands nor decision latitude. CONCLUSIONS: Our findings suggest that the prevention of unnecessary work tasks may benefit employee mental health, particularly among employees with pre-existing mental health problems.


Asunto(s)
Salud Mental , Salud Laboral , Servicio Social , Estrés Psicológico/psicología , Dinamarca/epidemiología , Femenino , Humanos , Masculino , Estudios Prospectivos , Autoimagen , Autoinforme , Encuestas y Cuestionarios , Análisis y Desempeño de Tareas
14.
Cancer Cell ; 23(3): 332-46, 2013 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-23518348

RESUMEN

Understanding the mechanism underlying the regulation of the androgen receptor (AR), a central player in the development of castration-resistant prostate cancer (CRPC), holds promise for overcoming the challenge of treating CRPC. We demonstrate that the ubiquitin ligase Siah2 targets a select pool of NCOR1-bound, transcriptionally-inactive AR for ubiquitin-dependent degradation, thereby promoting expression of select AR target genes implicated in lipid metabolism, cell motility, and proliferation. Siah2 is required for prostate cancer cell growth under androgen-deprivation conditions in vitro and in vivo, and Siah2 inhibition promotes prostate cancer regression upon castration. Notably, Siah2 expression is markedly increased in human CRPCs. Collectively, we find that selective regulation of AR transcriptional activity by the ubiquitin ligase Siah2 is important for CRPC development.


Asunto(s)
Proteínas Nucleares/metabolismo , Co-Represor 1 de Receptor Nuclear/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Transcripción Genética , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Castración , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Metabolismo de los Lípidos , Masculino , Ratones , Proteínas Nucleares/genética , Interferencia de ARN , ARN Interferente Pequeño , Transducción de Señal , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/genética
15.
Cell Adh Migr ; 6(3): 231-5, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22568983

RESUMEN

The major cellular components of tumor microenvironment, referred to as the cancer stroma, are composed of cancer-associated fibroblasts that support tumor epithelial growth, invasion and therapeutic resistance. Thus when we speak of developing therapies that address tumor heterogeneity it is not only a matter of different mutations within the tumor epithelia. While individual mutations in the stromal compartment are controversial, the heterogeneity in fibroblastic population in a single tumor is not up for debate. Cooperative interaction among heterotypic fibroblasts and tumor cells contribute to cancer progression. Therefore to tackle solid tumors, we need to understand its complex microenvironment. Here we review some seminal developments in the field of tumor microenvironment, mainly focusing on cancer-associated fibroblast.


Asunto(s)
Progresión de la Enfermedad , Fibroblastos/patología , Neoplasias/patología , Células del Estroma/patología , Animales , Fibroblastos/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neoplasias/metabolismo , Comunicación Paracrina , Transducción de Señal , Células del Estroma/metabolismo , Microambiente Tumoral
16.
Antonie Van Leeuwenhoek ; 100(3): 405-13, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21638110

RESUMEN

Plant growth promoting Pantoea agglomerans NBRISRM (NBRISRM) was able to produce 60.4 µg/ml indole acetic acid and solubilize 77.5 µg/ml tri-calcium phosphate under in vitro conditions. Addition of 2% NaCl (w/v) in the media induced the IAA production and phosphate solubilization by 11% and 7%, respectively. For evaluating the plant growth promotory effect of NBRISRM inoculation a micro plot trial was conducted using maize and chickpea as host plants. The results revealed significant increase in all growth parameters tested in NBRISRM inoculated maize and chickpea plants, which were further confirmed by higher macronutrients (N, P and K) accumulation as compared to un-inoculated controls. Throughout the growing season of maize and chickpea, rhizosphere population of NBRISRM were in the range 10(7)-10(8) CFU/g soil and competing with 10(7)-10(9) CFU/g soil with heterogeneous bacterial population. Functional richness, diversity, and evenness were found significantly higher in maize rhizosphere as compared to chickpea, whereas NBRISRM inoculation were not able to change it, in both crops as compared to their un-inoculated control. To the best of our knowledge this is first report where we demonstrated the effect of P. agglomerans strain for improving maize and chickpea growth without altering the functional diversity.


Asunto(s)
Biodiversidad , Cicer/microbiología , Pantoea/aislamiento & purificación , Pantoea/fisiología , Rizosfera , Microbiología del Suelo , Zea mays/microbiología , Cicer/crecimiento & desarrollo , Datos de Secuencia Molecular , Pantoea/clasificación , Pantoea/genética , Filogenia , Raíces de Plantas/crecimiento & desarrollo , Raíces de Plantas/microbiología , Zea mays/crecimiento & desarrollo
17.
Prostate ; 71(2): 184-96, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-20672321

RESUMEN

BACKGROUND: Matriptase, a type II transmembrane serine protease, has been linked to initiation and promotion of epidermal carcinogenesis in a murine model, suggesting that deregulation of its role in epithelia contributes to transformation. In human prostate cancer, matriptase expression correlates with progression. It is therefore of interest to determine how matriptase may contribute to epithelial neoplastic progression. One approach for studying this is to identify potential matriptase substrates involved in epithelial integrity and/or transformation like the extracellular matrix macromolecule, laminin-332 (Ln-332), which is found in the basement membrane of many epithelia, including prostate. Proteolytic processing of Ln-332 regulates cell motility of both normal and transformed cells, which has implications in cancer progression. METHODS: In vitro cleavage experiments were performed with purified Ln-332 protein and matriptase. Western blotting, enzyme inhibition assays, and mass spectrometry were used to confirm cleavage events. Matriptase overexpressing LNCaP prostate cancer cells were generated and included in Transwell migration assays and single cell motility assays, along with other prostate cells. RESULTS: We report that matriptase proteolytically cleaves Ln-332 in the ß3 chain. Substrate specificity was confirmed by blocking cleavage with the matriptase inhibitor, Kunitz domain-1. Transwell migration assays showed that DU145 cell motility was significantly enhanced when plated on matriptase-cleaved Ln-332. Similarly, Transwell migration of matriptase-overexpressing LNCaP cells was significantly increased on Ln-332 and, as determined by live single-cell microscopy, two motility parameters of this cell line, speed and directional persistence, were also higher. CONCLUSIONS: Proteolytic processing of Ln-332 by matriptase enhances speed and directional persistence of prostate cancer cells.


Asunto(s)
Movimiento Celular/fisiología , Laminina/metabolismo , Neoplasias de la Próstata/metabolismo , Serina Endopeptidasas/metabolismo , Línea Celular Tumoral , Humanos , Masculino , Microscopía de Contraste de Fase , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , Proteínas Inhibidoras de Proteinasas Secretoras/farmacología , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
18.
J Oncol ; 20102010.
Artículo en Inglés | MEDLINE | ID: mdl-20862207

RESUMEN

Lysophosphatidic acid (LPA) is a bioactive phospholipid that affects various biological functions, such as cell proliferation, migration, survival, wound healing, and tumor invasion through LPA receptors. Previously, we reported that LPA induces A431 colony dispersal, accompanied by disruption of cell-cell contacts and cell migration. However, it remains unclear how LPA affects cell migration and gene expression during A431 colony dispersal. In this paper, we performed cDNA microarray analysis to investigate this question by comparing gene expression between untreated and LPA-treated A431 cells. Interestingly, these results revealed that LPA treatment upregulates several TGF-ß1 target genes, including laminin-332 (Ln-332) components (α3, ß3, and γ2 chains). Western blot analysis also showed that LPA increased phosphorylation of Smad2, an event that is carried out by TGF-ß1 interactions. Among the genes upregulated, we further addressed the role of Ln-332. Real-time PCR analysis confirmed the transcriptional upregulation of all α3, ß3, and γ2 chains of Ln-332 by LPA, corresponding to the protein level increases revealed by western blot. Further, the addition of anti-Ln-332 antibody prevented LPA-treated A431 colonies from dispersing. Taken together, our results suggest that LPA-induced Ln-332 plays a significant role in migration of individual cells from A431 colonies.

19.
J Cell Physiol ; 223(3): 541-8, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20301201

RESUMEN

Laminin-332 (Ln-332) is an extracellular matrix molecule that regulates cell adhesion, spreading, and migration by interaction with cell surface receptors such as alpha3beta1 and alpha6beta4. Previously, we developed a function-blocking monoclonal antibody against rat Ln-332, CM6, which blocks hemidesmosome assembly induced by Ln-332-alpha6beta4 interactions. However, the location of its epitope on Ln-332 has remained unclear. In this study, we show that the CM6 epitope is located on the laminin G-like (LG)2 module of the Ln-332 alpha3 chain. To specify the residues involved in this epitope, we produced a series of GST-fused alpha3 LG2 mutant proteins in which rat-specific acids were replaced with human acids by a site-directed mutagenesis strategy. CM6 reactivity against these proteins showed that CM6 binds to the (1089)NERSVR(1094) sequence of rat Ln-332 LG2 module. In a structural model, this sequence maps to an LG2 loop sequence that is exposed to solvent according to predictions, consistent with its accessibility to antibody. CM6 inhibits integrin-dependent cell adhesion on Ln-332 and inhibits cell spreading on both Ln-332 and recombinant LG2 (rLG2; but not rLG3), suggesting the presence of an alpha3beta1 binding site on LG2. However, we were unable to show that rLG2 supports adhesion in standard assays, suggesting that LG2 may contain a "weak" integrin binding site, only detectable in spreading assays that do not require washes. These results, together with our previous findings, indicate that binding sites for alpha3beta1 and alpha6beta4 are closely spaced in the Ln-332 LG domains where they regulate alternative cell functions, namely adhesion/migration or hemidesmosome anchoring.


Asunto(s)
Anticuerpos Bloqueadores/metabolismo , Anticuerpos Monoclonales/metabolismo , Moléculas de Adhesión Celular/química , Moléculas de Adhesión Celular/metabolismo , Mapeo Epitopo , Integrinas/metabolismo , Secuencia de Aminoácidos , Animales , Especificidad de Anticuerpos , Sitios de Unión , Línea Celular , Movimiento Celular , Humanos , Laminina/metabolismo , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Péptidos/química , Estructura Terciaria de Proteína , Ratas , Kalinina
20.
Biomaterials ; 31(19): 5110-21, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20347131

RESUMEN

The extracellular matrix (ECM) is thought to be an essential component of tissue scaffolding and engineering because it fulfills fundamental functions related to cell adhesion, migration, and three-dimensional organization. Natural ECM preparations, however, are challenging to work with because they are comprised of macromolecules that are large and insoluble in their functional state. Functional fragments of ECM macromolecules are a viable answer to this challenge, as demonstrated by the RGD-based engineered scaffolds, where the tri-peptide, Arg-Gly-Asp (RGD), represents the minimal functional unit of fibronectin and related ECM. Laminins (Ln) are main components of epithelial tissues, since they enter into the composition of basement membranes. Application of Ln to epithelial tissue engineering would be desirable, since they could help mimic ideal functional conditions for both lining and glandular epithelial tissues. However, functional fragments of Ln that could be used in artificial settings have not been characterized in detail. In this paper, we describe the production and application of the recombinant LG4 (rLG4) fragment of laminin-332 (Ln-332), and show that it mimics three fundamental functional properties of Ln-332: integrin-mediated cell adhesion, spreading, and migration. Adhesive structures formed by cells on rLG4 closely resemble those formed on Ln-332, as judged by microscopy-based analyses of their molecular composition. As on Ln-332, focal adhesion kinase (FAK) is phosphorylated in cells adhering to rLG4, and colocalized with other focal adhesion components. We conclude that rLG4 could be a useful substitute to recapitulate, in vitro, the tissue scaffolding properties of Ln-332.


Asunto(s)
Moléculas de Adhesión Celular/administración & dosificación , Moléculas de Adhesión Celular/genética , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Integrina alfa3beta1/metabolismo , Proteínas Recombinantes/administración & dosificación , Línea Celular , Relación Dosis-Respuesta a Droga , Humanos , Ingeniería de Proteínas/métodos , Kalinina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...