Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Front Lupus ; 22024.
Article in English | MEDLINE | ID: mdl-38707772

ABSTRACT

Background/Purpose: Cutaneous lupus erythematosus (CLE) affects up to 70% of patients with systemic lupus erythematosus (SLE), and type I interferons (IFNs) are important promoters of SLE and CLE. Our previous work identified IFN-kappa (IFN-κ), a keratinocyte-produced type I IFN, as upregulated in non-lesional and lesional lupus skin and as a critical regulator for enhanced UVB-mediated cell death in SLE keratinocytes. Importantly, the molecular mechanisms governing regulation of IFN-κ expression have been relatively unexplored. Thus, this study sought to identify critical regulators of IFN-κ and identified a novel role for IFN-beta (IFN-ß). Methods: Human N/TERT keratinocytes were treated with the RNA mimic poly (I:C) or 50 mJ/cm2 ultraviolet B (UVB), followed by mRNA expression quantification by RT-qPCR in the presence or absence neutralizing antibody to the type I IFN receptor (IFNAR). IFNB and STAT1 knockout (KO) keratinocytes were generated using CRISPR/Cas9. Results: Time courses of poly(I:C) and UVB treatment revealed a differential expression of IFNB, which was upregulated between 3-6 hours and IFNK, which was upregulated 24 hours after stimulation. Intriguingly, only IFNK expression was substantially abrogated by neutralizing antibodies to IFNAR, suggesting that IFNK upregulation required type I IFN signaling for induction. Indeed, deletion of IFNB abrogated IFNK expression. Further exploration confirmed a role for type I IFN-triggered STAT1 activation. Conclusion: Collectively, our work describes a novel mechanistic paradigm in keratinocytes in which initial IFN-κ induction in response to poly(I:C) and UVB is IFNß1-dependent, thus describing IFNK as both an IFN gene and an interferon-stimulated gene.

3.
J Clin Invest ; 130(4): 1635-1652, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31874109

ABSTRACT

The incidence of human papillomavirus-positive (HPV+) head and neck squamous cell carcinoma (HNSCC) has surpassed that of cervical cancer and is projected to increase rapidly until 2060. The coevolution of HPV with transforming epithelial cells leads to the shutdown of host immune detection. Targeting proximal viral nucleic acid-sensing machinery is an evolutionarily conserved strategy among viruses to enable immune evasion. However, E7 from the dominant HPV subtype 16 in HNSCC shares low homology with HPV18 E7, which was shown to inhibit the STING DNA-sensing pathway. The mechanisms by which HPV16 suppresses STING remain unknown. Recently, we characterized the role of the STING/type I interferon (IFN-I) pathway in maintaining immunogenicity of HNSCC in mouse models. Here we extended those findings into the clinical domain using tissue microarrays and machine learning-enhanced profiling of STING signatures with immune subsets. We additionally showed that HPV16 E7 uses mechanisms distinct from those used by HPV18 E7 to antagonize the STING pathway. We identified NLRX1 as a critical intermediary partner to facilitate HPV16 E7-potentiated STING turnover. The depletion of NLRX1 resulted in significantly improved IFN-I-dependent T cell infiltration profiles and tumor control. Overall, we discovered a unique HPV16 viral strategy to thwart host innate immune detection that can be further exploited to restore cancer immunogenicity.


Subject(s)
Head and Neck Neoplasms/immunology , Human papillomavirus 16/immunology , Membrane Proteins/immunology , Mitochondrial Proteins/immunology , Proteolysis , Squamous Cell Carcinoma of Head and Neck/immunology , Tumor Escape , Animals , Cell Line, Tumor , Female , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/virology , Human papillomavirus 16/genetics , Humans , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , Mitochondrial Proteins/genetics , Papillomavirus E7 Proteins/genetics , Papillomavirus E7 Proteins/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/virology
4.
Methods Mol Biol ; 1960: 85-91, 2019.
Article in English | MEDLINE | ID: mdl-30798523

ABSTRACT

In addition to leukocytes, a variety of cells also participate in the innate immune response, including endothelial cells, epithelial cells, and fibroblasts. Thus, the study of these cells is highly relevant in broadening our understanding of mechanisms that modulate innate immunity. With the rise of genetically engineered animals, it is now common to confirm in vitro data acquired using immortalized cell lines with more physiologically relevant primary cells from these animals ex vivo. Indeed, many studies exploring innate immune system function employ mouse embryonic fibroblasts (MEFs). These cells are relatively simple to generate and are a powerful tool to explore regulatory networks, examine biochemical profiling of protein complexes, and investigate novel signaling pathways associated with innate immune system signaling. Here, we provide a robust protocol to isolate, maintain, and store primary MEFs. This protocol is designed for users with minimal experience using mouse models. We have also added precautions and common pitfalls associated with these procedures.


Subject(s)
Embryo, Mammalian/cytology , Fibroblasts/cytology , Animals , Cell Culture Techniques , Cell Differentiation/physiology , Embryo, Mammalian/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Mice , Signal Transduction/physiology
5.
Methods Mol Biol ; 1960: 93-99, 2019.
Article in English | MEDLINE | ID: mdl-30798524

ABSTRACT

With the renewed enthusiasm in immuno-oncology, characterization of the tumor immune microenvironment constitutes an essential and unique aspect to the assessment of therapeutics. The isolation of tumor-infiltrating lymphocytes (TILs) is a desirable approach toward the understanding of antitumor immune response. This chapter provides an effective protocol to mechanically dissociate tumor tissue and generate single-cell suspension from excised tumors. TILs are then isolated by Ficoll-Paque density gradient centrifugation. This protocol is applicable to both human and experimental tumors in immunocompetent murine models.


Subject(s)
Cell Separation/methods , Centrifugation, Density Gradient/methods , Lymphocytes, Tumor-Infiltrating/cytology , Lymphocytes, Tumor-Infiltrating/metabolism , Animals , Humans , Tumor Microenvironment/genetics , Tumor Microenvironment/physiology
6.
Clin Cancer Res ; 24(17): 4242-4255, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29769207

ABSTRACT

Purpose: The response rates of Head and Neck Squamous Cell Carcinoma (HNSCC) to checkpoint blockade are below 20%. We aim to develop a mechanism-based vaccine to prevent HNSCC immune escape.Experimental Design: We performed RNA-Seq of sensitive and resistant HNSCC cells to discover central pathways promoting resistance to immune killing. Using biochemistry, animal models, HNSCC microarray, and immune cell deconvolution, we assessed the role of SOX2 in inhibiting STING-type I interferon (IFN-I) signaling-mediated antitumor immunity. To bypass SOX2-potentiated STING suppression, we engineered a novel tumor antigen-targeted nanosatellite vehicle to enhance the efficacy of STING agonist and sensitize SOX2-expressing HNSCC to checkpoint blockade.Results: The DNA-sensing defense response is the most suppressed pathway in immune-resistant HNSCC cells. We identified SOX2 as a novel inhibitor of STING. SOX2 facilitates autophagy-dependent degradation of STING and inhibits IFN-I signaling. SOX2 potentiates an immunosuppressive microenvironment and promotes HNSCC growth in vivo in an IFN-I-dependent fashion. Our unique nanosatellite vehicle significantly enhances the efficacy of STING agonist. We show that the E6/E7-targeted nanosatellite vaccine expands the tumor-specific CD8+ T cells by over 12-fold in the tumor microenvironment and reduces tumor burden. A combination of nanosatellite vaccine with anti-PD-L1 significantly expands tumor-specific CTLs and limits the populations expressing markers for exhaustion, resulting in more effective tumor control and improved survival.Conclusions: SOX2 dampens the immunogenicity of HNSCC by targeting the STING pathway for degradation. The nanosatellite vaccine offers a novel and effective approach to enhance the adjuvant potential of STING agonist and break cancer tolerance to immunotherapy. Clin Cancer Res; 24(17); 4242-55. ©2018 AACR.


Subject(s)
Cancer Vaccines/immunology , Membrane Proteins/genetics , SOXB1 Transcription Factors/genetics , Squamous Cell Carcinoma of Head and Neck/immunology , Animals , Autophagy/immunology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Cancer Vaccines/pharmacology , Drug Resistance, Neoplasm/immunology , Gene Expression Regulation, Neoplastic , HEK293 Cells , High-Throughput Nucleotide Sequencing , Humans , Immune Tolerance , Immunotherapy , Interferon Type I/genetics , Interferon Type I/immunology , Membrane Proteins/immunology , Mice , Nanostructures/administration & dosage , Nanostructures/chemistry , SOXB1 Transcription Factors/immunology , Squamous Cell Carcinoma of Head and Neck/prevention & control , Squamous Cell Carcinoma of Head and Neck/therapy , Tumor Microenvironment/immunology
7.
Oncotarget ; 7(42): 68597-68613, 2016 Oct 18.
Article in English | MEDLINE | ID: mdl-27612423

ABSTRACT

Non-small cell lung cancer (NSCLC) patients carrying specific EGFR kinase activating mutations (L858R, delE746-A750) respond well to tyrosine kinase inhibitors (TKIs). However, drug resistance develops within a year. In about 50% of such patients, acquired drug resistance is attributed to the enrichment of a constitutively active point mutation within the EGFR kinase domain (T790M). To date, differential drug-binding and altered ATP affinities by EGFR mutants have been shown to be responsible for differential TKI response. As it has been reported that EGFR stability plays a role in the survival of EGFR driven cancers, we hypothesized that differential TKI-induced receptor degradation between the sensitive L858R and delE746-A750 and the resistant T790M may also play a role in drug responsiveness. To explore this, we have utilized an EGFR-null CHO overexpression system as well as NSCLC cell lines expressing various EGFR mutants and determined the effects of erlotinib treatment. We found that erlotinib inhibits EGFR phosphorylation in both TKI sensitive and resistant cells, but the protein half-lives of L858R and delE746-A750 were significantly shorter than L858R/T790M. Third generation EGFR kinase inhibitor (AZD9291) inhibits the growth of L858R/T790M-EGFR driven cells and also induces EGFR degradation. Erlotinib treatment induced polyubiquitination and proteasomal degradation, primarily in a c-CBL-independent manner, in TKI sensitive L858R and delE746-A750 mutants when compared to the L858R/T790M mutant, which correlated with drug sensitivity. These data suggest an additional mechanism of TKI resistance, and we postulate that agents that degrade L858R/T790M-EGFR protein may overcome TKI resistance.


Subject(s)
ErbB Receptors/genetics , Erlotinib Hydrochloride/pharmacology , Mutation , Protein Kinase Inhibitors/pharmacology , Animals , CHO Cells , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cricetinae , Cricetulus , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Phosphorylation/drug effects , Polyubiquitin/metabolism , Protein Stability/drug effects , Proteolysis/drug effects , Ubiquitination/drug effects
8.
Oral Oncol ; 61: 159-65, 2016 10.
Article in English | MEDLINE | ID: mdl-27553942

ABSTRACT

Evidence gleaned from recent studies on the role of tumor-infiltrating lymphocytes (TILs) suggests that cancer is not only a genetic disease but also an immunologic disease. Head and Neck Squamous Cell Carcinoma (HNSCC) has been a significant model to study cancer cell-immune cell interactions. First, immune cell infiltration is an important feature of these tumors. Second, HNSCC frequently develops resistance to immunogenic cytotoxicity, which provides a window to decipher how tumors engage the immune system to establish immune tolerance. Finally, chemoradiation therapy, as a central modality for HNSCC treatment, has been shown to elicit immune activation. The presence of effector immune cells in the tumor microenvironment is often associated with superior clinical response to adjuvant therapy. On the other hand, an activated immune system, in addition to limiting tumor initiation and progression, could also exert selective pressure to promote the growth of less immunogenic tumors, as a pivotal immunoediting process. But it remains unclear how cancer cell signaling regulates tumor immunogenicity and how to mitigate HNSCC-potentiated TIL suppression. In this review, we will revisit the prognostic role of TILs in HNSCC, and collectively discuss how cancer cell machinery impacts upon the plasticity of TILs.


Subject(s)
Head and Neck Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Mouth Neoplasms/immunology , Autophagy , Head and Neck Neoplasms/pathology , Humans , Mouth Neoplasms/pathology
9.
Br J Haematol ; 168(4): 583-97, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25312678

ABSTRACT

Expression levels of MIR144 and MIR451 increase during erythropoiesis, a pattern that is conserved from zebrafish to humans. As these two miRs are expressed from the same polycistronic transcript, we manipulated MIR144 and MIR451 in human erythroid cells individually and together to investigate their effects on human erythropoiesis. Inhibition of endogenous human MIR451 resulted in decreased numbers of erythroid (CD71(hi) CD235a(hi) CD34(-) ) cells, consistent with prior studies in zebrafish and mice. In addition, inhibition of MIR144 impaired human erythroid differentiation, unlike in zebrafish and mouse studies where the functional effect of MIR144 on erythropoiesis was minimal. In this study, we found RAB14 is a direct target of both MIR144 and MIR451. As MIR144 and MIR451 expression increased during human erythropoiesis, RAB14 protein expression decreased. Enforced RAB14 expression phenocopied the effect of MIR144 and/or MIR451 depletion, whereas shRNA-mediated RAB14 knockdown protected cells from MIR144 and/or MIR451 depletion-mediated erythropoietic inhibition. RAB14 knockdown increased the frequency and number of erythroid cells, increased ß-haemoglobin expression, and decreased CBFA2T3 expression during human erythropoiesis. In summary, we utilized MIR144 and MIR451 to identify RAB14 as a novel physiological inhibitor of human erythropoiesis.


Subject(s)
Erythropoiesis/physiology , MicroRNAs/physiology , rab GTP-Binding Proteins/physiology , Cell Line, Tumor , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/drug effects , Erythropoietin/pharmacology , Gene Expression Regulation , Genetic Vectors/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Lentivirus/genetics , Leukemia, Erythroblastic, Acute/pathology , MicroRNAs/antagonists & inhibitors , MicroRNAs/biosynthesis , MicroRNAs/genetics , RNA Interference , RNA, Small Interfering/pharmacology , Recombinant Proteins/pharmacology , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Transduction, Genetic , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/genetics , rab GTP-Binding Proteins/antagonists & inhibitors , rab GTP-Binding Proteins/biosynthesis , rab GTP-Binding Proteins/genetics
10.
PLoS One ; 9(11): e111777, 2014.
Article in English | MEDLINE | ID: mdl-25368993

ABSTRACT

MicroRNAs (miRs) regulate essentially all cellular processes, but few miRs are known to inhibit growth of precursor-B acute lymphoblastic leukemias (B-ALLs). We identified miR-509 via a human genome-wide gain-of-function screen for miRs that inhibit growth of the NALM6 human B-ALL cell line. MiR-509-mediated inhibition of NALM6 growth was confirmed by 3 independent assays. Enforced miR-509 expression inhibited 2 of 2 additional B-ALL cell lines tested, but not 3 non-B-ALL leukemia cell lines. MiR-509-transduced NALM6 cells had reduced numbers of actively proliferating cells and increased numbers of cells undergoing apoptosis. Using miR target prediction algorithms and a filtering strategy, RAB5C was predicted as a potentially relevant target of miR-509. Enforced miR-509 expression in NALM6 cells reduced RAB5C mRNA and protein levels, and RAB5C was demonstrated to be a direct target of miR-509. Knockdown of RAB5C in NALM6 cells recapitulated the growth inhibitory effects of miR-509. Co-expression of the RAB5C open reading frame without its 3' untranslated region (3'UTR) blocked the growth-inhibitory effect mediated by miR-509. These findings establish RAB5C as a target of miR-509 and an important regulator of B-ALL cell growth with potential as a therapeutic target.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , rab5 GTP-Binding Proteins/genetics , Cell Line, Tumor , Cell Proliferation , Humans , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , RNA, Messenger/genetics , S Phase Cell Cycle Checkpoints
11.
PLoS One ; 9(4): e94852, 2014.
Article in English | MEDLINE | ID: mdl-24747944

ABSTRACT

Several individual miRNAs (miRs) have been implicated as potent regulators of important processes during normal and malignant hematopoiesis. In addition, many miRs have been shown to fine-tune intricate molecular networks, in concert with other regulatory elements. In order to study hematopoietic networks as a whole, we first created a map of global miR expression during early murine hematopoiesis. Next, we determined the copy number per cell for each miR in each of the examined stem and progenitor cell types. As data is emerging indicating that miRs function robustly mainly when they are expressed above a certain threshold (∼100 copies per cell), our database provides a resource for determining which miRs are expressed at a potentially functional level in each cell type. Finally, we combine our miR expression map with matched mRNA expression data and external prediction algorithms, using a Bayesian modeling approach to create a global landscape of predicted miR-mRNA interactions within each of these hematopoietic stem and progenitor cell subsets. This approach implicates several interaction networks comprising a "stemness" signature in the most primitive hematopoietic stem cell (HSC) populations, as well as "myeloid" patterns associated with two branches of myeloid development.


Subject(s)
Gene Expression Profiling , MicroRNAs/genetics , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/metabolism , Algorithms , Animals , Cell Differentiation , Female , Mice , RNA, Messenger/genetics
12.
PLoS Biol ; 10(3): e1001290, 2012.
Article in English | MEDLINE | ID: mdl-22479149

ABSTRACT

The proteasome inhibitor MG132 had been shown to prevent galactose induction of the S. cerevisiae GAL1 gene, demonstrating that ubiquitin proteasome-dependent degradation of transcription factors plays an important role in the regulation of gene expression. The deletion of the gene encoding the F-box protein Mdm30 had been reported to stabilize the transcriptional activator Gal4 under inducing conditions and to lead to defects in galactose utilization, suggesting that recycling of Gal4 is required for its function. Subsequently, however, it was argued that Gal4 remains stably bound to the enhancer under inducing conditions, suggesting that proteolytic turnover of Gal4 might not be required for its function. We have performed an alanine-scanning mutagenesis of ubiquitin and isolated a galactose utilization-defective ubiquitin mutant. We have used it for an unbiased suppressor screen and identified the inhibitor Gal80 as a suppressor of the transcriptional defects of the ubiquitin mutant, indicating that the protein degradation of the inhibitor Gal80, and not of the activator Gal4, is required for galactose induction of the GAL genes. We also show that in the absence of Gal80, Mdm30 is not required for Gal4 function, strongly supporting this hypothesis. Furthermore, we have found that Mediator controls the galactose-induced protein degradation of Gal80, which places Mediator genetically upstream of the activator Gal4. Mediator had originally been isolated by its ability to respond to transcriptional activators, and here we have discovered a leading role for Mediator in the process of transcription. The protein kinase Snf1 senses the inducing conditions and transduces the signal to Mediator, which initiates the degradation of the inhibitor Gal80 with the help of the E3 ubiquitin ligase SCF(Mdm30). The ability of Mediator to control the protein degradation of transcriptional inhibitors indicates that Mediator is actually able to direct its own recruitment to gene promoters.


Subject(s)
DNA-Binding Proteins/metabolism , Mediator Complex/metabolism , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Transcription Factors/metabolism , Transcriptional Activation , Culture Media/metabolism , Cyclins/genetics , Cyclins/metabolism , DNA-Binding Proteins/genetics , F-Box Proteins/genetics , F-Box Proteins/metabolism , Galactose/metabolism , Gene Deletion , Gene Expression Regulation, Fungal , Genes, Fungal , HeLa Cells , Humans , Immunoprecipitation , Mediator Complex/genetics , Promoter Regions, Genetic , Protein Binding , Protein Stability , Proteolysis , Repressor Proteins/genetics , S-Phase Kinase-Associated Proteins/genetics , S-Phase Kinase-Associated Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Signal Transduction , Transcription Factors/genetics , Transfection , Ubiquitin/genetics , Ubiquitin/metabolism
13.
Cardiovasc Res ; 93(1): 162-9, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22028336

ABSTRACT

AIMS: Hypoxia-inducible factor 1 (HIF-1) is a heterodimer composed of HIF-1α and HIF-1ß subunits. HIF-1 is known to promote tissue vascularization by activating the transcription of genes encoding angiogenic factors, which bind to receptors on endothelial cells (ECs) and bone marrow-derived angiogenic cells (BMDACs). In this study, we analysed whether HIF-1 activity in the responding ECs and BMDACs is also required for cutaneous vascularization during burn wound healing. METHODS AND RESULTS: We generated mice with floxed alleles at the Hif1a or Arnt locus encoding HIF-1α and HIF-1ß, respectively. Expression of Cre recombinase was driven by the Tie2 gene promoter, which is expressed in ECs and bone marrow cells. Tie2Cre(+) and Tie2Cre(-) mice were subjected to burn wounds of reproducible diameter and depth. Deficiency of HIF-1α or HIF-1ß in Tie2-lineage cells resulted in delayed wound closure, reduced vascularization, decreased cutaneous blood flow, impaired BMDAC mobilization, and decreased BMDAC homing to burn wounds. CONCLUSION: HIF-1 activity in Tie2-lineage cells is required for the mobilization and homing of BMDACs to cutaneous burn wounds and for the vascularization of burn wound tissue.


Subject(s)
Aryl Hydrocarbon Receptor Nuclear Translocator/antagonists & inhibitors , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Burns/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Wound Healing/physiology , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator/deficiency , Base Sequence , Bone Marrow Cells/pathology , Burns/genetics , Burns/physiopathology , Cell Movement/genetics , Cell Movement/physiology , DNA Primers/genetics , Gene Knockout Techniques , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Male , Mice , Mice, Knockout , Neovascularization, Physiologic , Receptor Protein-Tyrosine Kinases/genetics , Receptor, TIE-2 , Skin/blood supply , Skin/injuries , Skin/pathology , Wound Healing/genetics
14.
Wound Repair Regen ; 18(2): 193-201, 2010.
Article in English | MEDLINE | ID: mdl-20163569

ABSTRACT

Hypoxia-inducible factor 1 (HIF-1) is a transcription factor that controls vascular responses to hypoxia and ischemia. In this study, mice that were heterozygous (HET) for a null allele at the locus encoding the HIF-1alpha subunit (HET mice) and their wild-type (WT) littermates were subjected to a thermal injury involving 10% of the body surface area. HIF-1alpha protein levels were increased in burn wounds of WT but not of HET mice on day 2. The serum levels of stromal-derived factor 1alpha, which binds to CXCR4, were increased on day 2 in WT but not in HET mice. Circulating angiogenic cells were also increased on day 2 in WT but not in HET mice and included CXCR4(+)Sca1(+) cells. Laser Doppler perfusion imaging demonstrated increased blood flow in burn wounds of WT but not HET mice on day 7. Immunohistochemistry on day 7 revealed a reduced number of CD31(+) vessels at the healing margin of burn wounds in HET as compared with WT mice. Vessel maturation was also impaired in wounds of HET mice as determined by the number of alpha-smooth muscle actin-positive vessels on day 21. The remaining wound area on day 14 was significantly increased in HET mice compared with WT littermates. The percentage of healed wounds on day 14 was significantly decreased in HET mice. These data delineate a signaling pathway by which HIF-1 promotes angiogenesis during burn wound healing.


Subject(s)
Burns/metabolism , Leukocytes, Mononuclear/metabolism , Neovascularization, Physiologic , Animals , Cell Movement , Chemokine CXCL12/blood , Heterozygote , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Laser-Doppler Flowmetry , Mice , Mice, Transgenic , Receptors, CXCR4/metabolism , Regional Blood Flow , Wound Healing
15.
Proc Natl Acad Sci U S A ; 105(50): 19579-86, 2008 Dec 16.
Article in English | MEDLINE | ID: mdl-19020076

ABSTRACT

A library of drugs that are in clinical trials or use was screened for inhibitors of hypoxia-inducible factor 1 (HIF-1). Twenty drugs inhibited HIF-1-dependent gene transcription by >88% at a concentration of 0.4 microM. Eleven of these drugs were cardiac glycosides, including digoxin, ouabain, and proscillaridin A, which inhibited HIF-1alpha protein synthesis and expression of HIF-1 target genes in cancer cells. Digoxin administration increased latency and decreased growth of tumor xenografts, whereas treatment of established tumors resulted in growth arrest within one week. Enforced expression of HIF-1alpha by transfection was not inhibited by digoxin, and xenografts derived from these cells were resistant to the anti-tumor effects of digoxin, demonstrating that HIF-1 is a critical target of digoxin for cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Cardiac Glycosides/pharmacology , Digoxin/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Animals , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Genes, Reporter/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Luciferases, Firefly/genetics , Mice , Mice, SCID , Protein Biosynthesis/drug effects , Transcription, Genetic/drug effects , Transfection , Xenograft Model Antitumor Assays
16.
J Biol Chem ; 283(16): 10892-903, 2008 Apr 18.
Article in English | MEDLINE | ID: mdl-18281291

ABSTRACT

Autophagy is a process by which cytoplasmic organelles can be catabolized either to remove defective structures or as a means of providing macromolecules for energy generation under conditions of nutrient starvation. In this study we demonstrate that mitochondrial autophagy is induced by hypoxia, that this process requires the hypoxia-dependent factor-1-dependent expression of BNIP3 and the constitutive expression of Beclin-1 and Atg5, and that in cells subjected to prolonged hypoxia, mitochondrial autophagy is an adaptive metabolic response which is necessary to prevent increased levels of reactive oxygen species and cell death.


Subject(s)
Autophagy , Hypoxia-Inducible Factor 1/metabolism , Hypoxia , Mitochondria/metabolism , Animals , Apoptosis Regulatory Proteins , Autophagy-Related Protein 5 , Beclin-1 , Cell Death , Cytoplasm/metabolism , Membrane Proteins/biosynthesis , Mice , Mice, Knockout , Microtubule-Associated Proteins/biosynthesis , Mitochondrial Proteins/biosynthesis , Models, Biological , Molecular Conformation , Proteins/metabolism , Reactive Oxygen Species
SELECTION OF CITATIONS
SEARCH DETAIL
...