Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Cell Rep ; 28(6): 1612-1622.e4, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31390573

ABSTRACT

Cachexia is a wasting syndrome characterized by pronounced skeletal muscle loss. In cancer, cachexia is associated with increased morbidity and mortality and decreased treatment tolerance. Although advances have been made in understanding the mechanisms of cachexia, translating these advances to the clinic has been challenging. One reason for this shortcoming may be the current animal models, which fail to fully recapitulate the etiology of human cancer-induced tissue wasting. Because pancreatic ductal adenocarcinoma (PDA) presents with a high incidence of cachexia, we engineered a mouse model of PDA that we named KPP. KPP mice, similar to PDA patients, progressively lose skeletal and adipose mass as a consequence of their tumors. In addition, KPP muscles exhibit a similar gene ontology as cachectic patients. We envision that the KPP model will be a useful resource for advancing our mechanistic understanding and ability to treat cancer cachexia.


Subject(s)
Cachexia/etiology , Disease Models, Animal , Pancreatic Neoplasms/complications , Animals , Cachexia/genetics , Cachexia/metabolism , Disease Progression , Female , Gene Ontology , Heterografts , Humans , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Neoplasm Transplantation , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , RNA-Seq , Transcriptome , Pancreatic Neoplasms
2.
Proc Natl Acad Sci U S A ; 115(16): E3798-E3807, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29610295

ABSTRACT

Sterile alpha motif and HD-domain-containing protein 1 (SAMHD1) blocks replication of retroviruses and certain DNA viruses by reducing the intracellular dNTP pool. SAMHD1 has been suggested to down-regulate IFN and inflammatory responses to viral infections, although the functions and mechanisms of SAMHD1 in modulating innate immunity remain unclear. Here, we show that SAMHD1 suppresses the innate immune responses to viral infections and inflammatory stimuli by inhibiting nuclear factor-κB (NF-κB) activation and type I interferon (IFN-I) induction. Compared with control cells, infection of SAMHD1-silenced human monocytic cells or primary macrophages with Sendai virus (SeV) or HIV-1, or treatment with inflammatory stimuli, induces significantly higher levels of NF-κB activation and IFN-I induction. Exogenous SAMHD1 expression in cells or SAMHD1 reconstitution in knockout cells suppresses NF-κB activation and IFN-I induction by SeV infection or inflammatory stimuli. Mechanistically, SAMHD1 inhibits NF-κB activation by interacting with NF-κB1/2 and reducing phosphorylation of the NF-κB inhibitory protein IκBα. SAMHD1 also interacts with the inhibitor-κB kinase ε (IKKε) and IFN regulatory factor 7 (IRF7), leading to the suppression of the IFN-I induction pathway by reducing IKKε-mediated IRF7 phosphorylation. Interactions of endogenous SAMHD1 with NF-κB and IFN-I pathway proteins were validated in human monocytic cells and primary macrophages. Comparing splenocytes from SAMHD1 knockout and heterozygous mice, we further confirmed SAMHD1-mediated suppression of NF-κB activation, suggesting an evolutionarily conserved property of SAMHD1. Our findings reveal functions of SAMHD1 in down-regulating innate immune responses to viral infections and inflammatory stimuli, highlighting the importance of SAMHD1 in modulating antiviral immunity.


Subject(s)
Immunity, Innate , Inflammation/immunology , Interferon-alpha/biosynthesis , NF-kappa B/metabolism , SAM Domain and HD Domain-Containing Protein 1/physiology , Virus Diseases/immunology , Animals , Cells, Cultured , Down-Regulation , Gene Expression Regulation/drug effects , Gene Silencing , HEK293 Cells , HIV/physiology , Humans , I-kappa B Kinase/antagonists & inhibitors , Interferon Regulatory Factor-7/antagonists & inhibitors , Interferon-alpha/genetics , Macrophages/immunology , Macrophages/virology , Male , Mice , NF-KappaB Inhibitor alpha/metabolism , Phosphorylation , Protein Processing, Post-Translational , Recombinant Proteins/immunology , Sendai virus/physiology , Signal Transduction/immunology , THP-1 Cells
3.
Front Oncol ; 8: 104, 2018.
Article in English | MEDLINE | ID: mdl-29696133

ABSTRACT

BACKGROUND: Metabolic reprogramming has emerged as a cancer hallmark, and one of the well-known cancer-associated metabolic alterations is the increase in the rate of glycolysis. Recent reports have shown that both the classical and alternative signaling pathways of nuclear factor κB (NF-κB) play important roles in controlling the metabolic profiles of normal cells and cancer cells. However, how these signaling pathways affect the metabolism of sarcomas, specifically rhabdomyosarcoma (RMS) and osteosarcoma (OS), has not been characterized. METHODS: Classical NF-κB activity was inhibited through overexpression of the IκBα super repressor of NF-κB in RMS and OS cells. Global gene expression analysis was performed using Affymetrix GeneChip Human Transcriptome Array 2.0, and data were interpreted using gene set enrichment analysis. Seahorse Bioscience XFe24 was used to analyze oxygen consumption rate as a measure of aerobic respiration. RESULTS: Inhibition of classical NF-κB activity in sarcoma cell lines restored alternative signaling as well as an increased oxidative respiratory metabolic phenotype in vitro. In addition, microarray analysis indicated that inhibition of NF-κB in sarcoma cells reduced glycolysis. We showed that a glycolytic gene, hexokinase (HK) 2, is a direct NF-κB transcriptional target. Knockdown of HK2 shifted the metabolic profile in sarcoma cells away from aerobic glycolysis, and re-expression of HK2 rescued the metabolic shift induced by inhibition of NF-κB activity in OS cells. CONCLUSION: These findings suggest that classical signaling of NF-κB plays a crucial role in the metabolic profile of pediatric sarcomas potentially through the regulation of HK2.

4.
Dis Model Mech ; 10(9): 1109-1115, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28883017

ABSTRACT

Alveolar rhabdomyosarcoma (aRMS) is a pediatric soft tissue cancer commonly associated with a chromosomal translocation that leads to the expression of a Pax3:Foxo1 or Pax7:Foxo1 fusion protein, the developmental underpinnings of which may give clues to its therapeutic approaches. In aRMS, the NFκB-YY1-miR-29 regulatory circuit is dysregulated, resulting in repression of miR-29 and loss of the associated tumor suppressor activity. To further elucidate the role of NFκB in aRMS, we first tested 55 unique sarcoma cell lines and primary cell cultures in a large-scale chemical screen targeting diverse molecular pathways. We found that pharmacological inhibition of NFκB activity resulted in decreased cell proliferation of many of the aRMS tumor cultures. Surprisingly, mice that were orthotopically allografted with aRMS tumor cells exhibited no difference in tumor growth when administered an NFκB inhibitor, compared to control. Furthermore, inhibition of NFκB by genetically ablating its activating kinase inhibitor, IKKß, by conditional deletion in a mouse model harboring the Pax3:Foxo1 chimeric oncogene failed to abrogate spontaneous tumor growth. Genetically engineered mice with conditionally deleted IKKß exhibited a paradoxical decrease in tumor latency compared with those with active NFκB. However, using a synthetic-lethal approach, primary cell cultures derived from tumors with inactivated NFκB showed sensitivity to the BCL-2 inhibitor navitoclax. When used in combination with an NFκB inhibitor, navitoclax was synergistic in decreasing the growth of both human and IKKß wild-type mouse aRMS cells, indicating that inactivation of NFκB alone may not be sufficient for reducing tumor growth, but, when combined with another targeted therapeutic, may be clinically beneficial.


Subject(s)
NF-kappa B/metabolism , Rhabdomyosarcoma, Alveolar/metabolism , Signal Transduction , Allografts/drug effects , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Dogs , Female , Gene Deletion , Genetic Complementation Test , Humans , I-kappa B Kinase/metabolism , Mice, SCID , Peptides/pharmacology , Phenotype , Proto-Oncogene Proteins c-bcl-2/metabolism , Rhabdomyosarcoma, Alveolar/pathology , Signal Transduction/drug effects
5.
J Clin Invest ; 127(10): 3796-3809, 2017 Oct 02.
Article in English | MEDLINE | ID: mdl-28891811

ABSTRACT

Macrophages are attracted to developing tumors and can participate in immune surveillance to eliminate neoplastic cells. In response, neoplastic cells utilize NF-κB to suppress this killing activity, but the mechanisms underlying their self-protection remain unclear. Here, we report that this dynamic interaction between tumor cells and macrophages is integrally linked by a soluble factor identified as growth and differentiation factor 15 (GDF-15). In vitro, tumor-derived GDF-15 signals in macrophages to suppress their proapoptotic activity by inhibiting TNF and nitric oxide (NO) production. In vivo, depletion of GDF-15 in Ras-driven tumor xenografts and in an orthotopic model of pancreatic cancer delayed tumor development. This delay correlated with increased infiltrating antitumor macrophages. Further, production of GDF-15 is directly regulated by NF-κB, and the colocalization of activated NF-κB and GDF-15 in epithelial ducts of human pancreatic adenocarcinoma supports the importance of this observation. Mechanistically, we found that GDF-15 suppresses macrophage activity by inhibiting TGF-ß-activated kinase (TAK1) signaling to NF-κB, thereby blocking synthesis of TNF and NO. Based on these results, we propose that the NF-κB/GDF-15 regulatory axis is important for tumor cells in evading macrophage immune surveillance during the early stages of tumorigenesis.


Subject(s)
Adenocarcinoma/immunology , Growth Differentiation Factor 15/immunology , Immunologic Surveillance , Macrophages/immunology , NF-kappa B/immunology , Neoplasm Proteins/immunology , Neoplasms, Experimental/immunology , Pancreatic Neoplasms/immunology , Signal Transduction/immunology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Female , Growth Differentiation Factor 15/genetics , Heterografts , MAP Kinase Kinase Kinases , Macrophages/pathology , Male , Mice , Mice, Knockout , NF-kappa B/genetics , Neoplasm Proteins/genetics , Neoplasm Transplantation , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Nitric Oxide/genetics , Nitric Oxide/immunology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Signal Transduction/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
6.
Cell Rep ; 17(2): 514-526, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27705798

ABSTRACT

MyoD is a key regulator of skeletal myogenesis that directs contractile protein synthesis, but whether this transcription factor also regulates skeletal muscle metabolism has not been explored. In a genome-wide ChIP-seq analysis of skeletal muscle cells, we unexpectedly observed that MyoD directly binds to numerous metabolic genes, including those associated with mitochondrial biogenesis, fatty acid oxidation, and the electron transport chain. Results in cultured cells and adult skeletal muscle confirmed that MyoD regulates oxidative metabolism through multiple transcriptional targets, including PGC-1ß, a master regulator of mitochondrial biogenesis. We find that PGC-1ß expression is cooperatively regulated by MyoD and the alternative NF-κB signaling pathway. Bioinformatics evidence suggests that this cooperativity between MyoD and NF-κB extends to other metabolic genes as well. Together, these data identify MyoD as a regulator of the metabolic capacity of mature skeletal muscle to ensure that sufficient energy is available to support muscle contraction.


Subject(s)
Mitochondria/metabolism , Muscle, Skeletal/metabolism , MyoD Protein/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Animals , Mice , Mitochondria/genetics , Muscle Contraction/genetics , Muscle Development/genetics , MyoD Protein/metabolism , Myoblasts/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Oxidation-Reduction , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Protein Binding , Signal Transduction , Transcription Factor RelB/genetics , Transcription Factor RelB/metabolism
7.
Lab Invest ; 96(4): 481-91, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26878133

ABSTRACT

Sarcomas are malignant heterogenous tumors of mesenchymal derivation. Emerging data suggest that miRNA might have a causal role in sarcomagenesis. Herein, we used a selective miRNA screening platform to study the comparative global miRNA expression signatures in a cohort of human sarcomas with the caveat that comparisons between tumor and non-tumor cells were performed from the same patients using formalin-fixed paraffin-embedded tissue. Five histologic types were examined that included: myxoid liposarcoma, well-differentiated liposarcoma, dedifferentiated liposarcoma, pleomorphic rhabdomyosarcoma, and synovial sarcoma. In addition, soft-tissue lipomas and normal fat were included as a separate set of controls for the lipogenic tumors. Clustering analysis showed a distinct global difference in expression patterns between the normal and sarcoma tissues. Expression signatures in an unsupervised hierarchical clustering analysis revealed tight clustering in synovial and myxoid liposarcomas, and the least clustering was observed in the pleomorphic rhabdomyosarcoma subtype. MiR-145 showed underexpression in pleomorphic rhabdomyosarcoma, well-differentiated liposarcoma, and synovial sarcoma. Unexpectedly, we found that a set of muscle-specific microRNAs (miRNAs; myomiRs): miR-133, miR-1, and miR-206 was significantly underexpressed in well-differentiated liposarcoma and synovial sarcoma, suggesting that they may function as tumor suppressors as described in muscle-relevant rhabdomyosarcomas. In addition, a tight linear progression of miRNA expression was identified from normal fat to dedifferentiated liposarcoma. These results suggest that miRNA expression profiles could elucidate classes of miRNAs that may elicit tumor-relevant activities in specific sarcoma subtypes.


Subject(s)
Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Sarcoma/genetics , Adult , Aged , Cluster Analysis , Cohort Studies , Diagnosis, Differential , Female , Humans , Liposarcoma/diagnosis , Liposarcoma/genetics , Liposarcoma, Myxoid/diagnosis , Liposarcoma, Myxoid/genetics , Male , MicroRNAs/classification , Middle Aged , Muscles/metabolism , Organ Specificity/genetics , Reverse Transcriptase Polymerase Chain Reaction , Rhabdomyosarcoma/diagnosis , Rhabdomyosarcoma/genetics , Sarcoma/diagnosis , Sarcoma, Synovial/diagnosis , Sarcoma, Synovial/genetics , Young Adult
8.
Neuron ; 81(5): 1009-1023, 2014 Mar 05.
Article in English | MEDLINE | ID: mdl-24607225

ABSTRACT

Neuroinflammation is one of the most striking hallmarks of amyotrophic lateral sclerosis (ALS). Nuclear factor-kappa B (NF-κB), a master regulator of inflammation, is upregulated in spinal cords of ALS patients and SOD1-G93A mice. In this study, we show that selective NF-κB inhibition in ALS astrocytes is not sufficient to rescue motor neuron (MN) death. However, the localization of NF-κB activity and subsequent deletion of NF-κB signaling in microglia rescued MNs from microglial-mediated death in vitro and extended survival in ALS mice by impairing proinflammatory microglial activation. Conversely, constitutive activation of NF-κB selectively in wild-type microglia induced gliosis and MN death in vitro and in vivo. Taken together, these data provide a mechanism by which microglia induce MN death in ALS and suggest a novel therapeutic target that can be modulated to slow the progression of ALS and possibly other neurodegenerative diseases by which microglial activation plays a role.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Cell Death/physiology , Microglia/cytology , Motor Neurons/cytology , NF-kappa B/metabolism , Age Factors , Amyotrophic Lateral Sclerosis/metabolism , Animals , Animals, Newborn , Astrocytes/cytology , Astrocytes/metabolism , Cell Communication/physiology , Coculture Techniques , Disease Models, Animal , Female , Mice , Mice, Inbred Strains , Mice, Transgenic , Microglia/metabolism , Motor Neurons/metabolism , NF-kappa B/antagonists & inhibitors , Primary Cell Culture , Signal Transduction/physiology , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
9.
Sci Signal ; 6(286): ra63, 2013 Jul 30.
Article in English | MEDLINE | ID: mdl-23901138

ABSTRACT

In sarcoma, the activity of NF-κB (nuclear factor κB) reduces the abundance of the microRNA (miRNA) miR-29. The tumor suppressor A20 [also known as TNFAIP3 (tumor necrosis factor-α-induced protein 3)] inhibits an upstream activator of NF-κB and is often mutated in lymphomas. In a panel of human sarcoma cell lines, we found that the activation of NF-κB was increased and, although the abundance of A20 protein and mRNA was decreased, the gene encoding A20 was rarely mutated. The 3' untranslated region (UTR) of A20 mRNA has conserved binding sites for both of the miRNAs miR-29 and miR-125. Whereas the expression of miR-125 was increased in human sarcoma tissue, that of miR-29 was decreased in most samples. Overexpression of miR-125 decreased the abundance of A20 mRNA, whereas reconstituting miR-29 in sarcoma cell lines increased the abundance of A20 mRNA and protein. By interacting directly with the RNA binding protein HuR (human antigen R; also known as ELAVL1), miR-29 prevented HuR from binding to the A20 3'UTR and recruiting the RNA degradation complex RISC (RNA-induced silencing complex), suggesting that miR-29 can act as a decoy for HuR, thus protecting A20 transcripts. Decreased miR-29 and A20 abundance in sarcomas correlated with increased activity of NF-κB and decreased expression of genes associated with differentiation. Together, the findings reveal a unique role of miR-29 and suggest that its absence may contribute to sarcoma tumorigenesis.


Subject(s)
DNA-Binding Proteins/metabolism , ELAV Proteins/metabolism , Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins/metabolism , MicroRNAs/genetics , MicroRNAs/physiology , Nuclear Proteins/metabolism , 3' Untranslated Regions , Animals , Binding Sites , Computational Biology , Gene Silencing , Genes, Reporter , Humans , Immunoprecipitation , Inflammation , Mice , Mutation , NF-kappa B/metabolism , Nuclear Pore Complex Proteins/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Sequence Analysis, DNA , Tumor Necrosis Factor alpha-Induced Protein 3
10.
Ann Neurol ; 69(3): 540-52, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21446026

ABSTRACT

OBJECTIVE: Facioscapulohumeral muscular dystrophy (FSHD) is associated with D4Z4 repeat contraction on human chromosome 4q35. This genetic lesion does not result in complete loss or mutation of any gene. Consequently, the pathogenic mechanisms underlying FSHD have been difficult to discern. In leading FSHD pathogenesis models, D4Z4 contractions are proposed to cause epigenetic changes, which ultimately increase expression of genes with myopathic potential. Although no gene has been conclusively linked to FSHD development, recent evidence supports a role for the D4Z4-encoded DUX4 gene in FSHD. In this study, our objective was to test the in vivo myopathic potential of DUX4. METHODS: We delivered DUX4 to zebrafish and mouse muscle by transposon-mediated transgenesis and adeno-associated viral vectors, respectively. RESULTS: Overexpression of DUX4, which encodes a transcription factor, caused abnormalities associated with muscular dystrophy in zebrafish and mice. This toxicity required DNA binding, because a DUX4 DNA binding domain mutant produced no abnormalities. Importantly, we found the myopathic effects of DUX4 were p53 dependent, as p53 inhibition mitigated DUX4 toxicity in vitro, and muscles from p53 null mice were resistant to DUX4-induced damage. INTERPRETATION: Our work demonstrates the myopathic potential of DUX4 in animal muscle. Considering previous studies showed DUX4 was elevated in FSHD patient muscles, our data support the hypothesis that DUX4 overexpression contributes to FSHD development. Moreover, we provide a p53-dependent mechanism for DUX4 toxicity that is consistent with previous studies showing p53 pathway activation in FSHD muscles. Our work justifies further investigation of DUX4 and the p53 pathway in FSHD pathogenesis.


Subject(s)
Homeodomain Proteins/genetics , Muscle, Skeletal/pathology , Muscular Diseases/genetics , Tumor Suppressor Protein p53/genetics , Animals , Female , Gene Transfer Techniques , Hand Strength/physiology , Homeodomain Proteins/metabolism , Immunohistochemistry , Male , Mice , Mice, Knockout , Muscle Strength/physiology , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Muscular Diseases/pathology , Muscular Diseases/physiopathology , Reverse Transcriptase Polymerase Chain Reaction , Zebrafish
11.
PLoS One ; 5(8): e12479, 2010 Aug 30.
Article in English | MEDLINE | ID: mdl-20814569

ABSTRACT

BACKGROUND: Classical NF-kappaB signaling functions as a negative regulator of skeletal myogenesis through potentially multiple mechanisms. The inhibitory actions of TNFalpha on skeletal muscle differentiation are mediated in part through sustained NF-kappaB activity. In dystrophic muscles, NF-kappaB activity is compartmentalized to myofibers to inhibit regeneration by limiting the number of myogenic progenitor cells. This regulation coincides with elevated levels of muscle derived TNFalpha that is also under IKKbeta and NF-kappaB control. METHODOLOGY/PRINCIPAL FINDINGS: Based on these findings we speculated that in DMD, TNFalpha secreted from myotubes inhibits regeneration by directly acting on satellite cells. Analysis of several satellite cell regulators revealed that TNFalpha is capable of inhibiting Notch-1 in satellite cells and C2C12 myoblasts, which was also found to be dependent on NF-kappaB. Notch-1 inhibition occurred at the mRNA level suggesting a transcriptional repression mechanism. Unlike its classical mode of action, TNFalpha stimulated the recruitment of Ezh2 and Dnmt-3b to coordinate histone and DNA methylation, respectively. Dnmt-3b recruitment was dependent on Ezh2. CONCLUSIONS/SIGNIFICANCE: We propose that in dystrophic muscles, elevated levels of TNFalpha and NF-kappaB inhibit the regenerative potential of satellite cells via epigenetic silencing of the Notch-1 gene.


Subject(s)
DNA Methylation/drug effects , Down-Regulation/drug effects , Histone-Lysine N-Methyltransferase/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/genetics , Receptor, Notch1/genetics , Tumor Necrosis Factor-alpha/pharmacology , Animals , Enhancer of Zeste Homolog 2 Protein , Humans , Mice , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , NF-kappa B/metabolism , Polycomb Repressive Complex 2 , Promoter Regions, Genetic/genetics , Receptor, Notch1/deficiency
12.
EMBO Rep ; 10(11): 1272-8, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19779484

ABSTRACT

Nuclear factor (NF)-kappaB is a positive regulator of tumour development and progression, but how it functions in normal cells leading to oncogenesis is not clear. As cellular senescence has proven to be an intrinsic tumour suppressor mechanism that cells must overcome to establish deregulated growth, we used primary fibroblasts to follow NF-kappaB function in cells transitioning from senescence to subsequent immortalization. Our findings show that RelA/p65(-/-) murine fibroblasts immortalize at considerably faster rates than RelA/p65(+/+) cells. The ability of RelA/p65(-/-) fibroblasts to escape senescence earlier is due to their genomic instability, characterized by high frequencies of DNA mutations, gene deletions and gross chromosomal translocations. This increase in genomic instability is closely related to a compromised DNA repair that occurs in both murine RelA/p65(-/-) fibroblasts and tissues. Significantly, these results can also be duplicated in human fibroblasts lacking NF-kappaB. Altogether, our findings present a fresh perspective on the role of NF-kappaB as a tumour suppressor, which acts in pre-neoplastic cells to maintain cellular senescence by promoting DNA repair and genomic stability.


Subject(s)
Cellular Senescence , DNA Repair , Genomic Instability , Transcription Factor RelA/metabolism , 3T3 Cells , Animals , Cell Line , DNA/genetics , Fibroblasts/metabolism , Gene Deletion , Hydrogen Peroxide/chemistry , Mice , Mutation , NF-kappa B/metabolism , Translocation, Genetic
13.
Cancer Cell ; 14(5): 369-81, 2008 Nov 04.
Article in English | MEDLINE | ID: mdl-18977326

ABSTRACT

Studies support the importance of microRNAs in physiological and pathological processes. Here we describe the regulation and function of miR-29 in myogenesis and rhabdomyosarcoma (RMS). Results demonstrate that in myoblasts, miR-29 is repressed by NF-kappaB acting through YY1 and the Polycomb group. During myogenesis, NF-kappaB and YY1 downregulation causes derepression of miR-29, which in turn accelerates differentiation by targeting its repressor YY1. However, in RMS cells and primary tumors that possess impaired differentiation, miR-29 is epigenetically silenced by an activated NF-kappaB-YY1 pathway. Reconstitution of miR-29 in RMS in mice inhibits tumor growth and stimulates differentiation, suggesting that miR-29 acts as a tumor suppressor through its promyogenic function. Together, these results identify a NF-kappaB-YY1-miR-29 regulatory circuit whose disruption may contribute to RMS.


Subject(s)
Gene Expression Regulation, Developmental , MicroRNAs/metabolism , Muscle Development/physiology , Myoblasts, Skeletal/cytology , NF-kappa B/metabolism , Rhabdomyosarcoma/metabolism , YY1 Transcription Factor/metabolism , Animals , Blotting, Western , Cell Cycle/physiology , Cell Differentiation/physiology , Cell Proliferation , Cells, Cultured , Chromatin Immunoprecipitation , Computational Biology , Down-Regulation , Feedback, Physiological , Fibroblasts , Humans , Mice , Mice, Inbred C57BL , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Myoblasts, Skeletal/metabolism , NF-kappa B/genetics , Nucleic Acid Conformation , Promoter Regions, Genetic , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/prevention & control , Signal Transduction , YY1 Transcription Factor/genetics
14.
Cancer Res ; 68(20): 8351-60, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18922907

ABSTRACT

We hypothesized that IFN-alpha would enhance the apoptotic activity of bortezomib on melanoma cells. Combined treatment with bortezomib and IFN-alpha induced synergistic apoptosis in melanoma and other solid tumor cell lines. Apoptosis was associated with processing of procaspase-3, procaspase-7, procaspase-8, and procaspase-9 and with cleavage of Bid and poly(ADP-ribose) polymerase. Bortezomib plus IFN-alpha was effective at inducing apoptosis in melanoma cells that overexpressed Bcl-2 or Mcl-1, suggesting that this treatment combination can overcome mitochondrial pathways of cell survival and resistance to apoptosis. The proapoptotic effects of this treatment combination were abrogated by a caspase-8 inhibitor, led to increased association of Fas and FADD before the onset of cell death, and were significantly reduced in cells transfected with a dominant-negative FADD construct or small interfering RNA targeting Fas. These data suggest that bortezomib and IFN-alpha act through the extrinsic pathway of apoptosis via FADD-induced caspase-8 activation to initiate cell death. Finally, bortezomib and IFN-alpha displayed statistically significant antitumor activity compared with either agent alone in both the B16 murine model of melanoma and in athymic mice bearing human A375 xenografts. These data support the future clinical development of bortezomib and IFN-alpha for malignant melanoma.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Boronic Acids/pharmacology , Interferon-alpha/pharmacology , Melanoma/drug therapy , Proto-Oncogene Proteins c-bcl-2/analysis , Pyrazines/pharmacology , Animals , Bortezomib , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/pathology , Caspase 3/metabolism , Caspase 8/metabolism , Fas-Associated Death Domain Protein/physiology , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Melanoma/chemistry , Melanoma/pathology , Mice , Mice, Inbred BALB C , Myeloid Cell Leukemia Sequence 1 Protein , Poly(ADP-ribose) Polymerases/metabolism
15.
J Cell Biol ; 180(4): 787-802, 2008 Feb 25.
Article in English | MEDLINE | ID: mdl-18299349

ABSTRACT

Nuclear factor kappaB (NF-kappaB) is involved in multiple skeletal muscle disorders, but how it functions in differentiation remains elusive given that both anti- and promyogenic activities have been described. In this study, we resolve this by showing that myogenesis is controlled by opposing NF-kappaB signaling pathways. We find that myogenesis is enhanced in MyoD-expressing fibroblasts deficient in classical pathway components RelA/p65, inhibitor of kappaB kinase beta (IKKbeta), or IKKgamma. Similar increases occur in myoblasts lacking RelA/p65 or IKKbeta, and muscles from RelA/p65 or IKKbeta mutant mice also contain higher fiber numbers. Moreover, we show that during differentiation, classical NF-kappaB signaling decreases, whereas the induction of alternative members IKKalpha, RelB, and p52 occurs late in myogenesis. Myotube formation does not require alternative signaling, but it is important for myotube maintenance in response to metabolic stress. Furthermore, overexpression or knockdown of IKKalpha regulates mitochondrial content and function, suggesting that alternative signaling stimulates mitochondrial biogenesis. Together, these data reveal a unique IKK/NF-kappaB signaling switch that functions to both inhibit differentiation and promote myotube homeostasis.


Subject(s)
I-kappa B Kinase/metabolism , Mitochondria/metabolism , Muscle Development/genetics , Muscle, Skeletal/embryology , Muscle, Skeletal/metabolism , Myoblasts, Skeletal/metabolism , NF-kappa B/metabolism , Animals , Animals, Newborn , Cell Differentiation/genetics , Cell Line , Cells, Cultured , Down-Regulation/genetics , Gene Expression Regulation, Developmental/genetics , I-kappa B Kinase/genetics , Mice , Mice, Transgenic , Mitochondria/ultrastructure , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/ultrastructure , Myoblasts, Skeletal/ultrastructure , Signal Transduction/genetics , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism
16.
Brain Behav Immun ; 22(2): 215-23, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17845840

ABSTRACT

Increased levels of proinflammatory cytokines, TNF-alpha and IL-6, predict mortality and morbidity. In cardiovascular disease patients, they are observed in atherosclerotic lesions and serum. Factors behind the increased levels of these cytokines are multifaceted and may include latent herpesviruses, such as Epstein-Barr virus (EBV) that can be reactivated by stress. Previously, we showed that the EBV-encoded deoxyuridine triphosphate nucleotidohydrolase (dUTPase), a protein synthesized in the early phase of virus replication, can induce human monocytes/macrophages to produce TNF-alpha and IL-6. In this study, we modeled the interactions that take place between macrophages and endothelial cells in vivo using human umbilical vein endothelial cells (HUVEC). HUVEC were stimulated by soluble factors induced by EBV dUTPase-treated monocyte-derived macrophages (MDM) that resulted in the upregulation of VCAM-1 and ICAM-1. These changes were related to MDM production of TNF-alpha following the activation of NF-kappaB. In a previous study, chronically stressed dementia caregivers had elevations in plasma IL-6 levels, a risk for cardiovascular disease. We found a relationship between plasma IL-6 levels and neutralizing antibody titers to EBV dUTPase suggesting that one source of the plasma IL-6 observed in our previous study could be related to the effect of EBV-encoded dUTPase on macrophages. The results suggest that EBV-encoded dUTPase can enhance production of proinflammatory cytokines by monocytes/macrophages in contact with endothelial cells of blood vessels, and may play a role in cardiovascular pathology and chronic inflammation.


Subject(s)
Atherosclerosis/immunology , Depressive Disorder/immunology , Epstein-Barr Virus Infections/immunology , Herpesvirus 4, Human/immunology , Pyrophosphatases/metabolism , Aged , Aged, 80 and over , Antibodies, Viral/blood , Atherosclerosis/epidemiology , Atherosclerosis/virology , Cell Communication/immunology , Cells, Cultured , Depressive Disorder/epidemiology , Depressive Disorder/virology , Endothelial Cells/cytology , Endothelial Cells/immunology , Endothelial Cells/metabolism , Epstein-Barr Virus Infections/epidemiology , Female , Herpesvirus 4, Human/enzymology , Herpesvirus 4, Human/genetics , Humans , Intercellular Adhesion Molecule-1/metabolism , Interleukin-6/metabolism , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Male , Middle Aged , Monocytes/cytology , Monocytes/immunology , Monocytes/metabolism , Pyrophosphatases/genetics , Risk Factors , Stress, Psychological/epidemiology , Stress, Psychological/immunology , Stress, Psychological/virology , Tumor Necrosis Factor-alpha/metabolism , Umbilical Veins/cytology , Vascular Cell Adhesion Molecule-1/metabolism
17.
Mol Cell Biol ; 25(12): 4956-68, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15923614

ABSTRACT

IkappaB inhibitor proteins are the primary regulators of NF-kappaB. In contrast to the defined regulatory interplay between NF-kappaB and IkappaBalpha, much less is known regarding the regulation of IkappaBbeta by NF-kappaB. Here, we describe in detail the regulation of IkappaBbeta by RelA/p65. Using p65(-/-) fibroblasts, we show that IkappaBbeta is profoundly reduced in these cells, but not in other NF-kappaB subunit knockouts. This regulation prevails during embryonic and postnatal development in a tissue-specific manner. Significantly, in both p65(-/-) cells and tissues, IkappaBalpha is also reduced, but not nearly to the same extent as IkappaBbeta, thus highlighting the degree to which IkappaBbeta is dependent on p65. This dependence is based on the ability of p65 to stabilize IkappaBbeta protein from the 26S proteasome, a process mediated in large part through the p65 carboxyl terminus. Furthermore, IkappaBbeta was found to exist in both a basally phosphorylated and a hyperphosphorylated form. While the hyperphosphorylated form is less abundant, it is also more stable and less dependent on p65 and its carboxyl domain. Finally, we show that in p65(-/-) fibroblasts, expression of a proteolysis-resistant form of IkappaBbeta, but not IkappaBalpha, causes a severe growth defect associated with apoptosis. Based on these findings, we propose that tight control of IkappaBbeta protein by p65 is necessary for the maintenance of cellular homeostasis.


Subject(s)
I-kappa B Proteins/metabolism , NF-kappa B/metabolism , Animals , Apoptosis , Cell Survival , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Fibroblasts/cytology , Fibroblasts/physiology , Gene Expression Regulation, Developmental , I-kappa B Kinase , I-kappa B Proteins/genetics , Mice , Mice, Knockout , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Serine/metabolism , Tissue Distribution , Transcription Factor RelA , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
18.
J Clin Invest ; 114(3): 370-8, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15286803

ABSTRACT

Cachexia is a syndrome characterized by wasting of skeletal muscle and contributes to nearly one-third of all cancer deaths. Cytokines and tumor factors mediate wasting by suppressing muscle gene products, but exactly which products are targeted by these cachectic factors is not well understood. Because of their functional relevance to muscle architecture, such targets are presumed to represent myofibrillar proteins, but whether these proteins are regulated in a general or a selective manner is also unclear. Here we demonstrate, using in vitro and in vivo models of muscle wasting, that cachectic factors are remarkably selective in targeting myosin heavy chain. In myotubes and mouse muscles, TNF-alpha plus IFN-gamma strongly reduced myosin expression through an RNA-dependent mechanism. Likewise, colon-26 tumors in mice caused the selective reduction of this myofibrillar protein, and this reduction correlated with wasting. Under these conditions, however, loss of myosin was associated with the ubiquitin-dependent proteasome pathway, which suggests that mechanisms used to regulate the expression of muscle proteins may be cachectic factor specific. These results shed new light on cancer cachexia by revealing that wasting does not result from a general downregulation of muscle proteins but rather is highly selective as to which proteins are targeted during the wasting state.


Subject(s)
Cachexia/metabolism , Colonic Neoplasms/complications , Gene Expression Regulation, Neoplastic , Muscle, Skeletal/metabolism , MyoD Protein/metabolism , Myosin Heavy Chains/metabolism , Animals , CHO Cells , Cell Line, Tumor , Colonic Neoplasms/metabolism , Cricetinae , Cricetulus , Cysteine Endopeptidases/metabolism , Disease Models, Animal , Down-Regulation , Drug Synergism , Gene Targeting , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Mice , Mice, Inbred Strains , Mice, Nude , Multienzyme Complexes/metabolism , Muscle, Skeletal/drug effects , MyoD Protein/drug effects , MyoD Protein/genetics , Myosin Heavy Chains/drug effects , Myosin Heavy Chains/genetics , Proteasome Endopeptidase Complex , Sensitivity and Specificity , Transcription, Genetic/drug effects , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Ubiquitins/metabolism
19.
J Biol Chem ; 278(4): 2294-303, 2003 Jan 24.
Article in English | MEDLINE | ID: mdl-12431991

ABSTRACT

NF-kappaB activation is classically defined as a transient response initiated by the degradation of IkappaB inhibitor proteins leading to nuclear import of NF-kappaB and culminating with the resynthesis of IkappaBalpha and subsequent inactivation of the transcription factor. Although this type of regulation is considered the paradigm for NF-kappaB activation, other regulatory profiles are known to exist. By far the most common of these is chronic or persistent activation of NF-kappaB. In comparison, regulation of NF-kappaB in a biphasic manner represents a profile that is scarcely documented and whose biological significance remains poorly understood. Here we show using differentiated skeletal muscle cells, that tumor necrosis factor (TNF) induces NF-kappaB activation in a biphasic manner. Unlike the first transient phase, which is terminated within 1 h of cytokine addition, the second phase persists for an additional 24-36 h. Biphasic activation is mediated at both the levels of NF-kappaB DNA binding and transactivation function, and both phases are dependent on the IKK/26 S proteasome pathway. We find that regulation of the first transient phase is mediated by the degradation and subsequent resynthesis of IkappaBalpha, as well as by a TNF-induced expression of A20. Second phase activity correlates with persistent down-regulation of both IkappaBalpha and IkappaBbeta proteins, derived from a continuous TNF signal. Finally, we demonstrate that inhibition of NF-kappaB prior to initiation of the second phase of activity inhibits cytokine-mediated loss of muscle proteins. We propose that the biphasic activation of NF-kappaB in response to TNF may play a key regulatory role in skeletal muscle wasting associated with cachexia.


Subject(s)
Cytokines/metabolism , Muscle, Skeletal/metabolism , NF-kappa B/metabolism , Proteasome Endopeptidase Complex , Tumor Necrosis Factor-alpha/metabolism , Animals , Blotting, Northern , Blotting, Western , Cachexia/metabolism , Cell Line , DNA/metabolism , Humans , I-kappa B Proteins/metabolism , Interferon-gamma/metabolism , Mice , Microscopy, Fluorescence , Muscle, Skeletal/cytology , NF-KappaB Inhibitor alpha , Peptide Hydrolases/metabolism , Protein Binding , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...