Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 279
Filter
1.
Proc Natl Acad Sci U S A ; 118(48)2021 11 30.
Article in English | MEDLINE | ID: mdl-34810256

ABSTRACT

Global inactivation of IκB kinase (IKK)-α results in defective lymph node (LN) formation and B cell maturation, and loss of IKK-α-dependent noncanonical NF-κB signaling in stromal organizer and hematopoietic cells is thought to underlie these distinct defects. We previously demonstrated that this pathway is also activated in vascular endothelial cells (ECs). To determine the physiologic function of EC-intrinsic IKK-α, we crossed IkkαF/F mice with Tie2-cre or Cdh5-cre mice to ablate IKK-α in ECs. Notably, the compound defects of global IKK-α inactivation were recapitulated in IkkαTie2 and IkkαCdh5 mice, as both lacked all LNs and mature follicular and marginal zone B cell numbers were markedly reduced. However, as Tie2-cre and Cdh5-cre are expressed in all ECs, including blood forming hemogenic ECs, IKK-α was also absent in hematopoietic cells (HC). To determine if loss of HC-intrinsic IKK-α affected LN development, we generated IkkαVav mice lacking IKK-α in only the hematopoietic compartment. While mature B cell numbers were significantly reduced in IkkαVav mice, LN formation was intact. As lymphatic vessels also arise during development from blood ECs, we generated IkkαLyve1 mice lacking IKK-α in lymphatic ECs (LECs) to determine if IKK-α in lymphatic vessels impacts LN development. Strikingly, while mature B cell numbers were normal, LNs were completely absent in IkkαLyve1 mice. Thus, our findings reveal that IKK-α in distinct EC-derived compartments is uniquely required to promote B cell homeostasis and LN development, and we establish that LEC-intrinsic IKK-α is absolutely essential for LN formation.


Subject(s)
B-Lymphocytes/metabolism , I-kappa B Kinase/physiology , Lymph Nodes/metabolism , Animals , B-Lymphocytes/physiology , Cell Line , Endothelial Cells/metabolism , Female , Homeostasis/physiology , I-kappa B Kinase/metabolism , I-kappa B Proteins/metabolism , Lymph Nodes/physiology , Lymphoid Tissue/metabolism , Male , Mice , Mice, Inbred C57BL , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B/metabolism , Organogenesis/physiology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/metabolism
2.
Sci Rep ; 11(1): 21697, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34737366

ABSTRACT

IKKα and IKKß are essential kinases for activating NF-κB transcription factors that regulate cellular differentiation and inflammation. By virtue of their small size, chemokines support the crosstalk between cartilage and other joint compartments and contribute to immune cell chemotaxis in osteoarthritis (OA). Here we employed shRNA retroviruses to stably and efficiently ablate the expression of each IKK in primary OA chondrocytes to determine their individual contributions for monocyte chemotaxis in response to chondrocyte conditioned media. Both IKKα and IKKß KDs blunted both the monocyte chemotactic potential and the protein levels of CCL2/MCP-1, the chemokine with the highest concentration and the strongest association with monocyte chemotaxis. These findings were mirrored by gene expression analysis indicating that the lowest levels of CCL2/MCP-1 and other monocyte-active chemokines were in IKKαKD cells under both basal and IL-1ß stimulated conditions. We find that in their response to IL-1ß stimulation IKKαKD primary OA chondrocytes have reduced levels of phosphorylated NFkappaB p65pSer536 and H3pSer10. Confocal microscopy analysis revealed co-localized p65 and H3pSer10 nuclear signals in agreement with our findings that IKKαKD effectively blunts their basal level and IL-1ß dependent increases. Our results suggest that IKKα could be a novel OA disease target.


Subject(s)
I-kappa B Kinase/metabolism , Interleukin-1beta/metabolism , Monocytes/metabolism , Cells, Cultured , Chemokine CCL2/metabolism , Chemokines/immunology , Chemokines/metabolism , Chemotaxis/physiology , Chondrocytes/metabolism , Female , Humans , I-kappa B Kinase/physiology , Inflammation , Interleukin-1beta/physiology , Male , Middle Aged , NF-kappa B/metabolism , Osteoarthritis/metabolism , Phosphorylation , Protein Serine-Threonine Kinases , Signal Transduction/physiology , Transcription Factor RelA
3.
Cell Metab ; 33(6): 1171-1186.e9, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33951476

ABSTRACT

Antihyperglycemic therapy is an important priority for the treatment of type 2 diabetes (T2D). Excessive hepatic glucose production (HGP) is a major cause of fasting hyperglycemia. Therefore, a better understanding of its regulation would be important to develop effective antihyperglycemic therapies. Using a gluconeogenesis-targeted kinome screening approach combined with transcriptome analyses, we uncovered Nemo-like kinase (NLK) as a potent suppressor of HGP. Mechanistically, NLK phosphorylates and promotes nuclear export of CRTC2 and FOXO1, two key regulators of hepatic gluconeogenesis, resulting in the proteasome-dependent degradation of the former and the inhibition of the self-transcriptional activity and expression of the latter. Importantly, the expression of NLK is downregulated in the liver of individuals with diabetes and in diabetic rodent models and restoring NLK expression in the mouse model ameliorates hyperglycemia. Therefore, our findings uncover NLK as a critical player in the gluconeogenic regulatory network and as a potential therapeutic target for T2D.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Forkhead Box Protein O1/metabolism , I-kappa B Kinase/physiology , Intracellular Signaling Peptides and Proteins/physiology , Transcription Factors/metabolism , Animals , Glucose Intolerance , HEK293 Cells , Humans , Hyperglycemia , Male , Mice , Mice, Inbred C57BL
4.
Shock ; 55(3): 338-348, 2021 03 01.
Article in English | MEDLINE | ID: mdl-32925605

ABSTRACT

ABSTRACT: Cell necroptosis, a form of regulated inflammatory cell death, is one of the mechanisms that controls cell release of inflammatory mediators from innate immune cells, such as polymorphonuclear neutrophils (PMNs), and critically regulates the progress of inflammation. Cell necroptosis features receptor-interacting protein (RIPK) 1 activation and necroptosome formation. This leads to loss of plasma membrane integrity, the release of cell contents into the extracellular space, and subsequent increased inflammation. Here, we report an intra-PMN mechanism of negative regulation of necroptosis mediated through TBK1/IKKε. Using an in vivo mouse model of intratracheal injection (i.t.) of LPS and in vitro LPS stimulation of mouse PMN, we found that LPS-TLR4 signaling in PMNs activates and phosphorylates TBK1 and IKKε, which in turn suppress LPS-induced formation of the RIPK1-RIPK3-MLKL (necrosome) complex. TBK1 dysfunction by knockdown or inhibitor significantly increases the phosphorylation of RIPK1 (∼67%), RIPK3 (∼68%), and MLKL (∼50%) and promotes RIPK1-RIPK3 and RIPK3-MLKL interactions and increases PMN necroptosis (∼83%) in response to LPS, with subsequent augmented lung inflammation. These findings suggest that the LPS-TLR4-TBK1 axis serves as a negative regulator for PMN necroptosis and might be a therapeutic target for modulating PMN death and inflammation.


Subject(s)
I-kappa B Kinase/physiology , Necroptosis/physiology , Neutrophils/physiology , Protein Serine-Threonine Kinases/physiology , Animals , Lipopolysaccharides/administration & dosage , Male , Mice , Mice, Inbred C57BL , Pneumonia
5.
Biomed Pharmacother ; 127: 110138, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32387861

ABSTRACT

With the improvement of people's living standards and the change of dietary habits, Non-alcoholic fatty liver disease (NAFLD) has gradually become one of the liver diseases that endanger human health around the world. However, there are no particularly effective drugs for NAFLD in the current market. Therefore, new drug candidates which could provide high efficacy and low toxicity are needed valuable for the prevention and treatment of NAFLD. 2,3,5,4'-tetrahydroxystilbence-2-O-ß-D-glucoside (TSG) is extracted from Polygonum multiflorum Thunb., and has been widely used to treat a variety of chronic diseases in China. Recently, TSG has been reported to exert various biological activities in many studies, such as lipid-lowering, anti-inflammatory and anti-oxidant activities, which indicate that TSG may have the effect of improving NAFLD. After feeding 5% high cholesterol diet to 5 days post fertilization larval zebrafish for 10 days, hepatic steatosis larval zebrafish model was established successfully. Then the effect of TSG on the improvement of hepatic steatosis larval zebrafish was studied. Moreover, the potential mechanism of TSG on anti-NAFLD effect were studied using RT-qPCR methods from multiple pathogenesis aspects of lipogenesis, lipid-lowering, inflammation, and oxidant stress. To conclude, TSG attenuates hepatic steatosis via regulating lipid metabolism related pathway, IKKß/NF-κB anti-inflammatory pathway and Keap1-Nrf2 anti-oxidant pathway.


Subject(s)
Glucosides/pharmacology , I-kappa B Kinase/physiology , Kelch-Like ECH-Associated Protein 1/physiology , NF-E2-Related Factor 2/physiology , NF-kappa B/physiology , Non-alcoholic Fatty Liver Disease/drug therapy , Stilbenes/pharmacology , Zebrafish Proteins/physiology , Animals , Cholesterol, Dietary/administration & dosage , Disease Models, Animal , Glucosides/therapeutic use , Lipid Metabolism/drug effects , Non-alcoholic Fatty Liver Disease/metabolism , Oxidative Stress/drug effects , Signal Transduction/drug effects , Stilbenes/therapeutic use , Zebrafish
6.
Nucleic Acids Res ; 48(10): 5366-5382, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32324216

ABSTRACT

Resistance to androgen receptor (AR) targeting therapeutics in prostate cancer (PC) is a significant clinical problem. Mechanisms by which this is accomplished include AR amplification and expression of AR splice variants, demonstrating that AR remains a key therapeutic target in advanced disease. For the first time we show that IKBKE drives AR signalling in advanced PC. Significant inhibition of AR regulated gene expression was observed upon siRNA-mediated IKBKE depletion or pharmacological inhibition due to inhibited AR gene expression in multiple cell line models including a LNCaP derivative cell line resistant to the anti-androgen, enzalutamide (LNCaP-EnzR). Phenotypically, this resulted in significant inhibition of proliferation, migration and colony forming ability suggesting that targeting IKBKE could circumvent resistance to AR targeting therapies. Indeed, pharmacological inhibition in the CWR22Rv1 xenograft mouse model reduced tumour size and enhanced survival. Critically, this was validated in patient-derived explants where enzymatic inactivation of IKBKE reduced cell proliferation and AR expression. Mechanistically, we provide evidence that IKBKE regulates AR levels via Hippo pathway inhibition to reduce c-MYC levels at cis-regulatory elements within the AR gene. Thus, IKBKE is a therapeutic target in advanced PC suggesting repurposing of clinically tested IKBKE inhibitors could be beneficial to castrate resistant PC patients.


Subject(s)
I-kappa B Kinase/physiology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Androgen/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Disease Progression , Gene Expression Regulation, Neoplastic , Hippo Signaling Pathway , Humans , I-kappa B Kinase/antagonists & inhibitors , Male , Mice, Nude , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Signal Transduction , Transcription Factors/metabolism , Transcription, Genetic , YAP-Signaling Proteins
7.
Cell Rep ; 31(1): 107492, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32268090

ABSTRACT

Stimulator of Interferon Genes (STING) is a critical component of host innate immune defense but can contribute to chronic autoimmune or autoinflammatory disease. Once activated, the cyclic guanosine monophosphate (GMP)-adenosine monophosphate (AMP) (cGAMP) synthase (cGAS)-STING pathway induces both type I interferon (IFN) expression and nuclear factor-κB (NF-κB)-mediated cytokine production. Currently, these two signaling arms are thought to be mediated by a single upstream kinase, TANK-binding kinase 1 (TBK1). Here, using genetic and pharmacological approaches, we show that TBK1 alone is dispensable for STING-induced NF-κB responses in human and mouse immune cells, as well as in vivo. We further demonstrate that TBK1 acts redundantly with IκB kinase ε (IKKε) to drive NF-κB upon STING activation. Interestingly, we show that activation of IFN regulatory factor 3 (IRF3) is highly dependent on TBK1 kinase activity, whereas NF-κB is significantly less sensitive to TBK1/IKKε kinase inhibition. Our work redefines signaling events downstream of cGAS-STING. Our findings further suggest that cGAS-STING will need to be targeted directly to effectively ameliorate the inflammation underpinning disorders associated with STING hyperactivity.


Subject(s)
I-kappa B Kinase/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Female , HEK293 Cells , Humans , I-kappa B Kinase/physiology , Immunity, Innate , Interferon Regulatory Factor-3/metabolism , Interferon-beta/metabolism , Male , Membrane Proteins/metabolism , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Myeloid Cells/metabolism , NF-kappa B/metabolism , Nucleotides, Cyclic/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/physiology , Signal Transduction/immunology
8.
Atherosclerosis ; 292: 23-30, 2020 01.
Article in English | MEDLINE | ID: mdl-31733453

ABSTRACT

BACKGROUND AND AIMS: IKKα is an important regulator of gene expression. As IKKα kinase inactivity in bone marrow-derived cells does not affect atherosclerosis, we here investigate the impact of a whole body-IKKα kinase inactivity on atherosclerosis. METHODS: Apolipoprotein E (Apoe)-deficient mice homozygous for an activation-resistant Ikkα-mutant (IkkαAA/AAApoe-/-) and Ikkα+/+Apoe-/- controls received a Western-type diet. Atherosclerotic lesion size and cellular content were analyzed using histology and immunofluorescence. Vascular protein expression and IKKα kinase activity were quantified by Luminex multiplex immuno-assay and ELISA. RESULTS: A vascular site-specific IKKα expression and kinase activation profile was revealed, with higher total IKKα protein levels in aortic root but increased IKKα phosphorylation, representing activated IKKα, in the aortic arch. This was associated with a vascular site-specific effect of IkkαAA/AA knock-in on atherosclerosis: in the aortic root, IkkαAA/AA knock-in decreased lesion size by 22.0 ±â€¯7.7% (p < 0.01), reduced absolute lesional smooth muscle cell numbers and lowered pro-atherogenic MMP2. In contrast, IkkαAA/AA knock-in increased lesion size in the aortic arch by 43.7 ±â€¯20.1% (p < 0.001), increased the abundance of lesional smooth muscle cells in brachiocephalic artery as main arch side branch and elevated MMP2. A similar profile was observed for MMP3. No effects were observed on necrotic core or collagen deposition in atherosclerotic lesions, nor on absolute lesional macrophage numbers. CONCLUSIONS: A non-activatable IKKα kinase differentially affects atherosclerosis in aortic root vs. aortic arch/brachiocephalic artery, associated with a differential vascular IKKα expression and kinase activation profile as well as with a vascular site-dependent impact on lesional smooth muscle cell accumulation and protein expression profiles.


Subject(s)
Atherosclerosis/etiology , I-kappa B Kinase/physiology , Animals , Apolipoproteins E/deficiency , Atherosclerosis/metabolism , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/genetics , Mice , Mutation
9.
Exp Gerontol ; 128: 110743, 2019 12.
Article in English | MEDLINE | ID: mdl-31629801

ABSTRACT

Parkinson's disease is the second most common neurodegenerative disease. Its main pathological feature is the substantial nigra-striatum dopaminergic neuronal dysfunction, which causes insufficient release of DA, induces motor symptoms, and is accompanied by nonmotor symptoms. Schisandrol A belongs to lignan components and has anti-inflammatory, antioxidant and neuroprotective effects. In this experiment, we injected 6-OHDA into medial forebrain bundle of C57BL/6J male mice to establish the model. The motor function of mice was examined by open field test and pole test, the depression-like behavior of mice was examined by sucrose preference test and the memory function was examined by Y maze. We found that schisandrol A (20 mg/kg/d) could significantly improve the motor symptoms, and alleviate the depression-like symptoms and memory dysfunction of PD mice induced by 6-OHDA. Then we studied the neuroprotective mechanism of schisandrol A by H.E., ELISA assay kits and Western blot. Results showed that schisandrol A may enhance the PI3K/AKT pathway, inhibit the IKK/IκBα/NF-κB pathway, reduce neuronal inflammation and oxidative stress, and enhance the survival of DA neurons in the brain of mice. These results indicate that schisandrol A is expected to be a potential drug for improving Parkinson's disease.


Subject(s)
Cyclooctanes/pharmacology , I-kappa B Kinase/physiology , Lignans/pharmacology , NF-KappaB Inhibitor alpha/physiology , NF-kappa B/physiology , Neuroprotective Agents/pharmacology , Parkinson Disease/drug therapy , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , Animals , Corpus Striatum/drug effects , Corpus Striatum/pathology , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , Oxidopamine , Signal Transduction/drug effects
10.
Proc Natl Acad Sci U S A ; 116(38): 19090-19097, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31481626

ABSTRACT

Aberrant T cell development is a pivotal risk factor for autoimmune disease; however, the underlying molecular mechanism of T cell overactivation is poorly understood. Here, we identified NF-κB-inducing kinase (NIK) and IkB kinase α (IKKα) in thymic epithelial cells (TECs) as essential regulators of T cell development. Mouse TEC-specific ablation of either NIK or IKKα resulted in severe T cell-mediated inflammation, injury, and fibrosis in the liver and lung, leading to premature death within 18 d of age. NIK or IKKα deficiency abrogated medullary TEC development, and led to breakdown of central tolerance, production of autoreactive T cells, and fatal autoimmune destruction in the liver and lung. TEC-specific ablation of NIK or IKKα also impaired thymic T cell development from the double-negative through the double-positive stages and inhibited peripheral B cell development. These results unravel a hitherto unrecognized essential role of TEC-intrinsic NIK and IKKα pathways in autoimmunity and T cell-instigated chronic liver and lung diseases.


Subject(s)
Autoimmunity/immunology , I-kappa B Kinase/physiology , Inflammation/immunology , Liver/immunology , Lung/immunology , Protein Serine-Threonine Kinases/physiology , Thymus Gland/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , Cell Differentiation , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Homeostasis , Inflammation/metabolism , Inflammation/pathology , Liver/metabolism , Liver/pathology , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Thymus Gland/metabolism , Thymus Gland/pathology , NF-kappaB-Inducing Kinase
11.
J Virol ; 93(12)2019 06 15.
Article in English | MEDLINE | ID: mdl-30944180

ABSTRACT

Equine arteritis virus (EAV) and porcine reproductive and respiratory syndrome virus (PRRSV) represent two members of the family Arteriviridae and pose major threats for the horse- and swine-breeding industries worldwide. A previous study suggested that PRRSV nsp4, a 3C-like protease, antagonizes interferon beta (IFN-ß) production by cleaving the NF-κB essential modulator (NEMO) at a single site, glutamate 349 (E349). Here, we demonstrated that EAV nsp4 also inhibited virus-induced IFN-ß production by targeting NEMO for proteolytic cleavage and that the scission occurred at four sites: E166, E171, glutamine 205 (Q205), and E349. Additionally, we found that, besides the previously reported cleavage site E349 in NEMO, scission by PRRSV nsp4 took place at two additional sites, E166 and E171. These results imply that while cleaving NEMO is a common strategy utilized by EAV and PRRSV nsp4 to antagonize IFN induction, EAV nsp4 adopts a more complex substrate recognition mechanism to target NEMO. By analyzing the abilities of the eight different NEMO fragments resulting from EAV or PRRSV nsp4 scission to induce IFN-ß production, we serendipitously found that a NEMO fragment (residues 1 to 349) could activate IFN-ß transcription more robustly than full-length NEMO, whereas all other NEMO cleavage products were abrogated for the IFN-ß-inducing capacity. Thus, NEMO cleavage at E349 alone may not be sufficient to completely inactivate the IFN response via this signaling adaptor. Altogether, our findings suggest that EAV and PRRSV nsp4 cleave NEMO at multiple sites and that this strategy is critical for disarming the innate immune response for viral survival.IMPORTANCE The arterivirus nsp4-encoded 3C-like protease (3CLpro) plays an important role in virus replication and immune evasion, making it an attractive target for antiviral therapeutics. Previous work suggested that PRRSV nsp4 suppresses type I IFN production by cleaving NEMO at a single site. In contrast, the present study demonstrates that both EAV and PRRSV nsp4 cleave NEMO at multiple sites and that this strategy is essential for disruption of type I IFN production. Moreover, we reveal that EAV nsp4 also cleaves NEMO at glutamine 205 (Q205), which is not targeted by PRRSV nsp4. Notably, targeting a glutamine in NEMO for cleavage has been observed only with picornavirus 3C proteases (3Cpro) and coronavirus 3CLpro In aggregate, our work expands knowledge of the innate immune evasion mechanisms associated with NEMO cleavage by arterivirus nsp4 and describes a novel substrate recognition characteristic of EAV nsp4.


Subject(s)
Equartevirus/metabolism , Interferon-beta/biosynthesis , Viral Nonstructural Proteins/metabolism , Animals , Arteriviridae/metabolism , Arterivirus/metabolism , Cell Line , Equartevirus/physiology , HEK293 Cells , Horses , Humans , I-kappa B Kinase/metabolism , I-kappa B Kinase/physiology , Immune Evasion , Immunity, Innate , Interferon-beta/metabolism , Porcine respiratory and reproductive syndrome virus/metabolism , Proteolysis , Signal Transduction , Swine , Virus Replication
12.
Nat Commun ; 10(1): 1729, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30988283

ABSTRACT

RIPK1 regulates cell death and inflammation through kinase-dependent and -independent mechanisms. As a scaffold, RIPK1 inhibits caspase-8-dependent apoptosis and RIPK3/MLKL-dependent necroptosis. As a kinase, RIPK1 paradoxically induces these cell death modalities. The molecular switch between RIPK1 pro-survival and pro-death functions remains poorly understood. We identify phosphorylation of RIPK1 on Ser25 by IKKs as a key mechanism directly inhibiting RIPK1 kinase activity and preventing TNF-mediated RIPK1-dependent cell death. Mimicking Ser25 phosphorylation (S > D mutation) protects cells and mice from the cytotoxic effect of TNF in conditions of IKK inhibition. In line with their roles in IKK activation, TNF-induced Ser25 phosphorylation of RIPK1 is defective in TAK1- or SHARPIN-deficient cells and restoring phosphorylation protects these cells from TNF-induced death. Importantly, mimicking Ser25 phosphorylation compromises the in vivo cell death-dependent immune control of Yersinia infection, a physiological model of TAK1/IKK inhibition, and rescues the cell death-induced multi-organ inflammatory phenotype of the SHARPIN-deficient mice.


Subject(s)
Apoptosis , Models, Immunological , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Animals , Caspase 8/genetics , Caspase 8/metabolism , Caspase 8/physiology , Cell Line , I-kappa B Kinase/metabolism , I-kappa B Kinase/physiology , Immunity/physiology , Mice , Phosphorylation , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Serine/chemistry , Serine/metabolism , Yersinia , Yersinia Infections/immunology
13.
FASEB J ; 33(6): 7451-7466, 2019 06.
Article in English | MEDLINE | ID: mdl-30884248

ABSTRACT

Development of physiologic cardiac hypertrophy has primarily been ascribed to the IGF-1 and its receptor, IGF-1 receptor (IGF-1R), and subsequent activation of the protein kinase B (Akt) pathway. However, regulation of endosome-mediated recycling and degradation of IGF-1R during physiologic hypertrophy has not been investigated. In a physiologic hypertrophy model of treadmill-exercised mice, we observed that levels of tumor susceptibility gene 101 (Tsg101), a key member of the endosomal sorting complex required for transport, were dramatically elevated in the heart compared with sedentary controls. To determine the role of Tsg101 on physiologic hypertrophy, we generated a transgenic (TG) mouse model with cardiac-specific overexpression of Tsg101. These TG mice exhibited a physiologic-like cardiac hypertrophy phenotype at 8 wk evidenced by: 1) the absence of cardiac fibrosis, 2) significant improvement of cardiac function, and 3) increased total and plasma membrane levels of IGF-1R and increased phosphorylation of Akt. Mechanistically, we identified that Tsg101 interacted with family-interacting protein 3 (FIP3) and IGF-1R, thereby stabilizing FIP3 and enhancing recycling of IGF-1R. In vitro, adenovirus-mediated overexpression of Tsg101 in neonatal rat cardiomyocytes resulted in cell hypertrophy, which was blocked by addition of monensin, an inhibitor of endosome-mediated recycling, and by small interfering RNA-mediated knockdown (KD) of FIP3. Furthermore, cardiac-specific KD of Tsg101 showed a significant reduction in levels of endosomal recycling compartment members (Rab11a and FIP3), IGF-1R, and Akt phosphorylation. Most interestingly, Tsg101-KD mice failed to develop cardiac hypertrophy after intense treadmill training. Taken together, our data identify Tsg101 as a novel positive regulator of physiologic cardiac hypertrophy through facilitating the FIP3-mediated endosomal recycling of IGF-1R.-Essandoh, K., Deng, S., Wang, X., Jiang, M., Mu, X., Peng, J., Li, Y., Peng, T., Wagner, K.-U., Rubinstein, J., Fan, G.-C. Tsg101 positively regulates physiologic-like cardiac hypertrophy through FIP3-mediated endosomal recycling of IGF-1R.


Subject(s)
Cardiomegaly/physiopathology , DNA-Binding Proteins/physiology , Endosomal Sorting Complexes Required for Transport/physiology , Endosomes/metabolism , I-kappa B Kinase/physiology , Receptor, IGF Type 1/metabolism , Transcription Factors/physiology , Animals , Female , Gene Expression Profiling , Male , Mice , Rats
14.
Arterioscler Thromb Vasc Biol ; 39(4): 719-730, 2019 04.
Article in English | MEDLINE | ID: mdl-30816805

ABSTRACT

Objective- TFEB (transcription factor EB) was recently reported to be induced by atheroprotective laminar flow and play an anti-atherosclerotic role by inhibiting inflammation in endothelial cells (ECs). This study aims to investigate whether TFEB regulates endothelial inflammation in diabetic db/db mice and the molecular mechanisms involved. Approach and Results- Endothelial denudation shows that TFEB is mainly expressed in ECs in mouse aortas. Western blotting shows TFEB total protein level decreases whereas the p-TFEB S142 (phosphorylated form of TFEB) increases in db/db mouse aortas, suggesting a decreased TFEB activity. Adenoviral TFEB overexpression reduces endothelial inflammation as evidenced by decreased expression of vascular inflammatory markers in db/db mouse aortas, and reduced expression of a wide range of adhesion molecules and chemokines in human umbilical vein ECs. Monocyte attachment assay shows TFEB suppresses monocyte adhesion to human umbilical vein ECs. RNA sequencing of TFEB-overexpressed human umbilical vein ECs suggested TFEB inhibits NF-κB (nuclear factor-kappa B) signaling. Indeed, luciferase assay shows TFEB suppresses NF-κB transcriptional activity. Mechanistically, TFEB suppresses IKK (IκB kinase) activity to protect IκB-α from degradation, leading to reduced p65 nuclear translocation. Inhibition of IKK by PS-1145 abolished TFEB silencing-induced inflammation in human umbilical vein ECs. Lastly, we identified KLF2 (Krüppel-like factor 2) upregulates TFEB expression and promoter activity. Laminar flow experiment showed that KLF2 is required for TFEB induction by laminar flow and TFEB is an anti-inflammatory effector downstream of laminar flow-KLF2 signaling in ECs. Conclusions- These findings suggest that TFEB exerts anti-inflammatory effects in diabetic mice and such function in ECs is achieved by inhibiting IKK activity and increasing IκBα level to suppress NF-κB activity. KLF2 mediates TFEB upregulation in response to laminar flow.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , Diabetic Angiopathies/prevention & control , Endothelial Cells/metabolism , Gene Expression Regulation/physiology , I-kappa B Kinase/physiology , Signal Transduction/physiology , Transcription Factor RelA/physiology , Animals , Aorta/metabolism , Cell Adhesion , Diabetes Mellitus, Type 2/genetics , Diabetic Angiopathies/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Inflammation , Kruppel-Like Transcription Factors/physiology , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Physical Conditioning, Animal , Receptors, Leptin/deficiency , Recombinant Proteins/metabolism , Transcription, Genetic
15.
Actas Dermosifiliogr (Engl Ed) ; 110(4): 273-278, 2019 May.
Article in English, Spanish | MEDLINE | ID: mdl-30660327

ABSTRACT

Incontinentia pigmenti (Bloch-Sulzberger syndrome) is a rare neuroectodermal dysplasia. It is an X-linked dominant disorder caused by mutations in the IKBKG/NEMO gene on Xq28. Approximately 80% of patients have a deletion of exons 4 to 10. Incontinentia pigmenti has an estimated incidence of 0.7 cases per 100,000 births. In hemizygous males, it is usually lethal, while in females, it has a wide spectrum of clinical manifestations. Incontinentia pigmenti is a multisystemic disease that invariably features skin changes. These changes are the main diagnostic criteria and they evolve in 4 stages, in association with other abnormalities affecting the central nervous system, eyes, teeth, mammary glands, hair, nails, skin, and other parts of the body. The aim of this brief review is to highlight the clinical features of this genodermatosis and underline the importance of case-by-case interdisciplinary management, including genetic counseling.


Subject(s)
Incontinentia Pigmenti , Diagnosis, Differential , Disease Management , Female , Genes, X-Linked , Genotype , Humans , I-kappa B Kinase/deficiency , I-kappa B Kinase/physiology , Incontinentia Pigmenti/epidemiology , Incontinentia Pigmenti/genetics , Incontinentia Pigmenti/pathology , Incontinentia Pigmenti/therapy , Male , Organ Specificity , Phenotype , Precision Medicine , Sequence Deletion , Skin/pathology
16.
FASEB J ; 33(2): 2359-2371, 2019 02.
Article in English | MEDLINE | ID: mdl-30285578

ABSTRACT

Experimental nephrotoxic serum nephritis (NTN) is a model for T-cell-mediated human rapid progressive glomerulonephritis. T-cell receptor stimulation involves intracellular signaling events that ultimately lead to the activation of transcription factors, such as NF-κB. We explored the involvement of the NF-κB components IKK-2 and NEMO in NTN, by using cell-specific knockouts of IKK-2 and NEMO in CD4+ T lymphocytes. Our results demonstrate that although the course of disease was not grossly altered in CD4xIKK2Δ and CD4xNEMOΔ animals, renal regulatory T cells were significantly reduced and T helper (Th)1 and Th17 cells significantly increased in both knockout mouse groups. The expression of the renal cytokines and chemokines IL-1ß, CCL-2, and CCL-20 was also significantly altered in both knockout mice. Lymphocyte transcriptome analysis confirmed the increased expression of Th17-related cytokines in spleen CD4+ T cells. Moreover, our array data demonstrate an interrupted canonical NF-κB pathway and an increased expression of noncanonical NF-κB pathway-related genes in nephritic CD4xNEMOΔ mice, highlighting different downstream effects of deletion of IKK-2 or NEMO in T lymphocytes. We propose that better understanding of the role of IKK-2 and NEMO in nephritis is essential for the clinical application of kinase inhibitors in patients with glomerulonephritis.-Guo, L., Huang, J., Chen, M., Piotrowski, E., Song, N., Zahner, G., Paust, H.-J., Alawi, M., Geffers, R., Thaiss, F. T-lymphocyte-specific knockout of IKK-2 or NEMO induces Th17 cells in an experimental nephrotoxic nephritis mouse model.


Subject(s)
Disease Models, Animal , I-kappa B Kinase/physiology , Intracellular Signaling Peptides and Proteins/physiology , Nephritis/pathology , T-Lymphocytes/metabolism , Th17 Cells/immunology , Animals , Cells, Cultured , Cytokines/metabolism , Male , Mice , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/metabolism , Nephritis/chemically induced , Nephritis/immunology , Phosphorylation , Signal Transduction , Th17 Cells/metabolism , Th17 Cells/pathology
17.
Elife ; 72018 08 02.
Article in English | MEDLINE | ID: mdl-30070632

ABSTRACT

Reparative hepatocyte replication is impaired in chronic liver disease, contributing to disease progression; however, the underlying mechanism remains elusive. Here, we identify Map3k14 (also known as NIK) and its substrate Chuk (also called IKKα) as unrecognized suppressors of hepatocyte replication. Chronic liver disease is associated with aberrant activation of hepatic NIK pathways. We found that hepatocyte-specific deletion of Map3k14 or Chuk substantially accelerated mouse hepatocyte proliferation and liver regeneration following partial-hepatectomy. Hepatotoxin treatment or high fat diet feeding inhibited the ability of partial-hepatectomy to stimulate hepatocyte replication; remarkably, inactivation of hepatic NIK markedly increased reparative hepatocyte proliferation under these liver disease conditions. Mechanistically, NIK and IKKα suppressed the mitogenic JAK2/STAT3 pathway, thereby inhibiting cell cycle progression. Our data suggest that hepatic NIK and IKKα act as rheostats for liver regeneration by restraining overgrowth. Pathological activation of hepatic NIK or IKKα likely blocks hepatocyte replication, contributing to liver disease progression.


Subject(s)
End Stage Liver Disease/pathology , I-kappa B Kinase/physiology , Liver Failure, Acute/pathology , Liver Regeneration , Protein Serine-Threonine Kinases/physiology , Animals , Cells, Cultured , End Stage Liver Disease/etiology , End Stage Liver Disease/metabolism , Hepatectomy/adverse effects , Hepatocytes , Janus Kinase 2/metabolism , Liver Failure, Acute/etiology , Liver Failure, Acute/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Phosphorylation , STAT3 Transcription Factor/metabolism , Signal Transduction , NF-kappaB-Inducing Kinase
18.
Proc Natl Acad Sci U S A ; 115(4): E812-E821, 2018 01 23.
Article in English | MEDLINE | ID: mdl-29311298

ABSTRACT

Lung adenocarcinoma (ADC) and squamous cell carcinoma (SCC) are two distinct and predominant types of human lung cancer. IκB kinase α (IKKα) has been shown to suppress lung SCC development, but its role in ADC is unknown. We found inactivating mutations and homologous or hemizygous deletions in the CHUK locus, which encodes IKKα, in human lung ADCs. The CHUK deletions significantly reduced the survival time of patients with lung ADCs harboring KRAS mutations. In mice, lung-specific Ikkα ablation (IkkαΔLu ) induces spontaneous ADCs and promotes KrasG12D-initiated ADC development, accompanied by increased cell proliferation, decreased cell senescence, and reactive oxygen species (ROS) accumulation. IKKα deletion up-regulates NOX2 and down-regulates NRF2, leading to ROS accumulation and blockade of cell senescence induction, which together accelerate ADC development. Pharmacologic inhibition of NADPH oxidase or ROS impairs KrasG12D-mediated ADC development in IkkαΔLu mice. Therefore, IKKα modulates lung ADC development by controlling redox regulatory pathways. This study demonstrates that IKKα functions as a suppressor of lung ADC in human and mice through a unique mechanism that regulates tumor cell-associated ROS metabolism.


Subject(s)
Adenocarcinoma/genetics , I-kappa B Kinase/physiology , Lung Neoplasms/genetics , Acetophenones , Acetylcysteine , Adenocarcinoma/metabolism , Animals , Cell Proliferation , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Epigenesis, Genetic , Humans , Lung Neoplasms/metabolism , Mice , NADPH Oxidase 2/metabolism , NF-E2-Related Factor 2/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/metabolism
19.
Leukemia ; 31(7): 1532-1539, 2017 07.
Article in English | MEDLINE | ID: mdl-28232743

ABSTRACT

Drug resistance to BCR-ABL1 tyrosine kinase inhibitor (TKI) and disease progression to blast crisis (BC) are major clinical problems in chronic myeloid leukemia (CML); however, underlying mechanisms governing this process remain to be elucidated. Here, we report Cordon-bleu protein-like 1 (Cobll1) as a distinct molecular marker associated with drug resistance as well as progression to BC. In detail, Cobll1 increases IKKγ stability, leading to NF-κB activation and reduction of nilotinib-dependent apoptosis, suggesting Cobll1-mediated NF-κB could be involved in drug resistance. Recently, NF-κB signalling has been highlighted as a core mechanism for chronic phase (CP)-BC progression, stem cell survival and tyrosine kinase inhibitor resistance. We also demonstrated that high expression of Cobll1 confers drug resistance to tyrosine kinase inhibitors in CML cell line as well as patient samples. The analysis of large sets of primary CML samples (n=90) shows that Cobll1 expression is dramatically increased in BC but not in CP, which is correlated with a poor survival rate (P=0.002). Moreover, our studies show that Cobll1 is highly expressed in CD34+ primitive stem cell populations, and the zebrafish paralog Cobll1b is important for normal hematopoiesis during embryonic development. Based on these results, we propose that Cobll1 is a novel biomarker and potential therapeutic target for CML-BC.


Subject(s)
Blast Crisis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Transcription Factors/physiology , Cell Line, Tumor , Drug Resistance, Neoplasm , Fusion Proteins, bcr-abl/physiology , Humans , I-kappa B Kinase/physiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/mortality , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , MicroRNAs/physiology , NF-kappa B/physiology , Pyrimidines/therapeutic use
20.
J Exp Med ; 214(2): 423-437, 2017 02.
Article in English | MEDLINE | ID: mdl-28082356

ABSTRACT

Inhibition of the IκB kinase complex (IKK) has been implicated in the therapy of several chronic inflammatory diseases including inflammatory bowel diseases. In this study, using mice with an inactivatable IKKα kinase (IkkαAA/AA), we show that loss of IKKα function markedly impairs epithelial regeneration in a model of acute colitis. Mechanistically, this is caused by compromised secretion of cytoprotective IL-18 from IKKα-mutant intestinal epithelial cells because of elevated caspase 12 activation during an enhanced unfolded protein response (UPR). Induction of the UPR is linked to decreased ATG16L1 stabilization in IkkαAA/AA mice. We demonstrate that both TNF-R and nucleotide-binding oligomerization domain stimulation promote ATG16L1 stabilization via IKKα-dependent phosphorylation of ATG16L1 at Ser278. Thus, we propose IKKα as a central mediator sensing both cytokine and microbial stimulation to suppress endoplasmic reticulum stress, thereby assuring antiinflammatory function during acute intestinal inflammation.


Subject(s)
Carrier Proteins/metabolism , I-kappa B Kinase/physiology , Inflammation/metabolism , Animals , Autophagy-Related Proteins , Carrier Proteins/chemistry , Caspase 12/physiology , Colitis/prevention & control , Endoplasmic Reticulum Stress , Endoribonucleases/physiology , Interleukin-18/metabolism , Mice , NF-kappa B/physiology , Nod2 Signaling Adaptor Protein/physiology , Protein Serine-Threonine Kinases/physiology , Protein Stability , Unfolded Protein Response
SELECTION OF CITATIONS
SEARCH DETAIL