Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Oncoimmunology ; 6(11): e1356151, 2017.
Article in English | MEDLINE | ID: mdl-29147611

ABSTRACT

Autophagy is usually a pro-survival mechanism in cancer cells, especially in the course of chemotherapy, thus autophagy inhibition may enhance the chemotherapy-mediated anti-cancer effect. However, since autophagy is strongly involved in the immunogenicity of cell death by promoting ATP release, its inhibition may reduce the immune response against tumors, negatively influencing the overall outcome of chemotherapy. In this study, we evaluated the in vitro and in vivo anti-cancer effect of curcumin (CUR) against Her2/neu overexpressing breast cancer cells (TUBO) in the presence or in the absence of the autophagy inhibitor chloroquine (CQ). We found that TUBO cell death induced by CUR was increased in vitro by CQ and slightly in vivo in nude mice. Conversely, CQ counteracted the Cur cytotoxic effect in immune competent mice, as demonstrated by the lack of in vivo tumor regression and the reduction of overall mice survival as compared with CUR-treated mice. Immunohistochemistry analysis revealed the presence of a remarkable FoxP3 T cell infiltrate within the tumors in CUR/CQ treated mice and a reduction of T cytotoxic cells, as compared with single CUR treatment. These findings suggest that autophagy is important to elicit anti-tumor immune response and that autophagy inhibition by CQ reduces such response also by recruiting T regulatory (Treg) cells in the tumor microenvironment that may be pro-tumorigenic and might counteract CUR-mediated anti-cancer effects.

2.
Cell Death Dis ; 7(6): e2280, 2016 06 30.
Article in English | MEDLINE | ID: mdl-27362798

ABSTRACT

Hyperglycemia, the condition of high blood glucose, is typical of diabetes and obesity and represents a significant clinical problem. The relationship between hyperglycemia and cancer risk has been established by several studies. Moreover, hyperglycemia has been shown to reduce cancer cell response to therapies, conferring resistance to drug-induced cell death. Therefore, counteracting the negative effects of hyperglycemia may positively improve the cancer cell death induced by chemotherapies. Recent studies showed that zinc supplementation may have beneficial effects on glycemic control. Here we aimed at evaluating whether ZnCl2 could counteract the high-glucose (HG) effects and consequently restore the drug-induced cancer cell death. At the molecular level we found that the HG-induced expression of genes known to be involved in chemoresistance (such as HIF-1α, GLUT1, and HK2 glycolytic genes, as well as NF-κB activity) was reduced by ZnCl2 treatment. In agreement, the adryamicin (ADR)-induced apoptotic cancer cell death was significantly impaired by HG and efficiently re-established by ZnCl2 cotreatment. Mechanistically, the ADR-induced c-Jun N-terminal kinase/stress-activated protein kinase (JNK/SAPK) phosphorylation, inhibited by HG, was efficiently restored by ZnCl2. The JNK involvement in apoptotic cell death was assessed by the use of JNK dominant-negative expression vector that indeed impaired the ZnCl2 ability to restore drug-induced cell death in HG condition. Altogether, these findings indicate that ZnCl2 supplementation efficiently restored the drug-induced cancer cell death, inhibited by HG, by both sustaining JNK activation and counteracting the glycolytic pathway.


Subject(s)
Chlorides/pharmacology , Doxorubicin/pharmacology , Glucose/toxicity , Zinc Compounds/pharmacology , Cell Death/drug effects , Cell Hypoxia/drug effects , Cell Hypoxia/genetics , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Deoxyglucose/metabolism , Down-Regulation/drug effects , Glucose Transporter Type 1/metabolism , Glycolysis/drug effects , Glycolysis/genetics , HCT116 Cells , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Phosphorylation/drug effects , Protein Transport/drug effects
3.
Cell Death Dis ; 6: e1621, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25633290

ABSTRACT

Mitogen-activated protein kinase kinase 3 (MAP2K3, MKK3) is a member of the dual specificity protein kinase group that belongs to the MAP kinase kinase family. This kinase is activated by mitogenic or stress-inducing stimuli and participates in the MAP kinase-mediated signaling cascade, leading to cell proliferation and survival. Several studies highlighted a critical role for MKK3 in tumor progression and invasion, and we previously identified MKK3 as transcriptional target of mutant (mut) p53 to sustain cell proliferation and survival, thus rendering MKK3 a promising target for anticancer therapies. Here, we found that targeting MKK3 with RNA interference, in both wild-type (wt) and mutp53-carrying cells, induced endoplasmic reticulum stress and autophagy that, respectively, contributed to stabilize wtp53 and degrade mutp53. MKK3 depletion reduced cancer cell proliferation and viability, whereas no significant effects were observed in normal cellular context. Noteworthy, MKK3 depletion in combination with chemotherapeutic agents increased tumor cell response to the drugs, in both wtp53 and mutp53 cancer cells, as demonstrated by enhanced poly (ADP-ribose) polymerase cleavage and reduced clonogenic ability in vitro. In addition, MKK3 depletion reduced tumor growth and improved biological response to chemotherapeutic in vivo. The overall results indicate MKK3 as a novel promising molecular target for the development of more efficient anticancer treatments in both wtp53- and mutp53-carrying tumors.


Subject(s)
MAP Kinase Kinase 3/antagonists & inhibitors , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/enzymology , Animals , Autophagy , Cell Line , Cell Proliferation , Cell Survival , Endoplasmic Reticulum Stress , Female , Humans , MAP Kinase Kinase 3/metabolism , Mice, Nude , Mutant Proteins/metabolism , Neoplasms/pathology , Protein Stability , RNA, Small Interfering/metabolism , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
4.
Cell Death Dis ; 5: e1271, 2014 May 29.
Article in English | MEDLINE | ID: mdl-24874727

ABSTRACT

TP53, one of the most important oncosuppressors, is frequently mutated in cancer. Several p53 mutant proteins escape proteolytic degradation and are highly expressed in an aberrant conformation often acquiring pro-oncogenic activities that promote tumor progression and resistance to therapy. Therefore, it has been vastly proposed that reactivation of wild-type (wt) function(s) from mutant p53 (mutp53) may have therapeutic significance. We have previously reported that Zn(II) restores a folded conformation from mutp53 misfolding, rescuing wild-type (wt) p53/DNA-binding and transcription activities. However, whether Zn(II) affects mutp53 stability has never been investigated. Here we show that a novel Zn(II) compound induced mutp53 (R175H) protein degradation through autophagy, the proteolytic machinery specifically devoted to clearing misfolded proteins. Accordingly, pharmacological or genetic inhibition of autophagy prevented Zn(II)-mediated mutp53H175 degradation as well as the ability of the Zn(II) compound to restore wtp53 DNA-binding and transcription activity from this mutant. By contrast, inhibition of the proteasome failed to do so, suggesting that autophagy is the main route for p53H175 degradation. Mechanistically, Zn(II) restored the wtp53 ability to induce the expression of the p53 target gene DRAM (damage-regulated autophagy modulator), a key regulator of autophagy, leading to autophagic induction. Accordingly, inhibition of wtp53 transactivation by pifithrin-α (PFT-α) impaired both autophagy and mutp53H175 degradation induced by curcumin-based zinc compound (Zn(II)-curc). Viewed together, our results uncover a novel mechanism employed by Zn(II)-curc to reactivate mutp53H175, which involves, at least in part, induction of mutp53 degradation via wtp53-mediated autophagy.


Subject(s)
Autophagy/drug effects , Down-Regulation/drug effects , Tumor Suppressor Protein p53/metabolism , Zinc Compounds/pharmacology , Cell Line, Tumor , Curcumin/chemistry , HCT116 Cells , Humans , Microtubule-Associated Proteins/metabolism , Mutation , RNA Interference , RNA, Small Interfering/metabolism , Transcriptional Activation/drug effects , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics , Zinc Compounds/chemistry
5.
Cell Death Dis ; 4: e730, 2013 Jul 18.
Article in English | MEDLINE | ID: mdl-23868063

ABSTRACT

Heat-shock protein (HSP) 70 is aberrantly expressed in different malignancies and has a cancer-specific cell-protective effect. As such, it has emerged as a promising target for anticancer therapy. In this study, the effect of the HSP70-specific inhibitor (PES), also Pifitrin-µ, on primary effusion lymphoma (PEL) cell viability was analyzed. PES treatment induced a dose- and time-dependent cytotoxic effect in BC3 and BCBL1 PEL cells by inducing lysosome membrane permeabilization, relocation of cathepsin D in the cytosol, Bid cleavage, mitochondrial depolarization with release and nuclear translocation of apoptosis-activating factor. The PES-induced cell death in PEL cells was characterized by the appearance of Annexin-V/propidium iodide double-positive cells from the early times of treatment, indicating the occurrence of an additional type of cell death other than apoptosis, which, accordingly, was not efficiently prevented by the pan-caspase inhibitor Z-VAD-fmk. Conversely, PES-induced cell death was robustly reduced by pepstatin A, which inhibits Bid and caspase 8 processing. In addition, PES was responsible for a block of the autophagic process in PEL cells. Finally, we found that PES-induced cell death has immunogenic potential being able to induce dendritic cell activation.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy , Cathepsin D/metabolism , HSP70 Heat-Shock Proteins/antagonists & inhibitors , Lysosomes/drug effects , Sulfonamides/pharmacology , Active Transport, Cell Nucleus , Apoptosis Inducing Factor/metabolism , BH3 Interacting Domain Death Agonist Protein/metabolism , Cathepsin D/antagonists & inhibitors , Cell Line, Tumor , Cell Survival/drug effects , Dendritic Cells/drug effects , Dendritic Cells/immunology , Drug Screening Assays, Antitumor , HSP70 Heat-Shock Proteins/metabolism , Humans , Lymphoma, Primary Effusion , Lysosomes/enzymology , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Pepstatins/pharmacology , Permeability , Protease Inhibitors/pharmacology
6.
Cell Death Dis ; 4: e639, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23703384

ABSTRACT

Tumor cell tolerance to nutrient deprivation can be an important factor for tumor progression, and may depend on deregulation of both oncogenes and oncosuppressor proteins. Homeodomain-interacting protein kinase 2 (HIPK2) is an oncosuppressor that, following its activation by several cellular stress, induces cancer cell death via p53-dependent or -independent pathways. Here, we used genetically matched human RKO colon cancer cells harboring wt-HIPK2 (HIPK2(+/+)) or stable HIPK2 siRNA interference (siHIPK2) to investigate in vitro whether HIPK2 influenced cell death in glucose restriction. We found that glucose starvation induced cell death, mainly due to c-Jun NH2-terminal kinase activation, in HIPK2(+/+)cells compared with siHIPK2 cells that did not die. (1)H-nuclear magnetic resonance quantitative metabolic analyses showed a marked glycolytic activation in siHIPK2 cells. However, treatment with glycolysis inhibitor 2-deoxy-D-glucose induced cell death only in HIPK2(+/+) cells but not in siHIPK2 cells. Similarly, siGlut-1 interference did not re-establish siHIPK2 cell death under glucose restriction, whereas marked cell death was reached only after zinc supplementation, a condition known to reactivate misfolded p53 and inhibit the pseudohypoxic phenotype in this setting. Further siHIPK2 cell death was reached with zinc in combination with autophagy inhibitor. We propose that the metabolic changes acquired by cells after HIPK2 silencing may contribute to induce resistance to cell death in glucose restriction condition, and therefore be directly relevant for tumor progression. Moreover, elimination of such a tolerance might serve as a new strategy for cancer therapy.


Subject(s)
Apoptosis/drug effects , Carrier Proteins/metabolism , Deoxyglucose/pharmacology , Protein Serine-Threonine Kinases/metabolism , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Deoxyglucose/therapeutic use , Glucose Transporter Type 1/antagonists & inhibitors , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Metabolome , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , RNA Interference , RNA, Small Interfering/metabolism , Zinc/pharmacology
7.
Oncogene ; 29(31): 4378-87, 2010 Aug 05.
Article in English | MEDLINE | ID: mdl-20514025

ABSTRACT

The p53 protein is the most studied tumor suppressor and the p53 pathway has been shown to mediate cellular stress responses that are disrupted when cancer develops. After DNA damage, p53 is activated as transcription factor to directly induce the expression of target genes involved in cell-cycle arrest, DNA repair, senescence and, importantly, apoptosis. Post-translational modifications of p53 are essential for the activation of p53 and for selection of target genes. The tumor suppressor homeodomain-interacting protein kinase-2 (HIPK2) is a crucial regulator of p53 apoptotic function by phosphorylating its N-terminal serine 46 (Ser46) and facilitating Lys382 acetylation at the C-terminus. HIPK2 is activated by numerous genotoxic agents and can be deregulated in tumors by several conditions including hypoxia. Recent findings suggest that HIPK2 active/inactive protein can affect p53 function in multiple and unexpected ways. This makes p53 as well as HIPK2 interesting targets for cancer therapy. Hence, understanding the role of HIPK2 as p53 activator may provide important insights in the process of tumor progression, and may also serve as the crucial point in the diagnostic and therapeutical aspects of cancer.


Subject(s)
Carrier Proteins/physiology , Neoplasms/therapy , Protein Serine-Threonine Kinases/physiology , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Carrier Proteins/metabolism , Humans , Models, Biological , Neoplasms/genetics , Neoplasms/metabolism , Phosphorylation , Protein Conformation , Protein Processing, Post-Translational/physiology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/physiology
9.
Cell Death Differ ; 11(6): 596-607, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15150542

ABSTRACT

Stimulation of the Ras/MAPK cascade can either activate p53 and promote replicative senescence and apoptosis, or degrade p53 and promote cell survival. Here we show that p53 can directly counteract the Ras/MAPK signaling by inactivating ERK2/MAPK. This inactivation is due to a caspase cleavage of the ERK2 protein and contributes to p53-mediated growth arrest. We found that in Ras-transformed cells, growth arrest induced by p53, but not p21(Waf1), is associated with a strong reduction in ERK2 activity, phosphorylation, and protein half-life, and with the appearance of caspase activity. Likewise, DNA damage-induced cell cycle arrest correlates with p53-dependent ERK2 downregulation and caspase activation. Furthermore, caspase inhibitors or expression of a caspase-resistant ERK2 mutant interfere with ERK2 cleavage and restore proliferation in the presence of p53 activation, indicating that caspase-mediated ERK2 degradation contributes to p53-induced growth arrest. These findings strongly point to ERK2 as a novel p53 target in growth suppression.


Subject(s)
Caspases/metabolism , Cell Division/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Antibiotics, Antineoplastic/pharmacology , Caspase 3 , Cell Division/drug effects , Cell Line, Transformed , DNA Damage/physiology , Down-Regulation , Doxorubicin/pharmacology , Mice
10.
Anticancer Res ; 20(5B): 3497-502, 2000.
Article in English | MEDLINE | ID: mdl-11131653

ABSTRACT

It has recently been shown that tumor cells can retain the ability to undergo senescence, while the capacity of bypassing senescence has been associated with tumor progression. In this report, we showed that v-Ha-ras-mediated transformation of already immortal C2C12 myoblasts can be associated with senesence, in a low amount during in vitro passages and, to a higher extent, affer cellular stress (cell culture alkalinkation), or DNA damage (doxorubicin treatment). The capacity to undergo replicative senescence is associated with a strong increase of wt-p53 transcriptional activity and p21WAF1 up-regulation. These biochemical activities are down-modulated in the cells that evade the massive replicative senescence after stressing stimuli. Altogether, these findings show that active ras can cause senescence during the transformation of already immortal cells in associaton with p53/p21WAF1 pathway activation and support the hypothesis that p53/p21WAF1 functional activity is important in maintaining the integrity of the senescence pathway during cellular transformation.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cellular Senescence/genetics , Cyclins/physiology , Gene Expression Regulation/physiology , Genes, ras , Tumor Suppressor Protein p53/physiology , Animals , Cell Division/genetics , Cell Line, Transformed , Cell Transformation, Neoplastic/metabolism , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , Cyclins/genetics , Genes, p53 , Mice , Muscles/cytology , Muscles/metabolism , Muscles/physiology , Signal Transduction/genetics , Stress, Physiological/genetics , Stress, Physiological/pathology , Transcriptional Activation , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics , Up-Regulation
11.
J Gene Med ; 2(1): 11-21, 2000.
Article in English | MEDLINE | ID: mdl-10765501

ABSTRACT

BACKGROUND: Expression of exogenous wild-type p53 (wt-p53) protein in tumor cells can suppress the transformed phenotype whereas it does not apparently induce detrimental effects in non-transformed cells. This observation may provide a molecular basis for p53-mediated gene therapy of p53-sensitive cancers without the need for tumor targeting. METHODS: To understand the molecular mechanisms responsible for this different behavior in tumor versus normal cells, biochemical and functional analyses of exogenous wt-p53 protein were performed on non-transformed C2C12 myoblasts and their transformed counterparts, the C2-ras cells. RESULTS: The exogenous wt-p53 protein, which induced persistent growth arrest only in transformed C2-ras cells, was shown to be significantly more stable in transformed than in non-transformed cells. This different stability was due to different p53 proteolytic degradation. Moreover, constitutively, exogenous wt-p53 protein was found to be transcriptionally active only in C2-ras cells but it could also be activated in C2C12 cells by genotoxic damage. CONCLUSIONS: Non-transformed C2C12 cells present regulatory system(s) which control the expression and the activity of exogenously expressed wt-p53 protein probably through degradation and maintenance in a latent form. This regulatory system is lost/inactivated upon transformation.


Subject(s)
Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Nuclear Proteins , Tumor Suppressor Protein p53/genetics , Adenoviridae/genetics , Animals , Blotting, Western , Cell Division , Cell Line , Cell Line, Transformed , Cell Transformation, Neoplastic/metabolism , Chloramphenicol O-Acetyltransferase/genetics , Chloramphenicol O-Acetyltransferase/metabolism , Cysteine Endopeptidases/pharmacology , DNA Damage/physiology , DNA, Recombinant/genetics , Doxorubicin/pharmacology , Gene Transfer Techniques , Genes, Reporter/genetics , Genes, ras/genetics , Genetic Vectors/genetics , Leupeptins/pharmacology , Mice , Mice, Inbred C3H , Multienzyme Complexes/pharmacology , Muscles/cytology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Proteasome Endopeptidase Complex , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2 , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/metabolism , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
12.
Gene Ther ; 6(6): 1064-72, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10455409

ABSTRACT

In this article, we investigated the effect induced by the reintroduction of wild-type p53 (wt-p53) protein on BCNU sensitivity in the ADF glioblastoma line. Using a wt-p53 recombinant adenovirus (Ad-p53), we demonstrated that exogenous wt-p53 expression was able to increase the sensitivity to BCNU in ADF cells. Interestingly, this effect was more evident when Ad-p53 infection was performed after BCNU treatment compared with the opposite sequence. To understand the biological basis of these different behaviors, we analyzed the cell cycle of the differently treated cells. We found that Ad-p53 infection induced a persistent accumulation of cells in the G0/G1 phase while, as expected, BCNU induced a block in the G2-M phase. Ad-p53-->BCNU sequence did not significantly modify the cell cycle profile in respect of Ad-p53 infected cells. In contrast, BCNU-->Ad-p53 sequence provoked G2-M arrest similar to that observed after treatment with BCNU alone, but prevented the later recovery of the cells through the cell cycle, by driving the cells to apoptotic death. These results demonstrate that the administration sequence is important to increase drug sensitivity. To generalize the phenomenon observed on ADF line, the antiproliferative effect of the two different schedules was analyzed on other glioblastoma lines (A172, CRS-A2, U373MG) with different BCNU sensitivity and p53 status. The data obtained confirm that the wt-p53 gene transfer enhances BCNU sensitivity in glioblastoma cells depending on the administration sequence.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/therapy , Carmustine/therapeutic use , Genetic Therapy/methods , Glioblastoma/therapy , Tumor Suppressor Protein p53/genetics , Adenoviridae , Drug Administration Schedule , Drug Resistance, Neoplasm , Genes, p53/genetics , Humans , Tumor Cells, Cultured
13.
Br J Cancer ; 78(12): 1541-6, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9862562

ABSTRACT

We recently reported that rhabdomyosarcoma cell lines express and secrete interleukin 15 (IL-15), a tightly regulated cytokine with IL-2-like activity. To test whether the p53-impaired function that is frequently found in this tumour type could play a role in the IL-15 production, wild-type p53 gene was transduced in the human rhabdomyosarcoma cell line RD (which harbours a mutated p53 gene), and its effect on proliferation and expression of IL-15 was studied. Arrest of proliferation was induced by wild-type p53; increased proportions of G1-arrested cells and of apoptotic cells were observed. A marked down-modulation of IL-15 expression, at both the mRNA and protein level, was found in p53-transduced cells. Because a direct effect of IL-15 on normal muscle cells has been reported, the presence of IL-15 membrane receptors was studied by cytofluorometric analysis. Rhabdomyosarcoma cells showed IL-15 membrane receptors, which are down-modulated by wild-type p53 transfected gene. In conclusion, wild-type p53 transduction in human rhabdomyosarcoma cells induces the down-modulation of both IL-15 production and IL-15 receptor expression.


Subject(s)
Genes, p53 , Interleukin-15/metabolism , Receptors, Interleukin-2/metabolism , Rhabdomyosarcoma/metabolism , Cell Division/genetics , Cell Lineage , Down-Regulation , Humans , Receptors, Interleukin-15 , Reverse Transcriptase Polymerase Chain Reaction , Rhabdomyosarcoma/pathology , Transcription, Genetic , Transduction, Genetic , Transfection , Tumor Cells, Cultured
14.
Exp Dermatol ; 6(6): 321-7, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9412821

ABSTRACT

Psoriasis is histologically characterized by hyperkeratosis and papillomatosis with elongated vessels in the upper dermis. In order to evaluate the role of gelatinases in remodelling psoriatic skin in this study we examined the production of the 72-kDa (gelatinase A), 92-kDa collagenase (gelatinase B) and their tissue inhibitors TIMP-2 and TIMP-1. A total of 19 patients affected by different types of psoriasis were included in this study. An immunohistochemical study on cryosections was performed using antibodies to 72-kDa gelatinase, 92-kDa gelatinase, TIMP-1, TIMP-2, laminin, collagen types I, III, IV, VII. mRNA expression for gelatinases and their inhibitors were also analyzed by reverse transcriptase polymerase chain reaction (RT-PCR). In 14 of 19 patients there was a positivity in 92-kDa protein expression in keratinocytes. The 92-kDa gelatinase protein was also present in the upper dermis with prevalence around blood vessels. In 15 of 19 patients the 72-kDa was localized in the upper dermis, almost exclusively in the papillary dermis but absent in epidermis. TIMP-1 and TIMP-2 were both negative in all cases in immunoperoxidase and RT-PCR. Using RT-PCR we show that the 72-kDa mRNA is expressed exclusively in the dermis, on the contrary the 92-kDa was present in epidermis and dermis. Type I, III, IV and VII collagens did not show any alteration or disruption. Overexpression and production of gelatinases without inhibitory effects suggest a role of these proteins in remodelling the psoriatic skin probably inducing the typical histological pattern of papillomatosis.


Subject(s)
Gelatinases/genetics , Psoriasis/enzymology , Psoriasis/genetics , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-2/genetics , Enzyme Inhibitors/metabolism , Epidermis/chemistry , Gene Expression Regulation , Gene Expression Regulation, Enzymologic , Humans , Immunohistochemistry , Polymerase Chain Reaction , Psoriasis/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Skin/metabolism , Skin/pathology
15.
Endocrinology ; 135(6): 2479-87, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7988435

ABSTRACT

Primary cultures of prepubertal rat Sertoli cells secrete two major tissue inhibitors of metalloproteinases: TIMP-1 (M(r) 28K) and TIMP-2 (M(r) 21 K). FSH stimulated Sertoli cell TIMP-1 and TIMP-2 activity in a time- and dose-dependent manner and also stimulated TIMP-1 and TIMP-2 protein and messenger RNA levels. These effects were mimicked by the cAMP analog, 8-bromo-cAMP, and the phosphodiesterase inhibitor 3-isobutyl-1-methylxanthine. The protein kinase C activating phorbol ester phorbol myristate acetate (TPA) stimulated TIMP-1 but not TIMP-2 activity and messenger RNA levels. Cycloheximide and actinomycin-D inhibited basal TIMP-1 and TIMP-2 activity and inhibited the ability of FSH, 8-bromo-cAMP, and TPA to stimulate TIMP activity. The protein kinase A (PKA) inhibitor AMP Rp isomer did not affect basal TIMP-1 and TIMP-2 activity or TPA-stimulated TIMP-1 activity. However, the PKA inhibitor markedly reduced FSH and 3-isobutyl-1-methylxanthine stimulation of TIMP-1 and TIMP-2 activity. FSH, 8-bromo-cAMP, and TPA stimuli induced DNA binding complexes capable of binding to a TIMP-1 AP-1 site consensus sequence oligonucleotide. The AP-1 site binding complex(es) induced by all three treatments reacted with antibodies directed broadly against fos and jun protooncogene families and against the specific family members c-fos, junB, and junD but not c-jun proteins. Constitutive cAMP response element binding activity capable of binding an artificial cAMP response element binding site oligonucleotide was demonstrated in Sertoli cell nuclear extracts. This activity was minimally modulated by FSH, 8-bromo-cAMP, or TPA treatment. In summary, Sertoli cells secrete TIMP-1 and TIMP-2 that can be coordinately up-regulated by FSH through a cAMP, PKA-dependent pathway. a convergence of TPA, FSH, and cAMP mediated signals in prepubertal Sertoli cells may occur with the induction of specific AP-1 site binding complex(es) containing jun and fos proteins. Our data suggest that FSH stimulation of TIMP-2 expression may be regulated independently to that of TIMP-1. We propose that the ability of FSH to stimulate Sertoli cell TIMP activity suggests a central role for this hormone in the control of extracellular matrix turnover during testicular development at the level of metalloproteinase inhibition.


Subject(s)
Follicle Stimulating Hormone/pharmacology , Glycoproteins/antagonists & inhibitors , Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Sertoli Cells/metabolism , Animals , Base Sequence , Binding Sites , Blotting, Northern , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/metabolism , Glycoproteins/genetics , Male , Metalloendopeptidases/antagonists & inhibitors , Molecular Sequence Data , Oligonucleotide Probes/genetics , Proteins/genetics , RNA, Messenger/metabolism , Rats , Sexual Maturation , Stimulation, Chemical , Tissue Inhibitor of Metalloproteinase-2 , Tissue Inhibitor of Metalloproteinases
16.
J Cell Physiol ; 156(2): 235-46, 1993 Aug.
Article in English | MEDLINE | ID: mdl-8344982

ABSTRACT

It has been proposed that proteases are important in endothelial cell behavior. We examined the contribution of the gelatinase/type IV collagenase system in an in vitro model of endothelial differentiation. Human umbilical vein endothelial cells rapidly align and form networks of tubes when cultured on a basement membrane preparation, Matrigel. Zymograms of culture supernates demonstrate a 72-kD and a 92-kD gelatinase activity; the cells produce most of the 72-kD gelatinase, whereas the 92-kD activity is derived entirely from the Matrigel. Addition of antibodies against type IV gelatinase/collagenase decreases the area of the tube network. Both tissue inhibitors of metalloproteinases, TIMP-1 and TIMP-2, similarly decrease tube formation when added to cultures. Conversely, exogenous recombinant 72-kD gelatinase increases tube-forming activity. The effects of the anti-gelatinase antibodies and the TIMPs are not additive. Inhibition by either antibodies or TIMPs is greatest when they are added at culture initiation, suggesting that the protease activity is important in the early steps of morphogenesis. However, culture of the cells on Matrigel does not increase early expression of mRNA for the 72-kD gelatinase. Expression of message for the enzyme actually decreases during the course of the assay, while transcription of mRNAs for TIMPs increases, further supporting the concept that collagenases facilitate an early event in tube formation. These data demonstrate that gelatinase/type IV collagenase activity is important in endothelial cell morphogenesis on Matrigel, and suggest a role for collagenases in formation of new capillaries in vivo.


Subject(s)
Collagenases/physiology , Endothelium, Vascular/cytology , Glycoproteins/pharmacology , Metalloendopeptidases/antagonists & inhibitors , Neoplasm Proteins/pharmacology , Amino Acid Sequence , Antibodies/immunology , Antibodies/pharmacology , Basement Membrane/enzymology , Blotting, Northern , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Collagen/pharmacology , Collagenases/genetics , Collagenases/immunology , Drug Combinations , Drug Interactions , Electrophoresis, Polyacrylamide Gel , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Humans , Laminin/pharmacology , Matrix Metalloproteinase 9 , Molecular Sequence Data , Molecular Weight , Morphogenesis/physiology , Proteoglycans/pharmacology , RNA, Messenger/analysis , RNA, Messenger/genetics , Recombinant Proteins/pharmacology , Time Factors , Tissue Inhibitor of Metalloproteinase-2 , Tissue Inhibitor of Metalloproteinases , Umbilical Veins/cytology
17.
Int J Cancer ; 53(6): 988-93, 1993 Apr 01.
Article in English | MEDLINE | ID: mdl-8473057

ABSTRACT

The ultrastructural pattern of the anti-tumor response elicited by interleukin-4 (IL-4) was investigated by using a spontaneous mammary adenocarcinoma (TS/A) unable to elicit protective immunity in syngeneic BALB/c mice as suggested by a variety of preimmunization-challenge experiments. A subcutaneous lethal challenge of TS/A tumor cells was inhibited in a significant number of BALB/c mice receiving recombinant murine IL-4 injected daily for 10 days around the tumor-draining lymph node. Tumor rejection was mainly the result of direct membrane and cytoplasmic damage to tumor cells by eosinophils, neutrophils and macrophages that deeply penetrated the proliferating tumor mass. Lymphocytes and fibroblasts participated in the reaction by interacting with tumor cells, granulocytes and each other. The most frequent cell interactions in the peri- and intra-tumoral areas and in the tumor-draining lymph nodes are illustrated. The efficiency with which the IL-4-activated reaction leads to tumor inhibition and induction of a T-lymphocyte-dependent tumor-specific immune memory appears to depend on interactions between distinct leukocytes.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/ultrastructure , Interleukin-4/pharmacology , Lymphocytes, Tumor-Infiltrating/physiology , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/ultrastructure , Adenocarcinoma/pathology , Animals , Cell Division/drug effects , Dose-Response Relationship, Drug , Extracellular Matrix/physiology , Female , Injections, Intralymphatic , Isotonic Solutions/pharmacology , Lymph Nodes/cytology , Lymph Nodes/drug effects , Lymph Nodes/physiology , Lymphocytes, Tumor-Infiltrating/drug effects , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Necrosis , Recombinant Proteins/pharmacology
18.
Cancer Lett ; 64(1): 31-7, 1992 May 30.
Article in English | MEDLINE | ID: mdl-1596874

ABSTRACT

A comparable pattern of morphological neuronal differentiation was induced in the human neuroblastoma cell line SMS-KCNR by treatment with either retinoic acid (RA) or exogenous laminin (LM). LM expression and synthesis by SMS-KCNR was increased upon RA treatment which involved the cell bound, rather than the secreted protein. These data suggest an involvement of LM in the neuroblastoma differentiation process manifested both as an ability of LM to induce a morphological neuronal differentiation and as a selective control on LM metabolism during RA induced neuronal differentiation.


Subject(s)
Laminin/biosynthesis , Neuroblastoma/metabolism , Tretinoin/pharmacology , Cell Differentiation/drug effects , Collagen/biosynthesis , Fibronectins/biosynthesis , Fluorescent Antibody Technique , Humans , Laminin/pharmacology , Microscopy, Phase-Contrast , Tumor Cells, Cultured/drug effects
19.
J Neurosurg Sci ; 34(1): 1-6, 1990.
Article in English | MEDLINE | ID: mdl-2401908

ABSTRACT

A group of 10 rats underwent spinal trauma by epidural spinal compression according to Tator's procedure. After 4 months from injury cortical and spinal SEP were recorded, and the spinal injured tract was examined by electronic microscope. We studied morphologic and functional changes showed after some months behind the trauma. This report demonstrates and compares the varying sensitivity of cortical and spinal SEP for revealing functional spinal damage; ultrastructural investigations testify to the existence of reparative processes of the nervous tissue.


Subject(s)
Evoked Potentials, Somatosensory , Spinal Cord Injuries/physiopathology , Animals , Electric Stimulation , Female , Microscopy, Electron , Rats , Rats, Inbred Strains , Reaction Time , Sciatic Nerve/physiology , Somatosensory Cortex/physiopathology , Spinal Cord/physiopathology , Spinal Cord/ultrastructure , Spinal Cord Injuries/pathology
20.
J Immunol ; 143(7): 2415-21, 1989 Oct 01.
Article in English | MEDLINE | ID: mdl-2778321

ABSTRACT

The ability of NK cells to synthesize and secrete fibronectin (FN), an extracellular matrix glycoprotein which plays a key role in many biologic processes including cellular adhesion, morphology, cytoskeletal organization, cell migration, and invasiveness, was studied. By using affinity-purified polyclonal antibodies directed against human cellular or plasma FN, the presence of FN was evidentiated on Percoll-purified rat large granular lymphocyte or on a large granular lymphocyte tumor cell line (CRC) by flow cytometry and immunoelectron microscopy. Its expression increased after NK cell activation by poly I:C administration. Biochemical analysis by immunoprecipitation and SDS-PAGE indicated that FN was associated to cell surface and secreted in the supernatant in a molecular form similar to that of FN from L929 fibroblasts. In an attempt to understand the role of FN in the NK cell function, we found that an antibody against human plasma FN and its F(ab')2 fragment inhibited NK cytotoxicity against YAC-1 target at the effector cell level. Inhibition occurred at the postbinding level, because F(ab')2 anti-FN inhibited induction of phosphatidylinositol hydrolysis by YAC-1 target cells, whereas binding to target cells was not affected. The possible role of FN in the NK cytotoxic function is suggested.


Subject(s)
Cytotoxicity, Immunologic , Fibronectins/biosynthesis , Killer Cells, Natural/metabolism , Animals , Binding Sites, Antibody , Binding, Competitive , Fibronectins/immunology , Fibronectins/physiology , Immune Sera/pharmacology , Immunoglobulin Fab Fragments/physiology , Killer Cells, Natural/immunology , Lymphocyte Activation , Male , Poly I-C/pharmacology , Rats , Rats, Inbred F344
SELECTION OF CITATIONS
SEARCH DETAIL
...