Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Am J Pathol ; 179(5): 2559-68, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21907176

ABSTRACT

Integrins participate in multiple cellular processes, including cell adhesion, migration, proliferation, survival, and the activation of growth factor receptors. Recent studies have shown that expression of αv integrins is elevated in the prostate cancer stem/progenitor cell subpopulation compared with more differentiated, committed precursors. Here, we examine the functional role of αv integrin receptor expression in the acquisition of a metastatic stem/progenitor phenotype in human prostate cancer. Stable knockdown of αv integrins expression in PC-3M-Pro4 prostate cancer cells coincided with a significant decrease of prostate cancer stem/progenitor cell characteristics (α2 integrin, CD44, and ALDH(hi)) and decreased expression of invasion-associated genes Snail, Snail2, and Twist. Consistent with these observations, αv-knockdown strongly inhibited the clonogenic and migratory potentials of human prostate cancer cells in vitro and significantly decreased tumorigenicity and metastatic ability in preclinical models of orthotopic growth and bone metastasis. Our data indicate that integrin αv expression is functionally involved in the maintenance of a highly migratory, mesenchymal cellular phenotype as well as the acquisition of a stem/progenitor phenotype in human prostate cancer cells with metastasis-initiating capacity.


Subject(s)
Integrin alphaV/metabolism , Neoplastic Stem Cells/metabolism , Prostatic Neoplasms/metabolism , Animals , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Movement , Cell Transformation, Neoplastic/metabolism , Gene Knockdown Techniques , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation , Phenotype , Tumor Stem Cell Assay , Up-Regulation
2.
Clin Exp Metastasis ; 28(7): 615-25, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21647815

ABSTRACT

High aldehyde dehydrogenase (ALDH) activity can be used to identify tumor-initiating and metastasis-initiating cells in various human carcinomas, including prostate cancer. To date, the functional importance of ALDH enzymes in prostate carcinogenesis, progression and metastasis has remained elusive. Previously we identified strong expression of ALDH7A1 in human prostate cancer cell lines, primary tumors and matched bone metastases. In this study, we evaluated whether ALDH7A1 is required for the acquisition of a metastatic stem/progenitor cell phenotype in human prostate cancer. Knockdown of ALDH7A1 expression resulted in a decrease of the α2(hi)/αv(hi)/CD44(+) stem/progenitor cell subpopulation in the human prostate cancer cell line PC-3M-Pro4. In addition, ALDH7A1 knockdown significantly inhibited the clonogenic and migratory ability of human prostate cancer cells in vitro. Furthermore, a number of genes/factors involved in migration, invasion and metastasis were affected including transcription factors (snail, snail2, and twist) and osteopontin, an ECM molecule involved in metastasis. Knockdown of ALDH7A1 resulted in decreased intra-bone growth and inhibited experimentally induced (bone) metastasis, while intra-prostatic growth was not affected. In line with these observations, evidence is presented that TGF-ß, a key player in cancer invasiveness and bone metastasis, strongly induced ALDH activity while BMP7 (an antagonist of TGF-ß signaling) down-regulated ALDH activity. Our findings show, for the first time, that the ALDH7A1 enzyme is functionally involved in the formation of bone metastases and that the effect appeared dependent on the microenvironment, i.e., bone versus prostate.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Bone Neoplasms/secondary , Neoplasm Metastasis/pathology , Prostatic Neoplasms/metabolism , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Line, Tumor , Disease Progression , Epithelial-Mesenchymal Transition , Humans , Male , Prostatic Neoplasms/pathology
3.
Neoplasia ; 13(6): 516-25, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21677875

ABSTRACT

Acquisition of an invasive phenotype by cancer cells is a requirement for bone metastasis. Transformed epithelial cells can switch to a motile, mesenchymal phenotype by epithelial-mesenchymal transition (EMT). Recently, it has been shown that EMT is functionally linked to prostate cancer stem cells, which are not only critically involved in prostate cancer maintenance but also in bone metastasis. We showed that treatment with the non-peptide α(v)-integrin antagonist GLPG0187 dose-dependently increased the E-cadherin/vimentin ratio, rendering the cells a more epithelial, sessile phenotype. In addition, GLPG0187 dose-dependently diminished the size of the aldehyde dehydrogenase high subpopulation of prostate cancer cells, suggesting that α(v)-integrin plays an important role in maintaining the prostate cancer stem/progenitor pool. Our data show that GLPG0187 is a potent inhibitor of osteoclastic bone resorption and angiogenesis in vitro and in vivo. Real-time bioluminescent imaging in preclinical models of prostate cancer demonstrated that blocking α(v)-integrins by GLPG0187 markedly reduced their metastatic tumor growth according to preventive and curative protocols. Bone tumor burden was significantly lower in the preventive protocol. In addition, the number of bone metastases/mouse was significantly inhibited. In the curative protocol, the progression of bone metastases and the formation of new bone metastases during the treatment period was significantly inhibited. In conclusion, we demonstrate that targeting of integrins by GLPG0187 can inhibit the de novo formation and progression of bone metastases in prostate cancer by antitumor (including inhibition of EMT and the size of the prostate cancer stem cell population), antiresorptive, and antiangiogenic mechanisms.


Subject(s)
Antineoplastic Agents/pharmacology , Bone Neoplasms/prevention & control , Integrin alphaV/metabolism , Neoplastic Stem Cells/drug effects , Prostatic Neoplasms/drug therapy , Animals , Animals, Newborn , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/metabolism , Neovascularization, Pathologic/prevention & control , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoclasts/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Burden/drug effects , Tumor Microenvironment/drug effects , Twist-Related Protein 1/genetics , Xenograft Model Antitumor Assays
4.
Eur Urol ; 60(2): 337-43, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21616583

ABSTRACT

BACKGROUND: Bladder cancer is the fifth most common malignancy in the Western world and the second most frequently diagnosed genitourinary tumor. In the majority of cases, death from bladder cancer results from metastatic disease. Understanding the multistep process of carcinogenesis and metastasis in urothelial cancers is pivotal to the development of new therapeutic strategies. Molecular imaging of cancer growth and metastasis in preclinical models provides the essential link between cell-based experiments and clinical translation. OBJECTIVE: Develop preclinical models for sensitive bladder cancer cell tracking during tumor progression and metastasis. DESIGN, SETTING, AND PARTICIPANTS: A human transitional cell carcinoma UM-UC-3 cell line was generated that stably expresses luciferase 2 (UM-UC-3luc2), a mammalian codon-optimized firefly luciferase with superior expression. Preclinical models were developed with human UM-UC-3luc2 cells xenografted into the bladder (orthotopic model with metastases) or inoculated into the left cardiac ventricle (bone metastasis model) of immunocompromised mice. MEASUREMENTS: Noninvasive, sensitive bioluminescent imaging of human firefly luciferase 2-positive bladder cancer in mice using the IVIS100 imaging system. RESULTS AND LIMITATIONS: In the orthotopic model (intravesical inoculation), tumor growth could be followed directly after inoculation of UM-UC-3luc2 cells. Importantly, micrometastatic lesions originating from orthotopically implanted cancer cells could be detected in the locoregional lymph nodes and in distant organs. In addition, the superior bioluminescent indicator firefly luciferase 2 allows the detection and monitoring of micrometastatic lesions in real time after intracardiac inoculation of human bladder cancer cells in mice. The main disadvantage is the lack of T-cell immunity in the preclinical models. CONCLUSIONS: The new bioluminescence-based preclinical bladder cancer models enable superior, noninvasive, and real-time tracking of cancer cells, tumor progression, and micrometastasis. Because of the significant improvement in detection of small cell numbers, the presented models are ideally suited for functional studies dealing with minimal residual disease as well as real-time imaging of drug response.


Subject(s)
Bone Neoplasms/secondary , Carcinoma, Transitional Cell/secondary , Cell Tracking/methods , Luciferases, Firefly/biosynthesis , Luminescent Measurements , Lung Neoplasms/secondary , Urinary Bladder Neoplasms/pathology , Animals , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Carcinoma, Transitional Cell/genetics , Carcinoma, Transitional Cell/metabolism , Cell Line, Tumor , Female , Humans , Luciferases, Firefly/genetics , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lymphatic Metastasis , Mice , Mice, Inbred BALB C , Mice, Nude , Time Factors , Transfection , Tumor Burden , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism
5.
Cancer Res ; 70(12): 5163-73, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20516116

ABSTRACT

Metastatic progression of advanced prostate cancer is a major clinical problem. Identifying the cell(s) of origin in prostate cancer and its distant metastases may permit the development of more effective treatment and preventive therapies. In this study, aldehyde dehydrogenase (ALDH) activity was used as a basis to isolate and compare subpopulations of primary human prostate cancer cells and cell lines. ALDH-high prostate cancer cells displayed strongly elevated clonogenicity and migratory behavior in vitro. More strikingly, ALDH-high cells readily formed distant metastases with strongly enhanced tumor progression at both orthotopic and metastatic sites in preclinical models. Several ALDH isoforms were expressed in human prostate cancer cells and clinical specimens of primary prostate tumors with matched bone metastases. Our findings suggest that ALDH-based viable cell sorting can be used to identify and characterize tumor-initiating and, more importantly perhaps, metastasis-initiating cells in human prostate cancer.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Bone Neoplasms/enzymology , Bone Neoplasms/secondary , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Animals , Biomarkers, Tumor/metabolism , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic , Disease Progression , Flow Cytometry , Humans , Immunoenzyme Techniques , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Prostate/enzymology , Prostate/pathology
6.
Microb Pathog ; 38(2-3): 63-76, 2005.
Article in English | MEDLINE | ID: mdl-15748808

ABSTRACT

The enteric pathogens Shigella dysenteriae serotype 1 and Shiga toxin-producing Escherichia coli share the property of expressing the structurally and functionally related cytotoxins that comprise the Shiga toxin (Stx) family. Stx-producing bacteria are causative agents of bloody diarrheal diseases that may progress to life threatening complications involving the destruction of blood vessels in the kidneys and the central nervous system (CNS). The precise mechanisms of toxin transport across the gut epithelial barrier, and the role of innate immunity in the development of systemic complications, remain to be fully characterized. Earlier studies suggested that Stxs and lipopolysaccharides (LPS) induce the expression of proinflammatory cytokines from differentiated (macrophage-like) THP-1 cells. These cytokines may exacerbate vascular damage by up-regulating the expression of toxin receptors on endothelial cells. Purified Stxs have also been shown to induce apoptosis of epithelial and endothelial cells in vitro, but a comparative evaluation of Stx-induced apoptosis of monocytes and macrophages has not been reported. We used FACS, TUNEL, and DNA laddering analyses to show that Shiga toxin-1 (Stx1) and LPS induce apoptosis in undifferentiated and differentiated THP-1 cells, although the kinetics and extent of apoptosis induction differ between monocytic and macrophage-like cells. Stx1-induced apoptosis is A-subunit-dependent. Stx1 and LPS trigger DNA fragmentation and caspase-3 activation, as evidenced by the cleavage of poly(ADP-ribose) polymerase (PARP). Induction of apoptosis in response to Stx1 and/or LPS treatment occurs without the widespread transcriptional activation of apoptosis-related genes. Finally, we present a model of the role of macrophages and monocytes in the pathogenesis of disease caused by Stxs.


Subject(s)
Apoptosis , Escherichia coli , Lipopolysaccharides/toxicity , Macrophages/cytology , Monocytes/cytology , Shiga Toxin 1/toxicity , Shigella dysenteriae , Caspase 3 , Caspases/analysis , Cell Line , DNA Fragmentation , Flow Cytometry , Humans , In Situ Nick-End Labeling , Macrophages/microbiology , Monocytes/microbiology , Poly(ADP-ribose) Polymerases/metabolism
7.
Infect Immun ; 73(1): 403-12, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15618178

ABSTRACT

Infections with Shiga toxin (Stx)-producing bacteria are associated with bloody diarrhea and postdiarrheal sequelae, including hemolytic uremic syndrome and central nervous system (CNS) abnormalities. Stx-induced intestinal, renal, and CNS vascular lesions may involve a localized production of proinflammatory cytokines in target organs, as tumor necrosis factor-alpha (TNF-alpha) and interleukin-1beta (IL-1beta) up-regulate Stx receptor globotriaosylceramide (Gb(3)) expression on vascular endothelial cells. However, leukocyte recruitment to injured sites may also exacerbate vascular damage. A cytokine macroarray analysis of transcripts derived from macrophage-like THP-1 cells treated with Stx1, lipopolysaccharides (LPS), or both demonstrated a consistent up-regulation of TNF-alpha, IL-1beta, and four genes encoding the chemokines interleukin-8 (IL-8), macrophage inflammatory protein-1alpha (MIP-1alpha), MIP-1beta, and growth-related oncogene beta (GRO-beta). Real-time PCR analysis verified the macroarray results. Northern blot analyses after the addition of the transcriptional inhibitor actinomycin D revealed increased IL-8 mRNA stability in THP-1 cells treated with Stx1 or Stx1 plus LPS. Finally, enzyme-linked immunosorbent assay data for Stx1- plus LPS-treated cells demonstrated a poor correlation between IL-8, MIP-1alpha, MIP-1beta, and GRO-beta mRNA levels and protein production, indicating a posttranscriptional regulatory effect. Our data suggest that in response to Stx1 and LPS, macrophages may be a source of chemokines that promote tissue damage through leukocyte recruitment and activation.


Subject(s)
Chemokines/genetics , Lipopolysaccharides/pharmacology , Shiga Toxin 1/pharmacology , Cell Line, Tumor , Chemokine CCL3 , Chemokine CCL4 , Cytokines/genetics , Humans , Interleukin-8/biosynthesis , Interleukin-8/genetics , Macrophage Inflammatory Proteins/biosynthesis , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...