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










Publication year range
1.
Nature ; 612(7939): 347-353, 2022 12.
Article in English | MEDLINE | ID: mdl-36385525

ABSTRACT

Solid cancers exhibit a dynamic balance between cell death and proliferation ensuring continuous tumour maintenance and growth1,2. Increasing evidence links enhanced cancer cell apoptosis to paracrine activation of cells in the tumour microenvironment initiating tissue repair programs that support tumour growth3,4, yet the direct effects of dying cancer cells on neighbouring tumour epithelia and how this paracrine effect potentially contributes to therapy resistance are unclear. Here we demonstrate that chemotherapy-induced tumour cell death in patient-derived colorectal tumour organoids causes ATP release triggering P2X4 (also known as P2RX4) to mediate an mTOR-dependent pro-survival program in neighbouring cancer cells, which renders surviving tumour epithelia sensitive to mTOR inhibition. The induced mTOR addiction in persisting epithelial cells is due to elevated production of reactive oxygen species and subsequent increased DNA damage in response to the death of neighbouring cells. Accordingly, inhibition of the P2X4 receptor or direct mTOR blockade prevents induction of S6 phosphorylation and synergizes with chemotherapy to cause massive cell death induced by reactive oxygen species and marked tumour regression that is not seen when individually applied. Conversely, scavenging of reactive oxygen species prevents cancer cells from becoming reliant on mTOR activation. Collectively, our findings show that dying cancer cells establish a new dependency on anti-apoptotic programs in their surviving neighbours, thereby creating an opportunity for combination therapy in P2X4-expressing epithelial tumours.


Subject(s)
Colonic Neoplasms , Organoids , Humans , Reactive Oxygen Species , Cause of Death , Cell Death , Tumor Microenvironment , TOR Serine-Threonine Kinases
3.
Cancer Cell ; 40(2): 168-184.e13, 2022 02 14.
Article in English | MEDLINE | ID: mdl-35120600

ABSTRACT

Standard cancer therapy targets tumor cells without considering possible damage on the tumor microenvironment that could impair therapy response. In rectal cancer patients we find that inflammatory cancer-associated fibroblasts (iCAFs) are associated with poor chemoradiotherapy response. Employing a murine rectal cancer model or patient-derived tumor organoids and primary stroma cells, we show that, upon irradiation, interleukin-1α (IL-1α) not only polarizes cancer-associated fibroblasts toward the inflammatory phenotype but also triggers oxidative DNA damage, thereby predisposing iCAFs to p53-mediated therapy-induced senescence, which in turn results in chemoradiotherapy resistance and disease progression. Consistently, IL-1 inhibition, prevention of iCAFs senescence, or senolytic therapy sensitizes mice to irradiation, while lower IL-1 receptor antagonist serum levels in rectal patients correlate with poor prognosis. Collectively, we unravel a critical role for iCAFs in rectal cancer therapy resistance and identify IL-1 signaling as an attractive target for stroma-repolarization and prevention of cancer-associated fibroblasts senescence.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Drug Resistance, Neoplasm , Rectal Neoplasms/metabolism , Tumor Microenvironment , Animals , Biomarkers , Cancer-Associated Fibroblasts/pathology , Cell Line, Tumor , Cellular Senescence/drug effects , Cellular Senescence/genetics , Cytokines/genetics , Cytokines/metabolism , DNA Damage , Disease Models, Animal , Disease Susceptibility , Gene Expression Profiling , Heterografts , High-Throughput Nucleotide Sequencing , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Mice , Neoadjuvant Therapy , Prognosis , Rectal Neoplasms/drug therapy , Rectal Neoplasms/etiology , Rectal Neoplasms/pathology , Signal Transduction , Tumor Microenvironment/genetics
4.
Cell ; 174(1): 88-101.e16, 2018 06 28.
Article in English | MEDLINE | ID: mdl-29909986

ABSTRACT

In colorectal cancer patients, a high density of cytotoxic CD8+ T cells in tumors is associated with better prognosis. Using a Stat3 loss-of-function approach in two wnt/ß-catenin-dependent autochthonous models of sporadic intestinal tumorigenesis, we unravel a complex intracellular process in intestinal epithelial cells (IECs) that controls the induction of a CD8+ T cell based adaptive immune response. Elevated mitophagy in IECs causes iron(II)-accumulation in epithelial lysosomes, in turn, triggering lysosomal membrane permeabilization. Subsequent release of proteases into the cytoplasm augments MHC class I presentation and activation of CD8+ T cells via cross-dressing of dendritic cells. Thus, our findings highlight a so-far-unrecognized link between mitochondrial function, lysosomal integrity, and MHC class I presentation in IECs and suggest that therapies triggering mitophagy or inducing LMP in IECs may prove successful in shifting the balance toward anti-tumor immunity in colorectal cancer.


Subject(s)
Adaptive Immunity , Mitophagy , Adaptive Immunity/drug effects , Animals , Azoxymethane/toxicity , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cell Membrane Permeability , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Cytokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Ferrous Compounds/metabolism , Humans , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Lysosomes/metabolism , Male , Mice , Mice, Knockout , Mitophagy/drug effects , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Survival Rate
5.
Nature ; 514(7523): 508-12, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25174708

ABSTRACT

Several features common to a Western lifestyle, including obesity and low levels of physical activity, are known risk factors for gastrointestinal cancers. There is substantial evidence suggesting that diet markedly affects the composition of the intestinal microbiota. Moreover, there is now unequivocal evidence linking dysbiosis to cancer development. However, the mechanisms by which high-fat diet (HFD)-mediated changes in the microbial community affect the severity of tumorigenesis in the gut remain to be determined. Here we demonstrate that an HFD promotes tumour progression in the small intestine of genetically susceptible, K-ras(G12Dint), mice independently of obesity. HFD consumption, in conjunction with K-ras mutation, mediated a shift in the composition of the gut microbiota, and this shift was associated with a decrease in Paneth-cell-mediated antimicrobial host defence that compromised dendritic cell recruitment and MHC class II molecule presentation in the gut-associated lymphoid tissues. When butyrate was administered to HFD-fed K-ras(G12Dint) mice, dendritic cell recruitment in the gut-associated lymphoid tissues was normalized, and tumour progression was attenuated. Importantly, deficiency in MYD88, a signalling adaptor for pattern recognition receptors and Toll-like receptors, blocked tumour progression. The transfer of faecal samples from HFD-fed mice with intestinal tumours to healthy adult K-ras(G12Dint) mice was sufficient to transmit disease in the absence of an HFD. Furthermore, treatment with antibiotics completely blocked HFD-induced tumour progression, suggesting that distinct shifts in the microbiota have a pivotal role in aggravating disease. Collectively, these data underscore the importance of the reciprocal interaction between host and environmental factors in selecting a microbiota that favours carcinogenesis, and they suggest that tumorigenesis is transmissible among genetically predisposed individuals.


Subject(s)
Carcinogenesis/drug effects , Diet, High-Fat/adverse effects , Dietary Fats/adverse effects , Dysbiosis/chemically induced , Dysbiosis/microbiology , Intestinal Neoplasms/microbiology , Obesity , Animals , Anti-Bacterial Agents/pharmacology , Butyrates/pharmacology , Disease Progression , Intestinal Mucosa/immunology , Intestinal Neoplasms/chemically induced , Intestines/drug effects , Intestines/microbiology , Mice , Obesity/chemically induced , Obesity/microbiology , Prebiotics
6.
Cell Rep ; 7(6): 1914-25, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-24882009

ABSTRACT

The recruitment of immune cells into solid tumors is an essential prerequisite of tumor development. Depending on the prevailing polarization profile of these infiltrating leucocytes, tumorigenesis is either promoted or blocked. Here, we identify IκB kinase α (IKKα) as a central regulator of a tumoricidal microenvironment during intestinal carcinogenesis. Mice deficient in IKKα kinase activity are largely protected from intestinal tumor development that is dependent on the enhanced recruitment of interferon γ (IFNγ)-expressing M1-like myeloid cells. In IKKα mutant mice, M1-like polarization is not controlled in a cell-autonomous manner but, rather, depends on the interplay of both IKKα mutant tumor epithelia and immune cells. Because therapies aiming at the tumor microenvironment rather than directly at the mutated cancer cell may circumvent resistance development, we suggest IKKα as a promising target for colorectal cancer (CRC) therapy.


Subject(s)
Carcinogenesis/metabolism , I-kappa B Kinase/metabolism , Intestines/immunology , Killer Cells, Natural/pathology , Myeloid Cells/cytology , Myeloid Cells/enzymology , Animals , CD4-Positive T-Lymphocytes/enzymology , CD4-Positive T-Lymphocytes/pathology , Carcinogenesis/pathology , Cell Polarity , Cell Transformation, Neoplastic , HEK293 Cells , Humans , Killer Cells, Natural/enzymology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cells/pathology , Phosphorylation , Signal Transduction
7.
J Clin Invest ; 123(5): 2231-43, 2013 May.
Article in English | MEDLINE | ID: mdl-23563314

ABSTRACT

Chronic pancreatitis is an inflammatory disease that causes progressive destruction of pancreatic acinar cells and, ultimately, loss of pancreatic function. We investigated the role of IκB kinase α (IKKα) in pancreatic homeostasis. Pancreas-specific ablation of IKKα (Ikkα(Δpan)) caused spontaneous and progressive acinar cell vacuolization and death, interstitial fibrosis, inflammation, and circulatory release of pancreatic enzymes, clinical signs resembling those of human chronic pancreatitis. Loss of pancreatic IKKα causes defective autophagic protein degradation, leading to accumulation of p62-mediated protein aggregates and enhanced oxidative and ER stress in acinar cells, but none of these effects is related to NF-κB. Pancreas-specific p62 ablation prevented ER and oxidative stresses and attenuated pancreatitis in Ikkα(Δpan) mice, suggesting that cellular stress induced by p62 aggregates promotes development of pancreatitis. Importantly, downregulation of IKKα and accumulation of p62 aggregates were also observed in chronic human pancreatitis. Our studies demonstrate that IKKα, which may control autophagic protein degradation through its interaction with ATG16L2, plays a critical role in maintaining pancreatic acinar cell homeostasis, whose dysregulation promotes pancreatitis through p62 aggregate accumulation.


Subject(s)
Acinar Cells/cytology , Gene Expression Regulation, Enzymologic , I-kappa B Kinase/metabolism , Pancreatitis/metabolism , Animals , Autophagy , Carrier Proteins/metabolism , Cell Proliferation , Down-Regulation , Endoplasmic Reticulum/metabolism , Fibrosis , Immunohistochemistry , Inflammation , Mice , Mice, Transgenic , NF-kappa B/metabolism , Oxidative Stress , Transcription Factor TFIIH , Transcription Factors/metabolism
8.
Cell ; 152(1-2): 25-38, 2013 Jan 17.
Article in English | MEDLINE | ID: mdl-23273993

ABSTRACT

Cell-type plasticity within a tumor has recently been suggested to cause a bidirectional conversion between tumor-initiating stem cells and nonstem cells triggered by an inflammatory stroma. NF-κB represents a key transcription factor within the inflammatory tumor microenvironment. However, NF-κB's function in tumor-initiating cells has not been examined yet. Using a genetic model of intestinal epithelial cell (IEC)-restricted constitutive Wnt-activation, which comprises the most common event in the initiation of colon cancer, we demonstrate that NF-κB modulates Wnt signaling and show that IEC-specific ablation of RelA/p65 retards crypt stem cell expansion. In contrast, elevated NF-κB signaling enhances Wnt activation and induces dedifferentiation of nonstem cells that acquire tumor-initiating capacity. Thus, our data support the concept of bidirectional conversion and highlight the importance of inflammatory signaling for dedifferentiation and generation of tumor-initiating cells in vivo.


Subject(s)
Cell Dedifferentiation , Cell Transformation, Neoplastic , Colonic Neoplasms/pathology , Neoplastic Stem Cells/pathology , Animals , Colon/pathology , Epithelial Cells/pathology , Female , Humans , Male , Mice , NF-kappa B/metabolism , Wnt Signaling Pathway
9.
Curr Top Microbiol Immunol ; 349: 159-69, 2011.
Article in English | MEDLINE | ID: mdl-20845109

ABSTRACT

Since the initial cloning of RelA and its close relationship to c-Rel, the cellular homolog of the viral oncoprotein v-Rel, the nuclear factor κB (NF-κB) signaling pathway and its upstream activating kinase complex (IκB-kinase) have been suspected to play a major role in tumorigenesis. This was further corroborated by the discovery of oncogenic mutations in NF-κB proteins in certain lymphoid malignancies and the notion that NF-κB is persistently activated in a large variety of solid tumors. With the advent of conditional knockout mice allowing tissue-specific targeting of the various components of the NF-κB signaling pathway, it was possible to genetically test the cell autonomous and non-autonomous functions of NF-κB in inflammation-associated cancer as well as sporadic cancers. Here, we review molecular evidence that demonstrates the various functions of NF-κB during different tumor stages and that supports the rationale to target NF-κB in cancer prevention and therapy.


Subject(s)
I-kappa B Kinase/physiology , NF-kappa B/physiology , Neoplasms/etiology , Animals , Humans , Neoplasms/therapy , Neoplastic Stem Cells/pathology , Signal Transduction
10.
J Exp Med ; 207(12): 2621-30, 2010 Nov 22.
Article in English | MEDLINE | ID: mdl-20975042

ABSTRACT

Inhibitor of κB (IκB) ß (IκBß) represents one of the major primary regulators of NF-κB in mammals. In contrast to the defined regulatory interplay between NF-κB and IκBα, much less is known about the biological function of IκBß. To elucidate the physiological role of IκBß in NF-κB signaling in vivo, we generated IκBß-deficient mice. These animals proved to be highly refractory to LPS-induced lethality, accompanied by a strong reduction in sepsis-associated cytokine production. In response to LPS, IκBß is recruited to the IL-1ß promoter forming a complex with the NF-κB subunits RelA/c-Rel required for IL-1ß transcription. Further transcriptome analysis of LPS-stimulated wild-type and IκBß-deficient BM-derived macrophages revealed several other genes with known regulatory functions in innate immunity arguing that a subset of NF-κB target genes is under control of IκBß. Collectively, these findings provide an essential proinflammatory role for IκBß in vivo, and establish a critical function for IκBß as a transcriptional coactivator under inflammatory conditions.


Subject(s)
I-kappa B Proteins/physiology , Interleukin-1beta/genetics , Lipopolysaccharides/pharmacology , Transcription, Genetic , Animals , Cytokines/biosynthesis , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Shock, Septic/prevention & control , Transcription Factor RelA/physiology
11.
Cancer Cell ; 18(2): 135-46, 2010 Aug 09.
Article in English | MEDLINE | ID: mdl-20708155

ABSTRACT

Colonic cancers with a serrated morphology have been proposed to comprise a molecularly distinct tumor entity following an alternative pathway of genetic alterations independently of APC mutations. We demonstrate that intestinal epithelial cell specific expression of oncogenic K-ras(G12D) in mice induces serrated hyperplasia, which is characterized by p16(ink4a) overexpression and induction of senescence. Deletion of Ink4a/Arf in K-ras(G12D) expressing mice prevents senescence and leads to invasive, metastasizing carcinomas with morphological and molecular alterations comparable to human KRAS mutated serrated tumors. Thus, we suggest that oncogenic K-ras represents a key player during an alternative, serrated pathway to colorectal cancer and hence propose RAS-RAF-MEK signaling apart from APC as an additional gatekeeper in colorectal tumor development.


Subject(s)
Cellular Senescence/genetics , Colorectal Neoplasms/pathology , Cyclin-Dependent Kinase Inhibitor p16/physiology , Oncogenes , Animals , Cell Division , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Cyclin-Dependent Kinase Inhibitor p16/genetics , Disease Progression , Genes, APC , Genes, ras , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mutation , Signal Transduction , Wnt Proteins/metabolism
12.
Cancer Cell ; 15(2): 91-102, 2009 Feb 03.
Article in English | MEDLINE | ID: mdl-19185844

ABSTRACT

Although gastrointestinal cancers are frequently associated with chronic inflammation, the underlying molecular links have not been comprehensively deciphered. Using loss- and gain-of-function mice in a colitis-associated cancer model, we establish here a link comprising the gp130/Stat3 transcription factor signaling axis. Mutagen-induced tumor growth and multiplicity are reduced following intestinal epithelial cell (IEC)-specific Stat3 ablation, while its hyperactivation promotes tumor incidence and growth. Conversely, IEC-specific Stat3 deficiency enhances susceptibility to chemically induced epithelial damage and subsequent mucosal inflammation, while excessive Stat3 activation confers resistance to colitis. Stat3 has the capacity to mediate IL-6- and IL-11-dependent IEC survival and to promote proliferation through G1 and G2/M cell-cycle progression as the common tumor cell-autonomous mechanism that bridges chronic inflammation to tumor promotion.


Subject(s)
Cell Cycle/physiology , Cell Survival/physiology , Colitis , Cytokine Receptor gp130/metabolism , Enterocytes/physiology , Neoplasms , STAT3 Transcription Factor/metabolism , Animals , Apoptosis/physiology , Cell Cycle Proteins/metabolism , Cell Proliferation , Colitis/complications , Colitis/immunology , Colitis/pathology , Cytokine Receptor gp130/genetics , Enterocytes/cytology , Enterocytes/pathology , Humans , Inflammation/immunology , Interleukin-11/immunology , Interleukin-6/immunology , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Mice , Neoplasms/etiology , Neoplasms/immunology , Neoplasms/pathology , Regeneration/physiology , STAT3 Transcription Factor/genetics , Signal Transduction/physiology
13.
Proc Natl Acad Sci U S A ; 105(39): 15058-63, 2008 Sep 30.
Article in English | MEDLINE | ID: mdl-18815378

ABSTRACT

NF-kappaB is a key transcriptional regulator of inflammatory responses, but also controls expression of prosurvival genes, whose products protect tissues from damage and may thus act indirectly in an antiinflammatory fashion. The variable importance of these two distinct NF-kappaB-controlled responses impacts the potential utility of NF-kappaB inhibition as a treatment strategy for intractable inflammatory conditions, such as inflammatory bowel disease. Here, we show in murine models that inhibition of IKKbeta-dependent NF-kappaB activation exacerbates acute inflammation, but attenuates chronic inflammatory disease in the intestinal tract. Acute ulcerating inflammation is aggravated because of diminished NF-kappaB-mediated protection against epithelial cell apoptosis and delayed mucosal regeneration secondary to reduced NF-kappaB-dependent recruitment of inflammatory cells that secrete cytoprotective factors. In contrast, in IL-10-deficient mice, which serve as a model of chronic T cell-dependent colitis, ablation of IKKbeta in the intestinal epithelium has no impact, yet IKKbeta deficiency in myeloid cells attenuates inflammation and prolongs survival. These results highlight the striking context and tissue dependence of the proinflammatory and antiapoptotic functions of NF-kappaB. Our findings caution against the therapeutic use of IKKbeta/NF-kappaB inhibitors in acute inflammatory settings dominated by cell loss and ulceration.


Subject(s)
Colitis, Ulcerative/metabolism , I-kappa B Kinase/metabolism , Acute Disease , Animals , Chronic Disease , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/drug therapy , Dextran Sulfate/toxicity , Disease Models, Animal , Gene Expression , I-kappa B Kinase/antagonists & inhibitors , Interleukin-1/genetics , Intestinal Mucosa , Mice , Mice, Mutant Strains , NF-kappa B/metabolism , STAT3 Transcription Factor/metabolism
14.
Cell ; 130(5): 918-31, 2007 Sep 07.
Article in English | MEDLINE | ID: mdl-17803913

ABSTRACT

IKKbeta-dependent NF-kappaB activation plays a key role in innate immunity and inflammation, and inhibition of IKKbeta has been considered as a likely anti-inflammatory therapy. Surprisingly, however, mice with a targeted IKKbeta deletion in myeloid cells are more susceptible to endotoxin-induced shock than control mice. Increased endotoxin susceptibility is associated with elevated plasma IL-1beta as a result of increased pro-IL-1beta processing, which was also seen upon bacterial infection. In macrophages enhanced pro-IL-1beta processing depends on caspase-1, whose activation is inhibited by NF-kappaB-dependent gene products. In neutrophils, however, IL-1beta secretion is caspase-1 independent and depends on serine proteases, whose activity is also inhibited by NF-kappaB gene products. Prolonged pharmacologic inhibition of IKKbeta also augments IL-1beta secretion upon endotoxin challenge. These results unravel an unanticipated role for IKKbeta-dependent NF-kappaB signaling in the negative control of IL-1beta production and highlight potential complications of long-term IKKbeta inhibition.


Subject(s)
I-kappa B Kinase/metabolism , Interleukin-1beta/metabolism , Macrophages/metabolism , NF-kappa B/metabolism , Neutrophil Infiltration , Neutrophils/metabolism , Shock, Septic/metabolism , Animals , Apoptosis , Carbolines/pharmacology , Caspase 1/metabolism , Cell Line , Disease Models, Animal , Humans , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/genetics , Interleukin-1beta/blood , Interleukin-1beta/genetics , Interleukin-6/blood , Lipopolysaccharides , Macrophages/drug effects , Macrophages/enzymology , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Knockout , Neutrophils/drug effects , Neutrophils/enzymology , Neutrophils/immunology , Neutrophils/pathology , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/metabolism , Receptors, Interleukin-1/metabolism , Serine Endopeptidases/metabolism , Serpins/metabolism , Shock, Septic/chemically induced , Shock, Septic/enzymology , Shock, Septic/immunology , Shock, Septic/pathology , Time Factors , Transfection , Tumor Necrosis Factor-alpha/blood
15.
Drug Metab Dispos ; 35(3): 402-9, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17151194

ABSTRACT

Hepatic transporters are responsible for uptake and efflux of bile acids and xenobiotics as an essential aspect of liver function. When normal vectorial transport of bile acids by the apical uptake and canalicular excretion transporters is disrupted, cholestasis ensues, leading to accumulation of toxic bile constituents and considerable hepatocellular damage. The purpose of this study was to assess the role of cytokines and nuclear factor-kappaB (NF-kappaB) in the transcriptional regulation of transporters in two models of cholestasis, lipopolysaccharide (LPS) administration and bile duct ligation (BDL). In wild-type (WT) and knockout mouse strains lacking tumor necrosis factor (TNF) receptor-1, interleukin (IL)-1 receptor I, IL-6, or inhibitor of kappaB(IkappaB) kinase beta, transporter mRNA levels in liver were determined using branched DNA signal amplification 16 h after LPS administration or 3 days after BDL. In WT mice, LPS administration tended to decrease mRNA levels of organic anion-transporting polypeptide (Oatp) 2, Na(+)-taurocholate cotransporting polypeptide (Ntcp), Oatp1, Oatp4, bile salt excretory protein (Bsep), multidrug resistance-associated protein (Mrp) 2, and Mrp6 compared with saline treatment, whereas it increased Mrp1, 3, and 5 levels. Similar changes were observed in each knockout strain after LPS administration. Conversely, BDL decreased only Oatp1 expression in WT mice, meanwhile increasing expression of Mrp1, 3, and 5 and Oatp2 expression in both WT and knockout strains. Because the transcriptional effects of BDL- and LPS-induced cholestasis reflect dissimilarity in hepatic transporter regulation, we conclude that these disparities are not due to the individual activity of TNF-alpha, IL-1, IL-6, or NF-kappaB but to the differences in the mechanism of cholestasis.


Subject(s)
Cholestasis/metabolism , Liver/metabolism , Organic Anion Transporters/genetics , Animals , Bile Ducts/surgery , Cholestasis/chemically induced , Disease Models, Animal , I-kappa B Kinase/deficiency , I-kappa B Kinase/genetics , Interleukin-1beta/deficiency , Interleukin-6/deficiency , Interleukin-6/genetics , Ligation , Lipopolysaccharides , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/deficiency , RNA, Messenger/metabolism , Receptors, Interleukin-1 Type I/deficiency , Receptors, Interleukin-1 Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/deficiency , Receptors, Tumor Necrosis Factor, Type I/genetics , Tumor Necrosis Factor-alpha/deficiency
16.
Nat Med ; 11(2): 191-8, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15685170

ABSTRACT

Inflammation may underlie the metabolic disorders of insulin resistance and type 2 diabetes. IkappaB kinase beta (IKK-beta, encoded by Ikbkb) is a central coordinator of inflammatory responses through activation of NF-kappaB. To understand the role of IKK-beta in insulin resistance, we used mice lacking this enzyme in hepatocytes (Ikbkb(Deltahep)) or myeloid cells (Ikbkb(Deltamye)). Ikbkb(Deltahep) mice retain liver insulin responsiveness, but develop insulin resistance in muscle and fat in response to high fat diet, obesity or aging. In contrast, Ikbkb(Deltamye) mice retain global insulin sensitivity and are protected from insulin resistance. Thus, IKK-beta acts locally in liver and systemically in myeloid cells, where NF-kappaB activation induces inflammatory mediators that cause insulin resistance. These findings demonstrate the importance of liver cell IKK-beta in hepatic insulin resistance and the central role of myeloid cells in development of systemic insulin resistance. We suggest that inhibition of IKK-beta, especially in myeloid cells, may be used to treat insulin resistance.


Subject(s)
Inflammation/metabolism , Insulin Resistance/physiology , Obesity/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Cells, Cultured , Cytokines/genetics , Cytokines/immunology , Dietary Fats , Glucose Tolerance Test , Hepatocytes/cytology , Hepatocytes/metabolism , I-kappa B Kinase , Insulin/metabolism , Mice , Mice, Knockout , Myeloid Cells/cytology , Myeloid Cells/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Phenotype , Protein Serine-Threonine Kinases/genetics , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...