Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Pharm ; 21(2): 873-882, 2024 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-38229228

RESUMEN

Prostate cancer (PC), particularly its metastatic castration-resistant form (mCRPC), is a leading cause of cancer-related deaths among men in the Western world. Traditional systemic treatments, including hormonal therapy and chemotherapy, offer limited effectiveness due to tumors' inherent resistance to these therapies. Moreover, they often come with significant side effects. We have developed a delivery method using a tumor-cell-specific heptamethine carbocyanine dye (DZ) designed to transport therapeutic agents directly to tumor cells. This research evaluated simvastatin (SIM) as the antitumor payload because of its demonstrated chemopreventive effects on human cancers and its well-documented safety profile. We designed and synthesized a DZ-SIM conjugate for tumor cell targeting. PC cell lines and xenograft tumor models were used to assess tumor-cell targeting specificity and killing activity and to investigate the corresponding mechanisms. DZ-SIM treatment effectively killed PC cells regardless of their androgen receptor status or inherent therapeutic resistance. The conjugate targeted and suppressed xenograft tumor formation without harming normal cells of the host. In cancer cells, DZ-SIM was enriched in subcellular organelles, including mitochondria, where the conjugate formed adducts with multiple proteins and caused the loss of transmembrane potential, promoting cell death. The DZ-SIM specifically targets PC cells and their mitochondria, resulting in a loss of mitochondrial function and cell death. With a unique subcellular targeting strategy, the conjugate holds the potential to outperform existing chemotherapeutic drugs. It presents a novel strategy to circumvent therapeutic resistance, offering a more potent treatment for mCRPC.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración , Simvastatina , Masculino , Humanos , Simvastatina/farmacología , Simvastatina/uso terapéutico , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Próstata/metabolismo , Carbocianinas , Línea Celular Tumoral
2.
BMC Cancer ; 23(1): 499, 2023 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-37268911

RESUMEN

BACKGROUND: Clear cell renal cell carcinoma (ccRCC) is the most common type of kidney cancer and is notorious for its resistance to both chemotherapy and small-molecule inhibitor targeted therapies. Subcellular targeted cancer therapy may thwart the resistance to produce a substantial effect. METHODS: We tested whether the resistance can be circumvented by subcellular targeted cancer therapy with DZ-CIS, which is a chemical conjugate of the tumor-cell specific heptamethine carbocyanine dye (HMCD) with cisplatin (CIS), a chemotherapeutic drug with limited use in ccRCC treatment because of frequent renal toxicity. RESULTS: DZ-CIS displayed cytocidal effects on Caki-1, 786-O, ACHN, and SN12C human ccRCC cell lines and mouse Renca cells in a dose-dependent manner and inhibited ACHN and Renca tumor formation in experimental mouse models. Noticeably, in tumor-bearing mice, repeated DZ-CIS use did not cause renal toxicity, in contrast to the CIS-treated control animals. In ccRCC tumors, DZ-CIS treatment inhibited proliferation markers but induced cell death marker levels. In addition, DZ-CIS at half maximal inhibitory concentration (IC50) sensitized Caki-1 cells to small-molecule mTOR inhibitors. Mechanistically, DZ-CIS selectively accumulated in ccRCC cells' subcellular organelles, where it damages the structure and function of mitochondria, leading to cytochrome C release, caspase activation, and apoptotic cancer cell death. CONCLUSIONS: Results from this study strongly suggest DZ-CIS be tested as a safe and effective subcellular targeted cancer therapy.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Humanos , Animales , Ratones , Carcinoma de Células Renales/patología , Cisplatino/farmacología , Cisplatino/uso terapéutico , Neoplasias Renales/patología , Apoptosis , Muerte Celular , Línea Celular Tumoral , Proliferación Celular
3.
Breast Cancer Res ; 24(1): 7, 2022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-35078507

RESUMEN

BACKGROUND: Keratins (KRTs) are intermediate filament proteins that interact with multiple regulatory proteins to initiate signaling cascades. Keratin 13 (KRT13) plays an important role in breast cancer progression and metastasis. The objective of this study is to elucidate the mechanism by which KRT13 promotes breast cancer growth and metastasis. METHODS: The function and mechanisms of KRT13 in breast cancer progression and metastasis were assessed by overexpression and knockdown followed by examination of altered behaviors in breast cancer cells and in xenograft tumor formation in mouse mammary fat pad. Human breast cancer specimens were examined by immunohistochemistry and multiplexed quantum dot labeling analysis to correlate KRT13 expression to breast cancer progression and metastasis. RESULTS: KRT13-overexpressing MCF7 cells displayed increased proliferation, invasion, migration and in vivo tumor growth and metastasis to bone and lung. Conversely, KRT13 knockdown inhibited the aggressive behaviors of HCC1954 cells. At the molecular level, KRT13 directly interacted with plakoglobin (PG, γ-catenin) to form complexes with desmoplakin (DSP). This complex interfered with PG expression and nuclear translocation and abrogated PG-mediated suppression of c-Myc expression, while the KRT13/PG/c-Myc signaling pathway increased epithelial to mesenchymal transition and stem cell-like phenotype. KRT13 expression in 58 human breast cancer tissues was up-regulated especially at the invasive front and in metastatic specimens (12/18) (p < 0.05). KRT13 up-regulation in primary breast cancer was associated with decreased overall patient survival. CONCLUSIONS: This study reveals that KRT13 promotes breast cancer cell growth and metastasis via a plakoglobin/c-Myc pathway. Our findings reveal a potential novel pathway for therapeutic targeting of breast cancer progression and metastasis.


Asunto(s)
Neoplasias de la Mama , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Transición Epitelial-Mesenquimal , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Queratina-13/genética , Queratina-13/metabolismo , Ratones , Metástasis de la Neoplasia , Proteínas Proto-Oncogénicas c-myc , Transducción de Señal , gamma Catenina/genética , gamma Catenina/metabolismo
4.
Prostate ; 80(3): 274-283, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31846114

RESUMEN

PURPOSE: We previously determined that cancer-stromal interaction was a direct route to tumor cell heterogeneity progression, since cancer-stromal cell fusion in coculture resulted in the creation of heterogeneous clones of fusion hybrid progeny. In this report, we modified the cancer-stromal coculture system to establish optimal experimental conditions for investigating cell fusion machinery and the mechanism of heterogeneity progression. EXPERIMENTAL DESIGN: Red fluorescence protein-tagged LNCaP cells were cocultured with green fluorescence protein-labeled prostate stromal cells for cancer-stromal cell fusion, which was tracked as dual fluorescent cells by fluorescence microscopy. RESULTS: We identified the most efficient strategy to isolate clones of fusion hybrid progenies. From the coculture, mixed cells including fusion hybrids were subjected to low-density replating for colony formation by fusion hybrid progeny. These colonies could propagate into derivative cell populations. Compared to the parental LNCaP cells, clones of the fusion hybrid progeny displayed divergent behaviors and exhibited permanent genomic hybridization. CONCLUSIONS: Cancer-stromal cell fusion leads to cancer cell heterogeneity. The cancer-stromal coculture system characterized in this study can be used as a model for molecular characterization of cancer cell fusion as the mechanism behind the progression of heterogeneity observed in clinical prostate cancers.


Asunto(s)
Proteínas Fluorescentes Verdes/análisis , Proteínas Luminiscentes/análisis , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Células del Estroma/metabolismo , Células del Estroma/patología , Comunicación Celular/fisiología , Fusión Celular , Línea Celular Tumoral , Técnicas de Cocultivo , Técnica del Anticuerpo Fluorescente/métodos , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Proteínas Luminiscentes/metabolismo , Masculino , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Microscopía Fluorescente/métodos , Transporte de Proteínas , Proteína Fluorescente Roja
5.
Int J Cancer ; 145(8): 2249-2259, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31020641

RESUMEN

Though human prostate cancer (PCa) heterogeneity can best be studied using multiple cell types isolated from clinical specimens, the difficulty of establishing cell lines from clinical tumors has hampered this approach. In this proof-of-concept study, we established a human PCa cell line from a prostatectomy surgical specimen without the need for retroviral transduction. In a previous report, we characterized the stromal cells derived from PCa specimens. Here, we characterized the epithelial cells isolated from the same tumors. Compared to the ease of establishing prostate stromal cell lines, prostatic epithelial cell lines are challenging. From three matched pairs of normal and tumor tissues, we established one new PCa cell line, HPE-15. We confirmed the origin of HPE-15 cells by short tandem repeat microsatellite polymorphism analysis. HPE-15 cells are androgen-insensitive and express marginal androgen receptor, prostate-specific antigen and prostate-specific membrane antigen proteins. HPE-15 expresses luminal epithelial markers of E-cadherin and cytokeratin 18, basal cell markers of cytokeratin 5 and p63 and neuroendocrine marker of chromogranin A. Interestingly, HPE-15 Cells exhibited no tumorigenicity in different strains of immune-deficient mice but can become tumorigenic through interaction with aggressive cancer cell types. HPE-15 cells can thus serve as an experimental model for the study of PCa progression, metastasis and tumor cell dormancy.


Asunto(s)
Células Epiteliales/citología , Mesodermo/citología , Próstata/citología , Neoplasias de la Próstata/patología , Células del Estroma/citología , Animales , Carcinogénesis , Comunicación Celular , Línea Celular , Línea Celular Transformada , Línea Celular Tumoral , Células Epiteliales/metabolismo , Humanos , Calicreínas/metabolismo , Masculino , Mesodermo/metabolismo , Ratones , Próstata/metabolismo , Antígeno Prostático Específico/metabolismo , Prostatectomía , Neoplasias de la Próstata/metabolismo , Células del Estroma/metabolismo , Trasplante Heterólogo , Células Tumorales Cultivadas
6.
Cancer ; 125(13): 2222-2232, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-30840322

RESUMEN

BACKGROUND: Burkitt lymphoma is a fast-growing mature B cell malignancy, whose genetic hallmark is translocation and activation of the c-myc gene. Prompt multiagent immunochemotherapy regimens can have favorable outcomes, but prognosis is poor in refractory or relapsed disease. We previously identified a novel family of near-infrared heptamethine carbocyanine fluorescent dyes (HMCD or DZ) with tumor-homing properties via organic anion-transporting peptides. These membrane carriers have uptake in tumor cells but not normal cells in cell culture, mouse and dog tumor models, patient-derived xenografts, and perfused kidney cancers in human patients. METHODS: Here we report the cytotoxic effects of a synthesized conjugate of DZ with cisplatin (CIS) on B cell lymphoma CA46, Daudi, Namalwa, Raji, and Ramos cell lines in cell culture and in xenograft tumor formation. Impaired mitochondrial membrane permeability was examined as the mechanism of DZ-CIS-induced lymphoma cell death. RESULTS: The new conjugate, DZ-CIS, is cytotoxic against Burkitt lymphoma cell lines and tumor models. DZ-CIS retains tumor-homing properties to mitochondrial and lysosomal compartments, does not accumulate in normal cells and tissues, and has no nephrotoxicity in mice. DZ-CIS accumulated in Burkitt lymphoma cells and tumors induces apoptosis and retards tumor cell growth in culture and xenograft tumor growth in mice. CONCLUSION: DZ-CIS downregulated c-myc and overcame CIS resistance in myc-driven TP53-mutated aggressive B cell Burkitt lymphoma. We propose that DZ-CIS could be used to treat relapsed/refractory aggressive Burkitt lymphomas.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Linfoma de Burkitt/tratamiento farmacológico , Carbocianinas/química , Cisplatino/química , Animales , Apoptosis , Proliferación Celular , Composición de Medicamentos , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Prostate ; 75(12): 1312-21, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25990623

RESUMEN

BACKGROUND: We previously reported that the activation of RANK and c-Met signaling components in both experimental mouse models and human prostate cancer (PC) specimens predicts bone metastatic potential and PC patient survival. This study addresses whether a population of metastasis-initiating cells (MICs) known to express a stronger RANKL, phosphorylated c-Met (p-c-Met), and neuropilin-1 (NRP1) signaling network than bystander or dormant cells (BDCs) can be detected in PC tissues from patients subjected to transurethral resection of the prostate (TURP) for urinary obstruction prior to the diagnosis of PC with or without prior hormonal manipulation, and whether the relative abundance of MICs over BDCs could predict castration-resistant progression and PC patient survival. METHODS: We employed a multiplexed quantum-dot labeling (mQDL) protocol to detect and quantify MICs and BDCs at the single cell level in TURP tissues obtained from 44 PC patients with documented overall survival and castration resistance status. RESULTS: PC tissues with a higher number of MICs and an activated RANK signaling network, including increased expression of RANKL, p-c-Met, and NRP1 compared to BDCs, were found to correlate with the development of castration resistance and overall survival. CONCLUSIONS: The assessment of PC cells with MIC and BDC phenotypes in primary PC tissues from hormone-naïve patients can predict the progression to castration resistance and the overall survival of PC patients.


Asunto(s)
Neoplasias Óseas/secundario , Neoplasias de la Próstata Resistentes a la Castración/diagnóstico , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-met/metabolismo , Ligando RANK/metabolismo , Receptores del Factor Natriurético Atrial/metabolismo , Resección Transuretral de la Próstata/métodos , Anciano , Anciano de 80 o más Años , Neoplasias Óseas/mortalidad , Neoplasias Óseas/cirugía , Progresión de la Enfermedad , Humanos , Masculino , Persona de Mediana Edad , Fosforilación , Pronóstico , Neoplasias de la Próstata/mortalidad , Neoplasias de la Próstata/cirugía , Puntos Cuánticos , Transducción de Señal , Tasa de Supervivencia
8.
Int Immunopharmacol ; 131: 111878, 2024 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-38493693

RESUMEN

BACKGROUND: Kidney renal papillary cell carcinoma (KIRP) is frequently associated with an unfavorable prognosis for affected individuals. Unfortunately, there has been insufficient exploration in search for a reliable prognosis signature and predictive indicators to forecast outcomes for KIRP patients. AIM: The aim of this study is to employ a comprehensive analysis of data for the identification of prognosis genes, leading to the development of a nomogram with strong predictive capabilities. The objective is to provide a valuable statistical tool that, when implemented in a clinical setting, can offer patients an early opportunity for treatment and enhance their chances of ultimate recovery from this life-threatening disease. METHODS: Different packages in R were used to analyze RNA-seq data from the TCGA data portal. Multivariate Cox regression analysis and Kaplan-Meier analysis were also used to investigate the prognostic values of immune-related genes and construct the predictive model and nomogram. A p-value < 0.05 was considered to be significant. RESULTS: A total of 368 immune-related genes and 60 TFs were identified as differentially expressed in KIRP tissues compared with normal tissues. Of the 368, 23 were found to be related to overall survival. GO and KEGG analysis suggested that these prognostic immune-related genes mainly participated in the ERK1 and ERK2 cascades, Rap1 signaling pathway, and the PI3K-Akt signaling pathway. 9 genes were identified from Cox regression to be statistically significant prognostic-related genes. Survival analysis showed that a model based on these 9 prognostic-related genes has high predictive performance. Immunohistochemistry results show that APOH, BIRC5, CCL19, and GRN were significantly increased in kidney cancer. B cells and CD4 + T cells were positively correlated with risk score model. CONCLUSION: A prognostic model was successfully created based on 9 immune-related genes correlated with overall survival in KIRP. This work aims to provide some insight into therapeutic approaches and prognostic predictors of KIRP.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Humanos , Pronóstico , Nomogramas , Fosfatidilinositol 3-Quinasas , Carcinoma de Células Renales/genética , Neoplasias Renales/genética , Riñón
9.
Cancers (Basel) ; 16(5)2024 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-38473313

RESUMEN

Tumor cells gain advantages in growth and survival by acquiring genotypic and phenotypic heterogeneity. Interactions with bystander cells in the tumor microenvironment contribute to the progression of heterogeneity. We have shown that fusion between tumor and bystander cells is one form of interaction, and that tumor-bystander cell fusion has contrasting effects. By trapping fusion hybrids in the heterokaryon or synkaryon state, tumor-bystander cell fusion prevents the progression of heterogeneity. However, if trapping fails, fusion hybrids will resume replication to form derivative clones with diverse genomic makeups and behavioral phenotypes. To determine the characteristics of bystander cells that influence the fate of fusion hybrids, we co-cultured prostate mesenchymal stromal cell lines and their spontaneously transformed sublines with LNCaP as well as HPE-15 prostate cancer cells. Subclones derived from cancer-stromal fusion hybrids were examined for genotypic and phenotypic diversifications. Both stromal cell lines were capable of fusing with cancer cells, but only fusion hybrids with the transformed stromal subline generated large numbers of derivative subclones. Each subclone had distinct cell morphologies and growth behaviors and was detected with complete genomic hybridization. The health conditions of the bystander cell compartment play a crucial role in the progression of tumor cell heterogeneity.

10.
J Urol ; 189(2): 702-710, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23000848

RESUMEN

PURPOSE: We assessed the application of near infrared heptamethine carbocyanine dyes, including IR-783 and the synthetic analogue MHI-148, as optical imaging agents for the rapid detection of human kidney cancer. MATERIALS AND METHODS: The uptake, retention and subcellular localization of these organic dyes were investigated in cultured kidney cancer cells. Tumor specificity of dye uptake and retention was evaluated by whole body imaging of mice bearing human kidney cancer xenografts or freshly harvested clinical kidney cancer specimens. In addition, dye accumulation at the tissue and cellular levels was confirmed by ex vivo studies with results confirmed by fluorescence imaging of frozen tissue sections. Peripheral blood spiked with kidney cancer cells was stained to simulate the detection of circulating tumor cells. RESULTS: Preferential uptake and retention of carbocyanine near infrared dyes was observed in cultured human kidney cancer cells, human kidney cancer cell spiked whole blood, human kidney cancer xenografts and freshly harvested human kidney cancer tissues compared to normal kidney epithelial cells and normal host organs. CONCLUSIONS: We describe a new class of near infrared heptamethine carbocyanine dyes that show potential for detecting kidney cancer cells in circulating blood and kidney cancer cells in clinical specimens. Near infrared carbocyanine dyes can be further developed as dual modality agents for deep tissue imaging of localized and disseminated kidney cancer in patients.


Asunto(s)
Carbocianinas , Colorantes Fluorescentes , Neoplasias Renales/diagnóstico , Animales , Diagnóstico por Imagen , Humanos , Ratones , Ratones Desnudos , Células Tumorales Cultivadas
11.
Semin Cell Dev Biol ; 21(1): 26-32, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19948237

RESUMEN

Cancer development is complex and involves several layers of interactions and pleotropic signaling mechanisms leading to progression. Cancer cells associate with resident stromal fibroblasts, smooth muscle cells, macrophages, endothelium, neurons and migrating cells at metastatic sites and phenotypically and genotypically activate them. These become an integral part of the cancer cell community through activated cell signaling mechanisms. During this process, the cancer cells and cells in the cancer microenvironment "co-evolve" in part due to oxidative stress, and acquire the ability to mimic other cell types (which can be termed osteomimicry, vasculomimicry, neuromimicry and stem cell mimicry), and undergo transition from epithelium to mesenchyme with definitive morphologic and behavioral modifications. In our laboratory, we demonstrated that prostate cancer cells co-evolve in their genotypic and phenotypic characters with stroma and acquire osteomimetic properties allowing them to proliferate and survive in the skeleton as bone metastasis. Several signaling interactions in the bone microenvironment, mediated by reactive oxygen species, soluble and membrane bound factors, such as superoxide, beta2-microglobulin and RANKL have been described. Targeting the signaling pathways in the cancer-associated stromal microenvironment in combination with known conventional therapeutic modalities could have a synergistic effect on cancer treatment. Since cancer cells are constantly interacting and acquiring adaptive and survival changes primarily directed by their microenvironment, it is imperative to delineate these interactions and co-target both cancer and stroma to improve the treatment and overall survival of cancer patients.


Asunto(s)
Neoplasias de la Próstata/patología , Células del Estroma , Neoplasias Óseas/secundario , Progresión de la Enfermedad , Humanos , Masculino , Metástasis de la Neoplasia , Osteoblastos/metabolismo , Osteoblastos/patología , Osteoprotegerina/metabolismo , Estrés Oxidativo , Neoplasias de la Próstata/fisiopatología , Ligando RANK/metabolismo , Células del Estroma/metabolismo , Células del Estroma/patología
12.
Front Physiol ; 13: 827531, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35237181

RESUMEN

Human cancers are often complicated with increased incidences of blood vessel occlusion, which are mostly insensitive to anticoagulation therapy. We searched for causal factors of cancer-associated embolism. A total of 2,017 blood samples was examined for visible abnormalities. Examined were peripheral blood samples from cancer patients who were about to undergo surgical treatment for genitourinary, breast, gastrointestinal or abdominal tumors. Samples from ambulatory patients being treated for recurrent or castration-resistant prostate cancers were included in the study. The lipid-rich nature was studied with lipophilic stains and lipid panel analysis, while surface membrane was assessed with specific staining and antibody detection. We identified a new entity, lipid droplet-like objects or circulating fatty objects (CFOs), visible in the blood samples of many cancer patients, with the potential of causing embolism. CFOs were defined as lipid-rich objects with a membrane, capable of gaining in volume through interaction with peripheral blood mononuclear cells in ex vivo culture. Blood samples from pancreatic cancer patients were found to have the highest CFO incidence and largest CFO numbers. Most noticeably, CFOs from many pancreatic cancer samples presented as large clusters entangled in insoluble fiber networks, suggestive of intravascular clotting. This study identifies CFO as an abnormal entity in cancer patient blood, and a contributory factor to intravascular embolism during cancer development and progression.

13.
Adv Ther (Weinh) ; 5(10)2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36590644

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is a disease with no effective therapeutics. We have developed a novel targeted therapy drug consisting of a tumor-targeting ligand, near-infrared (NIR) organic heptamethine carbocyanine dye (HMCD), and HMG-CoA inhibitor simvastatin (SIM), and assessed its efficacy in PDAC. PDAC cell specific targeting of DZ-SIM was measured by determining the fluorescence in cells and animals. Mitochondrial bioenergetics and functions were measured by Seahorse and flow cytometry, respectively. Apoptosis was assessed by DNA fragmentation, AnnexinV/Propidium Iodide staining, and TUNEL. Markers of cell invasion, epithelial-to-mesenchymal transition, and cancer stemness were measured. The effect of DZ-SIM on survival, tumor growth and metastasis was measured in the Krasþ/LSLG12D;Trp53þ/LSLR172H;Pdx-1-Cre (KPC) transgenic mice and in syngeneic and subcutaneous PDAC models. NIR fluorescence imaging showed specific localization of DZ-SIM to cancer, but not to normal cells and tissues. DZ-SIM significantly inhibited tumor growth and re-sensitized therapeutically resistant PDAC cells to conventional therapies. DZ-SIM killed cancer cells through unique pathways involving decreasing mitochondrial bioenergetics, including oxygen consumption and ATP production, and increasing ROS production. Mitochondrial depletion prevented the effect of DZ-SIM. Administration of DZ-SIM in 3 PDAC animal models resulted in a marked increase in survival and a decrease in tumor growth and metastasis.

14.
Cells ; 11(22)2022 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-36428982

RESUMEN

During disease progression and bone metastasis, breast tumor cells interact with various types of bystander cells residing in the tumor microenvironment. Such interactions prompt tumor cell heterogeneity. We used successive co-culture as an experimental model to examine cancer-bystander cell interaction. RMCF7-2, a clone of the human breast cancer MCF-7 cells tagged with a red fluorescent protein, was tracked for morphologic, behavioral, and gene expression changes. Co-cultured with various types of hematopoietic cells, RMCF7-2 adopted stable changes to a rounded shape in suspension growth of red fluorescent cells, from which derivative clones displayed marked expressional changes of marker proteins, including reduced E-cadherin and estrogen receptor α, and loss of progesterone receptor. In a successive co-culture with bone marrow-derived mesenchymal stem/stromal cells, the red fluorescent clones in suspension growth changed once more, adopting an attachment growth, but in diversified shapes. Red fluorescent clones recovered from the second-round co-culture were heterogeneous in morphology, but retained the altered marker protein expression while displaying increased proliferation, migration, and xenograft tumor formation. Interaction with bystander cells caused permanent morphologic, growth behavioral, and gene expressional changes under successive co-culture, which is a powerful model for studying cancer cell heterogeneity during breast cancer progression and metastasis.


Asunto(s)
Neoplasias de la Mama , Células Madre Mesenquimatosas , Humanos , Femenino , Células MCF-7 , Técnicas de Cocultivo , Neoplasias de la Mama/patología , Médula Ósea/patología , Células Madre Mesenquimatosas/metabolismo , Microambiente Tumoral
15.
Fetal Pediatr Pathol ; 30(4): 244-51, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21434830

RESUMEN

Hypospadias is a common congenital anomalies, yet its molecular basis remains unknown. Recent studies have linked perturbations in the Hedgehog signaling pathway to hypospadias. However, the expression of Sonic hedgehog (Shh) has not been reported during genital development. Immunohistochemical staining for Shh and its receptors was applied to 10 human fetal penises ranging from 12 to 29 weeks gestation. The intensity of Shh staining was greatest in the urethral epithelium at 14 weeks gestation, correlating with the time of urethral tubularization. Results suggest a role for Shh in human male genital development.


Asunto(s)
Proteínas Hedgehog/metabolismo , Pene/embriología , Pene/fisiología , Transducción de Señal/fisiología , Animales , Femenino , Feto/anatomía & histología , Edad Gestacional , Humanos , Hipospadias/etiología , Hipospadias/fisiopatología , Inmunohistoquímica , Masculino , Organogénesis , Receptores Patched , Pene/anomalías , Embarazo , Receptores de Superficie Celular/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptor Smoothened , Factores de Transcripción/metabolismo , Proteína con Dedos de Zinc GLI1
16.
Mol Cancer Ther ; 20(12): 2527-2538, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34583981

RESUMEN

Cisplatin and tyrosine kinase inhibitors (TKI) are recommended to treat non-small cell lung cancer (NSCLC). However, ubiquitously acquired drug resistance in patients with NSCLC diminishes their therapeutic efficacy. Strategies for overcoming cisplatin and TKI resistance are an unmet medical need. We previously described a group of near-infrared heptamethine carbocyanine fluorescent dyes, referred to as DZ, with tumor-homing properties via differentially expressed organic anion-transporting polypeptides on cancer cells. This group of organic dyes can deliver therapeutic payloads specifically to tumor cells in the form of a chemical conjugate. We synthesized DZ-simvastatin (DZ-SIM) initially to target cholesterol biosynthesis in lung cancer cells. DZ-SIM killed both cisplatin-sensitive and cisplatin-resistant as well as EGFR-TKI-sensitive and EGFR-TKI-resistant lung cancer cells. This conjugate specifically accumulated in and effectively inhibited the growth of xenograft tumors formed by NSCLC cells resistant to first-generation (H1650) and third-generation (PC9AR) EGFR TKIs. DZ-SIM induced cell death by targeting mitochondrial structure and function. We concluded that DZ-SIM could be a promising novel therapy for overcoming drug resistance in patients with NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Mitocondrias/metabolismo , Humanos
17.
Mol Cancer ; 9: 9, 2010 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-20085644

RESUMEN

BACKGROUND: Myeloid cell leukemia-1 (Mcl-1) is a member of the Bcl-2 family, which inhibits cell apoptosis by sequestering pro-apoptotic proteins Bim and Bid. Mcl-1 overexpression has been associated with progression in leukemia and some solid tumors including prostate cancer (PCa). However, the regulatory mechanism for Mcl-1 expression in PCa cells remains elusive. RESULTS: Immunohistochemical analyses revealed that Mcl-1 expression was elevated in PCa specimens with high Gleason grades and further significantly increased in bone metastasis, suggesting a pivotal role of Mcl-1 in PCa metastasis. We further found that vascular endothelial growth factor (VEGF) is a novel regulator of Mcl-1 expression in PCa cells. Inhibition of endogenous Mcl-1 induced apoptosis, indicating that Mcl-1 is an important survival factor in PCa cells. Neuropilin-1 (NRP1), the "co-receptor" for VEGF165 isoform, was found to be highly expressed in PCa cells, and indispensible in the regulation of Mcl-1. Intriguingly, VEGF165 promoted physical interaction between NRP1 and hepatocyte growth factor (HGF) receptor c-MET, and facilitated c-MET phosphorylation via a NRP1-dependent mechanism. VEGF165 induction of Mcl-1 may involve rapid activation of Src kinases and signal transducers and activators of transcription 3 (Stat3). Importantly, NRP1 overexpression and c-MET activation were positively associated with progression and bone metastasis in human PCa specimens and xenograft tissues. CONCLUSIONS: This study demonstrated that Mcl-1 overexpression is associated with PCa bone metastasis. Activation of VEGF165-NRP1-c-MET signaling could confer PCa cells survival advantages by up-regulating Mcl-1, contributing to PCa progression.


Asunto(s)
Neuropilina-1/metabolismo , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo , Neoplasias Óseas/genética , Neoplasias Óseas/secundario , Supervivencia Celular/efectos de los fármacos , Progresión de la Enfermedad , Activación Enzimática/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Modelos Biológicos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Neoplasias de la Próstata/patología , Unión Proteica/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/farmacología , Familia-src Quinasas/metabolismo
18.
Prostate ; 70(9): 993-1001, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20166133

RESUMEN

BACKGROUND: The alpha2 chain of the interleukin-13 receptor (IL13Ralpha2) is a high-affinity receptor and a candidate target for cytotoxic killing of cancer cells. Availability of a human prostate cancer cell line with high level of IL13Ralpha2 expression will facilitate the development of therapeutic modalities. METHODS: ARCaP(E) and ARCaP(M) human prostate cancer cell lines were subjected to comparative analyses of gene expression. Expression of the IL13Ralpha2 protein was confirmed by Western blotting and immunostaining. IL13Ralpha2 proteins in xenograft tumors and clinical human prostate cancer specimens were detected by specific antibodies. LNCaP prostate cancer cells stably transfected with IL13Ralpha2 were examined for accelerated growth in athymic mice. RESULTS: We found that IL13Ralpha2 proteins could be detected in both the ARCaP(E) and ARCaP(M) cells, but the expression level in ARCaP(M) was more than 17-fold higher than in ARCaP(E) cells. Importantly, the ARCaP lineage represented the only human prostate cancer cell line that expresses IL13Ralpha2 proteins at the level detectable by Western blotting. Expression of IL13Ralpha2 was accompanied by resistance to the anti-tumor activity of interleukin-13 (IL-13). ARCaP cells were found to be insensitive to growth inhibition upon IL-13 treatment, while overexpression of IL13Ralpha2 in LNCaP cells promoted intratibial tumor growth in athymic mice. CONCLUSIONS: Differential IL13Ralpha2 expression may account for the high tumorigenic and metastatic potential of ARCaP(M) cells. The unique expression of IL13Ralpha2 makes ARCaP lineage an attractive model for evaluating the targeting efficacy of therapeutic agents based on IL13Ralpha2 protein expression.


Asunto(s)
Subunidad alfa2 del Receptor de Interleucina-13/genética , Interleucina-13/metabolismo , Próstata/metabolismo , Animales , Western Blotting , Línea Celular Tumoral , Proliferación Celular , Células Cultivadas , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Inmunohistoquímica , Interleucina-13/genética , Interleucina-13/farmacología , Subunidad alfa2 del Receptor de Interleucina-13/metabolismo , Masculino , Ratones , Ratones Desnudos , Próstata/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Trasplante Heterólogo
19.
Prostate ; 70(10): 1119-26, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20333698

RESUMEN

BACKGROUND: We have reported that human prostate cancer ARCaP(E) cells undertake epithelial to mesenchymal transition (EMT) when stimulated by certain soluble factors, and that EMT is regulated by surface receptor-elicited signaling pathways through protein phosphorylation. It is known that phorbol ester phorbol-12-myristate-13-acetate (PMA), a potent antagonist to both conventional and novel protein kinase C (PKC) isoenzymes, induces cancer cell scattering. METHODS: To assess the effect of PMA on EMT, ARCaP(E) cells were treated with PMA and were assayed for EMT-related morphologic and behavioral changes. Specific inhibitors were used to investigate the PMA-induced EMT. RESULTS: PMA at 100 nM induced EMT in a time-dependent manner, resulting in a complete change from epithelial to mesenchymal stromal morphology. Concurrently, PMA inhibited expression of epithelial marker E-cadherin and increased the level of stromal marker protein vimentin, while the treated cells showed increased migratory and invasive capacities. Using specific inhibitors, we confirmed that the effect of PMA was mediated by PKC, while isoenzymes of the novel PKC subfamily were implicated as the main mediator. Finally, we determined that the EMT was dependent on newly synthesized proteins, because inhibitors for gene transcription and protein translation could both inhibit the initiation of EMT. CONCLUSIONS: Although PMA is well known for its effects on cell migration and tumor formation, this work is the first to define PMA as an EMT inducer in prostate cancer cells. Further investigation in this experimental model may reveal important regulatory mechanisms and additional molecular changes underlying EMT.


Asunto(s)
Carcinógenos/farmacología , Neoplasias de la Próstata/patología , Acetato de Tetradecanoilforbol/farmacología , Western Blotting , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Humanos , Indoles/farmacología , Isoenzimas/antagonistas & inhibidores , Isoenzimas/metabolismo , Masculino , Maleimidas/farmacología , Mesodermo/efectos de los fármacos , Mesodermo/patología , Microscopía de Contraste de Fase , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/ultraestructura , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , Células del Estroma/efectos de los fármacos , Células del Estroma/patología
20.
Prostate ; 70(5): 518-28, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19918799

RESUMEN

BACKGROUND: The mechanism of epithelial to mesenchymal transition (EMT) could be adopted by tumor cells for migration and invasion. We have reported that ARCaP(E) human prostate cancer cells undergo EMT-like changes during xenograft growth in athymic mice. METHODS: In this report, we assessed the extent of EMT by tracking changes in cloned ARCaP(E) cells expressing red fluorescence protein during successive orthotopic prostate tumor formation. Cancer cells with stromal-like morphology were isolated and examined for EMT-like changes. RESULTS: EMT-like morphologic and expression changes were detected after one round of in vivo tumor formation. Importantly, when recovered tumor cells were used in second round xenograft tumor formation, a large fraction of ARCaP(E) cells showed drastic EMT-like changes, with markedly enlarged cell size and divergent cell shapes similar to those of mesenchymal stromal cells. The morphologic change was accompanied by increased growth and metastasis, as tumor incidence increased while red fluorescent tumor cells could be detected from circulating blood, bone marrow, peritoneal ascites, and lung of the tumor-bearing mice. Recovered clones from these samples had lost epithelial markers but many showed activated stromal marker vimentin expression. The EMT appeared permanent since the newly acquired morphology was sustained after continuous passages. CONCLUSIONS: Results from this study demonstrate that through interaction with the host tumor microenvironment, cancer cells acquire cellular plasticity. During xenograft tumor formation and metastasis, a single clone of cancer cells could yield a heterogeneous population, with a substantial number of tumor cells adopting mesenchymal stroma-like phenotypes.


Asunto(s)
Epitelio/patología , Mesodermo/patología , Neoplasias de la Próstata/patología , Animales , Humanos , Masculino , Ratones , Trasplante de Neoplasias , Fenotipo , Células del Estroma/patología , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA