Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Clin Immunol ; 251: 109638, 2023 06.
Article in English | MEDLINE | ID: mdl-37149118

ABSTRACT

According to epidemiological research, skin autoimmune diseases are more prevalent among black Americans. We postulated that pigment-producing melanocytes may contribute to local immune regulation in the microenvironment. We examined murine epidermal melanocytes in vitro to determine the role of pigment production in immune responses mediated by dendritic cell (DC) activation. Our study revealed that darkly pigmented melanocytes produce more IL-3 and the pro-inflammatory cytokines, IL-6 and TNF-α, and consequently induce plasmacytoid DC (pDC) maturation. Additionally, we demonstrate that low pigment-associated fibromodulin (FMOD) interferes with cytokine secretion and subsequent pDC maturation.


Subject(s)
Cytokines , Interleukin-3 , Humans , Animals , Mice , Interleukin-3/metabolism , Interleukin-3/pharmacology , Fibromodulin/metabolism , Cytokines/metabolism , Pigmentation , Dendritic Cells
2.
J Inflamm Res ; 15: 4515-4526, 2022.
Article in English | MEDLINE | ID: mdl-35966006

ABSTRACT

Introduction: Epidemiological studies have associated pigment production with protection against certain human diseases. In contrast to African Americans, European descendants are more likely to suffer from angiogenesis-dependent and inflammatory diseases, such as wet age-related macular degeneration (ARMD) and ulcerative colitis (UC), respectively. Methods: In a mouse model of dextran sulfate sodium (DSS)-induced acute colitis, the effect of fibromodulin (FMOD) depletion was examined on colitis severity. Results: In this study, albino mice that produce high levels of FMOD developed less severe acute colitis compared with mice lacking in FMOD as assessed by clinical symptoms and histopathological changes. FMOD depletion affected the expression of tight junction proteins, contributing to the destruction of the epithelial barrier. Furthermore, this study revealed a stronger inflammatory response after DSS treatment in the absence of FMOD, where FMOD depletion led to an increase in activated T cells, plasmacytoid dendritic cells (pDCs), and type I interferon (IFN) production. Discussion: These findings point to FMOD as a potential biomarker of disease severity in UC among light-skinned individuals of European descent.

3.
Clin Immunol ; 241: 109055, 2022 08.
Article in English | MEDLINE | ID: mdl-35640789

ABSTRACT

Skin pigmentation has been linked to the development, prevalence, and severity of several immune-mediated diseases such as SLE. Here, we asked whether fibromodulin (FMOD), which is highly expressed in skin with light complexion, can explain the known differences in the magnitude of inflammation. C57 mice with different levels of pigmentation and FMOD were injected with human lupus serum to induce skin inflammation. Histopathologic studies revealed that black C57 FMOD+/+ that produce low levels of FMOD and white C57 FMOD -/- mice develop more severe inflammation compared with white FMOD +/+ mice. This study also revealed that dark pigmentation and FMOD deletion correlates with the increased numbers of Langerhans cells. Altogether, we identify low pigmentation and FMOD are linked to low severity of inflammation and approaches to promote FMOD expression should offer clinical benefit.


Subject(s)
Fibromodulin , Inflammation , Melanocytes , Skin , Animals , Fibromodulin/metabolism , Humans , Inflammation/metabolism , Lupus Erythematosus, Systemic , Mice , Skin/metabolism , Skin/pathology , Skin Pigmentation
4.
Front Oncol ; 11: 696532, 2021.
Article in English | MEDLINE | ID: mdl-34381718

ABSTRACT

Forkhead box protein M1 (FOXM1) is a crucial regulator of cancer development and chemoresistance. It is often overexpressed in acute myeloid leukemia (AML) and is associated with poor survival and reduced efficacy of cytarabine therapy. Molecular mechanisms underlying high FOXM1 expression levels in malignant cells are still unclear. Here we demonstrate that AKT and FOXM1 constitute a positive autoregulatory loop in AML cells that sustains high activity of both pro-oncogenic regulators. Inactivation of either AKT or FOXM1 signaling results in disruption of whole loop, coordinated suppression of FOXM1 or AKT, respectively, and similar transcriptomic changes. AML cells with inhibited AKT activity or stable FOXM1 knockdown display increase in HOXA genes expression and BCL2L1 suppression that are associated with prominent sensitization to treatment with Bcl-2 inhibitor venetoclax. Taken together, our data indicate that AKT and FOXM1 in AML cells should not be evaluated as single independent regulators but as two parts of a common FOXM1-AKT positive feedback circuit. We also report for the first time that FOXM1 inactivation can overcome AML venetoclax resistance. Thus, targeting FOXM1-AKT loop may open new possibilities in overcoming AML drug resistance and improving outcomes for AML patients.

5.
Cell Death Dis ; 12(7): 704, 2021 07 14.
Article in English | MEDLINE | ID: mdl-34262016

ABSTRACT

FOXM1 transcription factor is an oncogene and a master regulator of chemoresistance in multiple cancers. Pharmacological inhibition of FOXM1 is a promising approach but has proven to be challenging. We performed a network-centric transcriptomic analysis to identify a novel compound STL427944 that selectively suppresses FOXM1 by inducing the relocalization of nuclear FOXM1 protein to the cytoplasm and promoting its subsequent degradation by autophagosomes. Human cancer cells treated with STL427944 exhibit increased sensitivity to cytotoxic effects of conventional chemotherapeutic treatments (platinum-based agents, 5-fluorouracil, and taxanes). RNA-seq analysis of STL427944-induced gene expression changes revealed prominent suppression of gene signatures characteristic for FOXM1 and its downstream targets but no significant changes in other important regulatory pathways, thereby suggesting high selectivity of STL427944 toward the FOXM1 pathway. Collectively, the novel autophagy-dependent mode of FOXM1 suppression by STL427944 validates a unique pathway to overcome tumor chemoresistance and improve the efficacy of treatment with conventional cancer drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Drug Resistance, Neoplasm/drug effects , Forkhead Box Protein M1/antagonists & inhibitors , Gene Expression Profiling , Neoplasms/drug therapy , Cell Line, Tumor , Forkhead Box Protein M1/genetics , Forkhead Box Protein M1/metabolism , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Protein Stability , Protein Transport , Proteolysis , RNA-Seq , Transcriptome
6.
PLoS One ; 16(5): e0251121, 2021.
Article in English | MEDLINE | ID: mdl-33983985

ABSTRACT

Several angiogenesis-dependent diseases, including age-related macular degeneration and infantile hemangioma, display differential prevalence among Black, as compared to White individuals. Although socioeconomic status and genetic architecture have been suggested as explaining these differences, we have recently shown that pigment production per se might be involved. For example, we have shown that the extracellular protein fibromodulin is a pro-angiogenic factor highly secreted by melanocytes in White but not Black individuals. Still, additional pigment-dependent angiogenic factors and their molecular mechanisms remain to be identified. Understanding the contribution of pigmentation to angiogenesis in health and disease is essential for precision medicine of angiogenesis-dependent diseases with racial disparity. Toward that goal, we compared the transcriptomes of Black and White individuals in three tissues with angiogenic activity, namely artery, whole blood, and skin. We identified several differentially expressed angiogenesis pathways, including artery morphogenesis, regulation of endothelial cell chemotaxis, and cellular response to vascular endothelial growth factor stimulus. We then demonstrated that the expression of key genes in these pathways is directly modulated by the degree of pigmentation. We further identified the precise pigment production pathway controlling the expression of these genes, namely melanocortin 1 receptor (MC1R) signaling. These results demonstrate pigment-mediated regulation of angiogenesis-related pathways and their driver genes across human tissues.


Subject(s)
Melanocytes/metabolism , Neovascularization, Physiologic/genetics , Neovascularization, Physiologic/physiology , Animals , Arteries/metabolism , Black People/genetics , Blood/metabolism , Databases, Genetic , Female , Gene Expression/genetics , Gene Expression Regulation/genetics , Humans , Male , Melanocytes/physiology , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/immunology , Organ Specificity/genetics , Receptor, Melanocortin, Type 1/genetics , Receptor, Melanocortin, Type 1/metabolism , Skin/metabolism , Transcriptome/genetics , White People/genetics
7.
JCI Insight ; 3(15)2018 08 09.
Article in English | MEDLINE | ID: mdl-30089730

ABSTRACT

Acute myeloid leukemia (AML) patients with NPM1 mutations demonstrate a superior response to standard chemotherapy treatment. Our previous work has shown that these favorable outcomes are linked to the cytoplasmic relocalization and inactivation of FOXM1 driven by mutated NPM1. Here, we went on to confirm the important role of FOXM1 in increased chemoresistance in AML. A multiinstitution retrospective study was conducted to link FOXM1 expression to clinical outcomes in AML. We establish nuclear FOXM1 as an independent clinical predictor of chemotherapeutic resistance in intermediate-risk AML in a multivariate analysis incorporating standard clinicopathologic risk factors. Using colony assays, we show a dramatic decrease in colony size and numbers in AML cell lines with knockdown of FOXM1, suggesting an important role for FOXM1 in the clonogenic activity of AML cells. In order to further prove a potential role for FOXM1 in AML chemoresistance, we induced an FLT3-ITD-driven myeloid neoplasm in a FOXM1-overexpressing transgenic mouse model and demonstrated significantly higher residual disease after standard chemotherapy. This suggests that constitutive overexpression of FOXM1 in this model induces chemoresistance. Finally, we performed proof-of-principle experiments using a currently approved proteasome inhibitor, ixazomib, to target FOXM1 and demonstrated a therapeutic response in AML patient samples and animal models of AML that correlates with the suppression of FOXM1 and its transcriptional targets. Addition of low doses of ixazomib increases sensitization of AML cells to chemotherapy backbone drugs cytarabine and the hypomethylator 5-azacitidine. Our results underscore the importance of FOXM1 in AML progression and treatment, and they suggest that targeting it may have therapeutic benefit in combination with standard AML therapies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Resistance, Neoplasm/genetics , Forkhead Box Protein M1/metabolism , Leukemia, Myeloid, Acute/drug therapy , Proteasome Inhibitors/pharmacology , Aged , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Boron Compounds/pharmacology , Boron Compounds/therapeutic use , Cell Line, Tumor , Cell Nucleus/metabolism , Drug Resistance, Neoplasm/drug effects , Female , Forkhead Box Protein M1/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Glycine/analogs & derivatives , Glycine/pharmacology , Glycine/therapeutic use , Humans , Leukemia, Myeloid, Acute/genetics , Male , Mice , Middle Aged , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleophosmin , Primary Cell Culture , Proteasome Inhibitors/therapeutic use , Retrospective Studies , Treatment Failure , Xenograft Model Antitumor Assays
8.
Cell Death Dis ; 9(2): 84, 2018 01 24.
Article in English | MEDLINE | ID: mdl-29367668

ABSTRACT

Honokiol is a natural product and an emerging drug for a wide variety of malignancies, including hematopoietic malignancies, sarcomas, and common epithelial tumors. The broad range of activity of honokiol against numerous malignancies with diverse genetic backgrounds suggests that honokiol is inhibiting an activity that is common to multiple malignancies. Oncogenic transcription factor FOXM1 is one of the most overexpressed oncoproteins in human cancer. Here we found that honokiol inhibits FOXM1-mediated transcription and FOXM1 protein expression. More importantly, we found that honokiol's inhibitory effect on FOXM1 is a result of binding of honokiol to FOXM1. This binding is specific to honokiol, a dimerized allylphenol, and was not observed in compounds that either were monomeric allylphenols or un-substituted dihydroxy phenols. This indicates that both substitution and dimerization of allylphenols are required for physical interaction with FOXM1. We thus demonstrate a novel and specific mechanism for FOXM1 inhibition by honokiol, which partially may explain its anticancer activity in cancer cells.


Subject(s)
Biphenyl Compounds/pharmacology , Forkhead Box Protein M1/antagonists & inhibitors , Lignans/pharmacology , Animals , Biphenyl Compounds/chemistry , Cell Line, Tumor , Down-Regulation/drug effects , Humans , Lignans/chemistry , Mice , Proteasome Inhibitors/pharmacology , Transcriptional Activation/drug effects
9.
J Biol Chem ; 291(1): 142-8, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26559972

ABSTRACT

The oncogenic transcription factor FOXM1 is overexpressed in the majority of human cancers, and it is a potential target for anticancer therapy. We identified proteasome inhibitors as the first type of drugs that target FOXM1 in cancer cells. Here we found that HSP90 inhibitor PF-4942847 and heat shock also suppress FOXM1. The common effector, which was induced after treatment with proteasome and HSP90 inhibitors or heat shock, was the molecular chaperone HSP70. We show that HSP70 binds to FOXM1 following proteotoxic stress and that HSP70 inhibits FOXM1 DNA-binding ability. Inhibition of FOXM1 transcriptional autoregulation by HSP70 leads to the suppression of FOXM1 protein expression. In addition, HSP70 suppression elevates FOXM1 expression, and simultaneous inhibition of FOXM1 and HSP70 increases the sensitivity of human cancer cells to anticancer drug-induced apoptosis. Overall, we determined the unique and novel mechanism of FOXM1 suppression by proteasome inhibitors.


Subject(s)
Forkhead Transcription Factors/metabolism , HSP70 Heat-Shock Proteins/metabolism , Proteasome Inhibitors/pharmacology , Stress, Physiological/drug effects , Apoptosis/drug effects , Cell Line, Tumor , DNA/metabolism , Down-Regulation/drug effects , Forkhead Box Protein M1 , Humans , Models, Biological , Protein Binding/drug effects , Regulatory Sequences, Nucleic Acid/genetics , Up-Regulation/drug effects
10.
Cell Cycle ; 13(20): 3202-6, 2014.
Article in English | MEDLINE | ID: mdl-25485499

ABSTRACT

Tumor suppressor p53 is one of the most frequently mutated genes in cancer, with almost 50% of all types of cancer expressing a mutant form of p53. p53 transactivates the expression of its primary negative regulator, HDM2. HDM2 is a ubiquitin ligase, which initiates the proteasomal degradation of p53 following ubiquitination. Proteasome inhibitors, by targeting the ubiquitin proteasome pathway inhibit the degradation of the majority of cellular proteins including wild-type p53. In contrast, in this study we found that the protein expression of mutant p53 was suppressed following treatment with established or novel proteasome inhibitors. Furthermore, for the first time we demonstrated that Arsenic trioxide, which was previously shown to suppress mutant p53 protein level, exhibits proteasome inhibitory activity. Proteasome inhibitor-mediated suppression of mutant p53 was partially rescued by the knockdown of HDM2, suggesting that the stabilization of HDM2 by proteasome inhibitors might be responsible for mutant p53 suppression to some extent. This study suggests that suppression of mutant p53 is a general property of proteasome inhibitors and it provides additional rationale to use proteasome inhibitors for the treatment of tumors with mutant p53.


Subject(s)
Proteasome Inhibitors/pharmacology , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Arsenic Trioxide , Arsenicals/pharmacology , Cell Line, Tumor , Humans , Oxides/pharmacology , Proto-Oncogene Proteins c-mdm2/genetics , RNA Interference , Tumor Suppressor Protein p53/genetics
11.
Biochem J ; 454(2): 201-8, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23772801

ABSTRACT

NAC (N-acetyl-L-cysteine) is commonly used to identify and test ROS (reactive oxygen species) inducers, and to inhibit ROS. In the present study, we identified inhibition of proteasome inhibitors as a novel activity of NAC. Both NAC and catalase, another known scavenger of ROS, similarly inhibited ROS levels and apoptosis associated with H2O2. However, only NAC, and not catalase or another ROS scavenger Trolox, was able to prevent effects linked to proteasome inhibition, such as protein stabilization, apoptosis and accumulation of ubiquitin conjugates. These observations suggest that NAC has a dual activity as an inhibitor of ROS and proteasome inhibitors. Recently, NAC was used as a ROS inhibitor to functionally characterize a novel anticancer compound, piperlongumine, leading to its description as a ROS inducer. In contrast, our own experiments showed that this compound depicts features of proteasome inhibitors including suppression of FOXM1 (Forkhead box protein M1), stabilization of cellular proteins, induction of ROS-independent apoptosis and enhanced accumulation of ubiquitin conjugates. In addition, NAC, but not catalase or Trolox, interfered with the activity of piperlongumine, further supporting that piperlongumine is a proteasome inhibitor. Most importantly, we showed that NAC, but not other ROS scavengers, directly binds to proteasome inhibitors. To our knowledge, NAC is the first known compound that directly interacts with and antagonizes the activity of proteasome inhibitors. Taken together, the findings of the present study suggest that, as a result of the dual nature of NAC, data interpretation might not be straightforward when NAC is utilized as an antioxidant to demonstrate ROS involvement in drug-induced apoptosis.


Subject(s)
Acetylcysteine/pharmacology , Free Radical Scavengers/pharmacology , Proteasome Endopeptidase Complex/drug effects , Proteasome Inhibitors/pharmacology , Reactive Oxygen Species/antagonists & inhibitors , Acetylcysteine/metabolism , Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Catalase/genetics , Catalase/metabolism , Cell Line, Tumor , Chromans/antagonists & inhibitors , Chromans/metabolism , Chromans/pharmacology , Cytomegalovirus/enzymology , Dioxolanes/antagonists & inhibitors , Dioxolanes/pharmacology , Forkhead Box Protein M1 , Forkhead Transcription Factors/antagonists & inhibitors , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Free Radical Scavengers/metabolism , Humans , Hydrogen Peroxide/antagonists & inhibitors , Hydrogen Peroxide/pharmacology , Oxidants/antagonists & inhibitors , Oxidants/pharmacology , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/chemistry , Proteasome Inhibitors/metabolism , Protein Stability/drug effects , Reactive Oxygen Species/metabolism , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Ubiquitinated Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
12.
Am J Pathol ; 183(1): 257-65, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23665201

ABSTRACT

Tumor cells accumulate high level of reactive oxygen species (ROS) because they are metabolically more active than normal cells. Elevated ROS levels increase tumorigenecity but also render cancer cells more vulnerable to oxidative stress than normal cells. The oncogenic transcription factor Forkhead Box M1 (FOXM1), which is overexpressed in a wide range of human cancers, was reported to protect cancer cells from the adverse effects of oxidative stress by up regulating the expression of scavenger enzymes. We therefore hypothesized that the combination of FOXM1 ablation and ROS inducers could selectively eradicate cancer cells. We show that RNA interference-mediated knockdown of FOXM1 further elevates intracellular ROS levels and increases sensitivity of cancer cells to ROS-mediated cell death after treatment with ROS inducers. We also demonstrate that the combination of ROS inducers with FOXM1/proteasome inhibitors induces robust apoptosis in different human cancer cells. In addition, we show evidence that FOXM1/proteasome inhibitor bortezomib in combination with the ROS inducer ß-phenylethyl isothiocyanate efficiently inhibits the growth of breast tumor xenografts in nude mice. We conclude that the combination of ROS inducers and FOXM1 inhibitors could be used as a therapeutic strategy to selectively eliminate cancer cells.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Biomarkers, Tumor/antagonists & inhibitors , Forkhead Transcription Factors/antagonists & inhibitors , Mammary Neoplasms, Experimental/drug therapy , Oxidative Stress/drug effects , 2-Methoxyestradiol , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Boronic Acids/administration & dosage , Bortezomib , Cell Line, Tumor , Drug Administration Schedule , Estradiol/administration & dosage , Estradiol/analogs & derivatives , Female , Forkhead Box Protein M1 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , Isothiocyanates/administration & dosage , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Nude , Pyrazines/administration & dosage , RNA Interference , Random Allocation , Reactive Oxygen Species/metabolism , Transplantation, Heterologous , Treatment Outcome
13.
Mol Cancer Ther ; 12(3): 245-54, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23443798

ABSTRACT

FOXM1 is an oncogenic transcription factor of the Forkhead family and it has a well-defined role in cell proliferation and cell-cycle progression. Expression of FOXM1 is excluded in quiescent or differentiated cells, but its level is highly elevated in proliferating and malignant cells. Overexpression of FOXM1 has been reported in more than 20 types of human cancer. In recent years, FOXM1 has been implicated in diverse cellular processes and also a growing body of experimental data has underlined the relevance of FOXM1 in tumorigenesis. Although FOXM1 is under the control of three major tumor suppressors (RB, p53, and p19(ARF)), it is still active in the majority of human cancers. The oncogenic potential of FOXM1 is mainly based on its ability to transcriptionally activate genes that are involved in different facets of cancer development. In this review, the contribution of FOXM1 to each of the hallmarks of cancer will be summarized and discussed.


Subject(s)
Cell Transformation, Neoplastic/genetics , Forkhead Transcription Factors/genetics , Neoplasms/genetics , Transcriptional Activation/genetics , Cell Cycle Checkpoints , Cell Proliferation , Forkhead Box Protein M1 , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Humans
14.
Biochem Pharmacol ; 85(5): 644-652, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23103567

ABSTRACT

Oncogenic transcription factor FOXM1 is overexpressed in the majority of human cancers. In addition, FOXM1 has been implicated in cell migration, invasion, angiogenesis and metastasis. The important role of FOXM1 in cancer affirms its significance for therapeutic intervention. Current data suggest that targeting FOXM1 in mono- or combination therapy may have promising therapeutic benefits for the treatment of cancer. However, challenges with the delivery of anti-FOXM1 siRNA to tumors and the absence of small molecules, which specifically inhibit FOXM1, are delaying the development of FOXM1 inhibitors as feasible anticancer drugs. In this review, we describe and summarize the efforts that have been made to target FOXM1 in cancer and the consequences of FOXM1 suppression in human cancer cells.


Subject(s)
Antineoplastic Agents/therapeutic use , Forkhead Transcription Factors/antagonists & inhibitors , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic/physiology , Neoplasms/therapy , Antineoplastic Agents/chemistry , Antineoplastic Agents/classification , Drug Design , Forkhead Box Protein M1 , Forkhead Transcription Factors/genetics , Humans , Neoplasms/metabolism , RNA Interference
15.
Cell Cycle ; 11(18): 3370-2, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22894930

ABSTRACT

Nanoparticle-encapsulated thiazole antibiotic, thiostrepton, has been shown to be an effective agent for inhibiting tumor growth in solid tumor models through the inhibition of proteasomal activity by the induction of apoptosis in cancer cells. Here, we show the efficacy of thiostrepton-micelles in inhibiting tumor growth in a DEN/PB-induced liver cancer model. We also demonstrate an enhanced anticancer effect of the combination treatment of thiostrepton with bortezomib, another proteasome inhibitor in this liver cancer model.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Boronic Acids/therapeutic use , Cell Transformation, Neoplastic/pathology , Liver Neoplasms/drug therapy , Pyrazines/therapeutic use , Thiostrepton/therapeutic use , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Boronic Acids/pharmacology , Bortezomib , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Cell Transformation, Neoplastic/drug effects , Diethylnitrosamine , Disease Models, Animal , Liver/drug effects , Liver/pathology , Liver Neoplasms/pathology , Mice , Phenobarbital , Pyrazines/pharmacology , Thiostrepton/pharmacology
16.
PLoS One ; 7(2): e31761, 2012.
Article in English | MEDLINE | ID: mdl-22393369

ABSTRACT

Irradiation and DNA-damaging chemotherapeutic agents are commonly used in anticancer treatments. Following DNA damage FOXM1 protein levels are often elevated. In this study, we sought to investigate the potential role of FOXM1 in programmed cell death induced by DNA-damage. Human cancer cells after FOXM1 suppression were subjected to doxorubicin or γ-irradiation treatment. Our findings indicate that FOXM1 downregulation by stable or transient knockdown using RNAi or by treatment with proteasome inhibitors that target FOXM1 strongly sensitized human cancer cells of different origin to DNA-damage-induced apoptosis. We showed that FOXM1 suppresses the activation of pro-apoptotic JNK and positively regulates anti-apoptotic Bcl-2, suggesting that JNK activation and Bcl-2 down-regulation could mediate sensitivity to DNA-damaging agent-induced apoptosis after targeting FOXM1. Since FOXM1 is widely expressed in human cancers, our data further support the fact that it is a valid target for combinatorial anticancer therapy.


Subject(s)
Apoptosis , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Antineoplastic Agents/pharmacology , Cell Death , Cell Line, Tumor , DNA Damage , Dose-Response Relationship, Radiation , Down-Regulation , Forkhead Box Protein M1 , Humans , MAP Kinase Kinase 4/metabolism , Protease Inhibitors/pharmacology , Proteasome Inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Stem Cells
17.
J Biol Chem ; 286(48): 41425-41433, 2011 Dec 02.
Article in English | MEDLINE | ID: mdl-21979956

ABSTRACT

Using mass spectrometric analysis we found that oncogenic transcription factor FOXM1 that is overexpressed in a majority of human cancers interacts with multifunctional protein NPM, which is also overexpressed in a variety of human tumors. Coimmunoprecipitation and glutathione S-transferase pull-down experiments demonstrated that NPM forms a complex with FOXM1 and also identified the regions responsible for their interaction. Immunofluorescence microscopy confirmed the interaction between FOXM1 and NPM in cancer and immortal cells. Furthermore, knockdown of NPM in immortal and cancer cells led to significant down-regulation of FOXM1 similar to its levels in normal cells, suggesting that NPM might modulate FOXM1 level. In addition, in OCI/AML3 leukemia cells where mutant NPM is localized in the cytoplasm we found that typically nuclear FOXM1 was predominantly co-localized with NPM in the cytoplasm, while NPM knockdown led to the disappearance of FOXM1 from the cytoplasm, suggesting that NPM may also determine intracellular localization of FOXM1. Knockdown of FOXM1 or NPM in MIA PaCa-2 pancreatic cancer cells inhibited anchorage-dependent and independent growth in cell culture, and tumor growth in nude mice. In addition, over-expression of FOXM1 reversed the effect of NPM knockdown in vitro. Our data suggest that in cancer cells NPM interacts with FOXM1 and their interaction is required for sustaining the level and localization of FOXM1. Targeting the interaction between FOXM1 and NPM by peptides or small molecules may represent a novel therapeutic strategy against cancer.


Subject(s)
Forkhead Transcription Factors/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Nuclear Proteins/metabolism , Animals , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cytoplasm/genetics , Cytoplasm/metabolism , Forkhead Box Protein M1 , Forkhead Transcription Factors/genetics , Gene Knockdown Techniques , HEK293 Cells , Humans , Male , Mice , Mice, Nude , Mutation , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/therapy , Nuclear Proteins/genetics , Nucleophosmin , Protein Transport/genetics
19.
Cell Cycle ; 8(20): 3425-7, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19806025

ABSTRACT

The Forkhead box M1 (FoxM1) oncogenic transcription factor is overexpressed in a majority of human tumors. p53 is a transcription factor and a major tumor suppressor that is mutated in 50% of human cancers. In this study, we compared the levels of FoxM1 in normal BJ human fibroblasts, BJ fibroblasts with p53 knockdown and corresponding BJ immortal/oncogenic cell lines with inactivated p53. We found that partial deletion or inactivation of p53 in these cells leads to upregulation of FoxM1 expression. Similarly, p53 knockdown in several human cancer cell lines with wt-p53 led to upregulation of FoxM1 mRNA and protein expression, while induction of p53 by DNA-damage led to downregulation of FoxM1. These data suggest that p53 negatively regulates FoxM1 expression and therefore inactivation of p53 in tumors could partially explain the phenomenon of FoxM1 overexpression in human cancers.


Subject(s)
Forkhead Transcription Factors/metabolism , Tumor Suppressor Protein p53/physiology , Cell Cycle Proteins/metabolism , Cell Line , DNA Damage , Forkhead Box Protein M1 , Humans , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , RNA, Small Interfering/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Up-Regulation , Polo-Like Kinase 1
20.
PLoS One ; 4(8): e6593, 2009 Aug 12.
Article in English | MEDLINE | ID: mdl-19672316

ABSTRACT

Proteasome inhibitors are currently in the clinic or in clinical trials, but the mechanism of their anticancer activity is not completely understood. The oncogenic transcription factor FoxM1 is one of the most overexpressed genes in human tumors, while its expression is usually halted in normal non-proliferating cells. Previously, we established that thiazole antibiotics Siomycin A and thiostrepton inhibit FoxM1 and induce apoptosis in human cancer cells. Here, we report that Siomycin A and thiostrepton stabilize the expression of a variety of proteins, such as p21, Mcl-1, p53 and hdm-2 and also act as proteasome inhibitors in vitro. More importantly, we also found that well-known proteasome inhibitors such as MG115, MG132 and bortezomib inhibit FoxM1 transcriptional activity and FoxM1 expression. In addition, overexpression of FoxM1 specifically protects against bortezomib-, but not doxorubicin-induced apoptosis. These data suggest that negative regulation of FoxM1 by proteasome inhibitors is a general feature of these drugs and it may contribute to their anticancer properties.


Subject(s)
Enzyme Inhibitors/pharmacology , Forkhead Transcription Factors/drug effects , Proteasome Inhibitors , Apoptosis/drug effects , Cell Line, Tumor , Forkhead Box Protein M1 , Humans , Peptides/pharmacology , Thiostrepton/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...