Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 109
Filter
Add more filters

Publication year range
1.
Genes Dev ; 32(21-22): 1367-1379, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30366905

ABSTRACT

The mammalian circadian clock is encoded by an autoregulatory transcription feedback loop that drives rhythmic behavior and gene expression in the brain and peripheral tissues. Transcriptomic analyses indicate cell type-specific effects of circadian cycles on rhythmic physiology, although how clock cycles respond to environmental stimuli remains incompletely understood. Here, we show that activation of the inducible transcription factor NF-κB in response to inflammatory stimuli leads to marked inhibition of clock repressors, including the Period, Cryptochrome, and Rev-erb genes, within the negative limb. Furthermore, activation of NF-κB relocalizes the clock components CLOCK/BMAL1 genome-wide to sites convergent with those bound by NF-κB, marked by acetylated H3K27, and enriched in RNA polymerase II. Abrogation of NF-κB during adulthood alters the expression of clock repressors, disrupts clock-controlled gene cycles, and impairs rhythmic activity behavior, revealing a role for NF-κB in both unstimulated and activated conditions. Together, these data highlight NF-κB-mediated transcriptional repression of the clock feedback limb as a cause of circadian disruption in response to inflammation.


Subject(s)
Circadian Rhythm/genetics , NF-kappa B/physiology , ARNTL Transcription Factors/metabolism , Animals , Behavior, Animal , CLOCK Proteins/metabolism , Cell Line , Chromatin/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/metabolism , Repressor Proteins/metabolism , Transcription, Genetic
2.
BMC Cancer ; 24(1): 414, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38570770

ABSTRACT

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) presents with a high mortality rate. Two important features of PDAC contribute to this poor outcome. The first is metastasis which occurs in ~ 80% of PDAC patients. The second is cachexia, which compromises treatment tolerance for patients and reduces their quality of life. Although various mouse models of PDAC exist, recapitulating both metastatic and cachectic features have been challenging. METHODS: Here, we optimize an orthotopic mouse model of PDAC by altering several conditions, including the subcloning of parental murine PDAC cells, implantation site, number of transplanted cells, and age of recipient mice. We perform spatial profiling to compare primary and metastatic immune microenvironments and RNA sequencing to gain insight into the mechanisms of muscle wasting in PDAC-induced cachexia, comparing non-metastatic to metastatic conditions. RESULTS: These modifications extend the time course of the disease and concurrently increase the rate of metastasis to approximately 70%. Furthermore, reliable cachexia endpoints are achieved in both PDAC mice with and without metastases, which is reminiscent of patients. We also find that cachectic muscles from PDAC mice with metastasis exhibit a similar transcriptional profile to muscles derived from mice and patients without metastasis. CONCLUSION: Together, this model is likely to be advantageous in both advancing our understanding of the mechanism of PDAC cachexia, as well as in the evaluation of novel therapeutics.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Mice , Animals , Cachexia/genetics , Quality of Life , Pancreatic Neoplasms/complications , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Phenotype , Tumor Microenvironment
3.
Proc Natl Acad Sci U S A ; 116(36): 17980-17989, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31434789

ABSTRACT

The fat mass and obesity-associated gene (FTO) encodes an m6A RNA demethylase that controls mRNA processing and has been linked to both obesity and bone mineral density in humans by genome-wide association studies. To examine the role of FTO in bone, we characterized the phenotype of mice lacking Fto globally (FtoKO ) or selectively in osteoblasts (FtoOcKO ). Both mouse models developed age-related reductions in bone volume in both the trabecular and cortical compartments. RNA profiling in osteoblasts following acute disruption of Fto revealed changes in transcripts of Hspa1a and other genes in the DNA repair pathway containing consensus m6A motifs required for demethylation by FtoFto KO osteoblasts were more susceptible to genotoxic agents (UV and H2O2) and exhibited increased rates of apoptosis. Importantly, forced expression of Hspa1a or inhibition of NF-κB signaling normalized the DNA damage and apoptotic rates in Fto KO osteoblasts. Furthermore, increased metabolic stress induced in mice by feeding a high-fat diet induced greater DNA damage in osteoblast of FtoOc KO mice compared to controls. These data suggest that FTO functions intrinsically in osteoblasts through Hspa1a-NF-κB signaling to enhance the stability of mRNA of proteins that function to protect cells from genotoxic damage.


Subject(s)
Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Apoptosis , Bone and Bones/metabolism , DNA Damage , Osteoblasts/metabolism , Signal Transduction , Stress, Physiological , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics , Animals , Bone and Bones/pathology , Dietary Fats/adverse effects , Dietary Fats/pharmacology , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Hydrogen Peroxide/adverse effects , Hydrogen Peroxide/pharmacology , Mice , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/metabolism , Osteoblasts/pathology , Ultraviolet Rays/adverse effects
4.
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
5.
Semin Cell Dev Biol ; 54: 82-91, 2016 06.
Article in English | MEDLINE | ID: mdl-26385617

ABSTRACT

While changes in muscle protein synthesis and degradation have long been known to contribute to muscle wasting, a body of literature has arisen which suggests that regulation of the satellite cell and its ensuing regenerative program are impaired in atrophied muscle. Lessons learned from cancer cachexia suggest that this regulation is simply not a consequence, but a contributing factor to the wasting process. In addition to satellite cells, evidence from mouse models of cancer cachexia also suggests that non-satellite progenitor cells from the muscle microenvironment are also involved. This chapter in the series reviews the evidence of dysfunctional muscle repair in multiple wasting conditions. Potential mechanisms for this dysfunctional regeneration are discussed, particularly in the context of cancer cachexia.


Subject(s)
Cachexia/etiology , Muscle, Skeletal/physiopathology , Neoplasms/complications , Regeneration , Animals , Cachexia/metabolism , Cachexia/pathology , Humans , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/pathology , Muscular Atrophy/physiopathology , Myoblasts/metabolism , Myoblasts/pathology , Neoplasms/metabolism , Neoplasms/pathology
6.
Cancer Cell Int ; 18: 89, 2018.
Article in English | MEDLINE | ID: mdl-29983640

ABSTRACT

BACKGROUND: Sarcomas are malignant heterogeneous tumors of mesenchymal derivation. Dedifferentiated liposarcoma (DDLPS) is aggressive with recurrence in 80% and metastasis in 20% of patients. We previously found that miR-133a was significantly underexpressed in liposarcoma tissues. As this miRNA has recently been shown to be a tumor suppressor in many cancers, the objective of this study was to characterize the biological and molecular consequences of miR-133a underexpression in DDLPS. METHODS: Real-time PCR was used to evaluate expression levels of miR-133a in human DDLPS tissue, normal fat tissue, and human DDLPS cell lines. DDLPS cells were stably transduced with miR-133a vector to assess the effects in vitro on proliferation, cell cycle, cell death, migration, and metabolism. A Seahorse Bioanalyzer system was also used to assess metabolism in vivo by measuring glycolysis and oxidative phosphorylation (OXPHOS) in subcutaneous xenograft tumors from immunocompromised mice. RESULTS: miR-133a expression was significantly decreased in human DDLPS tissue and cell lines. Enforced expression of miR-133a decreased cell proliferation, impacted cell cycle progression kinetics, decreased glycolysis, and increased OXPHOS. There was no significant effect on cell death or migration. Using an in vivo xenograft mouse study, we showed that tumors with increased miR-133a expression had no difference in tumor growth compared to control, but did exhibit an increase in OXPHOS metabolic respiration. CONCLUSIONS: Based on our collective findings, we propose that in DDPLS, loss of miR-133a induces a metabolic shift due to a reduction in oxidative metabolism favoring a Warburg effect in DDLPS tumors, but this regulation on metabolism was not sufficient to affect DDPLS.

7.
Physiol Rev ; 90(2): 495-511, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20393192

ABSTRACT

NF-kappaB is a ubiquitiously expressed transcription factor that plays vital roles in innate immunity and other processes involving cellular survival, proliferation, and differentiation. Activation of NF-kappaB is controlled by an IkappaB kinase (IKK) complex that can direct either canonical (classical) NF-kappaB signaling by degrading the IkappaB inhibitor and releasing p65/p50 dimers to the nucleus, or causes p100 processing and nuclear translocation of RelB/p52 via a noncanonical (alternative) pathway. Under physiological conditions, NF-kappaB activity is transiently regulated, whereas constitutive activation of this transcription factor typically in the classical pathway is associated with a multitude of disease conditions, including those related to skeletal muscle. How NF-kappaB functions in muscle diseases is currently under intense investigation. Insight into this role of NF-kappaB may be gained by understanding at a more basic level how this transcription factor contributes to skeletal muscle cell differentiation. Recent data from knockout mice support that the classical NF-kappaB pathway functions as an inhibitor of skeletal myogenesis and muscle regeneration acting through multiple mechanisms. In contrast, alternative NF-kappaB signaling does not appear to be required for myofiber conversion, but instead functions in myotube homeostasis by regulating mitochondrial biogenesis. Additional knowledge of these signaling pathways in skeletal myogenesis should aid in the development of specific inhibitors that may be useful in treatments of muscle disorders.


Subject(s)
Muscle Development/physiology , Muscle, Skeletal/growth & development , NF-kappa B/metabolism , Signal Transduction/physiology , Animals , Muscular Diseases/metabolism
8.
Proc Natl Acad Sci U S A ; 111(12): 4525-9, 2014 Mar 25.
Article in English | MEDLINE | ID: mdl-24616506

ABSTRACT

MicroRNAs (miRNAs) are small, noncoding RNAs that regulate gene expression and, in cancers, are often packaged within secreted microvesicles. The cachexia syndrome is a debilitating state of cancer that predominantly results from the loss of skeletal muscle mass, which is in part associated with apoptosis. How tumors promote apoptosis in distally located skeletal muscles has not been explored. Using both tumor cell lines and patient samples, we show that tumor-derived microvesicles induce apoptosis of skeletal muscle cells. This proapoptotic activity is mediated by a microRNA cargo, miR-21, which signals through the Toll-like 7 receptor (TLR7) on murine myoblasts to promote cell death. Furthermore, tumor microvesicles and miR-21 require c-Jun N-terminal kinase activity to regulate this apoptotic response. Together, these results describe a unique pathway by which tumor cells promote muscle loss, which might provide a great insight into elucidating the causes and treatment options of cancer cachexia.


Subject(s)
Apoptosis/genetics , Cachexia/pathology , MicroRNAs/physiology , Muscle, Skeletal/pathology , Neoplasms/complications , Organelles/genetics , Toll-Like Receptor 7/physiology , Animals , Cachexia/etiology , Cell Line, Tumor , Humans , Mice , Neoplasms/pathology
9.
Angew Chem Int Ed Engl ; 56(6): 1525-1529, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28035784

ABSTRACT

Therapeutic applications of peptides are currently limited by their proteolytic instability and impermeability to the cell membrane. A general, reversible bicyclization strategy is now reported to increase both the proteolytic stability and cell permeability of peptidyl drugs. A peptide drug is fused with a short cell-penetrating motif and converted into a conformationally constrained bicyclic structure through the formation of a pair of disulfide bonds. The resulting bicyclic peptide has greatly enhanced proteolytic stability as well as cell-permeability. Once inside the cell, the disulfide bonds are reduced to produce a linear, biologically active peptide. This strategy was applied to generate a cell-permeable bicyclic peptidyl inhibitor against the NEMO-IKK interaction.


Subject(s)
Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/pharmacology , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Protein Interaction Maps/drug effects , Amino Acid Sequence , Cell Membrane Permeability , Cell-Penetrating Peptides/metabolism , Cell-Penetrating Peptides/pharmacokinetics , Drug Discovery , Drug Stability , HeLa Cells , Humans , I-kappa B Kinase/metabolism , Peptides, Cyclic/metabolism , Peptides, Cyclic/pharmacokinetics , Pharmacokinetics , Proteolysis , Solid-Phase Synthesis Techniques
10.
J Physiol ; 594(15): 4371-88, 2016 08 01.
Article in English | MEDLINE | ID: mdl-26990577

ABSTRACT

KEY POINTS: Using an experimental rat intensive care unit (ICU) model, not limited by early mortality, we have previously shown that passive mechanical loading attenuates the loss of muscle mass and force-generation capacity associated with the ICU intervention. Mitochondrial dynamics have recently been shown to play a more important role in muscle atrophy than previously recognized. In this study we demonstrate that mitochondrial dynamics, as well as mitophagy, is affected by mechanosensing at the transcriptional level, and muscle changes induced by unloading are counteracted by passive mechanical loading. The recently discovered ubiquitin ligases Fbxo31 and SMART are induced by mechanical silencing, an induction that similarly is prevented by passive mechanical loading. ABSTRACT: The complete loss of mechanical stimuli of skeletal muscles, i.e. loss of external strain related to weight bearing and internal strain related to activation of contractile proteins, in mechanically ventilated, deeply sedated and/or pharmacologically paralysed intensive care unit (ICU) patients is an important factor triggering the critical illness myopathy (CIM). Using a unique experimental ICU rat model, mimicking basic ICU conditions, we have recently shown that mechanical silencing is a dominant factor triggering the preferential loss of myosin, muscle atrophy and decreased specific force in fast- and slow-twitch muscles and muscle fibres. The aim of this study is to gain improved understanding of the gene signature and molecular pathways regulating the process of mechanical activation of skeletal muscle that are affected by the ICU condition. We have focused on pathways controlling myofibrillar protein synthesis and degradation, mitochondrial homeostasis and apoptosis. We demonstrate that genes regulating mitochondrial dynamics, as well as mitophagy are induced by mechanical silencing and that these effects are counteracted by passive mechanical loading. In addition, the recently identified ubiquitin ligases Fbxo31 and SMART are induced by mechanical silencing, an induction that is reversed by passive mechanical loading. Thus, mechano-cell signalling events are identified which may play an important role for the improved clinical outcomes reported in response to the early mobilization and physical therapy in immobilized ICU patients.


Subject(s)
Critical Illness , Muscle, Skeletal/metabolism , Muscular Atrophy/genetics , Animals , Female , Gene Expression Profiling , Immunoglobulin G/metabolism , Intensive Care Units , Muscular Atrophy/metabolism , Pulmonary Ventilation , Rats, Sprague-Dawley , Signal Transduction
11.
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
12.
FASEB J ; 29(11): 4544-54, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26178166

ABSTRACT

Early treatment with heart failure drugs lisinopril and spironolactone improves skeletal muscle pathology in Duchenne muscular dystrophy (DMD) mouse models. The angiotensin converting enzyme inhibitor lisinopril and mineralocorticoid receptor (MR) antagonist spironolactone indirectly and directly target MR. The presence and function of MR in skeletal muscle have not been explored. MR mRNA and protein are present in all tested skeletal muscles from both wild-type mice and DMD mouse models. MR expression is cell autonomous in both undifferentiated myoblasts and differentiated myotubes from mouse and human skeletal muscle cultures. To test for MR function in skeletal muscle, global gene expression analysis was conducted on human myotubes treated with MR agonist (aldosterone; EC50 1.3 nM) or antagonist (spironolactone; IC50 1.6 nM), and 53 gene expression differences were identified. Five differences were conserved in quadriceps muscles from dystrophic mice treated with spironolactone plus lisinopril (IC50 0.1 nM) compared with untreated controls. Genes down-regulated more than 2-fold by MR antagonism included FOS, ANKRD1, and GADD45B, with known roles in skeletal muscle, in addition to NPR3 and SERPINA3, bona fide targets of MR in other tissues. MR is a novel drug target in skeletal muscle and use of clinically safe antagonists may be beneficial for muscle diseases.


Subject(s)
Aldosterone/pharmacology , Lisinopril/pharmacology , Muscle Fibers, Skeletal/metabolism , Muscle Proteins , Receptors, Melanocortin , Spironolactone/pharmacology , Animals , Cell Line , Humans , Mice , Muscle Proteins/agonists , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/metabolism , Muscular Diseases/drug therapy , Muscular Diseases/metabolism , Receptors, Melanocortin/agonists , Receptors, Melanocortin/antagonists & inhibitors , Receptors, Melanocortin/metabolism
13.
Int J Mol Sci ; 17(3): 323, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26938530

ABSTRACT

The increasing prevalence of inflammatory diseases and the adverse effects associated with the long-term use of current anti-inflammatory therapies prompt the identification of alternative approaches to reestablish immune balance. Apigenin, an abundant dietary flavonoid, is emerging as a potential regulator of inflammation. Here, we show that apigenin has immune-regulatory activity in vivo. Apigenin conferred survival to mice treated with a lethal dose of Lipopolysaccharide (LPS) restoring normal cardiac function and heart mitochondrial Complex I activity. Despite the adverse effects associated with high levels of splenocyte apoptosis in septic models, apigenin had no effect on reducing cell death. However, we found that apigenin decreased LPS-induced apoptosis in lungs, infiltration of inflammatory cells and chemotactic factors' accumulation, re-establishing normal lung architecture. Using NF-κB luciferase transgenic mice, we found that apigenin effectively modulated NF-κB activity in the lungs, suggesting the ability of dietary compounds to exert immune-regulatory activity in an organ-specific manner. Collectively, these findings provide novel insights into the underlying immune-regulatory mechanisms of dietary nutraceuticals in vivo.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Apigenin/pharmacology , Leukemic Infiltration/drug therapy , NF-kappa B/metabolism , Sepsis/drug therapy , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/therapeutic use , Apigenin/administration & dosage , Apigenin/therapeutic use , Apoptosis , Dietary Supplements , Leukemic Infiltration/immunology , Lipopolysaccharides/toxicity , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Sepsis/immunology , Spleen/drug effects , Spleen/metabolism , Spleen/pathology
14.
Mod Pathol ; 27(9): 1238-45, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24457462

ABSTRACT

Myxoid and round-cell liposarcoma is a frequently encountered liposarcoma subtype. The mainstay of treatment remains surgical excision with or without chemoradiation. However, treatment options are limited in the setting of metastatic disease. Cancer-testis antigens are immunogenic antigens with the expression largely restricted to testicular germ cells and various malignancies, making them attractive targets for cancer immunotherapy. Gene expression studies have reported the expression of various cancer-testis antigens in liposarcoma, with mRNA expression of CTAG1B, CTAG2, MAGEA9, and PRAME described specifically in myxoid and round-cell liposarcoma. Herein, we further explore the expression of the cancer-testis antigens MAGEA1, ACRBP, PRAME, and SSX2 in myxoid and round-cell liposarcoma by immunohistochemistry in addition to determining mRNA levels of CTAG2 (LAGE-1), PRAME, and MAGEA3 by quantitative real-time PCR. Samples in formalin-fixed paraffin-embedded blocks (n=37) and frozen tissue (n=8) were obtained for immunohistochemistry and quantitative real-time PCR, respectively. Full sections were stained with antibodies to MAGEA1, ACRBP, PRAME, and SSX2 and staining was assessed for intensity (1-2+) and percent tumor positivity. The gene expression levels of CTAG2, PRAME, and MAGEA3 were measured by quantitative real-time PCR. In total, 37/37 (100%) of the samples showed predominantly strong, homogenous immunoreactivity for PRAME. There was a variable, focal expression of MAGEA1 (11%) and SSX2 (16%) and no expression of ACRBP. Quantitative real-time PCR demonstrated PRAME and CTAG2 transcripts in all eight samples: six tumors with high mRNA levels; two tumors with low mRNA levels. The gene expression of MAGEA3 was not detected in the majority of cases. In conclusion, myxoid and round-cell liposarcomas consistently express PRAME by immunohistochemistry as well as CTAG2 and PRAME by qualitative real-time PCR. This supports the use of cancer-testis antigen-targeted immunotherapy in the treatment of this malignancy.


Subject(s)
Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Biomarkers, Tumor , Gene Expression Regulation/physiology , Liposarcoma, Myxoid/genetics , Liposarcoma, Myxoid/metabolism , RNA, Messenger/genetics , Antigens, Surface/genetics , Carrier Proteins/metabolism , Humans , Immunohistochemistry , Melanoma-Specific Antigens/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Real-Time Polymerase Chain Reaction , Repressor Proteins/metabolism
15.
Blood ; 119(23): 5467-77, 2012 Jun 07.
Article in English | MEDLINE | ID: mdl-22544702

ABSTRACT

Multiple transcription factors regulate B-cell commitment, which is coordinated with myeloid-erythroid lineage differentiation. NF-κB has long been speculated to regulate early B-cell development; however, this issue remains controversial. IκB kinase-α (IKKα) is required for splenic B-cell maturation but not for BM B-cell development. In the present study, we unexpectedly found defective BM B-cell development and increased myeloid-erythroid lineages in kinase-dead IKKα (KA/KA) knock-in mice. Markedly increased cytosolic p100, an NF-κB2-inhibitory form, and reduced nuclear NF-κB p65, RelB, p50, and p52, and IKKα were observed in KA/KA splenic and BM B cells. Several B- and myeloid-erythroid-cell regulators, including Pax5, were deregulated in KA/KA BM B cells. Using fetal liver and BM congenic transplantations and deleting IKKα from early hematopoietic cells in mice, this defect was identified as being B cell-intrinsic and an early event during hematopoiesis. Reintroducing IKKα, Pax5, or combined NF-κB molecules promoted B-cell development but repressed myeloid-erythroid cell differentiation in KA/KA BM B cells. The results of the present study demonstrate that IKKα regulates B-lineage commitment via combined canonical and noncanonical NF-κB transcriptional activities to target Pax5 expression during hematopoiesis.


Subject(s)
B-Lymphocytes/cytology , Gene Knock-In Techniques , I-kappa B Kinase/genetics , Lymphopoiesis , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Erythroid Cells/immunology , Erythroid Cells/metabolism , Gene Expression , Gene Expression Regulation , Hematopoiesis , I-kappa B Kinase/immunology , Interferon Regulatory Factors/genetics , Mice , Myeloid Cells/immunology , Myeloid Cells/metabolism , NF-kappa B/immunology , PAX5 Transcription Factor/genetics , Signal Transduction , Spleen/cytology
16.
Biogerontology ; 15(3): 217-32, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24652515

ABSTRACT

Mammalian skeletal muscles exhibit age-related adaptive and pathological remodeling. Several muscles in particular undergo progressive atrophy and degeneration beyond median lifespan. To better understand myocellular responses to aging, we used semi-quantitative global metabolomic profiling to characterize trends in metabolic changes between 15-month-old adult and 32-month-old aged Fischer 344 × Brown Norway (FBN) male rats. The FBN rat gastrocnemius muscle exhibits age-dependent atrophy, whereas the soleus muscle, up until 32 months, exhibits markedly fewer signs of atrophy. Both gastrocnemius and soleus muscles were analyzed, as well as plasma and urine. Compared to adult gastrocnemius, aged gastrocnemius showed evidence of reduced glycolytic metabolism, including accumulation of glycolytic, glycogenolytic, and pentose phosphate pathway intermediates. Pyruvate was elevated with age, yet levels of citrate and nicotinamide adenine dinucleotide were reduced, consistent with mitochondrial abnormalities. Indicative of muscle atrophy, 3-methylhistidine and free amino acids were elevated in aged gastrocnemius. The monounsaturated fatty acids oleate, cis-vaccenate, and palmitoleate also increased in aged gastrocnemius, suggesting altered lipid metabolism. Compared to gastrocnemius, aged soleus exhibited far fewer changes in carbohydrate metabolism, but did show reductions in several glycolytic intermediates, fumarate, malate, and flavin adenine dinucleotide. Plasma biochemicals showing the largest age-related increases included glycocholate, heme, 1,5-anhydroglucitol, 1-palmitoleoyl-glycerophosphocholine, palmitoleate, and creatine. These changes suggest reduced insulin sensitivity in aged FBN rats. Altogether, these data highlight skeletal muscle group-specific perturbations of glucose and lipid metabolism consistent with mitochondrial dysfunction in aged FBN rats.


Subject(s)
Aging/metabolism , Blood Proteins/metabolism , Metabolomics , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Sarcopenia/metabolism , Aging/pathology , Amino Acids/metabolism , Animals , Blood Glucose/metabolism , Citric Acid Cycle/physiology , Fatty Acids/metabolism , Male , Mice, Inbred C57BL , Muscle, Skeletal/pathology , Rats, Inbred BN , Rats, Inbred F344 , Sarcopenia/pathology
17.
Mod Pathol ; 26(2): 282-8, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22936067

ABSTRACT

Liposarcomas are a heterogenous group of fat-derived sarcomas, and surgery with or without chemoradiation therapy remains the main stay of treatment. NY-ESO-1 is a cancer-testis antigen expressed in various cancers where it can induce both cellular and humoral immunity. Immunotherapy has shown promise in clinical trials involving NY-ESO-1-expressing tumors. Gene expression studies have shown upregulation of the gene for NY-ESO-1, CTAG1B, in myxoid and round cell liposarcomas. Herein, we evaluated the expression of NY-ESO-1 among liposarcoma subtypes by quantitative real-time PCR, western blot analysis, and immunohistochemistry. Frozen tissue for quantitative real-time PCR and western blot analysis was obtained for the following liposarcoma subtypes (n=15): myxoid and round cell (n=8); well-differentiated (n=4), and dedifferentiated (n=3). Formalin-fixed paraffin-embedded blocks were obtained for the following liposarcoma subtypes (n=44): myxoid and round cell (n=18); well-differentiated (n=10); dedifferentiated (n=10); and pleomorphic (n=6). Full sections were stained with monoclonal antibody NY-ESO-1, and staining was assessed for intensity (1-3+), percentage of tumor positivity, and location. In all, 7/8 (88%) and 16/18 (89%) myxoid and round cell expressed CTAG1B and NY-ESO-1 by quantitative real-time PCR and immunohistochemistry, respectively. Western blot correlated with mRNA expression levels. By immunohistochemistry, 94% (15/16) of positive cases stained homogenously with 2-3+ intensity. Also, 3/6 (50%) pleomorphic liposarcomas demonstrated a range of staining: 1+ intensity in 50% of cells; 2+ intensity in 5% of cells; and 3+ intensity in 90% of cells. One case of dedifferentiated liposarcoma showed strong, diffuse staining (3+ intensity in 75% of cells). Our study shows that both CTAG1B mRNA and protein are overexpressed with high frequency in myxoid and round cell liposarcoma, enabling the potential use of targeted immunotherapy in the treatment of this malignancy.


Subject(s)
Antigens, Neoplasm/metabolism , Liposarcoma/metabolism , Membrane Proteins/metabolism , Testicular Neoplasms/metabolism , Testis/metabolism , Antigens, Neoplasm/genetics , Humans , Liposarcoma/genetics , Liposarcoma/pathology , Male , Membrane Proteins/genetics , Retrospective Studies , Testicular Neoplasms/genetics , Testicular Neoplasms/pathology , Testis/pathology
18.
Curr Opin Clin Nutr Metab Care ; 16(3): 251-7, 2013 May.
Article in English | MEDLINE | ID: mdl-23493016

ABSTRACT

PURPOSE OF REVIEW: Nuclear factor-kappaB (NF-κB) activation is associated with a wide range of muscle-related diseases. Here, we review the evidence implicating specific NF-κB components in different disease pathologies and discuss therapies designed to target aberrant NF-κB signaling for the treatment of those pathologies. RECENT FINDINGS: Many components of the NF-κB signaling pathway contribute to muscle pathologies, presumably through activation of the transcription factor. In addition, an increasing number of upstream factors have been connected to disease progression. Genetic models and therapeutic approaches affecting these upstream targets associate with ameliorating disease progression. SUMMARY: Dissecting the crosstalk between NF-κB, its upstream mediators, and other signaling pathways is vital to our understanding of how activation of this signaling pathway is mediated in various diseases. The strides made in therapeutically inhibiting the NF-κB pathway provide some promise for the treatment of these diseases.


Subject(s)
Muscle, Skeletal/pathology , Muscular Diseases/pathology , Signal Transduction , Transcription Factor RelA/metabolism , Animals , Cachexia/genetics , Cachexia/pathology , Disease Models, Animal , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Models, Genetic , Muscle, Skeletal/metabolism , Muscular Diseases/genetics , Muscular Diseases/metabolism , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/metabolism , Transcription Factor RelA/genetics , Transcription Factors , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
19.
Cancer Cell ; 8(5): 421-32, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16286249

ABSTRACT

Cachexia contributes to nearly a third of all cancer deaths, yet the mechanisms underlying skeletal muscle wasting in this syndrome remain poorly defined. We report that tumor-induced alterations in the muscular dystrophy-associated dystrophin glycoprotein complex (DGC) represent a key early event in cachexia. Muscles from tumor-bearing mice exhibited membrane abnormalities accompanied by reduced levels of dystrophin and increased glycosylation on DGC proteins. Wasting was accentuated in tumor mdx mice lacking a DGC but spared in dystrophin transgenic mice that blocked induction of muscle E3 ubiquitin ligases. Furthermore, DGC deregulation correlated positively with cachexia in patients with gastrointestinal cancers. Based on these results, we propose that, similar to muscular dystrophy, DGC dysfunction plays a critical role in cancer-induced wasting.


Subject(s)
Cachexia/metabolism , Dystrophin-Associated Proteins/physiology , Dystrophin/physiology , Glycoproteins/physiology , Neoplasms/metabolism , Adult , Aged , Animals , Cachexia/complications , Dystrophin/metabolism , Dystrophin-Associated Proteins/metabolism , Female , Founder Effect , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred mdx , Mice, Transgenic , Middle Aged , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Neoplasms/complications , Neoplasms/pathology , Ubiquitin-Protein Ligases/metabolism
20.
Mol Ther ; 20(6): 1222-33, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22434133

ABSTRACT

microRNAs (miRNAs) are noncoding RNAs that regulate gene expression in post-transcriptional fashion, and emerging studies support their importance in a multitude of physiological and pathological processes. Here, we describe the regulation and function of miR-29 in Duchenne muscular dystrophy (DMD) and its potential use as therapeutic target. Our results demonstrate that miR-29 expression is downregulated in dystrophic muscles of mdx mice, a model of DMD. Restoration of its expression by intramuscular and intravenous injection improved dystrophy pathology by both promoting regeneration and inhibiting fibrogenesis. Mechanistic studies revealed that loss of miR-29 in muscle precursor cells (myoblasts) promotes their transdifferentiation into myofibroblasts through targeting extracellular molecules including collagens and microfibrillar-associated protein 5 (Mfap5). We further demonstrated that miR-29 is under negative regulation by transforming growth factor-ß (TGF-ß) signaling. Together, these results not only identify TGF-ß-miR-29 as a novel regulatory axis during myoblasts conversion into myofibroblasts which constitutes a novel contributing route to muscle fibrogenesis of DMD but also implicate miR-29 replacement therapy as a promising treatment approach for DMD.


Subject(s)
MicroRNAs/genetics , Muscular Dystrophy, Duchenne/genetics , Myoblasts/metabolism , Animals , Cell Differentiation , Cell Transdifferentiation , Cells, Cultured , Down-Regulation/genetics , Extracellular Matrix/genetics , Fibrosis/genetics , Gene Expression Regulation , Mice , Mice, Inbred mdx , MicroRNAs/administration & dosage , Models, Biological , Muscle, Skeletal/metabolism , Myoblasts/cytology , Myofibroblasts/cytology , Regeneration , Signal Transduction , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL