Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
EBioMedicine ; 77: 103878, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35189575

ABSTRACT

BACKGROUND: Prominent early features of COVID-19 include severe, often clinically silent, hypoxia and a pronounced reduction in B cells, the latter important in defence against SARS-CoV-2. This presentation resembles the phenotype of mice with VHL-deficient B cells, in which Hypoxia-Inducible Factors are constitutively active, suggesting hypoxia might drive B cell abnormalities in COVID-19. METHODS: Detailed B cell phenotyping was undertaken by flow-cytometry on longitudinal samples from patients with COVID-19 across a range of severities (NIHR Cambridge BioResource). The impact of hypoxia on the transcriptome was assessed by single-cell and whole blood RNA sequencing analysis. The direct effect of hypoxia on B cells was determined through immunisation studies in genetically modified and hypoxia-exposed mice. FINDINGS: We demonstrate the breadth of early and persistent defects in B cell subsets in moderate/severe COVID-19, including reduced marginal zone-like, memory and transitional B cells, changes also observed in B cell VHL-deficient mice. These findings were associated with hypoxia-related transcriptional changes in COVID-19 patient B cells, and similar B cell abnormalities were seen in mice kept in hypoxic conditions. INTERPRETATION: Hypoxia may contribute to the pronounced and persistent B cell pathology observed in acute COVID-19 pneumonia. Assessment of the impact of early oxygen therapy on these immune defects should be considered, as their correction could contribute to improved outcomes. FUNDING: Evelyn Trust, Addenbrooke's Charitable Trust, UKRI/NIHR, Wellcome Trust.


Subject(s)
COVID-19 , Pneumonia , Animals , Humans , Hypoxia , Mice , Oxygen , SARS-CoV-2
4.
Nat Genet ; 53(7): 1022-1035, 2021 07.
Article in English | MEDLINE | ID: mdl-34155378

ABSTRACT

Hypoxia-inducible transcription factors (HIFs) are fundamental to cellular adaptation to low oxygen levels, but it is unclear how they interact with chromatin and activate their target genes. Here, we use genome-wide mutagenesis to identify genes involved in HIF transcriptional activity, and define a requirement for the histone H3 lysine 4 (H3K4) methyltransferase SET1B. SET1B loss leads to a selective reduction in transcriptional activation of HIF target genes, resulting in impaired cell growth, angiogenesis and tumor establishment in SET1B-deficient xenografts. Mechanistically, we show that SET1B accumulates on chromatin in hypoxia, and is recruited to HIF target genes by the HIF complex. The selective induction of H3K4 trimethylation at HIF target loci is both HIF- and SET1B-dependent and, when impaired, correlates with decreased promoter acetylation and gene expression. Together, these findings show SET1B as a determinant of site-specific histone methylation and provide insight into how HIF target genes are differentially regulated.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Expression Regulation , Histone-Lysine N-Methyltransferase/metabolism , Hypoxia/genetics , Acetylation , Animals , Humans , Hypoxia/metabolism , Methylation , Mice , Mice, Knockout , Models, Animal , Promoter Regions, Genetic , Protein Binding
5.
Nat Immunol ; 21(11): 1408-1420, 2020 11.
Article in English | MEDLINE | ID: mdl-32868930

ABSTRACT

B lymphocyte development and selection are central to adaptive immunity and self-tolerance. These processes require B cell receptor (BCR) signaling and occur in bone marrow, an environment with variable hypoxia, but whether hypoxia-inducible factor (HIF) is involved is unknown. We show that HIF activity is high in human and murine bone marrow pro-B and pre-B cells and decreases at the immature B cell stage. This stage-specific HIF suppression is required for normal B cell development because genetic activation of HIF-1α in murine B cells led to reduced repertoire diversity, decreased BCR editing and developmental arrest of immature B cells, resulting in reduced peripheral B cell numbers. HIF-1α activation lowered surface BCR, CD19 and B cell-activating factor receptor and increased expression of proapoptotic BIM. BIM deletion rescued the developmental block. Administration of a HIF activator in clinical use markedly reduced bone marrow and transitional B cells, which has therapeutic implications. Together, our work demonstrates that dynamic regulation of HIF-1α is essential for normal B cell development.


Subject(s)
B-Lymphocytes/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lymphopoiesis/genetics , Animals , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Biomarkers , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunoglobulin Light Chains/genetics , Immunophenotyping , Mice , Mice, Knockout , RNA Editing , Receptors, Antigen, B-Cell/metabolism , Signal Transduction , Transcriptional Activation
6.
Nat Cell Biol ; 19(9): 1093-1104, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28825697

ABSTRACT

Loss-of-function mutations of cyclic-AMP response element binding protein, binding protein (CREBBP) are prevalent in lymphoid malignancies. However, the tumour suppressor functions of CREBBP remain unclear. We demonstrate that loss of Crebbp in murine haematopoietic stem and progenitor cells (HSPCs) leads to increased development of B-cell lymphomas. This is preceded by accumulation of hyperproliferative lymphoid progenitors with a defective DNA damage response (DDR) due to a failure to acetylate p53. We identify a premalignant lymphoma stem cell population with decreased H3K27ac, which undergoes transcriptional and genetic evolution due to the altered DDR, resulting in lymphomagenesis. Importantly, when Crebbp is lost later in lymphopoiesis, cellular abnormalities are lost and tumour generation is attenuated. We also document that CREBBP mutations may occur in HSPCs from patients with CREBBP-mutated lymphoma. These data suggest that earlier loss of Crebbp is advantageous for lymphoid transformation and inform the cellular origins and subsequent evolution of lymphoid malignancies.


Subject(s)
CREB-Binding Protein/deficiency , CREB-Binding Protein/metabolism , Cell Transformation, Neoplastic/metabolism , Lymphoid Progenitor Cells/metabolism , Lymphoma/metabolism , Neoplastic Stem Cells/metabolism , Acetylation , Animals , CREB-Binding Protein/genetics , Cell Proliferation , Cell Self Renewal , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cells, Cultured , DNA Damage , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Histones/metabolism , Lymphangiogenesis , Lymphoid Progenitor Cells/pathology , Lymphoma/genetics , Lymphoma/pathology , Lymphopoiesis , Methylation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Neoplastic Stem Cells/pathology , Phenotype , Signal Transduction , Time Factors , Transcription, Genetic , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
7.
Kidney Int ; 92(4): 900-908, 2017 10.
Article in English | MEDLINE | ID: mdl-28506759

ABSTRACT

Complement C1q is part of the C1 macromolecular complex that mediates the classical complement activation pathway: a major arm of innate immune defense. C1q is composed of A, B, and C chains that require post-translational prolyl 4-hydroxylation of their N-terminal collagen-like domain to enable the formation of the functional triple helical multimers. The prolyl 4-hydroxylase(s) that hydroxylate C1q have not previously been identified. Recognized prolyl 4-hydroxylases include collagen prolyl-4-hydroxylases (CP4H) and the more recently described prolyl hydroxylase domain (PHD) enzymes that act as oxygen sensors regulating hypoxia-inducible factor (HIF). We show that several small-molecule prolyl hydroxylase inhibitors that activate HIF also potently suppress C1q secretion by human macrophages. However, reducing oxygenation to a level that activates HIF does not compromise C1q hydroxylation. In vitro studies showed that a C1q A chain peptide is not a substrate for PHD2 but is a substrate for CP4H1. Circulating levels of C1q did not differ between wild-type mice or mice with genetic deficits in PHD enzymes, but were reduced by prolyl hydroxylase inhibitors. Thus, C1q is hydroxylated by CP4H, but not the structurally related PHD hydroxylases. Hence, reduction of C1q levels may be an important off-target side effect of small molecule PHD inhibitors developed as treatments for renal anemia.


Subject(s)
Complement C1q/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Hypoxia/metabolism , Macrophages/metabolism , Procollagen-Proline Dioxygenase/metabolism , Prolyl-Hydroxylase Inhibitors/pharmacology , Anemia/drug therapy , Anemia/etiology , Animals , Cell Line , Complement C1q/analysis , Complement Pathway, Classical , Female , Humans , Hydroxylation , Kidney Diseases/blood , Kidney Diseases/drug therapy , Kidney Diseases/pathology , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Prolyl-Hydroxylase Inhibitors/therapeutic use , Protein Processing, Post-Translational
8.
Exp Cell Res ; 356(2): 197-203, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28300562

ABSTRACT

The ability of cells to sense and adapt to changes in oxygen is mediated by hypoxia-inducible factor (HIF). Immune cells function in physiologically complex and varying environments whereby oxygen, pH, nutrients, metabolites and cytokines are continuously fluctuating. HIF is well known to play an important role in coordinating the adaptation and function of both innate immune cells and T cells in these complex environments. This review summarises recent discoveries concerning how hypoxia and HIF control B cell behaviour, and regulate antibody quality and decisions concerning tolerance. Hypoxia and HIF activation may provide an important context; coordinating metabolism with variable demands for quiescence, rapid proliferation, and differentiation. Understanding when and how HIF is activated during B cell development and response is important as drugs targeting HIF could influence antibody responses, providing novel therapeutic opportunities for vaccine adjuvants and in treating autoimmunity.


Subject(s)
B-Lymphocytes/metabolism , Cell Differentiation/immunology , Lymphocyte Activation/immunology , Oxygen/metabolism , Animals , Cell Hypoxia , Humans , T-Lymphocytes/immunology
9.
Oncotarget ; 7(39): 63106-63123, 2016 Sep 27.
Article in English | MEDLINE | ID: mdl-27527858

ABSTRACT

Anaplastic (ATC) and certain follicular thyroid-carcinomas (FTCs) are radioresistant. The Phosphatidylinositide 3-kinase (PI3K) pathway is commonly hyperactivated in thyroid-carcinomas. PI3K can modify the PI3K-related kinases (PIKKs) in response to radiation: How PIKKs interact with PI3K and contribute to radioresistance in thyroid-carcinomas is unknown. Further uncertainties exist in how these interactions function under the radioresistant hypoxic microenvironment. Under normoxia/anoxia, ATC (8505c) and FTC (FTC-133) cells were irradiated, with PI3K-inhibition (via GDC-0941 and PTEN-reconstitution into PTEN-null FTC-133s) and effects on PIKK-activation, DNA-damage, clonogenic-survival and cell cycle, assessed. FTC-xenografts were treated with 5 × 2 Gy, ± 50 mg/kg GDC-0941 (twice-daily; orally) for 14 days and PIKK-activation and tumour-growth assessed. PIKK-expression was additionally assessed in 12 human papillary thyroid-carcinomas, 13 FTCs and 12 ATCs. GDC-0941 inhibited radiation-induced activation of Ataxia-telangiectasia mutated (ATM), ATM-and Rad3-related (ATR) and DNA-dependent protein kinase catalytic subunit (DNA-PKcs). Inhibition of ATM and DNA-PKcs was PI3K-dependent, since activation was reduced in PTEN-reconstituted FTC-133s. Inhibition of PIKK-activation was greater under anoxia: Consequently, whilst DNA-damage was increased and prolonged under both normoxia and anoxia, PI3K-inhibition only reduced clonogenic-survival under anoxia. GDC-0941 abrogated radiation-induced cell cycle arrest, an effect most likely linked to the marked inhibition of ATR-activation. Importantly, GDC-0941 inhibited radiation-induced PIKK-activation in FTC-xenografts leading to a significant increase in time taken for tumours to triple in size: 26.5 ± 5 days (radiation-alone) versus 31.5 ± 5 days (dual-treatment). PIKKs were highly expressed across human thyroid-carcinoma classifications, with ATM scoring consistently lower. Interestingly, some loss of ATM and DNA-PKcs was observed. These data provide new insight into the mechanisms of hypoxia-associated radioresistance in thyroid-carcinoma.


Subject(s)
Carcinoma/radiotherapy , Phosphatidylinositol 3-Kinases/metabolism , Thyroid Neoplasms/radiotherapy , Animals , Carcinoma/metabolism , Carcinoma, Papillary/metabolism , Carcinoma, Papillary/radiotherapy , Cell Cycle , Cell Cycle Proteins/metabolism , Cell Line, Tumor , DNA Damage , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Radiation , Enzyme Activation , Female , Histones/metabolism , Humans , Hypoxia , Indazoles/pharmacology , Mice , Mice, Nude , Neoplasm Transplantation , Oxygen/chemistry , Radiation Tolerance , Signal Transduction/physiology , Sulfonamides/pharmacology , Thyroid Cancer, Papillary , Thyroid Neoplasms/metabolism
10.
Sci Rep ; 6: 22950, 2016 Mar 14.
Article in English | MEDLINE | ID: mdl-26972697

ABSTRACT

The hypoxic tumour microenvironment represents an aggressive, therapy-resistant compartment. As arginine is required for specific hypoxia-induced processes, we hypothesised that arginine-deprivation therapy may be useful in targeting hypoxic cancer cells. We explored the effects of the arginine-degrading agent ADI-PEG20 on hypoxia-inducible factor (HIF) activation, the hypoxia-induced nitric oxide (NO) pathway and proliferation using HCT116 and UMUC3 cells and xenografts. The latter lack argininosuccinate synthetase (ASS1) making them auxotrophic for arginine. In HCT116 cells, ADI-PEG20 inhibited hypoxic-activation of HIF-1α and HIF-2α, leading to decreased inducible-nitric oxide synthase (iNOS), NO-production, and VEGF. Interestingly, combining hypoxia and ADI-PEG20 synergistically inhibited ASS1. ADI-PEG20 inhibited mTORC1 and activated the unfolded protein response providing a mechanism for inhibition of HIF and ASS1. ADI-PEG20 inhibited tumour growth, impaired hypoxia-associated NO-production, and decreased vascular perfusion. Expression of HIF-1α/HIF-2α/iNOS and VEGF were reduced, despite an increased hypoxic tumour fraction. Similar effects were observed in UMUC3 xenografts. In summary, ADI-PEG20 inhibits HIF-activated processes in two tumour models with widely different arginine biology. Thus, ADI-PEG20 may be useful in the clinic to target therapy-resistant hypoxic cells in ASS1-proficient tumours and ASS1-deficient tumours.


Subject(s)
Hydrolases/pharmacology , Neoplasms/drug therapy , Nitric Oxide/biosynthesis , Polyethylene Glycols/pharmacology , Xenograft Model Antitumor Assays , Animals , Arginine/metabolism , Argininosuccinate Synthase/antagonists & inhibitors , Argininosuccinate Synthase/genetics , Argininosuccinate Synthase/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blotting, Western , Cell Hypoxia , Cell Line, Tumor , HCT116 Cells , Humans , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mice, SCID , Multiprotein Complexes/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Nitric Oxide Synthase Type II/metabolism , Perfusion , TOR Serine-Threonine Kinases/metabolism , Tumor Burden/drug effects , Unfolded Protein Response/drug effects , Vascular Endothelial Growth Factor A/metabolism
11.
Oncotarget ; 7(17): 24111-24, 2016 Apr 26.
Article in English | MEDLINE | ID: mdl-26992212

ABSTRACT

Compared to normal kidney, renal clear cell carcinomas (ccRCC) contain increased numbers of interstitial, non-hematopoietic CD133+cells that express stem cell markers and exhibit low rates of proliferation. These cells fail to form tumors upon transplantation but support tumor formation by differentiated malignant cells. We hypothesized that killing of ccRCC CD133+ (RCCCD133+) cells by cytotoxic agents might be enhanced by inducing them to divide. Since tumor necrosis factor-alpha (TNF), signalling through TNFR2, induces proliferation of malignant renal tubular epithelial cells, we investigated whether TNFR2 might similarly affect RCCCD133+cells. We compared treating organ cultures of ccRCC vs adjacent nontumour kidney (NK) and RCCCD133+vs NK CD133+ (NKCD133+) cell cultures with wild-type TNF (wtTNF) or TNF muteins selective for TNFR1 (R1TNF) or TNFR2 (R2TNF). In organ cultures, R2TNF increased expression of TNFR2 and promoted cell cycle entry of both RCCCD133+ and NKCD133+ but effects were greater in RCCCD133+. In contrast, R1TNF increased TNFR1 expression and promoted cell death. Importantly, cyclophosphamide triggered much more cell death in RCCCD133+ and NKCD133+cells pre-treated with R2TNF as compared to untreated controls. We conclude that selective engagement of TNFR2 by TNF can drives RCCCD133+ proliferation and thereby increase sensitivity to cell cycle-dependent cytotoxicity.


Subject(s)
AC133 Antigen/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , Receptors, Tumor Necrosis Factor, Type II/metabolism , Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/metabolism , Cell Proliferation/drug effects , Cyclophosphamide/pharmacology , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/metabolism , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/metabolism
12.
BMC Cancer ; 14: 594, 2014 Aug 16.
Article in English | MEDLINE | ID: mdl-25128202

ABSTRACT

BACKGROUND: Regions within solid tumours often experience oxygen deprivation, which is associated with resistance to chemotherapy and irradiation. The aim of this study was to evaluate the radiosensitising effect of gemcitabine and its main metabolite dFdU under normoxia versus hypoxia and to determine whether hypoxia-inducible factor 1 (HIF-1) is involved in the radiosensitising mechanism. METHODS: Stable expression of dominant negative HIF-1α (dnHIF) in MDA-MB-231 breast cancer cells, that ablated endogenous HIF-1 transcriptional activity, was validated by western blot and functionality was assessed by HIF-1α activity assay. Cells were exposed to varying oxygen environments and treated with gemcitabine or dFdU for 24 h, followed by irradiation. Clonogenicity was then assessed. Using radiosensitising conditions, cells were collected for cell cycle analysis. RESULTS: HIF-1 activity was significantly inhibited in cells stably expressing dnHIF. A clear radiosensitising effect under normoxia and hypoxia was observed for both gemcitabine and dFdU. No significant difference in radiobiological parameters between HIF-1 proficient and HIF-1 deficient MDA-MB-231 cells was demonstrated. CONCLUSIONS: For the first time, radiosensitisation by dFdU, the main metabolite of gemcitabine, was demonstrated under low oxygen conditions. No major role for functional HIF-1 protein in radiosensitisation by gemcitabine or dFdU could be shown.


Subject(s)
Deoxycytidine/analogs & derivatives , Deoxyuridine/pharmacology , Hypoxia-Inducible Factor 1/metabolism , Radiation-Sensitizing Agents/pharmacology , Breast Neoplasms , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Hypoxia/drug effects , Cell Hypoxia/radiation effects , Cell Line, Tumor , Deoxycytidine/pharmacology , Deoxyuridine/analogs & derivatives , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , In Vitro Techniques , Gemcitabine
13.
Radiother Oncol ; 108(3): 548-53, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23891094

ABSTRACT

BACKGROUND AND PURPOSE: Undifferentiated follicular and anaplastic thyroid tumours often respond poorly to radiotherapy and show increased metastatic potential. We evaluated radiation-induced effects on metastasis in thyroid carcinoma cells and tumours, mechanistically focusing on phosphatidylinositide 3-kinase (PI3K) and associated pathways. MATERIAL AND METHODS: Migration was analysed in follicular (FTC133) and anaplastic (8505c) cells following radiotherapy (0-6 Gray) with concomitant pharmacological (GDC-0941) or genetic inhibition of PI3K. Hypoxia-inducible factor-1 (HIF-1)-activity was measured using luciferase reporter assays and was inhibited using a dominant-negative variant. Activation and subcellular localisation of target proteins were assessed via Western blot and immunofluorescence. In vivo studies used FTC133 xenografts with metastatic lung dissemination assessed ex vivo. RESULTS: Radiation induced migration in a HIF-dependent manner in FTC133 cells but decreased migration in 8505c's. Post-radiation HIF-activity correlated with migratory phenotype. PI3K-targeting inhibited migration under basal and irradiated conditions through inhibition of HIF-1α, Rho-GTPase expression/activity and localisation whilst having little effect on src/FAK. In vivo, radiation induced PI3K, HIF, Rho-GTPases and src but only PI3K, HIF and Rho-GTPases were inhibited by GDC-0941. Co-treatment with GDC-0941 and radiation significantly reduced metastatic dissemination versus radiotherapy alone. CONCLUSIONS: Radiation modifies metastatic characteristics of thyroid carcinoma cells, which can be successfully inhibited by targeting PI3K using GDC-0941 in vitro and in vivo.


Subject(s)
Hypoxia-Inducible Factor 1/physiology , Neoplasm Metastasis/prevention & control , Phosphoinositide-3 Kinase Inhibitors , Signal Transduction/physiology , Thyroid Neoplasms/radiotherapy , rho GTP-Binding Proteins/physiology , Animals , Cell Line, Tumor , Cell Movement , Female , Humans , Indazoles/pharmacology , Mice , Phosphatidylinositol 3-Kinases/physiology , Sulfonamides/pharmacology , Thyroid Neoplasms/pathology
14.
Mol Cancer Ther ; 12(5): 819-28, 2013 May.
Article in English | MEDLINE | ID: mdl-23427298

ABSTRACT

The phosphoinositide 3-kinase (PI3K) pathway is deregulated in a range of cancers, and several targeted inhibitors are entering the clinic. This study aimed to investigate whether the positron emission tomography tracer 3'-deoxy-3'-[(18)F]fluorothymidine ([(18)F]-FLT) is suitable to mark the effect of the novel PI3K inhibitor GDC-0941, which has entered phase II clinical trial. CBA nude mice bearing U87 glioma and HCT116 colorectal xenografts were imaged at baseline with [(18)F]-FLT and at acute (18 hours) and chronic (186 hours) time points after twice-daily administration of GDC-0941 (50 mg/kg) or vehicle. Tumor uptake normalized to blood pool was calculated, and tissue was analyzed at sacrifice for PI3K pathway inhibition and thymidine kinase (TK1) expression. Uptake of [(18)F]-FLT was also assessed in tumors inducibly overexpressing a dominant-negative form of the PI3K p85 subunit p85α, as well as HCT116 liver metastases after GDC-0941 therapy. GDC-0941 treatment induced tumor stasis in U87 xenografts, whereas inhibition of HCT116 tumors was more variable. Tumor uptake of [(18)F]-FLT was significantly reduced following GDC-0941 dosing in responsive tumors at the acute time point and correlated with pharmacodynamic markers of PI3K signaling inhibition and significant reduction in TK1 expression in U87, but not HCT116, tumors. Reduction of PI3K signaling via expression of Δp85α significantly reduced tumor growth and [(18)F]-FLT uptake, as did treatment of HCT116 liver metastases with GDC-0941. These results indicate that [(18)F]-FLT is a strong candidate for the noninvasive measurement of GDC-0941 action.


Subject(s)
Antineoplastic Agents/pharmacology , Dideoxynucleosides , Indazoles/pharmacology , Neoplasms/diagnosis , Phosphoinositide-3 Kinase Inhibitors , Positron-Emission Tomography , Sulfonamides/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Dideoxynucleosides/metabolism , Disease Models, Animal , Female , HCT116 Cells , Humans , Indazoles/administration & dosage , Mice , Neoplasm Metastasis , Neoplasms/drug therapy , Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Sulfonamides/administration & dosage , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
15.
J Med Chem ; 55(11): 5591-600, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22621623

ABSTRACT

A panel of compounds belonging to the underexposed sulfamate class of carbonic anhydrase (CA, EC 4.2.1.1) inhibitors was generated that displayed high specificity at nanomolar levels for the tumor-associated CA IX/XII isoforms. Three of the specific CA IX/XII inhibitors showed a positive response in in vitro assays for tumor cell migration and spreading. One of them, 4-(3'-(3″,5″-dimethylphenyl)ureido)phenyl sulfamate (S4), was taken forward into the orthotopic MDA-MB-231 (breast carcinoma) model in mice. Treatment with a 10 mg/kg maintenance dosage of S4 given daily on a "5 days on, 2 days off" regimen reduced metastatic tumor burden in the lung while not affecting primary tumor growth or mouse condition. CA inhibitors of the sulfamate class specifically targeting the tumor-associated isoforms are potential candidates in antimetastatic therapy.


Subject(s)
Antigens, Neoplasm/metabolism , Antineoplastic Agents/chemical synthesis , Breast Neoplasms/drug therapy , Carbonic Anhydrase Inhibitors/chemical synthesis , Carbonic Anhydrases/metabolism , Lung Neoplasms/drug therapy , Phenylurea Compounds/chemical synthesis , Sulfonamides/chemical synthesis , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Breast Neoplasms/pathology , Carbonic Anhydrase IX , Carbonic Anhydrase Inhibitors/chemistry , Carbonic Anhydrase Inhibitors/therapeutic use , Cell Line, Tumor , Cell Movement/drug effects , Female , Humans , Lung Neoplasms/secondary , Mice , Neoplasm Transplantation , Phenylurea Compounds/chemistry , Phenylurea Compounds/therapeutic use , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/therapeutic use , Transplantation, Heterologous
16.
J Clin Endocrinol Metab ; 96(12): E1934-43, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21994956

ABSTRACT

CONTEXT: Phosphoinositide 3-kinase (PI3K) regulates the transcription factor hypoxia-inducible factor-1 (HIF-1) in thyroid carcinoma cells. Both pathways are associated with aggressive phenotype in thyroid carcinomas. OBJECTIVE: Our objective was to assess the effects of the clinical PI3K inhibitor GDC-0941 and genetic inhibition of PI3K and HIF on metastatic behavior of thyroid carcinoma cells in vitro and in vivo. DESIGN: Vascular endothelial growth factor ELISA, HIF activity assays, proliferation studies, and scratch-wound migration and cell spreading assays were performed under various O(2) tensions [normoxia, hypoxia (1 and 0.1% O(2)), and anoxia] with or without GDC-0941 in a panel of four thyroid carcinoma cell lines (BcPAP, WRO, FTC133, and 8505c). Genetic inhibition was achieved by overexpressing phosphatase and tensin homolog (PTEN) into PTEN-null cells and by using a dominant-negative variant of HIF-1α (dnHIF). In vivo, human enhanced green fluorescence protein-expressing follicular thyroid carcinomas (FTC) were treated with GDC-0941 (orally). Spontaneous lung metastasis was confirmed by viewing enhanced green fluorescence protein-positive colonies cultured from lung tissue. RESULTS: GDC-0941 inhibited hypoxia/anoxia-induced HIF-1α and HIF-2α expression and HIF activity in thyroid carcinoma cells. Basal (three of four cell lines) and/or hypoxia-induced (four of four) secreted vascular endothelial growth factor was inhibited by GDC-0941, whereas selective HIF targeting predominantly affected hypoxia/anoxia-mediated secretion (P < 0.05-0.0001). Antiproliferative effects of GDC-0941 were more pronounced in PTEN mutant compared with PTEN-restored cells (P < 0.05). Hypoxia increased migration in papillary cells and cell spreading/migration in FTC cells (P < 0.01). GDC-0941 reduced spreading and migration in all O(2) conditions, whereas dnHIF had an impact only on hypoxia-induced migration (P < 0.001). In vivo, GDC-0941 reduced expression of HIF-1α, phospho-AKT, GLUT-1, and lactate dehydrogenase A in FTC xenografts. DnHIF expression and GDC-0941 reduced FTC tumor growth and metastatic lung colonization (P < 0.05). CONCLUSIONS: PI3K plays a prominent role in the metastatic behavior of thyroid carcinoma cells irrespective of O(2) tension and appears upstream of HIF activation. GDC-0941 significantly inhibited the metastatic phenotype, supporting the clinical development of PI3K inhibition in thyroid carcinomas.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma/secondary , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Indazoles/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Sulfonamides/pharmacology , Thyroid Neoplasms/pathology , Animals , Carcinoma/enzymology , Carcinoma/pathology , Cell Hypoxia , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Humans , Mice , PTEN Phosphohydrolase/metabolism , Thyroid Neoplasms/enzymology , Thyroid Neoplasms/metabolism , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/metabolism
17.
J Thyroid Res ; 2011: 762905, 2011.
Article in English | MEDLINE | ID: mdl-21765994

ABSTRACT

Intratumoural hypoxia (low oxygen tension) is associated with aggressive disease and poor prognosis. Hypoxia-inducible factor-1 is a transcription factor activated by hypoxia that regulates the expression of genes that promote tumour cell survival, progression, metastasis, and resistance to chemo/radiotherapy. In addition to hypoxia, HIF-1 can be activated by growth factor-signalling pathways such as the mitogen-activated protein kinases- (MAPK-) and phosphatidylinositol-3-OH kinases- (PI3K-) signalling cascades. Mutations in these pathways are common in thyroid carcinoma and lead to enhanced HIF-1 expression and activity. Here, we summarise current data that highlights the potential role of both hypoxia and MAPK/PI3K-induced HIF-1 signalling in thyroid carcinoma progression, metastatic characteristics, and the potential role of HIF-1 in thyroid carcinoma response to radiotherapy. Direct or indirect targeting of HIF-1 using an MAPK or PI3K inhibitor in combination with radiotherapy may be a new potential therapeutic target to improve the therapeutic response of thyroid carcinoma to radiotherapy and reduce metastatic burden.

SELECTION OF CITATIONS
SEARCH DETAIL
...