Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 19 de 19
1.
Biomedicines ; 9(12)2021 Dec 16.
Article En | MEDLINE | ID: mdl-34944746

Characterization of tumor associated lymphocytes (TILs) in tumor lesions is important to obtain a clear definition of their prognostic value and address novel therapeutic opportunities. In this work, we examined the presence of T helper (Th)17 lymphocytes in cutaneous melanoma. We performed an immunohistochemical analysis of a small cohort of primary melanomas, retrospectively selected. Thereafter, we isolated TILs from seven freshly surgically removed melanomas and from three basal cell carcinomas (BCC), as a comparison with a non-melanoma skin cancer known to retain a high amount of Th17 cells. In both studies, we found that, differently from BCC, melanoma samples showed a lower percentage of Th17 lymphocytes. Additionally, TIL clones could not be induced to differentiate towards the Th17 phenotype in vitro. The presence or absence of Th17 cells did not correlate with any patient characteristics. We only observed a lower amount of Th17 cells in samples from woman donors. We found a tendency towards an association between expression by melanoma cells of placenta growth factor, angiogenic factors able to induce Th17 differentiation, and presence of Th17 lymphocytes. Taken together, our data indicate the necessity of a deeper analysis of Th17 lymphocytes in cutaneous melanoma before correlating them with prognosis or proposing Th17-cell based therapeutic approaches.

2.
Eur J Immunol ; 45(3): 922-31, 2015 Mar.
Article En | MEDLINE | ID: mdl-25487261

Interleukin-17 (IL-17) and IL-22 have been reported to play critical roles in autoimmunity and inflammation but information about their role in cancer is limited. In this study, we investigated the role of IL-17 and IL-22 in the progression of human skin basal-cell carcinoma (BCC) and squamous-cell carcinoma (SCC). We found that both tumor types are infiltrated with an high number of IL-17(+) and IL-22(+) T lymphocytes, as demonstrated by immunohistochemistry and by FACS analysis performed on peritumoral T-cell lines isolated from skin biopsies. In vitro studies demonstrated that proliferation and migration of the BCC- and SCC-cell lines M77015 and CAL27 were increased by IL-17 and IL-22. Moreover, IL-17, alone or in combination with TNF-α, was able to induce the production of two cytokines important for tumor progression, IL-6 and IL-8, in CAL27. We also showed that IL-17 upregulated NF-κB signaling, while IL-22 activated the STAT3 pathway and the antiapoptotic AKT protein in M77015 and CAL27. Finally, in vivo experiments demonstrated that IL-17 and IL-22 enhanced tumor growth in nude mice injected with CAL27. Altogether, our findings indicate that high levels of IL-22 and IL-17 in the BCC and SCC microenvironment promote tumor progression.


Carcinoma, Basal Cell/immunology , Carcinoma, Squamous Cell/immunology , Interleukin-17/immunology , Interleukins/immunology , Neoplasm Proteins/immunology , Skin Neoplasms/immunology , Tumor Microenvironment/immunology , Animals , Carcinoma, Basal Cell/pathology , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Female , Humans , Interleukin-6/immunology , Interleukin-8/immunology , Male , Mice , Mice, Nude , NF-kappa B/immunology , Signal Transduction/immunology , Skin Neoplasms/pathology , Interleukin-22
3.
Cell Cycle ; 10(10): 1679-89, 2011 May 15.
Article En | MEDLINE | ID: mdl-21508668

Absence of p53 expression or expression of mutant p53 (mtp53) are common in human cancers and are associated with increased cancer resistance to chemo- and radiotherapy. Therefore, significant efforts towards pharmaceutical reactivation of defective p53 pathways are underway. We previously reported that, in HIPK2 knockdown background, p53 undergoes misfolding with inhibition of DNA binding and transcriptional activities that correlate with increased chemoresistance, and that zinc rescues wild-type p53 activity. Zinc has a crucial role in the biology of p53, in that p53 binds to DNA through a structurally complex domain stabilized by zinc atom. In this study, we explored the role of zinc in p53 reactivation in mutant p53-expressing cancer cells. We found that zinc re-established chemosensitivity in breast cancer SKBR3 (expressing R175H mutation) and glioblastoma U373MG (expressing R273H mutation) cell lines. Biochemical studies showed that zinc partly induced the transition of mutant p53 protein (reactive to conformation-sensitive PAb240 antibody for mutant conformation) into a functional conformation (reactive to conformation-sensitive PAb1620 antibody for wild-type conformation). Zinc-mediated p53 reactivation also reduced the mtp53/p73 interaction restoring both wtp53 and p73 binding to target gene promoters by ChIP assay with in vivo induction of wtp53 target gene expression, which rendered mutant p53 cells more prone to drug killing in vitro. Finally, zinc administration in U373MG tumor xenografts increased drug-induced tumor regression in vivo, which correlated with increased wild-type p53 protein conformation. These results show that the use of zinc might restore drug sensitivity and inhibit tumor growth by reactivating mutant p53.


Antineoplastic Agents/pharmacology , Tumor Suppressor Protein p53/chemistry , Zinc/physiology , Animals , Antineoplastic Agents/therapeutic use , Apoptosis , Breast Neoplasms/drug therapy , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA-Binding Proteins/metabolism , Female , Humans , Mice , Mice, Nude , Mutation , Nuclear Proteins/metabolism , Protein Conformation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Transplantation, Heterologous , Tumor Protein p73 , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Zinc/pharmacology
4.
Aging (Albany NY) ; 3(1): 33-43, 2011 Jan.
Article En | MEDLINE | ID: mdl-21248371

Many human diseases are characterized by the development of tissue hypoxia. Hypoxia-inducible factor (HIF) is a transcription factor that regulates fundamental cellular processes in response to changes in oxygen concentration, such as angiogenesis, survival, and alterations in metabolism. The levels of HIF-1α subunit are increased in most solid tumors not only by low oxygen but also by growth factors and oncogenes and correlate with patient prognosis and treatment failure. The link between HIF-1α and apoptosis, a major determinant of cancer progression and treatment outcome, is poorly understood. Here we show that HIF-1α protects against drug-induced apoptosis by antagonizing the function of the tumor suppressor p53. HIF-1α upregulation induced proteasomal degradation of homeodomain-interacting protein kinase-2 (HIPK2), the p53 apoptotic activator. Inhibition of HIF-1α by siRNA, HIF-1α-dominant negative or by zinc re-established the HIPK2 levels and the p53-mediated chemosensitivity in tumor cells. Our findings identify a novel circuitry between HIF-1α and p53, and provide a paradigm for HIPK2 dictating cell response to antitumor therapies.


Apoptosis/physiology , Carrier Proteins/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , Carrier Proteins/genetics , Cell Line, Tumor , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/metabolism , Signal Transduction/physiology , Tumor Suppressor Protein p53/genetics , Zinc/metabolism
5.
PLoS One ; 5(12): e15048, 2010 Dec 13.
Article En | MEDLINE | ID: mdl-21179202

BACKGROUND: Hypoxia inducible factor-1α (HIF-1α) is responsible for the majority of HIF-1-induced gene expression changes under hypoxia and for the "angiogenic switch" during tumor progression. HIF-1α is often upregulated in tumors leading to more aggressive tumor growth and chemoresistance, therefore representing an important target for antitumor intervention. We previously reported that zinc downregulated HIF-1α levels. Here, we evaluated the molecular mechanisms of zinc-induced HIF-1α downregulation and whether zinc affected HIF-1α also in vivo. METHODOLOGY/PRINCIPAL FINDINGS: Here we report that zinc downregulated HIF-1α protein levels in human prostate cancer and glioblastoma cells under hypoxia, whether induced or constitutive. Investigations into the molecular mechanisms showed that zinc induced HIF-1α proteasomal degradation that was prevented by treatment with proteasomal inhibitor MG132. HIF-1α downregulation induced by zinc was ineffective in human RCC4 VHL-null renal carcinoma cell line; likewise, the HIF-1αP402/P564A mutant was resistant to zinc treatment. Similarly to HIF-1α, zinc downregulated also hypoxia-induced HIF-2α whereas the HIF-1ß subunit remained unchanged. Zinc inhibited HIF-1α recruitment onto VEGF promoter and the zinc-induced suppression of HIF-1-dependent activation of VEGF correlated with reduction of glioblastoma and prostate cancer cell invasiveness in vitro. Finally, zinc administration downregulated HIF-1α levels in vivo, by bioluminescence imaging, and suppressed intratumoral VEGF expression. CONCLUSIONS/SIGNIFICANCE: These findings, by demonstrating that zinc induces HIF-1α proteasomal degradation, indicate that zinc could be useful as an inhibitor of HIF-1α in human tumors to repress important pathways involved in tumor progression, such as those induced by VEGF, MDR1, and Bcl2 target genes, and hopefully potentiate the anticancer therapies.


Down-Regulation , Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Zinc/pharmacology , Antineoplastic Agents/pharmacology , Brain Neoplasms/metabolism , Cell Line , Cell Line, Tumor , Disease Progression , Glioblastoma/metabolism , Humans , In Vitro Techniques , Male , Prostatic Neoplasms/metabolism , Proteasome Endopeptidase Complex/pharmacology , Vascular Endothelial Growth Factor A/biosynthesis
6.
FEBS Lett ; 584(19): 4253-8, 2010 Oct 08.
Article En | MEDLINE | ID: mdl-20849851

Homeodomain-interacting protein kinase-2 (HIPK2) is a crucial regulator of p53 apoptotic function by phosphorylating serine 46 (Ser46) in response to DNA damage. In tumors with wild-type p53, its tumor suppressor function is often impaired by MDM2 overexpression that targets p53 for proteasomal degradation. Likewise, MDM2 targets HIPK2 for protein degradation impairing p53-apoptotic function. Here we report that zinc antagonised MDM2-induced HIPK2 degradation as well as p53 ubiquitination. The zinc inhibitory effect on MDM2 activity leads to HIPK2-induced p53Ser46 phosphorylation and p53 pro-apoptotic transcriptional activity. These results suggest that zinc derivatives are potential molecules to target the MDM2-induced HIPK2/p53 inhibition.


Apoptosis/physiology , Carrier Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Carrier Proteins/genetics , Cell Line , Cell Line, Tumor , Down-Regulation/drug effects , Genes, p53 , HeLa Cells , Humans , Models, Biological , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/genetics , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Signal Transduction/drug effects , Tumor Suppressor Protein p53/genetics , Ubiquitination/drug effects , Zinc/pharmacology
7.
PLoS One ; 5(4): e10171, 2010 Apr 14.
Article En | MEDLINE | ID: mdl-20418953

BACKGROUND: Homeodomain interacting protein kinase 2 (HIPK2) is an evolutionary conserved serine/threonine kinase whose activity is fundamental in maintaining wild-type p53 function, thereby controlling the destiny of cells when exposed to DNA damaging agents. We recently reported an altered conformational state of p53 in tissues from patients with Alzheimer's Disease (AD) that led to an impaired and dysfunctional response to stressors. METHODOLOGY/PRINCIPAL FINDINGS: Here we examined the molecular mechanisms underlying the impairment of p53 activity in two cellular models, HEK-293 cells overexpressing the amyloid precursor protein and fibroblasts from AD patients, starting from recent findings showing that p53 conformation may be regulated by HIPK2. We demonstrated that beta-amyloid 1-40 induces HIPK2 degradation and alters HIPK2 binding activity to DNA, in turn regulating the p53 conformational state and vulnerability to a noxious stimulus. Expression of HIPK2 was analysed by western blot experiments, whereas HIPK2 DNA binding was examined by chromatin immunoprecipitation analysis. In particular, we evaluated the recruitment of HIPK2 onto some target promoters, including hypoxia inducible factor-1alpha and metallothionein 2A. CONCLUSIONS/SIGNIFICANCE: These results support the existence of a novel amyloid-based pathogenetic mechanism in AD potentially leading to the survival of injured dysfunctional cells.


Alzheimer Disease/pathology , Carrier Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/chemistry , Amyloid beta-Protein Precursor , Carrier Proteins/physiology , Cell Survival , Cells, Cultured , DNA/metabolism , Humans , Promoter Regions, Genetic , Protein Binding , Protein Folding , Protein Serine-Threonine Kinases/physiology
8.
Cell Cycle ; 9(7): 1270-5, 2010 Apr 01.
Article En | MEDLINE | ID: mdl-20234185

Oncosuppressor p53 is often inactivated by either mutations or deregulation of regulatory proteins. These include the homeodomain-interacting protein kinase 2 (HIPK2) that, by phosphorylating p53 at Ser46 modulates p53 response to DNA damage by inducing pro-apoptotic transcription. There is compelling evidence that HIPK2 is also involved in the response to hypoxia by acting as co-suppressor of hypoxia inducible factor 1α (HIF-1α), a major factor in cancer progression that activates the transcription of genes involved in angiogenesis, glucose metabolism and invasion. Hence conditions that induce HIPK2 deregulation would end up in a multifactorial response leading to tumor chemoresistance by affecting p53 activity on one hand and to angiogenesis and cell proliferation by affecting HIF-1 activity on the other hand. For these reasons, HIPK2 protein is a promising target for anti-cancer therapies. HIPK2 can be inhibited by hypoxia. In this respect, we have recently shown that hypoxia-driven HIPK2 downregulation is not irreversible. We found that, zinc supplementation reactivates the hypoxia-inhibited HIPK2, leading to repression of the HIF-1 pathway and restoration of p53Ser46 apoptotic activity. Here, we discuss about these findings and the potential relevance of zinc supplementation to chemotherapy in cancer treatment. The results will be also discussed in light of recent findings showing that cancer treatment with antiangiogenic agents may result in hypoxia and selection of cancer cells with increased tumor aggressiveness and metastasis.


Carrier Proteins/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Carrier Proteins/genetics , Cell Hypoxia/genetics , Cell Hypoxia/physiology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Models, Biological , Neoplasms/genetics , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Protein p53/genetics
9.
Free Radic Biol Med ; 48(10): 1338-46, 2010 May 15.
Article En | MEDLINE | ID: mdl-20171273

HIPK2 is a stress-induced kinase and a transcriptional corepressor that functionally cooperates with p53 to suppress cancer. Activation of the p53 proapoptotic function requires a cascade of phosphorylations and acetylations, and HIPK2 takes part in both modifications in that it phosphorylates p53 Ser46 and induces p53 Lys382 acetylation. Here, to further investigate the role of HIPK2 in p53 activation, we started with the finding that HIPK2 inhibition upregulated Nox1, a homolog of the catalytic subunit of the superoxide-generating NADPH oxidase, involved in tumor progression and ROS production. We found that Nox1 inhibited p53 Lys382 acetylation, which is a target of SIRT1 deacetylase, and impaired p53 proapoptotic transcriptional activity. By the use of either small interfering RNAs to target SIRT1 or the SIRT1 inhibitor nicotinamide we found that Nox1-dependent inhibition of p53 transcriptional activity was SIRT1-dependent. Thus, Nox1 was unable to inhibit p53 when coexpressed with a SIRT1 deacetylase-defective mutant (SIRT1HY), suggesting a link between Nox1 and SIRT1 activity. Finally, recovery of HIPK2 function downregulated Nox1 expression with rescue of p53 Lys382 acetylation and p53 activity. Together, our findings indicate that Nox1 upregulation may activate SIRT1 and inhibit p53 and that Lys382 is important for p53 proapoptotic function.


Breast Neoplasms/metabolism , Carcinoma/metabolism , Colonic Neoplasms/metabolism , Lung Neoplasms/metabolism , NADPH Oxidases/biosynthesis , Tumor Suppressor Protein p53/metabolism , Acetylation/drug effects , Apoptosis/drug effects , Apoptosis/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma/genetics , Carcinoma/pathology , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cell Line, Tumor , Cloning, Molecular , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mutant Proteins/genetics , Mutant Proteins/metabolism , NADPH Oxidase 1 , NADPH Oxidases/genetics , Niacinamide/pharmacology , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/genetics , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/genetics , Sirtuin 1/metabolism , Transcriptional Activation/drug effects , Transcriptional Activation/genetics , Tumor Suppressor Protein p53/genetics
10.
J Mol Recognit ; 23(4): 343-51, 2010.
Article En | MEDLINE | ID: mdl-19941302

Azurin, a bacterial protein, can be internalized in cancer cells and induce apoptosis. Such anticancer effect is coupled to the formation of a complex with the tumour-suppressor p53. The mechanism by which azurin stabilizes p53 and the binding sites of their complex are still under investigation. It is also known that the predominant mechanism for p53 down-regulation implies its association to Mdm2, the main ubiquitin ligase affecting its stability. However, the p53/Mdm2 interaction, occurring at the level of both their N-terminal domains, has been characterized so far by experiments involving only partial domains of these proteins. The relevance of the p53/Mdm2 complex as a possible target of the anticancer therapies requires a deeper study of this complex as made up of the two entire proteins. Moreover, the apparent antagonist action of azurin against Mdm2, with respect of p53 regulation, might suggest the possibility that azurin binds p53 at the same site of Mdm2, preventing in such a way p53 and Mdm2 from association and thus p53 from degradation. By following the interaction of the two entire proteins by atomic force spectroscopy, we have assessed the formation of a specific complex between p53 and Mdm2. We found for it a binding strength and a dissociation rate constant typical of dynamical protein-protein interactions and we observed that azurin, even if capable to bind p53, does not compete with Mdm2 for the same binding site on p53. The formation of the p53/Mdm2/azurin ternary complex might suggest an alternative anti-cancer mechanism adopted by azurin.


Azurin/metabolism , Microscopy, Atomic Force , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Azurin/genetics , Humans , Models, Biological , Protein Binding , Proto-Oncogene Proteins c-mdm2/genetics , Tumor Suppressor Protein p53/genetics
11.
Mol Cancer ; 8: 85, 2009 Oct 14.
Article En | MEDLINE | ID: mdl-19828042

BACKGROUND: Activation of p53-mediated gene transcription is a critical cellular response to DNA damage and involves a phosphorylation-acetylation cascade of p53. The discovery of differences in the response to different agents raises the question whether some of the p53 oncosuppressor functions might be exerted by different posttranslational modifications. Stress-induced homeodomain-interacting protein kinase-2 (HIPK2) phosphorylates p53 at serine-46 (Ser46) for p53 apoptotic activity; p53 acetylation at different C-terminus lysines including p300-mediated lysine-382 (Lys382) is also required for full activation of p53 transcriptional activity. The purpose of the current study was to evaluate the interplay among HIPK2, p300, and p53 in p53 acetylation and apoptotic transcriptional activity in response to drug by using siRNA interference, p300 overexpression or deacetylase inhibitors, in cancer cells. RESULTS: Knockdown of HIPK2 inhibited both adriamycin-induced Ser46 phosphorylation and Lys382 acetylation in p53 protein; however, while combination of ADR and zinc restored Ser46 phosphorylation it did not recover Lys382 acetylation. Chromatin immunoprecipitation studies showed that HIPK2 was required in vivo for efficient p300/p53 co-recruitment onto apoptotic promoters and that both p53 modifications at Ser46 and Lys382 were necessary for p53 apoptotic transcription. Thus, p53Lys382 acetylation in HIPK2 knockdown as well as p53 apoptotic activity in response to drug could be rescued by p300 overexpression. Similar effect was obtained with the Sirt1-inhibitor nicotinamide. Interestingly trichostatin A (TSA), the inhibitor of histone deacetylase complexes (HDAC) did not have effect, suggesting that Sirt1 was the deacetylase involved in p53 deacetylation in HIPK2 knockdown. CONCLUSION: These results reveal a novel role for HIPK2 in activating p53 apoptotic transcription. Our results indicate that HIPK2 may regulate the balance between p53 acetylation and deacetylation, by stimulating on one hand co-recruitment of p300 and p53Lys382 on apoptotic promoters and on the other hand by inhibiting Sirt1 deacetylase activity. We attempted to reactivate p53 apoptotic transcriptional activity by rescuing both Ser46 and Lys382 modification in response to drug. Our data propose combination strategies for the treatment of tumors with dysfunctional p53 and/or HIPK2 that include classical chemotherapy with pharmacological or natural agents such as Sirt1-deacetylase inhibitors or zinc, respectively.


Apoptosis/genetics , Carrier Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Acetylation/drug effects , Apoptosis/drug effects , Cell Line, Tumor , Doxorubicin/pharmacology , E1A-Associated p300 Protein/metabolism , Gene Deletion , Gene Knockdown Techniques , Humans , Hydroxamic Acids/pharmacology , Lysine/metabolism , Niacinamide/pharmacology , Phosphorylation/drug effects , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Sirtuin 1/metabolism , Transcription, Genetic/drug effects , Zinc/pharmacology , bcl-2-Associated X Protein/metabolism
12.
PLoS One ; 4(8): e6819, 2009 Aug 28.
Article En | MEDLINE | ID: mdl-19714248

BACKGROUND: The tumor suppressor homeodomain-interacting protein kinase-2 (HIPK2) by phosphorylating serine 46 (Ser46) is a crucial regulator of p53 apoptotic function. HIPK2 is also a transcriptional co-repressor of hypoxia-inducible factor-1alpha (HIF-1alpha) restraining tumor angiogenesis and chemoresistance. HIPK2 can be deregulated in tumors by several mechanisms including hypoxia. Here, we sought to target hypoxia by restoring HIPK2 function and suppressing HIF-1alpha, in order to provide evidence for the involvement of both HIPK2 and p53 in counteracting hypoxia-induced chemoresistance. METHODOLOGY/PRINCIPAL FINDINGS: Upon exposure of colon and lung cancer cells to hypoxia, by either low oxygen or cobalt, HIPK2 function was impaired allowing for increased HIF-1alpha expression and inhibiting the p53-apoptotic response to drug. Cobalt suppressed HIPK2 recruitment onto HIF-1alpha promoter. Hypoxia induced expression of the p53 target MDM2 that downregulates HIPK2, thus MDM2 inhibition by siRNA restored the HIPK2/p53Ser46 response to drug. Zinc supplementation to hypoxia-treated cells increased HIPK2 protein stability and nuclear accumulation, leading to restoration of HIPK2 binding to HIF-1alpha promoter, repression of MDR1, Bcl2, and VEGF genes, and activation of the p53 apoptotic response to drug. Combination of zinc and ADR strongly suppressed tumor growth in vivo by inhibiting HIF-1 pathway and upregulating p53 apoptotic target genes. CONCLUSIONS/SIGNIFICANCE: We show here for the first time that hypoxia-induced HIPK2 deregulation was counteracted by zinc that restored HIPK2 suppression of HIF-1 pathway and reactivated p53 apoptotic response to drug, underscoring the potential use of zinc supplementation in combination with chemotherapy to address hypoxia and improve tumor treatment.


Carrier Proteins/metabolism , Cell Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , Antineoplastic Agents/pharmacology , Cobalt/pharmacology , Doxorubicin/pharmacology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Promoter Regions, Genetic , Proto-Oncogene Proteins c-mdm2/genetics , Transcription, Genetic/drug effects , Zinc/pharmacology
13.
Mol Cancer ; 8: 1, 2009 Jan 07.
Article En | MEDLINE | ID: mdl-19128456

BACKGROUND: Homeodomain-interacting protein kinase-2 (HIPK2), a transcriptional co-repressor with apoptotic function, can affect hypoxia-inducible factor 1 (HIF-1) transcriptional activity, through downmodulation of its HIF-1alpha subunit, in normoxic condition. Under hypoxia, a condition often found in solid tumors, HIF-1alpha is activated to induce target genes involved in chemoresistance, inhibition of apoptosis and tumor progression. Here, we investigated whether the HIPK2 overexpression could downregulate HIF-1alpha expression and activity in tumor cells treated with hypoxia-mimicking condition, and evaluated whether HIPK2-dependent downregulation of HIF-1alpha could sensitize chemoresistant tumor cells to adriamycin (ADR)-induced apoptosis. METHODS: Tumor cell lines carrying wild-type p53, siRNA p53, or mutant p53 were overexpressed with HIPK2 (full length or catalytic inactive mutant) and treated with cobalt chloride (CoCl2) to mimic hypoxia, in the presence or absence of ADR treatment. HIF-1alpha expression was measured by semiquantitative reverse-transcriptase (RT)-PCR and Western immunoblotting and HIF-1 activity was evaluated by luciferase assay using reporter plasmid containing hypoxia response elements (HREs) upstream of luciferase gene. HIF-1 target genes, including multidrug resistance 1 (MDR1) and the antiapoptotic Bcl2 were determined by RT-PCR. Cell survival and apoptosis were measured by colony assay and cleavage of the caspase-3 substrate PARP, respectively. RESULTS: Overexpression of HIPK2 resulted in downmodulation of cobalt-stabilized HIF-1alpha protein and HIF-1alpha mRNA levels, with subsequent inhibition of HIF-1 transcriptional activity. MDR1 and Bcl-2 gene expression was downmodulated by HIPK2 overexpression in cobalt-treated cells. Inhibition of HIF-1 transcriptional activity was dependent on HIPK2 catalytic activity. HIPK2 overexpression did not induce per se apoptosis of cobalt-treated cells, on the contrary it sensitized cobalt-treated cells to ADR-induced apoptosis, regardless of their p53 status. CONCLUSION: The ability of HIPK2 to restore the apoptosis-inducing potential of chemotherapeutic drug in hypoxia-mimicking condition and therefore to sensitize chemoresistant tumor cells suggests that HIPK2 may induce fundamental alterations in cell signaling pathways, involving or not p53 function. Thus potential use of HIPK2 is promising for cancer treatment by potentiating cytotoxic therapies, regardless of p53 cell status.


Apoptosis/physiology , Carrier Proteins/metabolism , Drug Resistance, Neoplasm/physiology , Gene Expression Regulation, Neoplastic/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Protein Serine-Threonine Kinases/metabolism , Antibiotics, Antineoplastic/pharmacology , Blotting, Western , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Cell Line, Tumor , Doxorubicin/pharmacology , Humans , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Tumor Suppressor Protein p53/metabolism
14.
Exp Cell Res ; 315(1): 67-75, 2009 Jan 01.
Article En | MEDLINE | ID: mdl-18996371

The maintenance of p53 transactivation activity is important for p53 apoptotic function. We have shown that stable knockdown of HIPK2 induces p53 misfolding with inhibition of p53 target gene transcription. In this study we established a lentiviral-based system for doxycyclin (Dox)-induced conditional interference of HIPK2 expression to evaluate the molecular mechanisms involved in p53 deregulation. We found that HIPK2 knockdown induced metallothionein 2A (MT2A) upregulation as assessed by RT-PCR analysis, increased promoter acetylation, and increased promoter luciferase activity. The MT2A upregulation correlated with resistance to Adriamycin (ADR)-driven apoptosis and with p53 inhibition. Thus, acute knockdown of HIPK2 (HIPK2i) induced misfolded p53 protein in MCF7 breast cancer cells and inhibited p53 DNA-binding and transcription activities in response to ADR treatment. Previous works show that MT may modulate p53 activity through zinc exchange. Here, we found that inhibition of MT2A expression by siRNA in the HIPK2i cells restored p53 transcription activity. Similarly zinc supplementation to HIPK2i cells restored p53 transcription activity and drug-induced apoptosis. These data support the notion that MT2A is involved in p53 deregulation and strengthen the possibility that combination of chemotherapy and zinc might be useful to treat tumors with inactive wtp53.


Metallothionein/metabolism , Protein Serine-Threonine Kinases/deficiency , Tumor Suppressor Protein p53/metabolism , Zinc/metabolism , Apoptosis/drug effects , Carrier Proteins/metabolism , Cell Line, Tumor , DNA, Neoplasm/metabolism , Doxorubicin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lentivirus , Phosphorylation/drug effects , Phosphoserine/metabolism , Protein Binding/drug effects , Protein Conformation/drug effects , Protein Folding/drug effects , Protein Serine-Threonine Kinases/metabolism , Transcription, Genetic/drug effects , Transcriptional Activation/drug effects , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics , Up-Regulation/drug effects , Zinc/pharmacology
15.
Biochim Biophys Acta ; 1793(2): 368-77, 2009 Feb.
Article En | MEDLINE | ID: mdl-19046997

HIPK2 has been implicated in restraining tumor progression by more than one mechanism, involving both its catalytic and transcriptional co-repressor functions. Starting from the finding that HIPK2 knockdown by RNA-interference (HIPK2i) induced significant up-regulation of HIF-1alpha mRNA and of its target VEGF in tumor cells, we evaluated the role of HIPK2 in transcriptional regulation of HIF-1alpha. We found that HIPK2 overexpression downmodulated both HIF-1alpha reporter activity and mRNA levels and showed that HIPK2 was bound in vivo to the HIF-1alpha promoter likely in a multiprotein co-repressor complex with histone deacetylase 1 (HDAC1). Thus, the HIF-1alpha promoter was strongly acetylated following HIPK2 knockdown. The HIF-1alpha-dependent VEGF transcription was evaluated by co-transfection of a dominant negative (DN) construct of HIF-1alpha that inhibited VEGF reporter activity induced by HIPK2 knockdown. HIF-1alpha and VEGF up-regulation in HIPK2i cells correlated with increased vascularity of tumor xenografts in vivo and tube formation in HUVEC in vitro. These findings provide the first evidence of HIPK2-mediated transcriptional regulation of HIF-1alpha that might play a critical role in VEGF expression.


Carrier Proteins/metabolism , Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Neoplasms/genetics , Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Transcription, Genetic , Animals , Cell Line, Tumor , Cell Proliferation , Chromatin Assembly and Disassembly/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Neoplasms/blood supply , Neoplasms/enzymology , Neovascularization, Pathologic/genetics , Promoter Regions, Genetic/genetics , Protein Binding , Protein Serine-Threonine Kinases/deficiency , Repressor Proteins/metabolism , Vascular Endothelial Growth Factor A/genetics
16.
J Exp Clin Cancer Res ; 27: 22, 2008 Jul 21.
Article En | MEDLINE | ID: mdl-18644116

BACKGROUND: Homeodomain-interacting protein kinase-2 (HIPK2) plays an essential role in restraining tumor progression as it may regulate, by itself or within multiprotein complexes, many proteins (mainly transcription factors) involved in cell growth and apoptosis. This study takes advantage of the recent finding that HIPK2 may repress the beta-catenin transcription activity. Thus, we investigated whether HIPK2 overexpression may down-regulate vascular endothelial growth factor (VEGF) levels (a beta-catenin target gene) and the role of beta-catenin in this regulation, in order to consider HIPK2 as a tool for novel anti-tumoral therapeutical approaches. METHODS: The regulation of VEGF expression by HIPK2 was evaluated by using luciferase assay with VEGF reporter construct, after overexpression of the beta-catenin transcription factor. Relative quantification of VEGF and beta-catenin mRNAs were assessed by reverse-transcriptase-PCR (RT-PCR) analyses, following HIPK2 overexpression, while beta-catenin protein levels were evaluated by western immunoblotting. RESULTS: HIPK2 overexpression in tumor cells downregulated VEGF mRNA levels and VEGF promoter activity. The VEGF downregulation was partly depending on HIPK2-mediated beta-catenin regulation. Thus, HIPK2 could induce beta-catenin protein degradation that was prevented by cell treatment with proteasome inhibitor MG132. The beta-catenin degradation was dependent on HIPK2 catalytic activity and independent of p53 and glycogen synthase kinase 3beta (GSK-3beta) activities. CONCLUSION: These results suggest that VEGF might be a target of HIPK2, at least in part, through regulation of beta-catenin activity. These findings support the function of HIPK2 as tumor suppressor and hypothesise a role for HIPK2 as antiangiogenic tool in tumor therapy approaches.


Carrier Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Vascular Endothelial Growth Factor A/genetics , Cell Line, Tumor , Down-Regulation , Gene Expression , Humans , Proteasome Endopeptidase Complex/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Transfection , Vascular Endothelial Growth Factor A/metabolism , beta Catenin/genetics , beta Catenin/metabolism
17.
Cancer Res ; 68(10): 3707-14, 2008 May 15.
Article En | MEDLINE | ID: mdl-18483253

About half of cancers sustain mutations in the TP53 gene, whereas the other half maintain a wild-type p53 (wtp53) but may compromise the p53 response because of other alterations. Homeodomain-interacting protein kinase-2 (HIPK2) is a positive regulator of p53 oncosuppressor function. Here, we show, by microarray analysis, that wtp53 lost the target gene activation following stable knockdown of HIPK2 (HIPK2i) in colon cancer cell line. Our data show that the stable knockdown of HIPK2 led to wtp53 misfolding, as detected by p53 immunoprecipitation with conformation-specific antibodies, and that p53 protein misfolding impaired p53 DNA binding and transcription of target genes. We present evidence that zinc supplementation to HIPK2i cells increased p53 reactivity to conformation-sensitive PAb1620 (wild-type conformation) antibody and restored p53 sequence-specific DNA binding in vivo and transcription of target genes in response to Adriamycin treatment. Finally, combination of zinc and Adriamycin suppressed tumor growth in vivo and activated misfolded p53 that induced its target genes in nude mice tumor xenografts derived from HIPK2i cells. Bioinformatics analysis of microarray data from colon cancer patients showed significant association of poor survival with low HIPK2 expression only in tumors expressing wtp53. These results show a critical role of HIPK2 in maintaining the transactivation activity of wtp53 and further suggest that low expression of HIPK2 may impair the p53 function in tumors harboring wtp53.


Carrier Proteins/physiology , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Genes, p53 , Mutation , Protein Serine-Threonine Kinases/physiology , Tumor Suppressor Protein p53/physiology , Animals , Antibiotics, Antineoplastic/pharmacology , Carrier Proteins/genetics , Cell Line, Tumor , Doxorubicin/pharmacology , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Protein Serine-Threonine Kinases/genetics , Transcriptional Activation , Tumor Suppressor Protein p53/genetics
18.
Gynecol Oncol ; 109(3): 403-10, 2008 Jun.
Article En | MEDLINE | ID: mdl-18395248

OBJECTIVE: Chemoresistance, due to inhibition of apoptotic response, is the major reason for the failure of anticancer therapies. HIPK2 regulates p53-apoptotic function via serine-46 (Ser46) phosphorylation and activation of p53 is a key determinant in ovarian cancer cell death. In this study we determined whether HIPK2 overexpression restored apoptotic response in chemoresistant cancer cells. METHODS: Using cisplatin chemosensitive (2008) and chemoresistant (2008C13) ovarian cancer cell lines we compared drug-induced activation of the HIPK2/p53Ser46 apoptotic pathway. The levels of HIPK2, Ser46 phosphorylation, and PARP cleavage were detected by Western blotting. The p53Ser46 apoptotic commitment was evaluated by luciferase assay using the Ser46 specific AIP1 target gene promoter. The apoptotic pathway was detected by caspase-3, -8, and -9 activities. RESULTS: HIPK2 was expressed differently in sensitive versus chemoresistant cells in response to different chemotherapeutic drugs (i.e., cisplatin and adriamycin), though the p53Ser46 apoptotic pathway was not defective in chemoresistant 2008C13 cells. Thus, 2008C13 cells were resistant to cisplatin but sensitive to adriamycin-induced apoptosis through activation of the HIPK2/p53Ser46 pathway. HIPK2 knock-down inhibited the adriamycin-induced apoptosis in 2008C13 cells. Exogenous HIPK2 triggered apoptosis in chemoresistant cells, associated with induction of p53Ser46-target gene AIP1. CONCLUSIONS: HIPK2 is an important regulator of p53 activity in response to a chemotherapeutic drug. These results suggest that different drug-activated pathways may regulate HIPK2 and that HIPK2/p53Ser46 deregulation is involved in chemoresistance. Exogenous HIPK2 might represent a novel therapeutic approach to circumvent inhibition of apoptosis in treatment of chemoresistant ovarian cancers with wtp53.


Apoptosis/drug effects , Apoptosis/physiology , Carrier Proteins/biosynthesis , Cisplatin/pharmacology , Doxorubicin/pharmacology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Protein Serine-Threonine Kinases/biosynthesis , Carrier Proteins/genetics , Carrier Proteins/metabolism , Caspases/metabolism , Cell Line, Tumor , Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/metabolism , Cystadenocarcinoma, Serous/pathology , Drug Resistance, Neoplasm , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
19.
Biochem Biophys Res Commun ; 361(1): 249-55, 2007 Sep 14.
Article En | MEDLINE | ID: mdl-17658469

Homeodomain Interacting Protein Kinase-2 (HIPK2) is a protein with many functions and a modulator of p53 oncosuppressor functions. TP53 is the "guardian of the genome" thus, is the most critical tumor suppressor gene product that inhibits malignant transformation. P53R2 gene is directly induced by p53 in response to DNA damage and is involved in the p53 checkpoint for repairing damaged DNA to block genome instability. Here we wanted to explore the involvement of HIPK2 in damaged-DNA repair by regulating p53-induced p53R2 gene. We show that, induction of p53R2 expression, p53 recruitment onto p53R2 promoter, and its transcriptional activation was strongly impaired by HIPK2 knock-down, in response to drug. The failure of p53-induced p53R2 activation markedly compromised damaged-DNA repair efficiency. Finally, overexpression of exogenous p53 overcame the inability of endogenous p53 to activate p53R2-luc promoter in HIPK2 depleted cells. These data suggest that HIPK2 is involved in damaged-DNA repair taking part in restraining tumor progression, at least in part depending on p53 regulation.


Carrier Proteins/physiology , DNA Damage , DNA Repair , Neoplasms/enzymology , Protein Serine-Threonine Kinases/physiology , Tumor Suppressor Protein p53/metabolism , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Cell Line, Tumor , Humans , Neoplasms/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , RNA Interference , Ribonucleotide Reductases/biosynthesis , Ribonucleotide Reductases/genetics , Transcription, Genetic , Tumor Suppressor Protein p53/antagonists & inhibitors
...