Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 2755: 179-187, 2024.
Article in English | MEDLINE | ID: mdl-38319578

ABSTRACT

The hypoxia-inducible transcription factors HIF-1 and HIF-2 regulate the response to hypoxia. Both proteins are dimers of an alpha subunit and a shared beta subunit. Under hypoxic conditions, the alpha subunits are stabilized, and the transactivation ability of the HIF-1 transcription factor is induced. Accordingly, assessment of HIF-1α protein levels and HIF transcriptional activity serve as an indirect indicator of hypoxia. In this series of protocols, I describe three methods to probe the HIF pathway.


Subject(s)
Hypoxia , Polymers , Humans
2.
Gastroenterology ; 166(1): e17-e19, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37714363
5.
Cell Syst ; 13(12): 1048-1064.e7, 2022 12 21.
Article in English | MEDLINE | ID: mdl-36462504

ABSTRACT

Response to hypoxia is a highly regulated process, but little is known about single-cell responses to hypoxic conditions. Using fluorescent reporters of hypoxia response factor-1α (HIF-1α) activity in various cancer cell lines and patient-derived cancer cells, we show that hypoxic responses in individual cancer cells can be highly dynamic and variable. These responses fall into three classes, including oscillatory activity. We identify a molecular mechanism that can account for all three response classes, implicating reactive-oxygen-species-dependent chaperone-mediated autophagy of HIF-1α in a subset of cells. Furthermore, we show that oscillatory response is modulated by the abundance of extracellular lactate in a quorum-sensing-like mechanism. We show that oscillatory HIF-1α activity rescues hypoxia-mediated inhibition of cell division and causes broad suppression of genes downregulated in cancers and activation of genes upregulated in many cancers, suggesting a mechanism for aggressive growth in a subset of hypoxic tumor cells.


Subject(s)
Chaperone-Mediated Autophagy , Lactic Acid , Humans , Lactic Acid/metabolism , Cell Line, Tumor , Hypoxia/metabolism , Cell Proliferation
7.
J Investig Med High Impact Case Rep ; 8: 2324709620947256, 2020.
Article in English | MEDLINE | ID: mdl-32755251

ABSTRACT

Hypoxia-inducible factor-1 (HIF-1) is a key regulator of erythropoiesis. In this article, we report 3 novel mutations, P378S, A385T, and G206C, on the EGLN1 gene encoding the negative HIF-1α regulator prolyl hydroxylase domain-2 (PHD2) in 3 patients with isolated erythrocytosis. These mutations impair PHD2 protein stability and partially reduce PHD2 activity, leading to increased HIF-1α protein levels in cultured cells.


Subject(s)
Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Polycythemia/genetics , Female , Humans , Male , Middle Aged , Mutation, Missense , Procollagen-Proline Dioxygenase/genetics
8.
Nature ; 582(7811): 271-276, 2020 06.
Article in English | MEDLINE | ID: mdl-32499640

ABSTRACT

A major factor in the progression to heart failure in humans is the inability of the adult heart to repair itself after injury. We recently demonstrated that the early postnatal mammalian heart is capable of regeneration following injury through proliferation of preexisting cardiomyocytes1,2 and that Meis1, a three amino acid loop extension (TALE) family homeodomain transcription factor, translocates to cardiomyocyte nuclei shortly after birth and mediates postnatal cell cycle arrest3. Here we report that Hoxb13 acts as a cofactor of Meis1 in postnatal cardiomyocytes. Cardiomyocyte-specific deletion of Hoxb13 can extend the postnatal window of cardiomyocyte proliferation and reactivate the cardiomyocyte cell cycle in the adult heart. Moreover, adult Meis1-Hoxb13 double-knockout hearts display widespread cardiomyocyte mitosis, sarcomere disassembly and improved left ventricular systolic function following myocardial infarction, as demonstrated by echocardiography and magnetic resonance imaging. Chromatin immunoprecipitation with sequencing demonstrates that Meis1 and Hoxb13 act cooperatively to regulate cardiomyocyte maturation and cell cycle. Finally, we show that the calcium-activated protein phosphatase calcineurin dephosphorylates Hoxb13 at serine-204, resulting in its nuclear localization and cell cycle arrest. These results demonstrate that Meis1 and Hoxb13 act cooperatively to regulate cardiomyocyte maturation and proliferation and provide mechanistic insights into the link between hyperplastic and hypertrophic growth of cardiomyocytes.


Subject(s)
Calcineurin/metabolism , Cell Proliferation , Homeodomain Proteins/metabolism , Myeloid Ecotropic Viral Integration Site 1 Protein/metabolism , Myocytes, Cardiac/cytology , Animals , Animals, Newborn , Female , Gene Deletion , Gene Expression Regulation , Heart/physiology , Homeodomain Proteins/genetics , Male , Mice , Myocardium/cytology , Protein Binding , Regeneration
9.
Am J Physiol Cell Physiol ; 309(12): C775-82, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26491052

ABSTRACT

Hypoxia is a physiological cue that impacts diverse physiological processes, including energy metabolism, autophagy, cell motility, angiogenesis, and erythropoiesis. One of the key cell-autonomous effects of hypoxia is as a modulator of cell proliferation. For most cell types, hypoxia induces decreased cell proliferation, since an increased number of cells, with a consequent increase in O2 demand, would only exacerbate hypoxic stress. However, certain cell populations maintain cell proliferation in the face of hypoxia. This is a common pathological hallmark of cancers, but can also serve a physiological function, as in the maintenance of stem cell populations that reside in a hypoxic niche. This review will discuss major molecular mechanisms by which hypoxia regulates cell proliferation in different cell populations, with a particular focus on the role of hypoxia-inducible factors.


Subject(s)
Cell Hypoxia/physiology , Cell Proliferation/physiology , Animals , Humans
10.
Proc Natl Acad Sci U S A ; 112(31): 9751-6, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26195796

ABSTRACT

Huntington's disease (HD) is a progressive neurodegenerative disease caused by a glutamine repeat expansion in mutant huntingtin (mHtt). Despite the known genetic cause of HD, the pathophysiology of this disease remains to be elucidated. Inositol polyphosphate multikinase (IPMK) is an enzyme that displays soluble inositol phosphate kinase activity, lipid kinase activity, and various noncatalytic interactions. We report a severe loss of IPMK in the striatum of HD patients and in several cellular and animal models of the disease. This depletion reflects mHtt-induced impairment of COUP-TF-interacting protein 2 (Ctip2), a striatal-enriched transcription factor for IPMK, as well as alterations in IPMK protein stability. IPMK overexpression reverses the metabolic activity deficit in a cell model of HD. IPMK depletion appears to mediate neural dysfunction, because intrastriatal delivery of IPMK abates the progression of motor abnormalities and rescues striatal pathology in transgenic murine models of HD.


Subject(s)
Huntington Disease/enzymology , Huntington Disease/physiopathology , Neurons/pathology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Adult , Aged , Animals , Biocatalysis , Demography , Dependovirus/metabolism , Disease Models, Animal , Enzyme Stability , Female , Humans , Huntington Disease/genetics , Huntington Disease/pathology , Male , Mice, Transgenic , Middle Aged , Mitochondria/metabolism , Motor Activity , Neostriatum/enzymology , Neostriatum/pathology , Neostriatum/physiopathology , Neurons/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Postmortem Changes , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/metabolism , Signal Transduction , Survival Analysis , Transcription, Genetic , Tumor Suppressor Proteins/metabolism
11.
Autophagy ; 11(5): 850-1, 2015.
Article in English | MEDLINE | ID: mdl-25945892

ABSTRACT

Hypoxia has long been known to serve as a stimulus for cell cycle arrest. Hypoxia-mediated cell cycle arrest is mediated through the actions of HIF1α (hypoxia inducible factor 1, α subunit [basic helix-loop-helix transcription factor]), which has a nontranscriptional role as an inhibitor of MCM (minichromosome maintenance complex component) helicase activity. We identified chaperone-mediated autophagy as a pathway for selective degradation of HIF1α through lysosomes prior to the onset of DNA replication. CDK2 (cyclin-dependent kinase 2) mediates degradation of HIF1α at the G1/S transition, whereas CDK1 (cyclin-dependent kinase 1) increases HIF1α levels and transcriptional activity prior to the onset of G1 phase. Lysosomal inhibitors induce cell cycle arrest, which is recovered by knockdown of HIF1α and EPAS1/HIF2α. These findings establish lysosomes as essential regulators of cell cycle progression through the degradation of HIF1α.


Subject(s)
Autophagy , Cell Cycle , Molecular Chaperones/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Models, Biological
12.
Integr Biol (Camb) ; 7(3): 364-72, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25784457

ABSTRACT

Direct intercellular transfer of cellular components is a recently described general mechanism of cell­cell communication. It is a more non-specific mode of intercellular communication that is not actively controlled by the participating cells. Though membrane bound proteins and small non-protein cytosolic components have been shown to be transferred between cells, the possibility of transfer of cytosolic proteins has not been clearly established, and its mechanism remains unexplained. Using a cell­cell pair of metastatic melanoma and endothelial cells, known to interact at various stages during cancer progression, we show that cytosolic proteins can indeed be transferred between heterotypic cells. Using precise relative cell patterning we provide evidence that this transfer depends on extent of the interface between heterotypic cell populations. This result is further supported by a mathematical model capturing various experimental conditions. We further demonstrate that cytosolic protein transfer can have important functional consequences for the tumor­stroma interactions, e.g., in heterotypic transfer of constitutively activated BRAF, a common melanoma associated mutation, leading to an enhanced activation of the downstream MAPK pathway. Our results suggest that cytosolic protein transfer can have important consequences for regulation of processes involving physical co-location of heterotypic cell types, particularly in invasive cancer growth.


Subject(s)
Cell Communication , Endothelial Cells/metabolism , Endothelial Cells/pathology , Intracellular Signaling Peptides and Proteins/metabolism , Melanoma/metabolism , Melanoma/secondary , Cell Line , Coculture Techniques/methods , Humans , Melanoma/pathology , Protein Transport
13.
Proc Natl Acad Sci U S A ; 111(32): E3325-34, 2014 Aug 12.
Article in English | MEDLINE | ID: mdl-25071185

ABSTRACT

Hypoxia-inducible factor 1 (HIF-1) is a transcription factor that mediates adaptive responses to oxygen deprivation. In addition, the HIF-1α subunit has a nontranscriptional role as a negative regulator of DNA replication through effects on minichromosome maintenance helicase loading and activation. However, some cell types continue to replicate under hypoxic conditions. The mechanism by which these cells maintain proliferation in the presence of elevated HIF-1α levels is unclear. Here we report that HIF-1α physically and functionally interacts with cyclin-dependent kinase 1 (Cdk1) and Cdk2. Cdk1 activity blocks lysosomal degradation of HIF-1α and increases HIF-1α protein stability and transcriptional activity. By contrast, Cdk2 activity promotes lysosomal degradation of HIF-1α at the G1/S phase transition. Blocking lysosomal degradation by genetic or pharmacological means leads to HIF-1α-dependent cell-cycle arrest, demonstrating that lysosomal degradation of HIF-1α is an essential step for the maintenance of cell-cycle progression under hypoxic conditions.


Subject(s)
CDC2 Protein Kinase/metabolism , Cell Cycle Checkpoints/physiology , Cyclin-Dependent Kinase 2/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lysosomes/metabolism , Animals , Cell Line , Cell Line, Tumor , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 2/genetics , G1 Phase Cell Cycle Checkpoints/physiology , Gene Knockdown Techniques , HeLa Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice , Proteasome Endopeptidase Complex/metabolism , Protein Stability , Proteolysis
14.
Proc Natl Acad Sci U S A ; 111(3): E384-93, 2014 Jan 21.
Article in English | MEDLINE | ID: mdl-24324133

ABSTRACT

Overexpression of Rho kinase 1 (ROCK1) and the G protein RhoA is implicated in breast cancer progression, but oncogenic mutations are rare, and the molecular mechanisms that underlie increased ROCK1 and RhoA expression have not been determined. RhoA-bound ROCK1 phosphorylates myosin light chain (MLC), which is required for actin-myosin contractility. RhoA also activates focal adhesion kinase (FAK) signaling. Together, these pathways are critical determinants of the motile and invasive phenotype of cancer cells. We report that hypoxia-inducible factors coordinately activate RhoA and ROCK1 expression and signaling in breast cancer cells, leading to cell and matrix contraction, focal adhesion formation, and motility through phosphorylation of MLC and FAK. Thus, intratumoral hypoxia acts as an oncogenic stimulus by triggering hypoxia-inducible factor → RhoA → ROCK1 → MLC → FAK signaling in breast cancer cells.


Subject(s)
Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Cell Hypoxia , Cell Line, Tumor , Cell Movement , Cell Survival , Collagen/chemistry , Cytoskeleton/metabolism , Female , HEK293 Cells , Humans , Microscopy, Confocal , Mutation , Neoplasm Metastasis , Oligonucleotide Array Sequence Analysis , Oxygen/metabolism , Phosphorylation , Rats , Signal Transduction , Tumor Microenvironment
15.
J Biol Chem ; 288(29): 20768-20775, 2013 Jul 19.
Article in English | MEDLINE | ID: mdl-23750001

ABSTRACT

Hypoxia-inducible factor (HIF) 1 and HIF-2 are heterodimeric proteins composed of an oxygen-regulated HIF-1α or HIF-2α subunit, respectively, and a constitutively expressed HIF-1ß subunit, which mediate adaptive transcriptional responses to hypoxia. Here, we report that Sirt7 (sirtuin-7) negatively regulates HIF-1α and HIF-2α protein levels by a mechanism that is independent of prolyl hydroxylation and that does not involve proteasomal or lysosomal degradation. The effect of Sirt7 was maintained in the presence of the sirtuin inhibitor nicotinamide and upon deletion or mutation of its deacetylase domain, indicating a non-catalytic function. Knockdown of Sirt7 led to an increase in HIF-1α and HIF-2α protein levels and an increase in HIF-1 and HIF-2 transcriptional activity. Thus, we identify a novel molecular function of Sirt7 as a negative regulator of HIF signaling.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Sirtuins/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line, Tumor , Down-Regulation , Gene Knockdown Techniques , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lysosomes/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Proteolysis , Transcription, Genetic
16.
J Biol Chem ; 288(15): 10703-14, 2013 Apr 12.
Article in English | MEDLINE | ID: mdl-23457305

ABSTRACT

Hypoxia-inducible factor-1 (HIF-1) is a heterodimeric transcription factor that mediates adaptive responses to hypoxia. We demonstrate that lysosomal degradation of the HIF-1α subunit by chaperone-mediated autophagy (CMA) is a major regulator of HIF-1 activity. Pharmacological inhibitors of lysosomal degradation, such as bafilomycin and chloroquine, increased HIF-1α levels and HIF-1 activity, whereas activators of chaperone-mediated autophagy, including 6-aminonicotinamide and nutrient starvation, decreased HIF-1α levels and HIF-1 activity. In contrast, macroautophagy inhibitors did not increase HIF-1 activity. Transcription factor EB, a master regulator of lysosomal biogenesis, also negatively regulated HIF-1 activity. HIF-1α interacts with HSC70 and LAMP2A, which are core components of the CMA machinery. Overexpression of HSC70 or LAMP2A decreased HIF-1α protein levels, whereas knockdown had the opposite effect. Finally, hypoxia increased the transcription of genes involved in CMA and lysosomal biogenesis in cancer cells. Thus, pharmacological and genetic approaches identify CMA as a major regulator of HIF-1 activity and identify interplay between autophagy and the response to hypoxia.


Subject(s)
Autophagy/physiology , HSC70 Heat-Shock Proteins/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lysosomes/metabolism , Proteolysis , Animals , Antirheumatic Agents/pharmacology , Autophagy/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Chloroquine/pharmacology , Gene Knockdown Techniques , HSC70 Heat-Shock Proteins/genetics , HeLa Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lysosomes/genetics , Mice
17.
Cancer Res ; 73(11): 3285-96, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23539444

ABSTRACT

The presence of hypoxia and fibrosis within the primary tumor are two major risk factors for metastasis of human breast cancer. In this study, we demonstrate that hypoxia-inducible factor 1 activates the transcription of genes encoding collagen prolyl hydroxylases that are critical for collagen deposition by breast cancer cells. We show that expression of collagen prolyl hydroxylases promotes cancer cell alignment along collagen fibers, resulting in enhanced invasion and metastasis to lymph nodes and lungs. Finally, we establish the prognostic significance of collagen prolyl hydroxylase mRNA expression in human breast cancer biopsies and show that ethyl 3,4-dihydroxybenzoate, a prolyl hydroxylase inhibitor, decreases tumor fibrosis and metastasis in a mouse model of breast cancer.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Procollagen-Proline Dioxygenase/metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Hypoxia/physiology , Cell Line, Tumor , Collagen/metabolism , Female , Gene Knockdown Techniques , Humans , Hydroxybenzoates/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness , Neoplasm Metastasis , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Procollagen-Proline Dioxygenase/biosynthesis , Procollagen-Proline Dioxygenase/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
18.
Mol Cancer Res ; 11(5): 456-66, 2013 May.
Article in English | MEDLINE | ID: mdl-23378577

ABSTRACT

Metastasis is the leading cause of death among patients who have breast cancer. Understanding the role of the extracellular matrix (ECM) in the metastatic process may lead to the development of improved therapies to treat patients with cancer. Intratumoral hypoxia, found in the majority of breast cancers, is associated with an increased risk of metastasis and mortality. We found that in hypoxic breast cancer cells, hypoxia-inducible factor 1 (HIF-1) activates transcription of the PLOD1 and PLOD2 genes encoding procollagen lysyl hydroxylases that are required for the biogenesis of collagen, which is a major constituent of the ECM. High PLOD2 expression in breast cancer biopsies is associated with increased risk of mortality. We show that PLOD2 is critical for fibrillar collagen formation by breast cancer cells, increases tumor stiffness, and is required for metastasis to lymph nodes and lungs.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Hypoxia/physiology , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase/metabolism , Procollagen/metabolism , Animals , Breast Neoplasms/genetics , Cell Growth Processes/physiology , Cell Line, Tumor , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunohistochemistry , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Metastasis , Procollagen/genetics , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase/genetics
19.
Sci Signal ; 6(262): ra10, 2013 Feb 12.
Article in English | MEDLINE | ID: mdl-23405012

ABSTRACT

Cell cycle arrest in response to hypoxia is a fundamental physiological mechanism to maintain a balance between O(2) supply and demand. Many of the cellular responses to reduced O(2) availability are mediated through the transcriptional activity of hypoxia-inducible factor 1 (HIF-1). We report a role for the isolated HIF-1α subunit as an inhibitor of DNA replication, and this role was independent of HIF-1ß and transcriptional regulation. In response to hypoxia, HIF-1α bound to Cdc6, a protein that is essential for loading of the minichromosome maintenance (MCM) complex (which has DNA helicase activity) onto DNA, and promoted the interaction between Cdc6 and the MCM complex. Although the interaction between Cdc6 and the MCM complex increased the association of the MCM proteins with chromatin, the binding of HIF-1α to the complex decreased phosphorylation and activation of the MCM complex by the kinase Cdc7. As a result, HIF-1α inhibited firing of replication origins, decreased DNA replication, and induced cell cycle arrest in various cell types. These findings establish a transcription-independent mechanism by which the stabilization of HIF-1α leads to cell cycle arrest in response to hypoxia.


Subject(s)
DNA Replication , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation , Chromatin/metabolism , Humans , Nuclear Proteins/metabolism , Phosphorylation , Transcription, Genetic
20.
Sci Signal ; 5(227): ra41, 2012 Jun 05.
Article in English | MEDLINE | ID: mdl-22669846

ABSTRACT

Tissue development and regeneration involve tightly coordinated and integrated processes: selective proliferation of resident stem and precursor cells, differentiation into target somatic cell type, and spatial morphological organization. The role of the mechanical environment in the coordination of these processes is poorly understood. We show that multipotent cells derived from native cardiac tissue continually monitored cell substratum rigidity and showed enhanced proliferation, endothelial differentiation, and morphogenesis when the cell substratum rigidity closely matched that of myocardium. Mechanoregulation of these diverse processes required p190RhoGAP, a guanosine triphosphatase-activating protein for RhoA, acting through RhoA-dependent and -independent mechanisms. Natural or induced decreases in the abundance of p190RhoGAP triggered a series of developmental events by coupling cell-cell and cell-substratum interactions to genetic circuits controlling differentiation.


Subject(s)
Cell Differentiation , Morphogenesis , Myocardium/cytology , Animals , Catenins/metabolism , Cell Cycle Proteins , Endothelium/cytology , GTPase-Activating Proteins/metabolism , Nuclear Proteins/metabolism , Rats , Transcription Factors/metabolism , Delta Catenin
SELECTION OF CITATIONS
SEARCH DETAIL
...