Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Cell Sci ; 137(12)2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38856651

ABSTRACT

During acute viral infections, innate immune cells invade inflamed tissues and face hypoxic areas. Hypoxia-inducible factors (HIFs) adapt cellular responses towards these conditions. We wanted to investigate the effects of a loss of HIF-2α in macrophages during acute Friend murine leukemia retrovirus (FV) infection in C57BL/6 mice using a Cre/loxP system. Remarkably, mice with floxed Hif-2a (Hif-2afl; Hif-2a is also known as Epas1) did not show any signs of FV infection independent of Cre activity. This prevented a detailed analysis of the role of macrophage HIF-2α for FV infection but allowed us to study a model of unexpected FV resistance. Hif-2afl mice showed a significant decrease in the expression of the Atp6v1e2 gene encoding for the E2 subunit of the vacuolar H+-ATPase, which resulted in a decreased acidification of lysosomes and limited virus entry into the cell. These findings highlight that the insertion of loxP sites is not always without functional consequences and has established a phenotype in the floxed Hif-2a mouse, which is not only unexpected, but unwanted and is of relevance for the use of this mouse strain in (at least virus) experiments.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Friend murine leukemia virus , Mice, Inbred C57BL , Vacuolar Proton-Translocating ATPases , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Mice , Vacuolar Proton-Translocating ATPases/metabolism , Vacuolar Proton-Translocating ATPases/genetics , Friend murine leukemia virus/genetics , Macrophages/metabolism , Macrophages/virology , Macrophages/immunology , Retroviridae Infections/genetics , Retroviridae Infections/metabolism , Retroviridae Infections/virology , Tumor Virus Infections/genetics , Tumor Virus Infections/metabolism , Lysosomes/metabolism
2.
Cell Rep ; 42(11): 113285, 2023 11 28.
Article in English | MEDLINE | ID: mdl-37910505

ABSTRACT

Deciphering the mechanisms underlying viral persistence is critical to achieving a cure for human immunodeficiency virus (HIV) infection. Here, we implement a systems approach to discover molecular signatures of HIV latently infected CD4+ T cells, identifying the immunosuppressive, adenosine-producing ectonucleotidase CD73 as a key surface marker of latent cells. Hypoxic conditioning, reflecting the lymphoid tissue microenvironment, increases the frequency of CD73+ CD4+ T cells and promotes HIV latency. Transcriptomic profiles of CD73+ CD4+ T cells favor viral quiescence, immune evasion, and cell survival. CD73+ CD4+ T cells are capable of harboring a functional HIV reservoir and reinitiating productive infection ex vivo. CD73 or adenosine receptor blockade facilitates latent HIV reactivation in vitro, mechanistically linking adenosine signaling to viral quiescence. Finally, tissue imaging of lymph nodes from HIV-infected individuals on antiretroviral therapy reveals spatial association between CD73 expression and HIV persistence in vivo. Our findings warrant development of HIV-cure strategies targeting the hypoxia-CD73-adenosine axis.


Subject(s)
HIV Infections , HIV-1 , Humans , Adenosine/metabolism , CD4-Positive T-Lymphocytes , Virus Activation , Virus Latency/physiology , Virus Replication/physiology
3.
Sci Rep ; 13(1): 7530, 2023 05 09.
Article in English | MEDLINE | ID: mdl-37161046

ABSTRACT

Myoglobin (MB) is expressed in different cancer types and may act as a tumor suppressor in breast cancer. The mechanisms by which basal MB expression level impacts murine mammary tumorigenesis are unclear. We investigated how MB expression in breast cancer influences proliferation, metastasis, tumor hypoxia, and chemotherapy treatment in vivo. We crossed PyMT and WapCreTrp53flox mammary cancer mouse models that differed in tumor grade/type and onset of mammary carcinoma with MB knockout mice. The loss of MB in WapCre;Trp53flox mice did not affect tumor development and progression. On the other hand, loss of MB decreased tumor growth and increased tissue hypoxia as well as the number of lung metastases in PyMT mice. Furthermore, Doxorubicin therapy prevented the stronger metastatic propensity of MB-deficient tumors in PyMT mice. This suggests that, although MB expression predicts improved prognosis in breast cancer patients, MB-deficient tumors may still respond well to first-line therapies. We propose that determining the expression level of MB in malignant breast cancer biopsies will improve tumor stratification, outcome prediction, and personalized therapy in cancer patients.


Subject(s)
Carcinoma , Myoglobin , Animals , Mice , Myoglobin/genetics , Biopsy , Disease Models, Animal , Hypoxia/genetics , Mice, Knockout
4.
Nat Med ; 29(5): 1191-1200, 2023 05.
Article in English | MEDLINE | ID: mdl-37106166

ABSTRACT

Erythropoietin (Epo) is the master regulator of erythropoiesis and oxygen homeostasis. Despite its physiological importance, the molecular and genomic contexts of the cells responsible for renal Epo production remain unclear, limiting more-effective therapies for anemia. Here, we performed single-cell RNA and transposase-accessible chromatin (ATAC) sequencing of an Epo reporter mouse to molecularly identify Epo-producing cells under hypoxic conditions. Our data indicate that a distinct population of kidney stroma, which we term Norn cells, is the major source of endocrine Epo production in mice. We use these datasets to identify the markers, signaling pathways and transcriptional circuits characteristic of Norn cells. Using single-cell RNA sequencing and RNA in situ hybridization in human kidney tissues, we further provide evidence that this cell population is conserved in humans. These preliminary findings open new avenues to functionally dissect EPO gene regulation in health and disease and may serve as groundwork to improve erythropoiesis-stimulating therapies.


Subject(s)
Anemia , Erythropoietin , Animals , Humans , Mice , Anemia/genetics , Erythropoiesis/genetics , Erythropoietin/genetics , Kidney/metabolism , RNA/metabolism
5.
J Immunol ; 208(5): 1280-1291, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35121641

ABSTRACT

Inflammatory bowel disease such as chronic colitis promotes colorectal cancer, which is a common cause of cancer mortality worldwide. Hypoxia is a characteristic of inflammation as well as of solid tumors and enforces a gene expression response controlled by hypoxia-inducible factors (HIFs). Once established, solid tumors are immunosuppressive to escape their abatement through immune cells. Although HIF activity is known to 1) promote cancer development and 2) drive tumor immune suppression through the secretion of adenosine, both prolyl hydroxylases and an asparaginyl hydroxylase termed factor-inhibiting HIF (FIH) negatively regulate HIF. Thus, FIH may act as a tumor suppressor in colorectal cancer development. In this study, we examined the role of colon epithelial FIH in a mouse model of colitis-induced colorectal cancer. We recapitulated colitis-associated colorectal cancer development in mice using the azoxymethane/dextran sodium sulfate model in Vil1-Cre/FIH+f/+f and wild-type siblings. Colon samples were analyzed regarding RNA and protein expression and histology. Vil1-Cre/FIH+f/+f mice showed a less severe colitis progress compared with FIH+f/+f animals and a lower number of infiltrating macrophages in the inflamed tissue. RNA sequencing analyses of colon tissue revealed a lower expression of genes associated with the immune response in Vil1-Cre/FIH+f/+f mice. However, tumor occurrence did not significantly differ between Vil1-Cre/FIH+f/+f and wild-type mice. Thus, FIH knockout in colon epithelial cells did not modulate colorectal cancer development but reduced the inflammatory response in chronic colitis.


Subject(s)
Colitis-Associated Neoplasms/pathology , Colitis/pathology , Colorectal Neoplasms/pathology , Intestinal Mucosa/pathology , Mixed Function Oxygenases/metabolism , Adenosine/metabolism , Animals , Azoxymethane/toxicity , Cell Hypoxia/physiology , Colitis/chemically induced , Colitis/genetics , Colitis-Associated Neoplasms/genetics , Colon/pathology , Colorectal Neoplasms/genetics , Dextran Sulfate/toxicity , Disease Models, Animal , Epithelial Cells/pathology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mixed Function Oxygenases/genetics , Prolyl Hydroxylases/metabolism , Signal Transduction/physiology , Tumor Escape/immunology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
6.
Antioxid Redox Signal ; 37(13-15): 956-971, 2022 11.
Article in English | MEDLINE | ID: mdl-35088604

ABSTRACT

Significance: Oxygen deprivation (hypoxia) is a common feature at sites of inflammation. Immune cells and all other cells present at the inflamed site have to adapt to these conditions. They do so by stabilization and activation of hypoxia-inducible factor subunit α (HIF-1α and HIF-2α, respectively), enabling constant generation of adenosine triphosphate (ATP) under these austere conditions by the induction of, for example, glycolytic pathways. Recent Advances: During recent years, it has become evident that HIFs play a far more important role than initially believed because they shape the inflammatory phenotype of immune cells. They are indispensable for migration, phagocytosis, and the induction of inflammatory cytokines by innate immune cells and thereby enable a crosstalk between innate and adaptive immunity. In short, they ensure the survival and function of immune cells under critical conditions. Critical Issues: Up to now, there are still open questions regarding the individual roles of HIF-1 and HIF-2 for the different cell types. In particular, the loss of both HIF-1 and HIF-2 in myeloid cells led to unexpected and contradictory results in the mouse models analyzed so far. Similarly, the role of HIF-1 in dendritic cell maturation is unclear due to inconsistent results from in vitro experiments. Future Directions: The HIFs are indispensable for immune cell survival and action under inflammatory conditions, but they might also trigger over-activation of immune cells. Therefore, they might be excellent setscrews to adjust the inflammatory response by pharmaceuticals. China and Japan and very recently (August 2021) Europe have approved prolyl hydroxylase inhibitors (PHIs) to stabilize HIF such as roxadustat for clinical use to treat anemia by increasing the production of erythropoietin, the classical HIF target gene. Nonetheless, we need further work regarding the use of PHIs under inflammatory conditions, because HIFs show specific activation and distinct expression patterns in innate immune cells. The extent to which HIF-1 or HIF-2 as a transcription factor regulates the adaptation of immune cells to inflammatory hypoxia differs not only by the cell type but also with the inflammatory challenge and the surrounding tissue. Therefore, we urgently need isoform- and cell type-specific modulators of the HIF pathway. Antioxid. Redox Signal. 37, 956-971.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Oxygen , Mice , Animals , Oxygen/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Inflammation , Myeloid Cells/metabolism , Hypoxia , Immunity, Innate , Hypoxia-Inducible Factor 1, alpha Subunit
7.
Mol Metab ; 57: 101424, 2022 03.
Article in English | MEDLINE | ID: mdl-34954109

ABSTRACT

OBJECTIVES: Glucocorticoids (GCs) are one of the most widely prescribed anti-inflammatory drugs. By acting through their cognate receptor, the glucocorticoid receptor (GR), GCs downregulate the expression of pro-inflammatory genes and upregulate the expression of anti-inflammatory genes. Metabolic pathways have recently been identified as key parts of both the inflammatory activation and anti-inflammatory polarization of macrophages, immune cells responsible for acute inflammation and tissue repair. It is currently unknown whether GCs control macrophage metabolism, and if so, to what extent metabolic regulation by GCs confers anti-inflammatory activity. METHODS: Using transcriptomic and metabolomic profiling of macrophages, we identified GC-controlled pathways involved in metabolism, especially in mitochondrial function. RESULTS: Metabolic analyses revealed that GCs repress glycolysis in inflammatory myeloid cells and promote tricarboxylic acid (TCA) cycle flux, promoting succinate metabolism and preventing intracellular accumulation of succinate. Inhibition of ATP synthase attenuated GC-induced transcriptional changes, likely through stalling of TCA cycle anaplerosis. We further identified a glycolytic regulatory transcription factor, HIF1α, as regulated by GCs, and as a key regulator of GC responsiveness during inflammatory challenge. CONCLUSIONS: Our findings link metabolism to gene regulation by GCs in macrophages.


Subject(s)
Citric Acid Cycle , Glucocorticoids , Glucocorticoids/pharmacology , Humans , Inflammation/metabolism , Macrophages/metabolism , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism
8.
Int J Mol Sci ; 21(22)2020 Nov 13.
Article in English | MEDLINE | ID: mdl-33202783

ABSTRACT

(1) Background: Hypoxia is a common feature of inflammation when hypoxia inducible factors (HIFs) adapt cells to conditions of low oxygen tension and inflammation. We studied the role of HIF-1 and HIF-2 in cells of the myeloid lineage in a mouse model of acute colitis. (2) Methods: Mice with and without a conditional knockout for either Hif-1a or Hif-2a or Hif-1a and Hif-2a in cells of the myeloid lineage were treated with 2.5% dextran sodium sulfate (DSS) for 6 days to induce an acute colitis. We analyzed the course of inflammation with respect to macroscopic (disease activity index) and microscopic (histology score and immunohistochemical staining of immune cells) parameters and quantified the mRNA expression of cytokines and chemokines in the colon and the mesenteric lymph nodes. (3) Results: A conditional knockout of myeloid Hif-1a ameliorated whereas the knockout of Hif-2a aggravated murine DSS colitis by increased recruitment of neutrophils to deeper layers of the colon. This led to higher expression of Il6, Ifng, Cd11c, Cd4, and Cd8 in the colon but also induced anti-inflammatory mediators such as Foxp3 and Il10. A conditional knockout of Hif-1a and Hif-2a did not show any differences compared to wildtype mice. (4) Conclusions: Myeloid HIF-1α and HIF-2α play opposing roles in acute DSS colitis. Thus, not only a cell type specific, but also the isoform specific modulation of HIFs needs to be addressed in attempts to modify HIF for therapeutic purposes.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/immunology , Inflammatory Bowel Diseases , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , CD4 Antigens/genetics , CD4 Antigens/immunology , CD8 Antigens/genetics , CD8 Antigens/immunology , Dextran Sulfate/toxicity , Disease Models, Animal , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/physiopathology , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Mice , Mice, Knockout , Myeloid Cells/immunology , Myeloid Cells/pathology
9.
PLoS One ; 15(5): e0233261, 2020.
Article in English | MEDLINE | ID: mdl-32413092

ABSTRACT

The transcription factor Hypoxia-inducible factor 1 (HIF-1) plays a pivotal role in tissue regeneration. HIF-1 is negatively controlled by O2-dependent prolyl hydroxylases with a predominant role of prolyl hydroxylase 2 isoform (Phd2). Transgenic mice, hypomorphic for this isoform, accumulate more HIF-1 under normoxic conditions. Using these mice, we investigated the influence of Phd2 and HIF-1 on the regenerative capability of skeletal muscle tissue after myotrauma. Phd2-hypomorphic and wild type mice (on C57Bl/6 background) were grouped with regeneration times from 6 to 168 hours after closed mechanic muscle trauma to the hind limb. Tissue samples were analysed by immuno-staining and real-time PCR. Bone marrow derived macrophages of wild type and Phd2-hypomorphic mice were isolated and analysed via flow cytometry and quantitative real-time PCR. Phd2 reduction led to a higher regenerative capability due to enhanced activation of myogenic factors accompanied by induction of genes responsible for glucose and lactate metabolism in Phd2-hypomorphic mice. Macrophage infiltration into the trauma areas in hypomorphic mice started earlier and was more pronounced compared to wild type mice. Phd2-hypomorphic mice also showed higher numbers of macrophages in areas with sustained trauma 72 hours after myotrauma application. In conclusion, we postulate that the HIF-1 pathway is activated secondary to a Phd2 reduction which may lead to i) higher activation of myogenic factors, ii) increased number of positive stem cell proliferation markers, and iii) accelerated macrophage recruitment to areas of trauma, resulting in faster muscle tissue regeneration after myotrauma. With the current development of prolyl hydroxylase domain inhibitors, our findings point towards a potential clinical benefit after myotrauma.


Subject(s)
Hypoxia-Inducible Factor-Proline Dioxygenases/deficiency , Muscle, Skeletal/physiology , Regeneration/physiology , Soft Tissue Injuries/physiopathology , Animals , Cell Proliferation/physiology , Disease Models, Animal , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Random Allocation , Vascular Endothelial Growth Factor A/metabolism
10.
JCI Insight ; 4(12)2019 06 20.
Article in English | MEDLINE | ID: mdl-31217351

ABSTRACT

During chronic HIV infection, immune cells become increasingly dysfunctional and exhausted. Little is known about how immune functions are restored after initiation of antiretroviral therapy (ART). In this study, we assessed cellular and metabolic activity and evaluated the effect of individual antiretrovirals on cellular subsets ex vivo in ART-treated and treatment-naive chronically HIV-infected individuals. We observed that cellular respiration was significantly decreased in most immune cells in chronic HIV infection. The respiration was correlated to immune activation and the inhibitory receptor programmed cell death 1 on CD8+ T cells. ART restored the metabolic phenotype, but the respiratory impairment persisted in CD4+ T cells. This was particularly the case for individuals receiving integrase strand transfer inhibitors. CD4+ T cells from these individuals showed a significant reduction in ex vivo proliferative capacity compared with individuals treated with protease inhibitors or nonnucleoside reverse transcriptase inhibitors. We noticed a significant decrease in respiration of cells treated with dolutegravir (DLG) or elvitegravir (EVG) and a switch from polyfunctional to TNF-α-dominated "stress" immune response. There was no effect on glycolysis, consistent with impaired mitochondrial function. We detected increased levels of mitochondrial ROS and mitochondrial mass. These findings indicate that EVG and DLG use is associated with slow proliferation and impaired respiration with underlying mitochondrial dysfunction, resulting in overall decreased cellular function in CD4+ T cells.


Subject(s)
Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , HIV Infections/immunology , Immune System/drug effects , Antiretroviral Therapy, Highly Active , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation/drug effects , Cell Respiration/drug effects , Glycolysis/drug effects , Humans
11.
PLoS One ; 12(12): e0190074, 2017.
Article in English | MEDLINE | ID: mdl-29261815

ABSTRACT

Inflammation and hypoxia are hallmarks of inflammatory bowel disease. Low oxygen levels activate hypoxia-inducible factors as central transcriptional regulators of cellular responses to hypoxia, particularly in myeloid cells where hypoxia-inducible factors control immune cell function and survival. Still, the role of myeloid hypoxia-inducible factor-1 during inflammatory bowel disease remains poorly defined. We therefore investigated the role of hypoxia-inducible factor-1 for myeloid cell function and immune response during colitis. Experimental colitis was induced by administration of 2.5% dextran sulfate sodium to mice with a conditional knockout of hypoxia-inducible factor-1α in myeloid cells and their wild type siblings. Murine colon tissue was examined by histologic analysis, immunohistochemistry, and quantitative polymerase chain reaction. Induction of experimental colitis increased levels of hypoxia and accumulation of hypoxia-inducible factor-1α positive cells in colon tissue of both treated groups. Myeloid hypoxia-inducible factor-1α knockout reduced weight loss and disease activity index when compared to wild type mice. Knockout mice displayed less infiltration of macrophages into intestinal mucosa and reduced mRNA expression of markers for dendritic cells and interleukin-17 secreting T helper cells. Expression of inflammatory and anti-inflammatory cytokines also showed a reduced and delayed induction in myeloid hypoxia-inducible factor-1α knockout mice. Our results show a disease promoting role of myeloid hypoxia-inducible factor-1 during intestinal inflammation. This might result from a hypoxia-inducible factor-1 dependent increase in pro-inflammatory interleukin-17 secreting T helper cells in the absence of obvious changes in regulatory T cells. In contrast, knockout mice appear to shift the balance to anti-inflammatory signals and cells resulting in milder intestinal inflammation.


Subject(s)
Colitis/chemically induced , Colitis/pathology , Gene Knockdown Techniques , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Myeloid Cells/metabolism , Myeloid Cells/pathology , Animals , Cell Hypoxia , Cell Polarity , Colitis/complications , Colon/pathology , Cytokines/genetics , Cytokines/metabolism , Dextran Sulfate , Female , Hypoxia/complications , Hypoxia/metabolism , Hypoxia/pathology , Inflammation/complications , Inflammation/pathology , Macrophages/pathology , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/genetics , RNA, Messenger/metabolism , T-Lymphocytes, Regulatory/immunology
13.
J Immunol Res ; 2016: 5134329, 2016.
Article in English | MEDLINE | ID: mdl-26966693

ABSTRACT

Dendritic cells (DCs) are considered as one of the main regulators of immune responses. They collect antigens, process them, and present typical antigenic structures to lymphocytes, thereby inducing an adaptive immune response. All these processes take place under conditions of oxygen shortage (hypoxia) which is often not considered in experimental settings. This review highlights how deeply hypoxia modulates human as well as mouse immature and mature dendritic cell functions. It tries to link in vitro results to actual in vivo studies and outlines how hypoxia-mediated shaping of dendritic cells affects the activation of (innate) immunity.


Subject(s)
Adaptive Immunity/drug effects , Dendritic Cells/drug effects , Hypoxia/immunology , Immunity, Innate/drug effects , Oxygen/pharmacology , Animals , Antigen Presentation/drug effects , Antigens/immunology , Cytokines/genetics , Cytokines/immunology , Dendritic Cells/immunology , Dendritic Cells/pathology , Gene Expression Regulation , Humans , Hypoxia/pathology , Inflammation , Lymphocytes/drug effects , Lymphocytes/immunology , Lymphocytes/pathology , Mice , Signal Transduction
14.
Anesthesiology ; 118(6): 1426-36, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23449494

ABSTRACT

BACKGROUND: Hypoxia-inducible factor-1 (HIF-1) is a molecular key player in response to hypoxemic/inflammatory conditions prevailing in sepsis. In a prospective observational study, we tested the hypotheses that sepsis affects HIF-1α messenger ribonucleic acid (mRNA) expression (primary hypothesis) and also (secondary hypotheses) the expression of the HIF-1α target genes adrenomedullin and ß2-integrins. Furthermore, we tested that lipopolysaccharide administration increases HIF-1α mRNA and protein in naive and endotoxin-tolerant monocytes. METHODS: In 99 patients with sepsis and 48 healthy volunteers, leukocyte HIF-1α mRNA expression (real-time polymerase chain reaction), cytokine concentrations (enzymelinked immunosorbent assay), and intracellular distribution of HIF-1α protein (immunofluorescence staining) were assessed. In vitro, HIF-1α mRNA expression and protein were measured in naive or endotoxin-tolerant (48 h; 0.05 ng/ml lipopolysaccharide) monocytes, with/without additional lipopolysaccharide (6h; 1 µg/ml). RESULTS: In comparison to healthy volunteers, HIF-1α mRNA expression (-67%; P = 0.0001) and HIF-1α protein positive cells (-66.7%; P = 0.01) were decreased in sepsis. mRNA expression of adrenomedullin (-75%), CD11a (-85%), and CD11b (-86%; all P = 0.0001) was also decreased. In contrast, interleukin 6 (P = 0.0001), interleukin 10 (P = 0.0001), and tumor necrosis factor-α (P = 0.0002) concentrations were increased. Of note, HIF-1α mRNA expression was inversely associated with illness severity (Simplified Acute Physiology Score II; r = -0.29; P = 0.0001). In vitro, acute lipopolysaccharide administration of naive monocytic cells increased HIF-1α mRNA expression, whereas HIF-1α mRNA and protein (-60%; P = 0.001) were decreased in endotoxin-tolerant cells, which still up regulated cytokines. CONCLUSIONS: In sepsis, HIF-1α mRNA expression was suppressed and inversely associated with illness severity. While acute lipopolysaccharide administration increased HIF-1α mRNA expression, prolonged stimulation suppressed HIF-1α expression. The clinical implications of decreased HIF-1α may include maladaption to tissue hypoxia or depressed immune function.


Subject(s)
Gene Expression , Hypoxia-Inducible Factor 1/metabolism , Sepsis/metabolism , Aged , Blotting, Western/methods , Female , Humans , Male , Middle Aged , Prospective Studies , Reverse Transcriptase Polymerase Chain Reaction/methods
15.
Biol Chem ; 394(4): 495-505, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23362200

ABSTRACT

Dendritic cells (DCs) are an important link between innate and adaptive immunity. DCs get activated in inflamed tissues where oxygen tension is usually low, which requires the transcription factor hypoxia inducible factor (HIF)-1 for cellular adaptation. To investigate whether the HIF-1 transcriptional complex plays a pivotal role in the function of DCs, we compared the effects of exogenous inflammatory stimuli and hypoxia on HIF-1α in bone marrow-derived DCs from wild type and myeloid-specific HIF-1α knock-out mice. We showed that the Toll-like receptor ligands lipopolysaccharides and cytosine-phosphatidyl-guanines significantly induce HIF-1α mRNA and protein, leading to elevated HIF-1 target gene expression of vascular endothelial growth factor. In contrast, polyinosinic:polycytidylic acid did not show comparable effects. Furthermore the potential to up-regulate inflammatory cytokines critically influences DC function. Our data demonstrate that HIF-1α protein is needed for adequate production of interferon-α and -ß. In co-cultures of DCs and cytotoxic T cells, we observed that DCs lacking active HIF-1α protein induce significantly less CD278 and granzyme B mRNA in T cells. We conclude that HIF-1α plays a crucial role in DC interferon production and T cell activation, linking the innate and adaptive immune system.


Subject(s)
Dendritic Cells/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interferons/biosynthesis , Animals , Blotting, Western , Cells, Cultured , Hypoxia/genetics , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Interferon Type I/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction
16.
Anesthesiology ; 118(1): 123-33, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23249930

ABSTRACT

BACKGROUND: Because the nuclear factor-κB (NF-κB) coupled pathway is believed to amplify inflammation prevailing in sepsis, the authors tested the hypotheses that the insertion-deletion polymorphism (-94ins/delATTG) (1) alters nuclear translocation of nuclear factor-κB and activator protein-1 (NF-κB1) in monocytes after lipopolysaccharide stimulation; (2) affects lipopolysaccharide-induced NF-κB1 messenger RNA expression, tumor necrosis factor α concentrations, and tissue factor activity; and (3) may be associated with increased 30-day mortality in patients with sepsis. METHODS: Nuclear translocation of NF-κB1 in monocytes after lipopolysaccharide stimulation from healthy blood donors was performed with immunofluorescence staining (n = 5 each). Lipopolysaccharide-induced NF-κB1 messenger RNA expression was measured with real-time polymerase chain reaction (PCR; n = 60), tumor necrosis factor α concentrations with a multiplexing system kit (n = 60), and tissue factor activity with thromboelastometry (n = 105). In a prospective study, multivariate proportional hazard analysis tested 30-day mortality in patients with sepsis (n = 143). METHODS AND RESULTS: The homozygous deletion genotype compared with the homozygous insertion genotype was associated with a nearly twofold increase in nuclear translocation of NF-κB1 (P = 0.001), a threefold difference in NF-κB1 messenger RNA expression (P = 0.001), and a twofold increase in tissue factor expression (P = 0.021). The deletion allele in adults with severe sepsis was tested as an independent prognostic factor for 30-day mortality (hazard ratio, 2.3; 95% CI, 1.13-4.8; P = 0.022). Mortality was 25% for homozygous insertion genotypes but 41% for combined heterozygous deletion/homozygous deletion genotypes (P = 0.034). CONCLUSION: The deletion allele of the NFκB1 insertion-deletion (-94ins/delATTG) polymorphism is associated with increased 30-day mortality in patients with severe sepsis and increased reaction of the innate immune system.


Subject(s)
Lipopolysaccharides/metabolism , NF-kappa B/genetics , Polymorphism, Genetic/genetics , Promoter Regions, Genetic/genetics , Sepsis/mortality , Translocation, Genetic/genetics , Alleles , Female , Gene Deletion , Humans , Inflammation/genetics , Lipopolysaccharides/genetics , Male , Middle Aged , Monocytes , Prospective Studies , Real-Time Polymerase Chain Reaction , Sepsis/genetics , Sepsis/metabolism
17.
J Immunol ; 185(3): 1786-93, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20574001

ABSTRACT

Myeloid cells recruited to sites of bacterial inflammation are exposed to low oxygen tension, hypoxia, and high concentrations of inflammatory cytokines that significantly affect myeloid cell function. Therefore, we analyzed the direct consequences of acute and severe hypoxia on monocytic adhesion to the endothelium in coculture experiments. Marked upregulation of monocytic ICAM-1, but no other monocytic adhesion molecule, was responsible for an approximately 50-fold increase in adhesion of the monocytic cells THP-1 to human and rat endothelial cells. ICAM-1 expression was rapidly induced after the onset of severe hypoxia, but it decreased after 4 h. Knockdown of ICAM-1 by siRNA in endothelial and monocytic cells abolished the adhesion, indicating that ICAM-1 expression on both cell types was indispensable for hypoxia-induced adhesion of monocytes to the endothelium. siRNA-mediated knockdown of hypoxia inducible factor (HIF)-1alpha, HIF-2alpha, and the NF-kappaB family member p65 revealed that hypoxic upregulation of ICAM-1 resulted from hypoxic NF-kappaB induction but not from activation of HIFs. Within the leukocyte-adhesion cascade, our results provide evidence for prolyl hydroxylase-dependent but HIF-independent activation of hypoxia-induced monocyte-endothelial adhesion and assign a new function to monocytic ICAM-1 under acute hypoxic conditions.


Subject(s)
Cell Hypoxia/immunology , Endothelium, Vascular/cytology , Endothelium, Vascular/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Intercellular Adhesion Molecule-1/biosynthesis , Monocytes/immunology , NF-kappa B/biosynthesis , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Acute Disease , Amino Acids, Dicarboxylic/pharmacology , Animals , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/physiology , Cell Adhesion/immunology , Cell Line , Cell Line, Tumor , Coculture Techniques , Endothelium, Vascular/enzymology , Enzyme Activation/drug effects , Enzyme Activation/immunology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/physiology , Monocytes/cytology , Monocytes/enzymology , Procollagen-Proline Dioxygenase/metabolism , RNA, Messenger/biosynthesis , Rats , Up-Regulation/immunology
18.
J Immunol ; 182(10): 6470-6, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19414801

ABSTRACT

The oxygen-sensitive transcription factor hypoxia-inducible factor 1 (HIF-1) is known as the key regulator of hypoxia-induced gene expression. In addition to hypoxia, endotoxins such as bacterial LPS as well as proinflammatory cytokines have been shown to induce HIF-1, suggesting an integrative role for HIF-1 in conditions of hypoxia and inflammation. Cells can become tolerant to endotoxins by repetitive exposure to LPS. Herein, we studied the effect of endotoxin tolerance on HIF-1alpha accumulation and expression of HIF target genes in human monocytic cells and primary mouse peritoneal macrophages. Tolerant cells had reduced levels of HIF-1alpha under hypoxia, which was due to lowered levels of HIF-1alpha mRNA. HIF-1alpha expression is under control of NF-kappaB and increased DNA binding of the p52 subunit of NF-kappaB was found in tolerant cells. Knock down of p52 abolished the effects of endotoxin tolerance on HIF-1alpha expression, which suggest a negative regulatory role of p52 on HIF-1alpha transcription during endotoxin tolerance. Endotoxin tolerant cells showed diminished expression of the HIF target genes phosphoglycerate kinase 1 and adrenomedullin and reduced viability under hypoxic conditions, as well as a significantly reduced invasion. Peritoneal macrophages from endotoxin-tolerant mice made showed significantly reduced HIF-1alpha protein accumulation and subsequent HIF target gene expression. We conclude that endotoxin tolerance impairs HIF-1alpha induction which reduces the ability of monocytic cells to survive and function under hypoxic conditions.


Subject(s)
Cell Hypoxia/immunology , Endotoxins/immunology , Gene Expression/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/immunology , Immune Tolerance/immunology , Monocytes/immunology , Animals , Blotting, Western , Cell Line , Cell Survival , Enzyme-Linked Immunosorbent Assay , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Monocytes/metabolism , NF-kappa B p52 Subunit/immunology , NF-kappa B p52 Subunit/metabolism , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...