Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 8 de 8
1.
Sci Immunol ; 9(95): eadi4191, 2024 May 10.
Article En | MEDLINE | ID: mdl-38728412

Conventional dendritic cells (DCs) are essential mediators of antitumor immunity. As a result, cancers have developed poorly understood mechanisms to render DCs dysfunctional within the tumor microenvironment (TME). After identification of CD63 as a specific surface marker, we demonstrate that mature regulatory DCs (mregDCs) migrate to tumor-draining lymph node tissues and suppress DC antigen cross-presentation in trans while promoting T helper 2 and regulatory T cell differentiation. Transcriptional and metabolic studies showed that mregDC functionality is dependent on the mevalonate biosynthetic pathway and its master transcription factor, SREBP2. We found that melanoma-derived lactate activates SREBP2 in tumor DCs and drives conventional DC transformation into mregDCs via homeostatic or tolerogenic maturation. DC-specific genetic silencing and pharmacologic inhibition of SREBP2 promoted antitumor CD8+ T cell activation and suppressed melanoma progression. CD63+ mregDCs were found to reside within the lymph nodes of several preclinical tumor models and in the sentinel lymph nodes of patients with melanoma. Collectively, this work suggests that a tumor lactate-stimulated SREBP2-dependent program promotes CD63+ mregDC development and function while serving as a promising therapeutic target for overcoming immune tolerance in the TME.


Dendritic Cells , Lactic Acid , Mice, Inbred C57BL , Signal Transduction , Sterol Regulatory Element Binding Protein 2 , Dendritic Cells/immunology , Animals , Mice , Humans , Sterol Regulatory Element Binding Protein 2/immunology , Lactic Acid/metabolism , Signal Transduction/immunology , Melanoma/immunology , Melanoma/pathology , Disease Progression , Immune Tolerance/immunology , Female , Cell Line, Tumor , Tumor Microenvironment/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology
2.
Signal Transduct Target Ther ; 5(1): 186, 2020 09 03.
Article En | MEDLINE | ID: mdl-32883951

Sterol regulatory element binding protein-2 (SREBP-2) is activated by cytokines or pathogen, such as virus or bacteria, but its association with diminished cholesterol levels in COVID-19 patients is unknown. Here, we evaluated SREBP-2 activation in peripheral blood mononuclear cells of COVID-19 patients and verified the function of SREBP-2 in COVID-19. Intriguingly, we report the first observation of SREBP-2 C-terminal fragment in COVID-19 patients' blood and propose SREBP-2 C-terminal fragment as an indicator for determining severity. We confirmed that SREBP-2-induced cholesterol biosynthesis was suppressed by Sestrin-1 and PCSK9 expression, while the SREBP-2-induced inflammatory responses was upregulated in COVID-19 ICU patients. Using an infectious disease mouse model, inhibitors of SREBP-2 and NF-κB suppressed cytokine storms caused by viral infection and prevented pulmonary damages. These results collectively suggest that SREBP-2 can serve as an indicator for severity diagnosis and therapeutic target for preventing cytokine storm and lung damage in severe COVID-19 patients.


Betacoronavirus/pathogenicity , Cholesterol/biosynthesis , Coronavirus Infections/genetics , Cytokine Release Syndrome/genetics , Host-Pathogen Interactions/genetics , Leukocytes, Mononuclear/immunology , Pneumonia, Viral/genetics , Sterol Regulatory Element Binding Protein 2/genetics , Betacoronavirus/immunology , COVID-19 , Case-Control Studies , Coronavirus Infections/immunology , Coronavirus Infections/mortality , Coronavirus Infections/virology , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/mortality , Cytokine Release Syndrome/virology , Gene Expression Regulation , Heat-Shock Proteins/genetics , Heat-Shock Proteins/immunology , Host-Pathogen Interactions/immunology , Humans , Intensive Care Units , Interleukin-1beta/genetics , Interleukin-1beta/immunology , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/immunology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/virology , Lung/immunology , Lung/metabolism , Lung/virology , NF-kappa B/genetics , NF-kappa B/immunology , Pandemics , Pneumonia, Viral/immunology , Pneumonia, Viral/mortality , Pneumonia, Viral/virology , Primary Cell Culture , Proprotein Convertase 9/genetics , Proprotein Convertase 9/immunology , SARS-CoV-2 , Signal Transduction , Sterol Regulatory Element Binding Protein 2/immunology , Survival Analysis , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
3.
J Biol Chem ; 291(37): 19651-60, 2016 09 09.
Article En | MEDLINE | ID: mdl-27471270

MicroRNAs (miRNAs) are short non-coding RNAs that regulate gene expression by promoting degradation and/or repressing translation of specific target mRNAs. Several miRNAs have been identified that regulate the amplitude of the innate immune response by directly targeting Toll-like receptor (TLR) pathway members and/or cytokines. miR-33a and miR-33b (the latter present in primates but absent in rodents and lower species) are located in introns of the sterol regulatory element-binding protein (SREBP)-encoding genes and control cholesterol/lipid homeostasis in concert with their host gene products. These miRNAs regulate macrophage cholesterol by targeting the lipid efflux transporters ATP binding cassette (ABC)A1 and ABCG1. We and others have previously reported that Abca1(-/-) and Abcg1(-/-) macrophages have increased TLR proinflammatory responses due to augmented lipid raft cholesterol. Given this, we hypothesized that miR-33 would augment TLR signaling in macrophages via a raft cholesterol-dependent mechanism. Herein, we report that multiple TLR ligands down-regulate miR-33 in murine macrophages. In the case of lipopolysaccharide, this is a delayed, Toll/interleukin-1 receptor (TIR) domain-containing adapter-inducing interferon-ß-dependent response that also down-regulates Srebf-2, the host gene for miR-33. miR-33 augments macrophage lipid rafts and enhances proinflammatory cytokine induction and NF-κB activation by LPS. This occurs through an ABCA1- and ABCG1-dependent mechanism and is reversible by interventions upon raft cholesterol and by ABC transporter-inducing liver X receptor agonists. Taken together, these findings extend the purview of miR-33, identifying it as an indirect regulator of innate immunity that mediates bidirectional cross-talk between lipid homeostasis and inflammation.


ATP Binding Cassette Transporter 1/immunology , ATP Binding Cassette Transporter, Subfamily G, Member 1/immunology , Immunity, Innate , Macrophages/immunology , Membrane Microdomains/immunology , MicroRNAs/immunology , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 1/genetics , Animals , Membrane Microdomains/genetics , Mice , Mice, Knockout , MicroRNAs/genetics , RAW 264.7 Cells , Sterol Regulatory Element Binding Protein 2/genetics , Sterol Regulatory Element Binding Protein 2/immunology
4.
PLoS One ; 10(8): e0136187, 2015.
Article En | MEDLINE | ID: mdl-26291618

The angiotensin converting enzyme 2-angiotensin-(1-7)-Mas axis (ACE2-Ang-(1-7)-Mas axis) is reported to participate in lipid metabolism in kidney, but its precise effects and underlying mechanisms remain unknown. We hypothesized that Ang-(1-7) reduces lipid accumulation and improves renal injury through the low density lipoprotein receptor-sterol regulatory element binding proteins 2-SREBP cleavage activating protein (LDLr-SREBP2-SCAP) system by suppressing inflammation in high fat diet (HFD)-fed mice. In this study, male C57BL/6 mice were randomized into four groups: STD (standard diet)+saline, HFD+saline, HFD+Ang-(1-7) and STD+Ang-(1-7). After 10 weeks of feeding, mice were administered Ang-(1-7) or saline for two weeks. We found that high inflammation status induced by HFD disrupted the LDLr-SREBP2-SCAP feedback system. Treatment of mice fed a high-fat diet with Ang-(1-7) induced significant improvement in inflammatory status, following the downregulation of LDLr, SREBP2 and SCAP, and then, decreased lipid deposition in kidney and improved renal injury. In conclusion, the anti-inflammatory effect of Ang-(1-7) alleviates renal injury triggered by lipid metabolic disorders through a LDLr- SREBP2-SCAP pathway.


Acute Kidney Injury/drug therapy , Angiotensin I/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Diet, High-Fat/adverse effects , Peptide Fragments/therapeutic use , Receptors, LDL/immunology , Sterol Regulatory Element Binding Protein 2/immunology , Acute Kidney Injury/etiology , Acute Kidney Injury/immunology , Acute Kidney Injury/pathology , Angiotensin I/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Dyslipidemias/drug therapy , Dyslipidemias/etiology , Dyslipidemias/immunology , Dyslipidemias/pathology , Inflammation/drug therapy , Inflammation/etiology , Inflammation/immunology , Inflammation/pathology , Kidney/drug effects , Kidney/immunology , Kidney/pathology , Lipid Metabolism/drug effects , Male , Mice, Inbred C57BL , Peptide Fragments/pharmacology , Signal Transduction/drug effects
5.
Int J Biochem Cell Biol ; 55: 196-208, 2014 Oct.
Article En | MEDLINE | ID: mdl-25218172

Establishment of infection by an intracellular pathogen depends on successful internalization with a concomitant neutralization of host defense machinery. Leishmania donovani, an intramacrophage pathogen, targets host SREBP2, a critical transcription factor, to regulate macrophage plasma membrane cholesterol and mitochondrial reactive oxygen species generation, favoring parasite invasion and persistence. Leishmania infection triggered membrane-raft reorientation-dependent Lyn-PI3K/Akt pathway activation which in turn deactivated GSK3ß to stabilize nuclear SREBP2. Moreover, cells perceiving less available intracellular cholesterol due to its sequestration at the plasma membrane resulted in the deregulation of the ER-residing SCAP-SREBP2-Insig circuit thereby assisting increased nuclear translocation of SREBP2. Both increased nuclear transport and stabilization of SREBP2 caused HMGCR-catalyzed cholesterol biosynthesis-mediated plasma membrane cholesterol enrichment leading to decreased membrane-fluidity and plausibly assisting delay in phagosomal acidification. Parasite survival ensuing entry was further ensured by SREBP2-dependent transcriptional up-regulation of UCP2, which suppressed mitochondrial ROS generation, one of the primary microbicidal molecules in macrophages recognized for its efficacy against Leishmania. Functional knock-down of SREBP2 both in vitro and in vivo was associated with reduction in macrophage plasma membrane cholesterol, increased ROS production and lower parasite survival. To our knowledge, this study, for the first time, reveals that Leishmania exploits macrophage cholesterol-dependent SREBP2 circuit to facilitate its entry and survival within the host.


Cholesterol/immunology , Leishmania donovani/immunology , Macrophages/immunology , Mitochondria/immunology , Oxidants/immunology , Sterol Regulatory Element Binding Protein 2/immunology , Animals , Blotting, Western , Cell Membrane/immunology , Cell Membrane/metabolism , Cells, Cultured , Cholesterol/metabolism , Female , Host-Parasite Interactions/immunology , Humans , Hydroxymethylglutaryl CoA Reductases/genetics , Hydroxymethylglutaryl CoA Reductases/immunology , Hydroxymethylglutaryl CoA Reductases/metabolism , Ion Channels/genetics , Ion Channels/metabolism , Leishmania donovani/physiology , Leishmaniasis, Visceral/immunology , Leishmaniasis, Visceral/metabolism , Leishmaniasis, Visceral/parasitology , Macrophages/metabolism , Macrophages/parasitology , Mice, Inbred BALB C , Mitochondria/metabolism , Mitochondria/parasitology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Oxidants/metabolism , Phosphatidylinositol 3-Kinases/immunology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/immunology , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference/immunology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/immunology , Sterol Regulatory Element Binding Protein 2/genetics , Sterol Regulatory Element Binding Protein 2/metabolism , Uncoupling Protein 2 , src-Family Kinases/immunology , src-Family Kinases/metabolism
6.
Circulation ; 128(6): 632-42, 2013 Aug 06.
Article En | MEDLINE | ID: mdl-23838163

BACKGROUND: The molecular basis for the focal nature of atherosclerotic lesions is poorly understood. Here, we explored whether disturbed flow patterns activate an innate immune response to form the NLRP3 inflammasome scaffold in vascular endothelial cells via sterol regulatory element binding protein 2 (SREBP2). METHODS AND RESULTS: Oscillatory flow activates SREBP2 and induces NLRP3 inflammasome in endothelial cells. The underlying mechanisms involve SREBP2 transactivating NADPH oxidase 2 and NLRP3. Consistently, SREBP2, NADPH oxidase 2, and NLRP3 levels were elevated in atheroprone areas of mouse aortas, suggesting that the SREBP2-activated NLRP3 inflammasome causes functionally disturbed endothelium with increased inflammation. Mimicking the effect of atheroprone flow, endothelial cell-specific overexpression of the activated form of SREBP2 synergized with hyperlipidemia to increase atherosclerosis in the atheroresistant areas of mouse aortas. CONCLUSIONS: Atheroprone flow induces NLRP3 inflammasome in endothelium through SREBP2 activation. This increased innate immunity in endothelium synergizes with hyperlipidemia to cause topographical distribution of atherosclerotic lesions.


Atherosclerosis/immunology , Carrier Proteins/immunology , Sterol Regulatory Element Binding Protein 2/immunology , Vasculitis/immunology , Animals , Apolipoproteins E/genetics , Atherosclerosis/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Endothelial Cells/immunology , Endothelial Cells/metabolism , Hemodynamics/immunology , Human Umbilical Vein Endothelial Cells , Humans , Immunity, Innate/immunology , Inflammasomes/immunology , Inflammasomes/metabolism , Male , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Mice , Mice, Transgenic , MicroRNAs/immunology , MicroRNAs/metabolism , NADPH Oxidase 2 , NADPH Oxidases/immunology , NADPH Oxidases/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , RNA, Small Interfering/genetics , Sterol Regulatory Element Binding Protein 2/genetics , Sterol Regulatory Element Binding Protein 2/metabolism , Stress, Mechanical , Vasculitis/genetics , Vasculitis/metabolism
8.
Inflamm Res ; 58(11): 809-18, 2009 Nov.
Article En | MEDLINE | ID: mdl-19533020

OBJECTIVE: To investigate if inflammatory stress increases intracellular accumulation of unmodified low-density lipoprotein (LDL) in human monocyte cell line (THP-1) macrophages by disrupting the sterol regulatory element binding proteins (SREBPs) cleavage-activating protein (SCAP)-SREBP2-mediated feedback regulation of LDL receptor. MATERIALS AND METHODS: THP-1 macrophages were incubated in serum-free medium in the absence or presence of LDL alone, LDL plus lipopolysaccharide (LPS) and LPS alone, then intracellular cholesterol content, tumor necrosis factor alpha level in the supernatants, mRNA and protein expression of LDL receptor, and SREBP2 and SCAP in the treated cells were assessed by Oil Red O staining, cholesterol enzymatic assay, enzyme-linked immunosorbent assay, real-time quantitative polymerase chain reaction, and Western blotting analysis, respectively. RESULTS: We demonstrated that LPS enhanced transformation of THP-1 macrophages into foam cells by increased uptake of unmodified LDL as evidenced by Oil Red O staining and direct assay of intracellular cholesterol. In the absence of LPS, 25 microg/ml LDL decreased LDL receptor mRNA and protein expression (p < 0.05). However, LPS enhanced LDL receptor expression, overcoming the suppression of LDL receptor induced by 25 microg/ml LDL and inappropriately increasing LDL uptake (p < 0.05). Exposure to LPS also caused overexpression of mRNA and protein of SCAP and SREBP2 (p < 0.05). These observations indicate that LPS disrupts cholesterol-mediated LDL receptor feedback regulation, permitting intracellular accumulation of unmodified LDL and causing foam-cell formation. CONCLUSION: The implication of these findings is that inflammatory stress may contribute to intracellular LDL accumulation in THP-1 macrophages without previous modification of LDL.


Foam Cells/metabolism , Inflammation/physiopathology , Lipoproteins, LDL/metabolism , Macrophages/physiology , Receptors, LDL/metabolism , Stress, Physiological , Animals , Cell Line , Foam Cells/cytology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/drug effects , Membrane Proteins/metabolism , Receptors, LDL/genetics , Sterol Regulatory Element Binding Protein 2/immunology , Tumor Necrosis Factor-alpha/immunology
...