Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Immunotherapy ; 13(12): 977-987, 2021 08.
Article in English | MEDLINE | ID: mdl-34184542

ABSTRACT

Aim: Graft-versus-host disease (GvHD) is a major complication arising in patients undergoing allogenic hematopoietic stem cell transplantation. Material & methods: We tested ruxolitinib (a selective JAK1/2 inhibitor) efficacy in three different preclinical models of GvHD. Results: Ruxolitinib, at doses that mimic clinically achievable human JAK/signal transducers and activators of transcription target inhibition, significantly reduced alloreactive T-cell activation and infiltration in the lung and skin, leading to improved outcomes in two experimental models of steroid-refractory acute and chronic GvHD. Additionally, we describe a novel humanized GvHD model in which immunodeficient NOG animals are engineered to produce human IL-15 to facilitate enhanced T- and NK cell engraftment, leading to severe GvHD. Conclusion: Ruxolitinib treatment ameliorated disease symptoms resulting from targeted immune modulation via JAK/signal transducers and activators of transcription signaling inhibition.


Subject(s)
Graft vs Host Disease/pathology , Hematopoietic Stem Cell Transplantation/adverse effects , Janus Kinase Inhibitors/pharmacology , Nitriles/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Animals , Disease Models, Animal , Heterografts , Humans , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic
2.
Clin Cancer Res ; 26(23): 6299-6309, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32998963

ABSTRACT

PURPOSE: T cells engineered to express a chimeric antigen receptor (CAR) are a promising cancer immunotherapy. Such targeted therapies have shown long-term relapse-free survival in patients with B-cell leukemia and lymphoma. However, cytokine release syndrome (CRS) represents a serious, potentially life-threatening side effect often associated with CAR T-cell therapy. CRS manifests as a rapid (hyper)immune reaction driven by excessive inflammatory cytokine release, including IFNγ and IL6. EXPERIMENTAL DESIGN: Many cytokines implicated in CRS are known to signal through the JAK-STAT pathway. Here we study the effect of blocking JAK pathway signaling on CAR T-cell proliferation, antitumor activity, and cytokine levels in in vitro and in vivo models. RESULTS: We report that itacitinib, a potent, selective JAK1 inhibitor, was able to significantly and dose-dependently reduce levels of multiple cytokines implicated in CRS in several in vitro and in vivo models. Importantly, we also report that at clinically relevant doses that mimic human JAK1 pharmacologic inhibition, itacitinib did not significantly inhibit proliferation or antitumor killing capacity of three different human CAR T-cell constructs (GD2, EGFR, and CD19). Finally, in an in vivo model, antitumor activity of CD19-CAR T cells adoptively transferred into CD19+ tumor-bearing immunodeficient animals was unabated by oral itacitinib treatment. CONCLUSIONS: Together, these data suggest that itacitinib has potential as a prophylactic agent for the prevention of CAR T cell-induced CRS, and a phase II clinical trial of itacitinib for prevention of CRS induced by CAR T-cell therapy has been initiated (NCT04071366).


Subject(s)
Azetidines/pharmacology , Cytokine Release Syndrome/drug therapy , Cytokines/antagonists & inhibitors , Immunotherapy, Adoptive/adverse effects , Isonicotinic Acids/pharmacology , Janus Kinase 1/antagonists & inhibitors , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Animals , Apoptosis , Cell Proliferation , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/pathology , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Eur J Pharmacol ; 885: 173505, 2020 Oct 15.
Article in English | MEDLINE | ID: mdl-32861662

ABSTRACT

Pharmacological modulation of the Janus kinase (JAK) family has achieved clinically meaningful therapeutic outcomes for the treatment of inflammatory and hematopoietic diseases. Several JAK1 selective compounds are being investigated clinically to determine their anti-inflammatory potential. We used recombinant enzymes and primary human lymphocytes to assess the JAK1 specificity of itacitinib (INCB039110) and study inhibition of signal transducers and activators of transcription (STAT) signaling. Rodent models of arthritis and inflammatory bowel disease were subsequently explored to elucidate the efficacy of orally administered itacitinib on inflammatory pathogenesis. Itacitinib is a potent and selective JAK1 inhibitor when profiled against the other JAK family members. Upon oral administration in rodents, itacitinib achieved dose-dependent pharmacokinetic exposures that highly correlated with STAT3 pharmacodynamic pathway inhibition. Itacitinib ameliorated symptoms and pathology of established experimentally-induced arthritis in a dose-dependent manner. Furthermore, itacitinib effectively delayed disease onset, reduced symptom severity, and accelerated recovery in three distinct mouse models of inflammatory bowel disease. Low dose itacitinib administered via cannula directly into the colon was highly efficacious in TNBS-induced colitis but with minimal systemic drug exposure, suggesting localized JAK1 inhibition is sufficient for disease amelioration. Itacitinib treatment in an acute graft-versus-host disease (GvHD) model rapidly reduced inflammatory markers within lymphocytes and target tissue, resulting in a marked improvement in disease symptoms. This is the first manuscript describing itacitinib as a potent and selective JAK1 inhibitor with anti-inflammatory activity across multiple preclinical disease models. These data support the scientific rationale for ongoing clinical trials studying itacitinib in select GvHD patient populations.


Subject(s)
Azetidines/pharmacology , Inflammation/drug therapy , Isonicotinic Acids/pharmacology , Janus Kinase 1/antagonists & inhibitors , Animals , Arthritis, Experimental/drug therapy , Azetidines/pharmacokinetics , Azetidines/therapeutic use , Chemokine CCL2/drug effects , Colitis/chemically induced , Colitis/drug therapy , Dose-Response Relationship, Drug , Graft vs Host Disease/drug therapy , Humans , Inflammatory Bowel Diseases/drug therapy , Isonicotinic Acids/pharmacokinetics , Isonicotinic Acids/therapeutic use , Lymphocytes/drug effects , Mice , Mice, Inbred BALB C , Primary Cell Culture , Rats , Rats, Inbred Lew , STAT Transcription Factors/drug effects , STAT3 Transcription Factor/drug effects , Signal Transduction/drug effects , T-Lymphocytes/drug effects
4.
Clin Immunol ; 218: 108517, 2020 09.
Article in English | MEDLINE | ID: mdl-32585295

ABSTRACT

Approximately 15% of patients with coronavirus disease 2019 (COVID-19) experience severe disease, and 5% progress to critical stage that can result in rapid death. No vaccines or antiviral treatments have yet proven effective against COVID-19. Patients with severe COVID-19 experience elevated plasma levels of pro-inflammatory cytokines, which can result in cytokine storm, followed by massive immune cell infiltration into the lungs leading to alveolar damage, decreased lung function, and rapid progression to death. As many of the elevated cytokines signal through Janus kinase (JAK)1/JAK2, inhibition of these pathways with ruxolitinib has the potential to mitigate the COVID-19-associated cytokine storm and reduce mortality. This is supported by preclinical and clinical data from other diseases with hyperinflammatory states, where ruxolitinib has been shown to reduce cytokine levels and improve outcomes. The urgent need for treatments for patients with severe disease support expedited investigation of ruxolitinib for patients with COVID-19.


Subject(s)
Betacoronavirus/pathogenicity , Coronavirus Infections/drug therapy , Cytokine Release Syndrome/prevention & control , Cytokines/antagonists & inhibitors , Pneumonia, Viral/drug therapy , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Severe Acute Respiratory Syndrome/prevention & control , Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/pharmacology , Betacoronavirus/immunology , COVID-19 , Coronavirus Infections/complications , Coronavirus Infections/immunology , Coronavirus Infections/virology , Cytokine Release Syndrome/complications , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/virology , Cytokines/genetics , Cytokines/immunology , Drug Dosage Calculations , Gene Expression Regulation , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/immunology , Humans , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/genetics , Janus Kinase 1/immunology , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/genetics , Janus Kinase 2/immunology , Lung/drug effects , Lung/immunology , Lung/pathology , Lung/virology , Nitriles , Pandemics , Pneumonia, Viral/complications , Pneumonia, Viral/immunology , Pneumonia, Viral/virology , Protein Kinase Inhibitors/pharmacokinetics , Pyrazoles/pharmacokinetics , Pyrimidines , SARS-CoV-2 , Severe Acute Respiratory Syndrome/complications , Severe Acute Respiratory Syndrome/immunology , Severe Acute Respiratory Syndrome/virology , Severity of Illness Index , Signal Transduction/drug effects
5.
Proc Natl Acad Sci U S A ; 113(30): E4338-47, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27402769

ABSTRACT

We previously reported that combining a phosphoinositide 3-kinase (PI3K) inhibitor with a poly-ADP Rib polymerase (PARP)-inhibitor enhanced DNA damage and cell death in breast cancers that have genetic aberrations in BRCA1 and TP53. Here, we show that enhanced DNA damage induced by PI3K inhibitors in this mutational background is a consequence of impaired production of nucleotides needed for DNA synthesis and DNA repair. Inhibition of PI3K causes a reduction in all four nucleotide triphosphates, whereas inhibition of the protein kinase AKT is less effective than inhibition of PI3K in suppressing nucleotide synthesis and inducing DNA damage. Carbon flux studies reveal that PI3K inhibition disproportionately affects the nonoxidative pentose phosphate pathway that delivers Rib-5-phosphate required for base ribosylation. In vivo in a mouse model of BRCA1-linked triple-negative breast cancer (K14-Cre BRCA1(f/f)p53(f/f)), the PI3K inhibitor BKM120 led to a precipitous drop in DNA synthesis within 8 h of drug treatment, whereas DNA synthesis in normal tissues was less affected. In this mouse model, combined PI3K and PARP inhibition was superior to either agent alone to induce durable remissions of established tumors.


Subject(s)
DNA Damage , Nucleosides/metabolism , Phosphoinositide-3 Kinase Inhibitors , Aminopyridines/administration & dosage , Aminopyridines/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Female , Humans , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Morpholines/administration & dosage , Morpholines/pharmacology , Phosphatidylinositol 3-Kinase/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/administration & dosage , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism
6.
Nat Cell Biol ; 18(5): 572-8, 2016 05.
Article in English | MEDLINE | ID: mdl-27088857

ABSTRACT

Cancer cells often select for mutations that enhance signalling through pathways that promote anabolic metabolism. Although the PI(3)K/Akt signalling pathway, which is frequently dysregulated in breast cancer, is a well-established regulator of central glucose metabolism and aerobic glycolysis, its regulation of other metabolic processes required for tumour growth is not well defined. Here we report that in mammary epithelial cells, oncogenic PI(3)K/Akt stimulates glutathione (GSH) biosynthesis by stabilizing and activating NRF2 to upregulate the GSH biosynthetic genes. Increased NRF2 stability is dependent on the Akt-mediated accumulation of p21(Cip1/WAF1) and GSK-3ß inhibition. Consistently, in human breast tumours, upregulation of NRF2 targets is associated with PI(3)K pathway mutation status and oncogenic Akt activation. Elevated GSH biosynthesis is required for PI(3)K/Akt-driven resistance to oxidative stress, initiation of tumour spheroids, and anchorage-independent growth. Furthermore, inhibition of GSH biosynthesis with buthionine sulfoximine synergizes with cisplatin to selectively induce tumour regression in PI(3)K pathway mutant breast cancer cells, both in vitro and in vivo. Our findings provide insight into GSH biosynthesis as a metabolic vulnerability associated with PI(3)K pathway mutant breast cancers.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Glutathione/biosynthesis , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Biosynthetic Pathways/genetics , Buthionine Sulfoximine/pharmacology , Cell Line, Tumor , Cisplatin/pharmacology , Female , Humans , Mutation/genetics , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Signal Transduction/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology
7.
Cell ; 164(3): 433-46, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-26824656

ABSTRACT

The phosphoinositide 3-kinase (PI3K) pathway regulates multiple steps in glucose metabolism and also cytoskeletal functions, such as cell movement and attachment. Here, we show that PI3K directly coordinates glycolysis with cytoskeletal dynamics in an AKT-independent manner. Growth factors or insulin stimulate the PI3K-dependent activation of Rac, leading to disruption of the actin cytoskeleton, release of filamentous actin-bound aldolase A, and an increase in aldolase activity. Consistently, PI3K inhibitors, but not AKT, SGK, or mTOR inhibitors, cause a significant decrease in glycolysis at the step catalyzed by aldolase, while activating PIK3CA mutations have the opposite effect. These results point toward a master regulatory function of PI3K that integrates an epithelial cell's metabolism and its form, shape, and function, coordinating glycolysis with the energy-intensive dynamics of actin remodeling.


Subject(s)
Fructose-Bisphosphate Aldolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Cytoskeleton/metabolism , Cytosol/metabolism , Disease Models, Animal , Epithelial Cells/metabolism , Glycolysis , Humans , Insulin/metabolism , Mice , Phosphoinositide-3 Kinase Inhibitors , Signal Transduction
8.
Breast Cancer Res ; 15(2): 308, 2013 Apr 08.
Article in English | MEDLINE | ID: mdl-23566381

ABSTRACT

The phosphoinositide 3-kinase (PI3K) pathway serves as a relay where signals that emanate from the cell membrane are received and converted into intracellular signals that promote proliferation and survival. Inhibitors of PI3K hold promise for the treatment of breast cancer because activation of this pathway is highly prevalent. However, as is increasingly observed with inhibitors of cell signaling, there appear to be mechanisms of primary and secondary resistance. Britschgi and colleagues report that compensatory activation of the IL-8 signaling axis is a mechanism of primary resistance to PI3K inhibitors in some triple-negative breast cancers. In a set of experiments that carefully emulate the clinical scenario in a mouse model, they show that simultaneous inhibition of Janus kinase 2 enhances the efficacy of PI3K/mammalian target of rapamycin inhibition. Their paper lends further support to the concept that successful design of treatments with signal transduction inhibitors must anticipate potential escape routes - and include agents to simultaneously block them.


Subject(s)
Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Interleukin-8/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , Humans , Mice
9.
Cancer Discov ; 2(11): 1048-63, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22915751

ABSTRACT

UNLABELLED: There is a need to improve treatments for metastatic breast cancer. Here, we show the activation of the phosphoinositide 3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) pathways in a MMTV-CreBrca1(f/f)Trp53(+/-) mouse model of breast cancer. When treated with the pan-class IA PI3K inhibitor NVP-BKM120, tumor doubling was delayed from 5 to 26 days. NVP-BKM120 reduced AKT phosphorylation, tumor cell proliferation, and angiogenesis. Resistant tumors maintained suppression of AKT phosphorylation but exhibited activation of the MAPK pathway at the "pushing margin." Surprisingly, PI3K inhibition increased indicators of DNA damage, poly-ADP-ribosylation (PAR), and γ-H2AX, but decreased Rad51 focus formation, suggesting a critical role of PI3K activity for Rad51 recruitment. The PARP inhibitor olaparib alone attenuated tumor growth modestly; however, the combination of NVP-BKM120 and olaparib delayed tumor doubling to more than 70 days in the mouse model and more than 50 days in xenotransplants from human BRCA1-related tumors, suggesting that combined PI3K and PARP inhibition might be an effective treatment of BRCA1-related tumors. SIGNIFICANCE: Current treatment options for triple-negative breast cancer are limited to chemotherapeutic regimens that have considerable toxicity and are not curative. We report here that the combination of a PI3K inhibitor with a PARP inhibitor provides in vivo synergy for treatment of an endogenous mouse model for BRCA1-related breast cancers, making this a candidate combination to be tested in human clinical trials.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Genes, BRCA1 , Phosphoinositide-3 Kinase Inhibitors , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Synergism , Enzyme Inhibitors/administration & dosage , Female , Humans , Mice , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism
10.
Methods Mol Biol ; 809: 49-62, 2012.
Article in English | MEDLINE | ID: mdl-22113267

ABSTRACT

Transcription factor NFκB is a key regulator of genes involved in immune and inflammatory responses, as well as genes regulating cell proliferation and survival. In addition to many inflammatory disorders, NFκB is constitutively activated in a variety of human cancers and leukemia. Thus, inhibition of NFκB DNA binding activity represents an important therapeutic approach for disorders characterized by high levels of constitutive NFκB activity. We have previously shown that NFκB DNA binding activity is suppressed by the nuclear translocation and accumulation of IκBα, which is induced by inhibition of the 26S proteasome. In this chapter, we describe a protocol that uses small inhibitory RNA (si RNA) interference followed by electrophoretic mobility shift assay (EMSA) to analyze the regulation of NFκB DNA binding by nuclear IκBα induced by the proteasome inhibitor MG132. Using this protocol, we show that in human leukemia Hut-78 cells that exhibit high levels of NFκB DNA binding activity, MG132 induces nuclear translocation and accumulation of IκBα, which then specifically inhibits NFκB DNA binding. This protocol uses human leukemia Hut-78 cells; however, it can be easily adapted for other cells exhibiting high levels of constitutive NFκB DNA binding.


Subject(s)
Electrophoretic Mobility Shift Assay/methods , I-kappa B Proteins/metabolism , NF-kappa B/metabolism , Blotting, Western , Cell Line , Electrophoresis, Polyacrylamide Gel , Humans , I-kappa B Proteins/genetics , NF-KappaB Inhibitor alpha , NF-kappa B/genetics , Proteasome Endopeptidase Complex/metabolism
11.
Methods Mol Biol ; 809: 121-34, 2012.
Article in English | MEDLINE | ID: mdl-22113272

ABSTRACT

Transcription factor NFκB comprises a family of proteins that serve as crucial regulators of genes involved in host immune and inflammatory responses, cell survival, proliferation, and differentiation. Since transcription of NFκB-dependent genes is increased in numerous inflammatory disorders as well as in many types of cancer and leukemia, inhibition of NFκB-dependent transcription thus represents an important therapeutic target. We have previously shown that in human leukocytes, transcription of NFκB-dependent genes is inhibited by the nuclear translocation and accumulation of IκBα, which can be induced by an inhibitor of CRM1-dependent nuclear export, leptomycin B (LMB). In this chapter, we describe a protocol that uses chromatin immunoprecipitation (ChIP) to analyze the regulation of NFκB recruitment to NFκB-dependent promoters by nuclear IκBα induced by LMB. We show that in lipopolysaccharide (LPS)-stimulated human U-937 macrophages, recruitment of NFκB p65 and p50 proteins to NFκB-dependent promoters of IκBα and cIAP2 genes is suppressed by the LMB-induced nuclear IκBα. Even though in this study we use U-937 macrophages, this protocol should be readily adaptable to analyze the regulation of NFκB recruitment by nuclear IκBα also in other cell types.


Subject(s)
Chromatin Immunoprecipitation/methods , I-kappa B Proteins/metabolism , Macrophages/metabolism , NF-kappa B/metabolism , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Humans , I-kappa B Proteins/genetics , Lipopolysaccharides/pharmacology , Macrophages/drug effects , NF-KappaB Inhibitor alpha , NF-kappa B/genetics , NF-kappa B p50 Subunit/genetics , NF-kappa B p50 Subunit/metabolism , Protein Transport/drug effects , Protein Transport/genetics , Real-Time Polymerase Chain Reaction , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism
12.
Breast Cancer Res ; 13(2): R30, 2011 Mar 11.
Article in English | MEDLINE | ID: mdl-21396117

ABSTRACT

INTRODUCTION: Women who carry a BRCA1 mutation typically develop "triple-negative" breast cancers (TNBC), defined by the absence of estrogen receptor (ER), progesterone receptor and Her2/neu. In contrast to ER-positive tumors, TNBCs frequently express high levels of epidermal growth factor receptor (EGFR). Previously, we found a disproportionate fraction of progenitor cells in BRCA1 mutation carriers with EGFR overexpression. Here we examine the role of EGFR in mammary epithelial cells (MECs) in the emergence of BRCA1-related tumors and as a potential target for the prevention of TNBC. METHODS: Cultures of MECs were used to examine EGFR protein levels and promoter activity in response to BRCA1 suppression with inhibitory RNA. EGFR was assessed by immunoblot and immunofluorescence analysis, real-time reverse transcriptase-polymerase chain reaction assay (RT-PCR) and flow cytometry. Binding of epidermal growth factor (EGF) to subpopulations of MECs was examined by Scatchard analysis. The responsiveness of MECs to the EGFR inhibitor erlotinib was assessed in vitro in three-dimensional cultures and in vivo. Mouse mammary tumor virus-Cre recombinase (MMTV-Cre) BRCA1flox/flox p53⁺/⁻ mice were treated daily with erlotinib or vehicle control, and breast cancer-free survival was analyzed using the Kaplan-Meier method. RESULTS: Inhibition of BRCA1 in MECs led to upregulation of EGFR with an inverse correlation of BRCA1 with cellular EGFR protein levels (r² = 0.87) and to an increase in cell surface-expressed EGFR. EGFR upregulation in response to BRCA1 suppression was mediated by transcriptional and posttranslational mechanisms. Aldehyde dehydrogenase 1 (ALDH1)-positive MECs expressed higher levels of EGFR than ALDH1-negative MECs and were expanded two- to threefold in the BRCA1-inhibited MEC population. All MECs were exquisitely sensitive to EGFR inhibition with erlotinib in vitro. EGFR inhibition in MMTV-Cre BRCA1flox/flox p53⁺/⁻ female mice starting at age 3 months increased disease-free survival from 256 days in the controls to 365 days in the erlotinib-treated cohort. CONCLUSIONS: We propose that even partial loss of BRCA1 leads to an overall increase in EGFR expression in MECs and to an expansion of the highly EGFR-expressing, ALDH1-positive fraction. Increased EGFR expression may confer a growth advantage to MECs with loss of BRCA1 at the earliest stages of transformation. Employing EGFR inhibition with erlotinib specifically at this premalignant stage was effective in decreasing the incidence of ER-negative breast tumors in this mouse model.


Subject(s)
BRCA1 Protein/metabolism , Breast/metabolism , Epithelial Cells/metabolism , ErbB Receptors/metabolism , Genes, BRCA1 , Mammary Neoplasms, Experimental/prevention & control , Aldehyde Dehydrogenase 1 Family , Animals , Breast/cytology , Cell Line, Tumor , Cell Transformation, Neoplastic , Cells, Cultured , Epidermal Growth Factor/metabolism , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride , Female , Genes, p53 , Humans , Isoenzymes/metabolism , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mammary Tumor Virus, Mouse , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , RNA Interference , Receptors, Estrogen/metabolism , Retinal Dehydrogenase/metabolism
13.
Mol Cancer Res ; 9(2): 183-94, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21224428

ABSTRACT

Cutaneous T-cell lymphoma (CTCL) is characterized by constitutive activation of nuclear factor κB (NF-κB), which plays a crucial role in the survival of CTCL cells and their resistance to apoptosis. NF-κB activity in CTCL is inhibited by the proteasome inhibitor bortezomib; however, the mechanisms remained unknown. In this study, we investigated mechanisms by which bortezomib suppresses NF-κB activity in CTCL Hut-78 cells. We demonstrate that bortezomib and MG132 suppress NF-κB activity in Hut-78 cells by a novel mechanism that consists of inducing nuclear translocation and accumulation of IκBα (nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha), which then associates with NF-κB p65 and p50 in the nucleus and inhibits NF-κB DNA binding activity. Surprisingly, however, while expression of NF-κB-dependent antiapoptotic genes cIAP1 and cIAP2 is inhibited by bortezomib, expression of Bcl-2 is not suppressed. Chromatin immunoprecipitation indicated that cIAP1 and cIAP2 promoters are occupied by NF-κB p65/50 heterodimers, whereas Bcl-2 promoter is occupied predominantly by p50/50 homodimers. Collectively, our data reveal a novel mechanism of bortezomib function in CTCL and suggest that the inhibition of NF-κB-dependent gene expression by bortezomib is gene specific and depends on the subunit composition of NF-κB dimers recruited to NF-κB-responsive promoters.


Subject(s)
Apoptosis/genetics , Boronic Acids/pharmacology , Cell Nucleus/metabolism , I-kappa B Proteins/metabolism , Lymphoma, T-Cell, Cutaneous/genetics , NF-kappa B/metabolism , Pyrazines/pharmacology , Transcription, Genetic/drug effects , Base Sequence , Bortezomib , Cell Line, Tumor , Cell Nucleus/drug effects , DNA, Neoplasm/metabolism , Drug Screening Assays, Antitumor , Gene Expression Regulation, Neoplastic/drug effects , Genes, Neoplasm/genetics , Humans , Leupeptins/pharmacology , Lymphoma, T-Cell, Cutaneous/pathology , Molecular Sequence Data , NF-KappaB Inhibitor alpha , NF-kappa B/genetics , NF-kappa B p50 Subunit/metabolism , Protein Binding/drug effects , Protein Subunits/metabolism , Protein Transport/drug effects , Transcription Factor RelA/metabolism
14.
J Immunol ; 185(6): 3685-93, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20696864

ABSTRACT

We have previously shown that increased nuclear accumulation of IkappaBalpha inhibits NF-kappaB activity and induces apoptosis in human leukocytes. In this study, we wanted to explore the possibility that the nucleocytoplasmic distribution of IkappaBalpha can be used as a therapeutic target for the regulation of NF-kappaB-dependent cytokine synthesis. Treatment of LPS-stimulated human U937 macrophages with an inhibitor of chromosome region maintenance 1-dependent nuclear export, leptomycin B, resulted in the increased nuclear accumulation of IkappaBalpha and inhibition of NF-kappaB DNA binding activity, caused by the nuclear IkappaBalpha-p65 NF-kappaB interaction. Surprisingly, however, whereas mRNA expression and cellular release of TNF-alpha, the beta form of pro-IL-1 (IL-1beta), and IL-6 were inhibited by the leptomycin B-induced nuclear IkappaBalpha, IL-8 mRNA expression and cellular release were not significantly affected. Analysis of in vivo recruitment of p65 NF-kappaB to NF-kappaB-regulated promoters by chromatin immunoprecipitation in U937 cells and human PBMCs indicated that although the p65 recruitment to TNF-alpha, IL-1beta, and IL-6 promoters was inhibited by the nuclear IkappaBalpha, p65 recruitment to IL-8 promoter was not repressed. Chromatin immunoprecipitation analyses using IkappaBalpha and S536 phosphospecific p65 NF-kappaB Abs demonstrated that although the newly synthesized IkappaBalpha induced by postinduction repression is recruited to TNF-alpha, IL-1beta, and IL-6 promoters but not to the IL-8 promoter, S536-phosphorylated p65 is recruited to IL-8 promoter, but not to TNF-alpha, IL-1beta, or IL-6 promoters. Together, these data indicate that the inhibition of NF-kappaB-dependent transcription by nuclear IkappaBalpha in LPS-stimulated macrophages is gene specific and depends on the S536 phosphorylation status of the recruited p65 NF-kappaB.


Subject(s)
Cytokines/antagonists & inhibitors , Down-Regulation/immunology , Gene Expression Regulation/immunology , I-kappa B Proteins/physiology , Inflammation Mediators/antagonists & inhibitors , Macrophage Activation/immunology , Nuclear Proteins/physiology , Adult , Cytokines/physiology , Humans , I-kappa B Proteins/metabolism , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Macrophage Activation/genetics , NF-KappaB Inhibitor alpha , Promoter Regions, Genetic/immunology , Transcription Factor RelA/antagonists & inhibitors , Transcription Factor RelA/metabolism , Transcription Factor RelA/physiology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , U937 Cells
15.
Arch Biochem Biophys ; 475(2): 156-63, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18468507

ABSTRACT

Proteasome inhibitors are known to suppress the proteasome-mediated degradation of IkappaBalpha in stimulated cells. This results in the cytoplasmic retention of NFkappaB and its reduced nuclear transcriptional activity. In this study, we show that in the metastatic prostate cancer cells, the proteasome inhibitors exhibit a novel, previously unrecognized effect: they increase the cellular levels of IkappaBalpha, which then translocates to the nucleus, associates with the nuclear p65 NFkappaB, thus inhibiting the constitutive NFkappaB DNA binding activity and inducing apoptosis. The proteasome inhibition-induced nuclear translocation of IkappaBalpha is dependent on de novo protein synthesis, occurs also in other cell types, and does not require IkappaBalpha phosphorylation on Ser-32. Since NFkappaB activity is constitutively increased in many human cancers as well as in inflammatory disorders, the proteasome inhibition-induced nuclear translocation of IkappaBalpha could thus provide a new therapeutic strategy aimed at the specific inhibition of NFkappaB activity by the nuclear IkappaBalpha.


Subject(s)
Apoptosis/drug effects , Cell Nucleus/metabolism , Cysteine Proteinase Inhibitors/pharmacology , I-kappa B Proteins/metabolism , Prostatic Neoplasms/drug therapy , Cell Line, Tumor , Humans , I-kappa B Proteins/genetics , Leupeptins/pharmacology , Male , NF-KappaB Inhibitor alpha , Prostatic Neoplasms/pathology , Proteasome Endopeptidase Complex/metabolism , Protein Transport
SELECTION OF CITATIONS
SEARCH DETAIL
...