Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Anticancer Res ; 42(6): 2859-2867, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35641280

ABSTRACT

BACKGROUND/AIM: Tumor vasculature is an important component of the tumor microenvironment and deeply affects anticancer immune response. Eribulin is a non-taxane inhibitor of the mitotic spindle. However, off-target effects interfering with the tumor vasculature have been reported. The mechanisms responsible of this effect are still unclear. MATERIALS AND METHODS: We designed an in vitro study to investigate the effect of eribulin, with or without TGF-ß, on neo-angiogenesis, and on the expression of the adhesion molecules ICAM-1 and VCAM-1. We also investigated the effects of paclitaxel and vinorelbine under the same experimental conditions. RESULTS: Eribulin up-regulated the epithelial markers VE-cadherin and CD-31 in HUVEC and inhibited tube formation in HUVEC cells cultured in Matrigel. The drug effectively arrested tube formation even in the presence of TGF-ß and counteracted the TGF-ß-induced change in cell shape from the endothelial cobblestone-like morphology to an elongated spindle-shaped morphology. We also observed that eribulin was able to upregulate ICAM-1 and to counteract its down-regulation induced by TGF-ß. CONCLUSION: Eribulin exerts different off-label effects: increases vascular remodeling, counteracts the endothelial-to-mesenchymal transition (EndMT) mediated by TGF-ß and promotes tumor infiltration by immune cells via increasing the expression of ICAM-1 and transcription of CD31 and VE-cadherin. Moreover, eribulin was able to inhibit vasculature remodeling and the induction of EndMT mediated by TGF-ß better than vinorelbine and paclitaxel. The effects observed in this study might have important therapeutic consequence if the drug is combined with immunotherapy.


Subject(s)
Intercellular Adhesion Molecule-1 , Neoplasms , Furans , Humans , Intercellular Adhesion Molecule-1/genetics , Ketones , Paclitaxel/pharmacology , Transforming Growth Factor beta , Tumor Microenvironment , Vinorelbine
2.
Carcinogenesis ; 41(12): 1695-1702, 2020 12 31.
Article in English | MEDLINE | ID: mdl-32614387

ABSTRACT

Arginine metabolism plays a significant role in regulating cell function, affecting tumor growth and metastatization. To study the effect of the arginine-catabolizing enzyme Arginase1 (ARG1) on tumor microenvironment, we generated a mouse model of mammary carcinogenesis by crossbreeding a transgenic mouse line overexpressing ARG1 in macrophages (FVBArg+/+) with the MMTV-Neu mouse line (FVBNeu+/+). This double transgenic line (FVBArg+/-;Neu+/+) showed a significant shortening in mammary tumor latency, and an increase in the number of mammary nodules. Transfer of tumor cells from FVBNeu+/+ into either FVB wild type or FVBArg+/+ mice resulted in increase regulatory T cells in the tumor infiltrate, suggestive of an impaired antitumor immune response. However, we also found increased frequency of tumor stem cells in tumors from FVBArg+/-;Neu+/+ transgenic compared with FVBNeu+/+ mice, suggesting that increased arginine metabolism in mammary tumor microenvironment may supports the cancer stem cells niche. We provide in vivo evidence of a novel, yet unexploited, mechanism through which ARG1 may contribute to tumor development.


Subject(s)
Arginase/metabolism , Cell Transformation, Neoplastic/pathology , Mammary Neoplasms, Experimental/pathology , Tumor Microenvironment/immunology , Animals , Apoptosis , Arginase/genetics , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Female , Humans , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Transgenic , Tumor Cells, Cultured
3.
Sci Rep ; 10(1): 1454, 2020 01 29.
Article in English | MEDLINE | ID: mdl-31996711

ABSTRACT

Abscisic acid (ABA) is a plant hormone active also in mammals where it regulates, at nanomolar concentrations, blood glucose homeostasis. Here we investigated the mechanism through which low-dose ABA controls glycemia and glucose fate. ABA stimulated uptake of the fluorescent glucose analog 2-NBDG by L6, and of [18F]-deoxy-glucose (FDG) by mouse skeletal muscle, in the absence of insulin, and both effects were abrogated by the specific AMPK inhibitor dorsomorphin. In L6, incubation with ABA increased phosphorylation of AMPK and upregulated PGC-1α expression. LANCL2 silencing reduced all these ABA-induced effects. In vivo, low-dose oral ABA stimulated glucose uptake and storage in the skeletal muscle of rats undergoing an oral glucose load, as detected by micro-PET. Chronic treatment with ABA significantly improved the AUC of glycemia and muscle glycogen content in CD1 mice exposed to a high-glucose diet. Finally, both acute and chronic ABA treatment of hypoinsulinemic TRPM2-/- mice ameliorated the glycemia profile and increased muscle glycogen storage. Altogether, these results suggest that low-dose oral ABA might be beneficial for pre-diabetic and diabetic subjects by increasing insulin-independent skeletal muscle glucose disposal through an AMPK-mediated mechanism.


Subject(s)
Abscisic Acid/metabolism , Diabetes Mellitus/metabolism , Glucose/metabolism , Muscle, Skeletal/pathology , Myoblasts/metabolism , 4-Chloro-7-nitrobenzofurazan/analogs & derivatives , 4-Chloro-7-nitrobenzofurazan/metabolism , AMP-Activated Protein Kinase Kinases , Animals , Cell Line , Deoxyglucose/analogs & derivatives , Deoxyglucose/metabolism , Disease Models, Animal , Insulin/metabolism , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Myoblasts/pathology , Protein Kinases/metabolism , RNA, Small Interfering/genetics , Rats , Rats, Wistar , TRPM Cation Channels/genetics
4.
Int J Mol Sci ; 20(23)2019 Dec 03.
Article in English | MEDLINE | ID: mdl-31816863

ABSTRACT

BACKGROUND: Prostate cancer (PCa) is a significant health concern throughout the world. Standard therapy for advanced disease consists of anti-androgens, however, almost all prostate tumors become castration resistant (CRPC). Progression from androgen-sensitive PCa to CRPC is promoted by inflammatory signaling through cyclooxygenase-2 (COX-2) expression and ErbB family receptors/AKT activation, compensating androgen receptor inactivity. METHODS: Making use of CRPC cell lines, we investigated the effects of the anti-inflammatory drug celecoxib. Biochemical data obtained using immunoblotting, enzyme-linked immunosorbent assay (ELISA), invasion, and xenografts were further integrated by bioinformatic analyses. RESULTS: Celecoxib reduced cell growth and induced apoptosis through AKT blockade, cleavage of poly (ADP-ribose) polymerase-1 (PARP-1), and proteasomal degradation of the anti-apoptotic protein Mcl-1. Epidermal growth factor receptor (EGFR), ErbB2, and ErbB3 degradation, and heterogeneous nuclear ribonucleoprotein K (hnRNP K) downregulation, further amplified the inhibition of androgen signaling. Celecoxib reduced the invasive phenotype of CRPC cells by modulating NF-κB activity and reduced tumor growth in mice xenografts when administered in association with the anti-EGFR receptor antibody cetuximab. Bioinformatic analyses on human prostate cancer datasets support the relevance of these pathways in PCa progression. CONCLUSIONS: Signaling nodes at the intersection of pathways implicated in PCa progression are simultaneously modulated by celecoxib treatment. In combination therapies with cetuximab, celecoxib could represent a novel therapeutic strategy to curb signal transduction during CRPC progression.


Subject(s)
Celecoxib/therapeutic use , Prostatic Neoplasms, Castration-Resistant/drug therapy , Signal Transduction , Amphiregulin/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Celecoxib/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cetuximab/pharmacology , Cetuximab/therapeutic use , Down-Regulation/drug effects , Drug Resistance, Neoplasm/drug effects , Epidermal Growth Factor/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Humans , Male , Mice, SCID , NF-kappa B/metabolism , Neoplasm Invasiveness , Phosphorylation/drug effects , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/metabolism , Receptors, Androgen/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays , p38 Mitogen-Activated Protein Kinases/metabolism
5.
J Exp Clin Cancer Res ; 38(1): 108, 2019 Mar 01.
Article in English | MEDLINE | ID: mdl-30823889

ABSTRACT

BACKGROUND: Cancer cells are characterized by chromosomal instability (CIN) and it is thought that errors in pathways involved in faithful chromosome segregation play a pivotal role in the genesis of CIN. Cohesin forms a large protein ring that binds DNA strands by encircling them. In addition to this central role in chromosome segregation, cohesin is also needed for DNA repair, gene transcription regulation and chromatin architecture. Though mutations in both cohesin and cohesin-regulator genes have been identified in many human cancers, the contribution of cohesin to cancer development is still under debate. METHODS: Normal mucosa, early adenoma, and carcinoma samples deriving from 16 subjects affected by colorectal cancer (CRC) were analyzed by OncoScan for scoring both chromosome gains and losses (CNVs) and loss of heterozygosity (LOH). Then the expression of SMC1A was analyzed by immunochemistry in 66 subjects affected by CRC. The effects of SMC1A overexpression and mutated SMC1A were analyzed in vivo using immunocompromised mouse models. Finally, we measured global gene expression profiles in induced-tumors by RNA-seq. RESULTS: Here we showed that SMC1A cohesin core gene was present as extra-copies, mutated, and overexpressed in human colorectal carcinomas. We then demonstrated that cohesin overexpression led to the development of aggressive cancers in immunocompromised mice through gene expression dysregulation. CONCLUSION: Collectively, these results support a role of defective cohesin in the development of human colorectal cancer.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/pathology , Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Adenoma/genetics , Adenoma/pathology , Adult , Aged , Animals , Cell Cycle Proteins/biosynthesis , Chromosomal Instability , Chromosomal Proteins, Non-Histone/biosynthesis , Female , Heterografts , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged
6.
J Enzyme Inhib Med Chem ; 32(1): 363-368, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28095711

ABSTRACT

The microenvironment of solid tumours is extremely acidic and this condition arises since the precancerous stage. This acidic milieu could therefore provide a useful target for both prophylactic and therapeutic approaches. In TRAMP transgenic mice, an in vivo model of prostate adenocarcinoma (AC), oral administration of alkaline water was devoid of unwanted side effects, and when started from an early age was as effective as NaHCO3 in significantly delaying tumour progression, while when started when prostate tumours were already present, a nonstatistically significant trend in the same direction was detected. These findings indicate that the use of alkalinizing drugs should be considered for chemoprevention and, in association with standard chemotherapy, for treatment of human prostate AC.


Subject(s)
Hydrogen-Ion Concentration , Prostatic Neoplasms/pathology , Animals , Cell Line, Tumor , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Tumor Necrosis Factor, Member 25/genetics , Tumor Microenvironment
7.
Hum Mol Genet ; 25(1): 97-108, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26512061

ABSTRACT

Dlx5 and Dlx6 are two closely associated homeobox genes which code for transcription factors involved in the control of steroidogenesis and reproduction. Inactivation of Dlx5/6 in the mouse results in a Leydig cell defect in the male and in ovarian insufficiency in the female. DLX5/6 are also strongly expressed by the human endometrium but their function in the uterus is unknown. The involvement of DLX5/6 in human uterine pathology is suggested by their strong downregulation in endometriotic lesions and upregulation in endometrioïd adenocarcinomas. We first show that Dlx5/6 expression begins in Müllerian ducts epithelia and persists then in the uterine luminal and glandular epithelia throughout post-natal maturation and in the adult. We then use a new mouse model in which Dlx5 and Dlx6 can be simultaneously inactivated in the endometrium using a Pgr(cre/+) allele. Post-natal inactivation of Dlx5/6 in the uterus results in sterility without any obvious ovarian involvement. The uteri of Pgr(cre/+); Dlx5/6(flox/flox) mice present very few uterine glands and numerous abnormally large and branched invaginations of the uterine lumen. In Dlx5/6 mutant uteri, the expression of genes involved in gland formation (Foxa2) and in epithelial remodelling during implantation (Msx1) is significantly reduced. Furthermore, we show that DLX5 is highly expressed in human endometrial glandular epithelium and that its expression is affected in endometriosis. We conclude that Dlx5 and Dlx6 expression determines uterine architecture and adenogenesis and is needed for implantation. Given their importance for female reproduction, DLX5 and DLX6 must be regarded as interesting targets for future clinical research.


Subject(s)
Endometriosis/genetics , Genes, Homeobox , Homeodomain Proteins/genetics , Uterus/growth & development , Animals , Embryo Implantation , Endometriosis/metabolism , Epithelium/growth & development , Female , Gene Expression Regulation , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/physiology , Mice , Transcriptome , Uterus/metabolism
8.
Carcinogenesis ; 36(11): 1354-62, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26363032

ABSTRACT

Arginase (ARG) is a metabolic enzyme present in two isoforms that hydrolyze l-arginine to urea and ornithine. In humans, ARG isoform 1 is also expressed in cells of the myeloid lineage. ARG activity promotes tumour growth and inhibits T lymphocyte activation. However, the two ARG transgenic mouse lines produced so far failed to show such effects. We have generated, in two different genetic backgrounds, transgenic mice constitutively expressing ARG1 under the control of the CD68 promoter in macrophages and monocytes. Both heterozygous and homozygous transgenic mice showed a relevant increase in mortality at early age, compared with wild-type siblings (67/267 and 48/181 versus 8/149, respectively, both P < 0.005). This increase was due to high incidence of haematologic malignancies, in particular myeloid leukaemia, myeloid dysplasia, lymphomas and disseminated intravascular coagulation (DIC), diseases that were absent in wild-type mice. Atrophy of lymphoid organs due to reduction in T-cell compartment was also detected. Our results indicate that ARG activity may participate in the pathogenesis of lymphoproliferative and myeloproliferative disorders, suggest the involvement of alterations of L-arginine metabolism in the onset of DIC and confirm a role for the enzyme in regulating T-cell homeostasis.


Subject(s)
Arginase/metabolism , Lymphoproliferative Disorders/enzymology , Monocytes/enzymology , Myeloproliferative Disorders/enzymology , Animals , Arginase/genetics , Cell Lineage , Female , Gene Expression , Lymphocyte Activation , Lymphoproliferative Disorders/pathology , Male , Mice, Transgenic , Myeloproliferative Disorders/pathology , T-Lymphocytes/enzymology , T-Lymphocytes/immunology
9.
Dis Model Mech ; 7(9): 1083-91, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25147298

ABSTRACT

Glycogen storage disease type 1a (GSD-1a) is caused by a deficiency in glucose-6-phosphatase-α (G6Pase-α), and is characterized by impaired glucose homeostasis and a high risk of developing hepatocellular adenomas (HCAs). A globally G6Pase-α-deficient (G6pc(-/-)) mouse model that shows pathological features similar to those of humans with GSD-1a has been developed. These mice show a very severe phenotype of disturbed glucose homeostasis and rarely live beyond weaning. We generated liver-specific G6Pase-α-deficient (LS­G6pc(-/-)) mice as an alternative animal model for studying the long-term pathophysiology of the liver and the potential treatment strategies, such as cell therapy. LS­G6pc(-/-) mice were viable and exhibited normal glucose profiles in the fed state, but showed significantly lower blood glucose levels than their control littermates after 6 hours of fasting. LS­G6pc(-/-) mice developed hepatomegaly with glycogen accumulation and hepatic steatosis, and progressive hepatic degeneration. Ninety percent of the mice analyzed developed amyloidosis by 12 months of age. Finally, 25% of the mice sacrificed at age 10-20 months showed the presence of multiple HCAs and in one case late development of hepatocellular carcinoma (HCC). In conclusion, LS­G6pc(-/-) mice manifest hepatic symptoms similar to those of human GSD-1a and, therefore, represent a valid model to evaluate long-term liver pathogenesis of GSD-1a.


Subject(s)
Adenoma/pathology , Carcinoma, Hepatocellular/pathology , Glucosephosphate Dehydrogenase/genetics , Liver Neoplasms/pathology , Adenoma/enzymology , Animals , Carcinoma, Hepatocellular/enzymology , Liver Neoplasms/enzymology , Mice
10.
Mol Oncol ; 8(3): 581-95, 2014 May.
Article in English | MEDLINE | ID: mdl-24484937

ABSTRACT

Chronic inflammation is a major risk factor for the development and metastatic progression of cancer. We have previously reported that the chemopreventive polyphenol Curcumin inhibits the expression of the proinflammatory cytokines CXCL1 and -2 leading to diminished formation of breast and prostate cancer metastases. In the present study, we have analyzed the effects of Curcumin on miRNA expression and its correlation to the anti-tumorigenic properties of this natural occurring polyphenol. Using microarray miRNA expression analyses, we show here that Curcumin modulates the expression of a series of miRNAs, including miR181b, in metastatic breast cancer cells. Interestingly, we found that miR181b down-modulates CXCL1 and -2 through a direct binding to their 3'-UTR. Overexpression or inhibition of miR181b in metastatic breast cancer cells has a significant impact on CXCL1 and -2 and is required for the effect of Curcumin on these two cytokines. miR181b also mediates the effects of Curcumin on inhibition of proliferation and invasion as well as induction of apoptosis. Importantly, over-expression of miR181b in metastatic breast cancer cells inhibits metastasis formation in vivo in immunodeficient mice. Finally, we demonstrated that Curcumin up-regulates miR181b and down-regulates CXCL1 and -2 in cells isolated from several primary human breast cancers. Taken together, these data show that Curcumin provides a simple bridge to bring metastamir modulation into the clinic, placing it in a primary and tertiary preventive, as well as a therapeutic, setting.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Chemokine CXCL1/genetics , Chemokine CXCL2/genetics , Curcumin/pharmacology , MicroRNAs/genetics , Neoplasm Metastasis/drug therapy , Aged , Animals , Breast/drug effects , Breast/metabolism , Breast/pathology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Down-Regulation/drug effects , Female , Humans , Mice , Middle Aged , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Tumor Cells, Cultured , Up-Regulation/drug effects
11.
Haematologica ; 99(3): 448-57, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24179150

ABSTRACT

Decidual natural killer cells accumulate at the fetal-maternal interface and play a key role in a successful pregnancy. However, their origin is still unknown. Do they derive from peripheral natural killer cells recruited in decidua or do they represent a distinct population that originates in situ? Here, we identified natural killer precursors in decidua and uterus of pregnant mice. These precursors underwent rapid in situ differentiation and large proportions of proliferating immature natural killer cells were present in decidua and uterus as early as gestation day 4.5. Here, we investigated the origin of decidua- and uterus-natural killer cells by performing transfer experiments of peripheral mature natural killer cells or precursors from EGFP(+) mice. Results showed that mature natural killer cells did not migrate into decidua and uterus, while precursors were recruited in these organs and differentiated towards natural killer cells. Moreover, decidua- and uterus-natural killer cells displayed unique phenotypic and functional features. They expressed high levels of the activating Ly49D receptor in spite of their immature phenotype. In addition, decidua- and uterus-natural killer cells were poorly cytolytic and produced low amounts of IFN-γ, while they released factors (GM-CSF, VEGF, IP-10) involved in neo-angiogenesis and tissue remodeling. Our data reveal in situ generation of decidual natural killer cells and provide an important correlation between mouse and human decidual natural killer cells, allowing further studies to be carried out on their role in pregnancy-related diseases.


Subject(s)
Cell Differentiation , Decidua/cytology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Animals , Antigens, Surface/metabolism , Cell Proliferation , Cytotoxicity, Immunologic , Decidua/immunology , Decidua/metabolism , Female , Hematopoiesis , Immunomodulation , Immunophenotyping , Killer Cells, Natural/immunology , Mice , Mice, Transgenic , NK Cell Lectin-Like Receptor Subfamily A/metabolism , Pregnancy , Uterus/cytology , Uterus/immunology
12.
PLoS One ; 8(1): e51700, 2013.
Article in English | MEDLINE | ID: mdl-23382810

ABSTRACT

The Dlx and Msx homeodomain transcription factors play important roles in the control of limb development. The combined disruption of Msx1 and Msx2, as well as that of Dlx5 and Dlx6, lead to limb patterning defects with anomalies in digit number and shape. Msx1;Msx2 double mutants are characterized by the loss of derivatives of the anterior limb mesoderm which is not observed in either of the simple mutants. Dlx5;Dlx6 double mutants exhibit hindlimb ectrodactyly. While the morphogenetic action of Msx genes seems to involve the BMP molecules, the mode of action of Dlx genes still remains elusive. Here, examining the limb phenotypes of combined Dlx and Msx mutants we reveal a new Dlx-Msx regulatory loop directly involving BMPs. In Msx1;Dlx5;Dlx6 triple mutant mice (TKO), beside the expected ectrodactyly, we also observe the hallmark morphological anomalies of Msx1;Msx2 double mutants suggesting an epistatic role of Dlx5 and Dlx6 over Msx2. In Msx2;Dlx5;Dlx6 TKO mice we only observe an aggravation of the ectrodactyly defect without changes in the number of the individual components of the limb. Using a combination of qPCR, ChIP and bioinformatic analyses, we identify two Dlx/Msx regulatory pathways: 1) in the anterior limb mesoderm a non-cell autonomous Msx-Dlx regulatory loop involves BMP molecules through the AER and 2) in AER cells and, at later stages, in the limb mesoderm the regulation of Msx2 by Dlx5 and Dlx6 occurs also cell autonomously. These data bring new elements to decipher the complex AER-mesoderm dialogue that takes place during limb development and provide clues to understanding the etiology of congenital limb malformations.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Extremities/growth & development , Homeodomain Proteins/genetics , MSX1 Transcription Factor/genetics , Animals , Ectoderm/growth & development , Ectoderm/metabolism , Female , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Mesoderm/growth & development , Mesoderm/metabolism , Mice , Morphogenesis , Mutation
13.
Carcinogenesis ; 33(12): 2507-19, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23042094

ABSTRACT

In America and Western Europe, prostate cancer is the second leading cause of death in men. Emerging evidence suggests that chronic inflammation is a major risk factor for the development and metastatic progression of prostate cancer. We previously reported that the chemopreventive polyphenol curcumin inhibits the expression of the proinflammatory cytokines CXCL1 and -2 leading to diminished formation of breast cancer metastases. In this study, we analyze the effects of curcumin on prostate carcinoma growth, apoptosis and metastasis. We show that curcumin inhibits translocation of NFκB to the nucleus through the inhibition of the IκB-kinase (IKKß, leading to stabilization of the inhibitor of NFκB, IκBα, in PC-3 prostate carcinoma cells. Inhibition of NFκB activity reduces expression of CXCL1 and -2 and abolishes the autocrine/paracrine loop that links the two chemokines to NFκB. The combination of curcumin with the synthetic IKKß inhibitor, SC-541, shows no additive or synergistic effects indicating that the two compounds share the target. Treatment of the cells with curcumin and siRNA-based knockdown of CXCL1 and -2 induce apoptosis, inhibit proliferation and downregulate several important metastasis-promoting factors like COX2, SPARC and EFEMP. In an orthotopic mouse model of hematogenous metastasis, treatment with curcumin inhibits statistically significantly formation of lung metastases. In conclusion, chronic inflammation can induce a metastasis prone phenotype in prostate cancer cells by maintaining a positive proinflammatory and prometastatic feedback loop between NFκB and CXCL1/-2. Curcumin disrupts this feedback loop by the inhibition of NFκB signaling leading to reduced metastasis formation in vivo.


Subject(s)
Antineoplastic Agents/therapeutic use , Chemokine CXCL1/antagonists & inhibitors , Chemokine CXCL2/antagonists & inhibitors , Curcumin/therapeutic use , Prostatic Neoplasms/drug therapy , Animals , Apoptosis , Cell Line, Tumor , Chemokine CXCL1/genetics , Chemokine CXCL2/genetics , Humans , Male , Mice , NF-kappa B/antagonists & inhibitors , NF-kappa B/physiology , Neoplasm Metastasis , Prostatic Neoplasms/pathology , Signal Transduction , Xenograft Model Antitumor Assays
14.
Biochem Pharmacol ; 83(12): 1634-42, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22445931

ABSTRACT

Although the vast majority of patients with acute lymphocytic leukemia (ALL) attain remission with modern therapies, relapsed leukemia will continue to be a common malignancy both in childhood and in adults, until new treatments are available. Therapeutic options for advanced B-cell acute lymphocytic leukemia are still limited and acquired drug resistance and extramedullary tissue infiltration are two major obstacles during treatment. The prenylflavonoid xanthohumol (XN) has shown in vitro and in vivo therapeutic potential against a range of tumors. In the present study we investigated the effects of XN on B-ALL cells in vitro and in an ALL-like xenograft mouse model. Treatment of ALL cell lines with XN resulted in growth arrest and apoptosis induction. XN retained its cytotoxicity when adriamycin resistant cells were examined while ALL cell clones adapted to long-term exposure to XN resulted highly responsive to cytotoxic drugs. Administration of 50µg XN/mouse (5 days/week) significantly increased animal life span by delaying the insurgence of neurological disorders due to leukemic cells dissemination. In agreement with a less invasive phenotype, cell migration and invasion were impaired by XN and basal levels of FAK, AKT and NF-κB signaling pathways were down-regulated in ALL cells upon XN exposure. Our data indicate that XN has significant antileukemic activity both in vitro and in vivo, which associates with impaired cell migration and invasion. Interestingly, this activity overcomes mechanisms leading to drug-resistance. XN represents a promising agent perspective for ALL therapy and recurrence prevention and would deserve clinical testing in the near future.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Flavonoids/pharmacology , Leukemic Infiltration/pathology , NF-kappa B/antagonists & inhibitors , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Propiophenones/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Animals , Cell Line, Tumor , Humans , Mice , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy
15.
Int J Cancer ; 131(3): 582-90, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22015967

ABSTRACT

Current diagnostic tools cannot predict clinical failure and androgen-independent disease progression for patients with prostate cancer (PC). The survival signaling pathways of prostate cells play a central role in the progression of tumors to a neuroendocrine (NE) phenotype. NE cells demonstrate attributes that suggest that they are an integral part of the signaling cascade leading to castration-resistant PC. In this study, making use of in vitro neuroendocrine differentiation (NED) of human LNCaP and mouse TRAMP-C2 cells after androgen withdrawal, and of the transgenic adenocarcinoma of mouse prostate (TRAMP) model, we characterized a sequence of molecular events leading to NED and identified a number of markers that could be detectable by routine analyses not only in castration resistant PC but also in hormone naïve PC at the time of initial diagnosis. We found that NED associates with AKT activation that in turn regulates heterogeneous nuclear ribonucleoprotein K (hnRNP K), androgen receptor (AR) and ß-catenin levels. Addition of molecules targeting membrane-bound receptors and protein kinases blocks NE differentiation in LNCaP and TRAMP-C2 cells. The extent of AKT phosphorylation and hnRNP K, AR and ß-catenin levels may have a potential value as prognostic indicators discriminating between androgen-responsive and unresponsive cells and could be used as molecular targets to monitor the anti-tumor action of new therapeutic protocols based on antireceptor agents and/or neuroendocrine hormone antagonists.


Subject(s)
Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Neuroendocrine Cells/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Signal Transduction , Androgens/metabolism , Animals , Cell Differentiation , Cell Line, Tumor , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuroendocrine Cells/pathology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Androgen/metabolism , beta Catenin/metabolism
16.
Cell Cycle ; 10(19): 3253-6, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21946516

ABSTRACT

Studies on oocyte transcriptome are important to understand the biological pathways involved in oogenesis, totipotence and early embryonic development. Moreover, genes regulating physiological pathways in gametes could represent potential candidates for reproductive disorders. In addition to oocyte specific transcription factors, also the members of the p53 family could be etiologically involved due to their biological functions. In fact, their role in the control of cell cycle, apoptosis, and germ-line genome stability is well known. Female reproductive aging is one of the causes of fertility reduction and it is often associated with egg aneuploidy increase. In order to verify the potential involvement of p73 in reproductive aging, we determined its expression in single mature MII oocytes from two groups of women, younger than 35 or older than 38 years, respectively. We found that TAp73 isoforms are down regulated in oocytes from women older than 38 years. We confirmed these data in pools of mouse oocytes. TAp73 down regulation in oocytes from women of advanced reproductive age could explain both the reduction of fertility and the increase of newborns with chromosomal abnormalities.


Subject(s)
DNA-Binding Proteins/metabolism , Down-Regulation , Nuclear Proteins/metabolism , Oocytes/metabolism , Reproduction , Tumor Suppressor Proteins/metabolism , Adult , Aneuploidy , Animals , Apoptosis , Cell Cycle Checkpoints , DNA Methylation , DNA-Binding Proteins/genetics , Female , Genomic Instability , Humans , Mice , Nuclear Proteins/genetics , Promoter Regions, Genetic , Protein Isoforms/genetics , Protein Isoforms/metabolism , Transcriptome , Tumor Protein p73 , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics
17.
J Hepatol ; 55(6): 1263-71, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21703205

ABSTRACT

BACKGROUND & AIMS: Several studies have shown that bone marrow-derived committed myelomonocytic cells can repopulate diseased livers by fusing with host hepatocytes and can restore normal liver function. These data suggest that myelomonocyte transplantation could be a promising approach for targeted and well-tolerated cell therapy aimed at liver regeneration. We sought to determine whether bone marrow-derived myelomonocytic cells could be effective for liver reconstitution in newborn mice knock-out for glucose-6-phosphatase-α. METHODS: Bone marrow-derived myelomonocytic cells obtained from adult wild type mice were transplanted in newborn knock-out mice. Tissues of control and treated mice were frozen for histochemical analysis, or paraffin-embedded and stained with hematoxylin and eosin for histological examination or analyzed by immunohistochemistry or fluorescent in situ hybridization. RESULTS: Histological sections of livers of treated knock-out mice revealed areas of regenerating tissue consisting of hepatocytes of normal appearance and partial recovery of normal architecture as early as 1 week after myelomonocytic cells transplant. FISH analysis with X and Y chromosome paints indicated fusion between infused cells and host hepatocytes. Glucose-6-phosphatase activity was detected in treated mice with improved profiles of liver functional parameters. CONCLUSIONS: Our data indicate that bone marrow-derived myelomonocytic cell transplant may represent an effective way to achieve liver reconstitution of highly degenerated livers in newborn animals.


Subject(s)
Glycogen Storage Disease Type I/pathology , Glycogen Storage Disease Type I/therapy , Liver/pathology , Myeloid Cells/transplantation , Animals , Animals, Newborn , Bone Marrow Transplantation , Female , Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/metabolism , Glycogen Storage Disease Type I/genetics , Glycogen Storage Disease Type I/physiopathology , Liver/physiopathology , Liver Regeneration , Male , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Pregnancy
18.
Hum Mol Genet ; 20(13): 2642-50, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21505076

ABSTRACT

Primary ovarian insufficiency (POI) is characterized by the loss of ovarian function before the age of 40 in humans. Although most cases of POI are idiopathic, many are familial, underlying a genetic origin of the disease. Mutations in genes involved in the control of steroidogenesis, such as NR5A1 (SF-1, Steroidogenic Factor 1), CYP17, CYP19A1 (aromatase), StAR (Steroidogenic Acute Regulatory), and the forkhead transcription factor FOXL2 have been associated with different forms of POI. In males, the homeobox transcription factors Dlx5 and Dlx6 are involved in the control of steroidogenesis through the activation of GATA4-induced-StAR transcription. Here, we analyze the potential involvement of Dlx5 and Dlx6 in female reproduction. To this end, we make use of an existing mouse model in which Dlx5 and Dlx6 are simultaneously disrupted. We show that: (i) allelic reduction of Dlx5 and Dlx6 in the mouse results in a POI-like phenotype, characterized by reduced fertility and early follicular exhaustion; (ii) in granulosa cell lines, a reciprocal regulation exists between Dlx5 and Foxl2; (iii) in the mouse ovary, allelic reduction of Dlx5/6 results in the upregulation of Foxl2. We propose that the mutual regulation between Dlx5/6 and Foxl2 and their opposite effects on StAR expression might contribute to determine the homeostatic control of steroidogenesis within the ovary. Dysregulation of this homeostatic control would result in abnormal follicular maturation and reduced fertility. Our results bring new elements to our conceptual model of follicle maturation and maintenance and provide new potential genetic targets for cases of familial POI.


Subject(s)
Disease Models, Animal , Homeodomain Proteins/genetics , Ovarian Follicle/metabolism , Ovarian Follicle/pathology , Primary Ovarian Insufficiency/genetics , Primary Ovarian Insufficiency/pathology , Alleles , Animals , Cell Line , Female , Fertility/genetics , Forkhead Box Protein L2 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Gene Order , Granulosa Cells/metabolism , Homeodomain Proteins/metabolism , Humans , Male , Mice , Mice, Knockout , Primary Ovarian Insufficiency/metabolism
19.
J Leukoc Biol ; 89(5): 721-7, 2011 May.
Article in English | MEDLINE | ID: mdl-21330347

ABSTRACT

ARG1, expressed by human PMNs, inhibits T cell proliferation by depleting extracellular L-arginine. Here, we report that ARG1, released from gelatinase granules by PMNs, is inactive at physiological pH unless activated by factor(s) stored in azurophil granules. Whereas ARG1 exocytosis was induced by TNF-α or ionomycin, only the latter mediated the release of both granules, resulting in extracellular ARG enzyme activity at physiological pH. Furthermore, after fractionation of the different classes of granules, only the mixture of gelatinase and azurophil granules resulted in ARG1 activity at physiological pH. The use of protease inhibitors indicated the involvement of a PMSF- and leupeptin-susceptible serine protease in ARG1 processing and activation. Finally, the supernatant of viable PMNs undergoing frustrated phagocytosis, which mediates gelatinase and azurophil granule release, inhibited T cell proliferation through ARG-dependent mechanisms. In vivo, high ARG1 concentrations and increased ARG enzyme activity, sufficient to inhibit T cell proliferation, were observed in synovial fluids from RA. These findings suggest that PMNs, recruited at sites of immune complex deposition, induce ARG1-dependent immune suppression through concomitant exocytosis of gelatinase and azurophil granules.


Subject(s)
Arginase/metabolism , Arthritis, Rheumatoid/metabolism , Cytoplasmic Granules/metabolism , Exocytosis/physiology , Neutrophils/metabolism , Synovial Fluid/metabolism , T-Lymphocytes/metabolism , Arginine/metabolism , Azure Stains , Blotting, Western , Case-Control Studies , Cell Proliferation , Enzyme-Linked Immunosorbent Assay , Female , Gelatinases/metabolism , Humans , Lymphocyte Activation , Male , Middle Aged , Phagocytosis/physiology
20.
J Mol Med (Berl) ; 89(5): 493-504, 2011 May.
Article in English | MEDLINE | ID: mdl-21221514

ABSTRACT

Dbl is the prototype of a large family of GDP-GTP exchange factors for small GTPases of the Rho family. In vitro, Dbl is known to activate Rho, Rac, and Cdc42 and to induce a transformed phenotype in murine fibroblasts. We previously reported that Dbl-null mice are viable and fertile but display defective dendrite elongation of distinct subpopulations of cortical neurons, suggesting a role of Dbl in controlling dendritic growth. To gain deeper insights into the role of Dbl in development and disease, we attempted a knock-in approach to create an endogenous allele that encodes a missense-mutation-mediated loss of function in the DH domain. We generated, by gene targeting technology, a mutant mouse strain by inserting a mutagenized human proto-Dbl cDNA clone expressing only the Dbl N terminus regulatory sequence at the starting codon of murine exon 1. Animals were monitored over a 21-month period, and necropsy specimens were collected for histological examination and immunohistochemistry analysis. Dbl knock-in mice are viable and did not manifest either decreased reproductive performances or gross developmental phenotype but revealed a reduced lifespan compared to wild-type (w.t.) mice and showed, with aging, a B cell lymphoproliferation that often has features of a frank diffuse large B cell lymphoma. Moreover, Dbl knock-in male mice displayed an increased incidence of lung adenoma compared to w.t. mice. These data indicate that Dbl is a tumor susceptibility gene in mice and that loss of function of Dbl DH domain by genetic missense mutations is responsible for induction of diffuse large B cell lymphoma.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Lymphoma, B-Cell/metabolism , Lymphoma, Large B-Cell, Diffuse/metabolism , Animals , Blotting, Western , Guanine Nucleotide Exchange Factors/genetics , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Lymphoma, B-Cell/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , Male , Mice , NIH 3T3 Cells , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...