Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters

Publication year range
1.
Sensors (Basel) ; 20(2)2020 Jan 14.
Article in English | MEDLINE | ID: mdl-31947584

ABSTRACT

Reactive oxygen species (ROS) are central effectors of inflammation and play a key role in cell signaling. Previous reports have described an association between oxidative events and the modulation of innate immunity. However, the role of redox signaling in adaptive immunity is still not well understood. This work is based on a novel investigation of diamide, a specific oxidant of sulfhydryl groups, and it is the first performed in purified T cell tyrosine phosphorylation signaling. Our data show that ex vivo T cells respond to -SH group oxidation with a distinctive tyrosine phosphorylation response and that these events elicit specific cellular responses. The expression of two essential T-cell receptors, CD25 and CD62L, and T-cell cytokine release is also affected in a specific way. Experiments with Syk inhibitors indicate a major contribution of this kinase in these phenomena. This pilot work confirms the presence of crosstalk between oxidation of cysteine residues and tyrosine phosphorylation changes, resulting in a series of functional events in freshly isolated T cells. Our experiments show a novel role of Syk inhibitors in applying their anti-inflammatory action through the inhibition of a ROS-generated reaction.


Subject(s)
L-Selectin/metabolism , Receptors, Interleukin-2/metabolism , Signal Transduction/physiology , Syk Kinase/metabolism , T-Lymphocytes , Cell Survival , Cells, Cultured , Diamide , Humans , Oxidation-Reduction , Phosphorylation , T-Lymphocytes/metabolism , T-Lymphocytes/physiology
2.
Br J Cancer ; 120(5): 527-536, 2019 03.
Article in English | MEDLINE | ID: mdl-30723303

ABSTRACT

BACKGROUND: Interferon-induced expression of programmed cell death ligands (PD-L1/PD-L2) may sustain tumour immune-evasion. Patients featuring MET amplification, a genetic lesion driving transformation, may benefit from anti-MET treatment. We explored if MET-targeted therapy interferes with Interferon-γ modulation of PD-L1/PD-L2 in MET-amplified tumours. METHODS: PD-L1/PD-L2 expression and signalling pathways downstream of MET or Interferon-γ were analysed in MET-amplified tumour cell lines and in patient-derived tumour organoids, in basal condition, upon Interferon-γ stimulation, and after anti-MET therapy. RESULTS: PD-L1 and PD-L2 were upregulated in MET-amplified tumour cells upon Interferon-γ treatment. This induction was impaired by JNJ-605, a selective inhibitor of MET kinase activity, and MvDN30, an antibody inducing MET proteolytic cleavage. We found that activation of JAKs/ STAT1, signal transducers downstream of the Interferon-γ receptor, was neutralised by MET inhibitors. Moreover, JAK2 and MET associated in the same signalling complex depending on MET phosphorylation. Results were confirmed in MET-amplified organoids derived from human colorectal tumours, where JNJ-605 treatment revoked Interferon-γ induced PD-L1 expression. CONCLUSIONS: These data show that in MET-amplified cancers, treatment with MET inhibitors counteracts the induction of PD-1 ligands by Interferon-γ. Thus, therapeutic use of anti-MET drugs may provide additional clinical benefit over and above the intended inhibition of the target oncogene.


Subject(s)
B7-H1 Antigen/drug effects , Interferon-gamma/pharmacology , Neoplasms/genetics , Programmed Cell Death 1 Ligand 2 Protein/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Tumor Escape/drug effects , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Cell Line, Tumor , Colorectal Neoplasms/pathology , Humans , Janus Kinases/drug effects , Janus Kinases/metabolism , Liver Neoplasms/secondary , Molecular Targeted Therapy , Neoplasms/metabolism , Organoids , Programmed Cell Death 1 Ligand 2 Protein/genetics , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Proto-Oncogene Proteins c-met/genetics , Receptors, Interferon , STAT1 Transcription Factor/drug effects , STAT1 Transcription Factor/metabolism , Signal Transduction , Tumor Escape/genetics , Interferon gamma Receptor
3.
Proc Natl Acad Sci U S A ; 111(32): E3343-52, 2014 Aug 12.
Article in English | MEDLINE | ID: mdl-25071179

ABSTRACT

Glial cells are an integral part of functional communication in the brain. Here we show that astrocytes contribute to the fast dynamics of neural circuits that underlie normal cognitive behaviors. In particular, we found that the selective expression of tetanus neurotoxin (TeNT) in astrocytes significantly reduced the duration of carbachol-induced gamma oscillations in hippocampal slices. These data prompted us to develop a novel transgenic mouse model, specifically with inducible tetanus toxin expression in astrocytes. In this in vivo model, we found evidence of a marked decrease in electroencephalographic (EEG) power in the gamma frequency range in awake-behaving mice, whereas neuronal synaptic activity remained intact. The reduction in cortical gamma oscillations was accompanied by impaired behavioral performance in the novel object recognition test, whereas other forms of memory, including working memory and fear conditioning, remained unchanged. These results support a key role for gamma oscillations in recognition memory. Both EEG alterations and behavioral deficits in novel object recognition were reversed by suppression of tetanus toxin expression. These data reveal an unexpected role for astrocytes as essential contributors to information processing and cognitive behavior.


Subject(s)
Astrocytes/physiology , Recognition, Psychology/physiology , Animals , Astrocytes/drug effects , Brain Waves/drug effects , Brain Waves/physiology , Calcium Signaling , Carbachol/pharmacology , Electroencephalography , Gene Expression , Glutamic Acid/metabolism , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/physiology , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Neurological , Nerve Net/cytology , Nerve Net/physiology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Synaptic Transmission , Tetanus Toxin/genetics , Tetanus Toxin/metabolism , Tissue Culture Techniques , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
4.
Cancer Cell ; 9(2): 81-94, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16473276

ABSTRACT

RARA becomes an acute promyelocytic leukemia (APL) oncogene by fusion with any of five translocation partners. Unlike RARalpha, the fusion proteins homodimerize, which may be central to oncogenic activation. This model was tested by replacing PML with dimerization domains from p50NFkappaB (p50-RARalpha) or the rapamycin-sensitive dimerizing peptide of FKBP12 (F3-RARalpha). The X-RARalpha fusions recapitulated in vitro activities of PML-RARalpha. For F3-RARalpha, these properties were rapamycin sensitive. Although in vivo the artificial fusions alone are poor initiators of leukemia, p50-RARalpha readily cooperates with an activated mutant CDw131 to induce APL-like disease. These results demonstrate that the dimerization interface of RARalpha fusion partners is a critical element in APL pathogenesis while pointing to other features of PML for enhancing penetrance and progression.


Subject(s)
Leukemia, Promyelocytic, Acute/metabolism , Leukemia, Promyelocytic, Acute/pathology , Receptors, Retinoic Acid/chemistry , Receptors, Retinoic Acid/metabolism , Animals , Bone Marrow/pathology , Carcinogens/metabolism , Cell Line , DNA/metabolism , DNA-Binding Proteins/metabolism , Dimerization , Down-Regulation/genetics , Mice , Mice, Transgenic , Mutation/genetics , Myeloid Cells/metabolism , Myeloid Cells/pathology , Neoplasm Proteins/metabolism , Oncogene Proteins, Fusion/metabolism , Protein Binding , Protein Structure, Quaternary , Receptors, Cytokine/metabolism , Receptors, Retinoic Acid/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repressor Proteins/chemistry , Repressor Proteins/genetics , Repressor Proteins/metabolism , Retinoic Acid Receptor alpha , Retinoid X Receptors/metabolism , Transcription, Genetic/genetics
5.
Cancer ; 119(24): 4290-8, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24122767

ABSTRACT

BACKGROUND: In patients with previously untreated metastatic colorectal cancer (mCRC), we conducted a phase 1b/randomized phase 2 trial to define the safety, tolerability, and efficacy of mFOLFOX6 plus bevacizumab (mFOLFOX6/bev) with conatumumab, an investigational, fully human monoclonal IgG1 antibody that specifically activates death receptor 5 (DR5). METHODS: Twelve patients were enrolled in a phase 1b open-label dose-escalation trial of conatumumab with mFOLFOX6/bev; thereafter, 190 patients were randomized 1:1:1 to receive mFOLFOX6/bev in combination with 2 mg/kg conatumumab, 10 mg/kg conatumumab, or placebo. Therapy cycles were repeated every 2 weeks until disease progression or the occurrence of unacceptable toxicity. RESULTS: In phase 1b, conatumumab with mFOLFOX6/bev was tolerated without apparent added toxicity over mFOLFOX6/bev alone. In phase 2, conatumumab with mFOLFOX6/bev did not confer a benefit in progression-free survival when compared with placebo with mFOLFOX6/bev. Toxicity was similar in all treatment arms. Following treatment, similar increases in circulating caspase-3 levels were observed in all arms. CONCLUSIONS: Conatumumab with mFOLFOX6/bev did not offer improved efficacy over the same chemotherapy with placebo in first-line treatment of patients with mCRC. These data do not support further development of conatumumab in advanced CRC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bevacizumab , Biomarkers, Tumor/metabolism , Caspase 3/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Disease Progression , Disease-Free Survival , Dose-Response Relationship, Drug , Double-Blind Method , Female , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Humans , Leucovorin/administration & dosage , Leucovorin/adverse effects , Male , Middle Aged , Neoplasm Metastasis , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/adverse effects , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism
6.
Ann Surg ; 257(6): 1089-95, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23665971

ABSTRACT

OBJECTIVE: Upon colon cancer metastasis resection in liver, disease outcome is heterogeneous, ranging from indolent to very aggressive, with early recurrence. The aim of this study is to investigate the capability of metastasis associated in colon cancer 1 (MACC1) levels measured in liver metastasis specimens to predict further recurrence of the disease. METHODS: Gene expression and gene dosage of MACC1, hepatocyte growth factor (HGF), and hepatocyte growth factor receptor (MET) were assessed using quantitative realtime polymerase chain reaction on a cohort of 64 liver metastasis samples from patients with complete follow-up of 36 months and detailed clinical annotation. The most relevant mutations associated to prognosis in colorectal cancer, KRAS, and PIK3CA were assessed on the same specimens with Sanger sequencing. RESULTS: Receiver operating characteristic (ROC) analysis revealed that MACC1 mRNA abundance is a good indicator of metastatic recurrence (AUC = 0.65, P < 0.05), whereas no such results were obtained with MET and HGF, nor with gene dosage. Generation of MACC1-based risk classes was capable of successfully separating patients into poor and good prognosis subgroups [hazard ratio (HR) = 5.236, 95% confidence interval (CI) = 1.2068-22.715, P < 0.05]. Also KRAS mutation was significantly associated with higher risk of recurrence (HR = 2.07, 95% CI = 1.048-4.09, P < 0.05). Cox regression multivariate analysis supported the independence of MACC1, but not KRAS, from known prognostic clinical information (Node Size HR = 3.155, 95% CI = 1.4418-6.905, P < 0.001, Preoperative carcinoembryonic antigen HR = 2.359, 95% CI = 1.0203-5.452, P < 0.05, MACC1 HR = 7.2739, 95% CI = 1.6584-31.905, P < 0.01). CONCLUSIONS: MACC1, a new easily detectable biomarker in cancer, is an independent prognostic factor of recurrence after liver resection of colorectal cancer metastasis.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Liver Neoplasms/genetics , Liver Neoplasms/secondary , Liver Neoplasms/surgery , Transcription Factors/genetics , Aged , Biomarkers, Tumor/genetics , Carcinoembryonic Antigen/metabolism , Class I Phosphatidylinositol 3-Kinases , Diagnostic Imaging , Female , Hepatocyte Growth Factor/genetics , Humans , Male , Monte Carlo Method , Mutation , Neoadjuvant Therapy , Neoplasm Recurrence, Local , Neoplasm Staging , Phosphatidylinositol 3-Kinases/genetics , Polymerase Chain Reaction , Predictive Value of Tests , Prognosis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins p21(ras) , RNA, Messenger , ROC Curve , Trans-Activators , ras Proteins/genetics
7.
Int J Cancer ; 130(6): 1357-66, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-21500189

ABSTRACT

The MET oncogene is amplified in a fraction of human gastric carcinoma cell lines, with consequent overexpression and constitutive activation of the corresponding protein product, the Met tyrosine kinase receptor. This genetically driven hyperactivation of Met is necessary for cancer cell growth and survival, so that Met pharmacological blockade results in cell-cycle arrest or apoptosis (oncogene addiction). MET gene amplification also occurs in vivo in a number of human gastric carcinomas, and clinical trials are now ongoing to assess the therapeutic efficacy of Met inhibitors in this type of malignancy. The aim of our study was to identify a preclinical algorithm of soluble surrogate biomarkers indicative of response to Met inhibition in gastric tumors, as a potential tool to integrate imaging criteria during patient follow-up. We started from a survey of candidate molecules based on antibody proteomics and gene expression profiling; after ELISA validation and analytical quantification, four biomarkers were identified that appeared to be strongly and consistently modulated by Met inhibition in a panel of Met-addicted gastric carcinoma cell lines, but not in Met-independent cell lines. Pharmacologic blockade of Met using specific small-molecule inhibitors led to reduced secretion of IL-8, GROα and the soluble form of uPAR and to increased production of IL-6 both in vitro (in culture supernatants) and in vivo (in the plasma of xenografted mice). If confirmed in patients, this information might prove useful to monitor clinical response to Met-targeted therapies in MET-amplified gastric carcinomas.


Subject(s)
Biomarkers, Tumor/metabolism , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Animals , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Cell Line, Tumor , Chemokine CXCL1/blood , Chemokine CXCL1/genetics , Chemokine CXCL1/metabolism , Female , Gene Expression Profiling , Humans , Indoles/pharmacology , Interleukin-6/blood , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-8/blood , Interleukin-8/genetics , Interleukin-8/metabolism , Mannose-Binding Lectins/blood , Mannose-Binding Lectins/genetics , Mannose-Binding Lectins/metabolism , Membrane Glycoproteins/blood , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Nude , Proteomics , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Receptors, Cell Surface/blood , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Stomach Neoplasms/blood , Stomach Neoplasms/genetics , Sulfones/pharmacology , Xenograft Model Antitumor Assays
8.
Antioxidants (Basel) ; 10(2)2021 Feb 02.
Article in English | MEDLINE | ID: mdl-33540918

ABSTRACT

Diabetes-associated long-term hyperglycaemia leads to oxidative stress-mediated fibrosis in different tissues and organs. Endothelial-to-mesenchymal-transition (EndMT) appears to play a role in diabetes-associated fibrotic conditions. Here, we investigate whether EndMT is implicated in the diabetic retinopathy fibrotic process and evaluate the possibility that resveratrol could counteract EndMT by inhibiting high glucose (HG)-induced increases in ROS. Primary Human Retinal Endothelial Cells (HRECs) were either pre-treated for 24 h with 1 µM resveratrol or left untreated, then glucose (30 mM) was applied at 3-day intervals for 10 days. qRT-PCR and ELISA were used to detect mRNA or protein expression of endothelial markers (CD31, CDH5, vWF) or mesenchymal markers (VIM, αSMA and collagen I), respectively. Intracellular ROS levels were measured with carboxy-DCFDA, while NOX-associated ROS levels were evaluated using the NADPH-specific redox biosensor p47-roGFP. Treatment of HRECs with HG increased intracellular ROS levels and promoted phenotype shifting towards EndMT, evidenced by decreased expression of endothelial markers concomitant with increased expression of mesenchymal ones. HG-induced EndMT appears to be mediated by NADPH-associated ROS generation as pre-treatment of HRECs with resveratrol or the NADPH inhibitor, diphenyleneiodonium chloride (DPI), attenuated ROS production and EndMT transition, suggesting that the effect of resveratrol on HG-induced ROS occurs via down-regulation of NADPH oxidase. It is worth noting that resveratrol or Chelerythrine, a Protein kinase C (PKC) inhibitor, reduce ROS and EndMT in HG-exposed cells, suggesting that NADPH activation occurs via a PKC-dependent mechanism. Taken together, our results provide the basis for a resveratrol-based potential protective therapy to prevent diabetic-associated complications.

9.
Nat Genet ; 53(1): 86-99, 2021 01.
Article in English | MEDLINE | ID: mdl-33414553

ABSTRACT

Patient-derived xenografts (PDXs) are resected human tumors engrafted into mice for preclinical studies and therapeutic testing. It has been proposed that the mouse host affects tumor evolution during PDX engraftment and propagation, affecting the accuracy of PDX modeling of human cancer. Here, we exhaustively analyze copy number alterations (CNAs) in 1,451 PDX and matched patient tumor (PT) samples from 509 PDX models. CNA inferences based on DNA sequencing and microarray data displayed substantially higher resolution and dynamic range than gene expression-based inferences, and they also showed strong CNA conservation from PTs through late-passage PDXs. CNA recurrence analysis of 130 colorectal and breast PT/PDX-early/PDX-late trios confirmed high-resolution CNA retention. We observed no significant enrichment of cancer-related genes in PDX-specific CNAs across models. Moreover, CNA differences between patient and PDX tumors were comparable to variations in multiregion samples within patients. Our study demonstrates the lack of systematic copy number evolution driven by the PDX mouse host.


Subject(s)
DNA Copy Number Variations/genetics , Xenograft Model Antitumor Assays , Animals , Databases, Genetic , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasm Metastasis , Polymorphism, Single Nucleotide/genetics , Exome Sequencing
10.
J Clin Invest ; 117(8): 2145-54, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17627301

ABSTRACT

The initiation of mammalian puberty and the maintenance of female reproductive cycles are events controlled by hypothalamic neurons that secrete the decapeptide gonadotropin-releasing hormone (GnRH). GnRH secretion is, in turn, controlled by changes in neuronal and glial inputs to GnRH-producing neurons. The hierarchical control of the process is unknown, but it requires coordinated regulation of these cell-cell interactions. Here we report the functional characterization of a gene (termed enhanced at puberty 1 [EAP1]) that appears to act as an upstream transcriptional regulator of neuronal networks controlling female reproductive function. EAP1 expression increased selectively at puberty in both the nonhuman primate and rodent hypothalamus. EAP1 encoded a nuclear protein expressed in neurons involved in the inhibitory and facilitatory control of reproduction. EAP1 transactivated genes required for reproductive function, such as GNRH1, and repressed inhibitory genes, such as preproenkephalin. It contained a RING finger domain of the C3HC4 subclass required for this dual transcriptional activity. Inhibition of EAP1 expression, targeted to the rodent hypothalamus via lentivirus-mediated delivery of EAP1 siRNAs, delayed puberty, disrupted estrous cyclicity, and resulted in ovarian abnormalities. These results suggest that EAP1 is a transcriptional regulator that, acting within the neuroendocrine brain, contributes to controlling female reproductive function.


Subject(s)
Estrous Cycle/metabolism , Gonadotropin-Releasing Hormone/biosynthesis , Hypothalamo-Hypophyseal System/metabolism , Neoplasm Proteins/biosynthesis , Neurons/metabolism , Protein Precursors/biosynthesis , Sexual Maturation , Transcription Factors/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Down-Regulation/genetics , Estrous Cycle/genetics , Female , Gonadotropin-Releasing Hormone/genetics , Humans , Hypothalamo-Hypophyseal System/cytology , Lentivirus , Macaca mulatta , Neoplasm Proteins/genetics , Neuroglia/cytology , Neuroglia/pathology , Neurons/cytology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Ovary/cytology , Ovary/growth & development , Ovary/metabolism , Protein Precursors/genetics , Protein Structure, Tertiary/genetics , Rats , Rats, Sprague-Dawley , Securin , Sexual Maturation/genetics , Transcription Factors/genetics , Transduction, Genetic
11.
Methods ; 49(1): 70-7, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19559089

ABSTRACT

Non-human primates (NHPs) are an invaluable resource for the study of genetic regulation of disease mechanisms. The main disadvantage of using NHPs as a preclinical model of human disease is the difficulty of manipulating the monkey genome using conventional gene modifying strategies. Lentiviruses offer the possibility of circumventing this difficulty because they can infect and transduce either dividing or nondividing cells, without producing an immune response. In addition, lentiviruses can permanently integrate into the genome of host cells, and are able to maintain long-term expression. In this article we describe the lentiviral vectors that we use to both express transgenes and suppress expression of endogenous genes via RNA interference (RNAi) in NHPs. We also discuss the safety features of currently available vectors that are especially important when lentiviral vectors are used in a species as closely related to humans as NHPs. Finally, we describe in detail the lentiviral vector production protocol we use and provide examples of how the vector can be employed to target peripheral tissues and the brain.


Subject(s)
Gene Expression Regulation , Gene Transfer Techniques , Genetic Vectors , Lentivirus/genetics , Primates/genetics , Primates/metabolism , Animals , Humans
12.
Nature ; 426(6963): 190-3, 2003 Nov 13.
Article in English | MEDLINE | ID: mdl-14614508

ABSTRACT

Fasting triggers a series of hormonal cues that promote energy balance by inducing glucose output and lipid breakdown in the liver. In response to pancreatic glucagon and adrenal cortisol, the cAMP-responsive transcription factor CREB activates gluconeogenic and fatty acid oxidation programmes by stimulating expression of the nuclear hormone receptor coactivator PGC-1 (refs 2-5). In parallel, fasting also suppresses lipid storage and synthesis (lipogenic) pathways, but the underlying mechanism is unknown. Here we show that mice deficient in CREB activity have a fatty liver phenotype and display elevated expression of the nuclear hormone receptor PPAR-gamma, a key regulator of lipogenic genes. CREB inhibits hepatic PPAR-gamma expression in the fasted state by stimulating the expression of the Hairy Enhancer of Split (HES-1) gene, a transcriptional repressor that is shown here to be a mediator of fasting lipid metabolism in vivo. The coordinate induction of PGC-1 and repression of PPAR-gamma by CREB during fasting provides a molecular rationale for the antagonism between insulin and counter-regulatory hormones, and indicates a potential role for CREB antagonists as therapeutic agents in enhancing insulin sensitivity in the liver.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression Regulation , Lipid Metabolism , Liver/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors , Cell Line, Tumor , Cyclic AMP Response Element-Binding Protein/genetics , Fasting , Fatty Liver/genetics , Fatty Liver/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , Phenotype , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factor HES-1 , Transcription Factors/genetics , Triglycerides/metabolism
13.
Sci Transl Med ; 12(555)2020 08 05.
Article in English | MEDLINE | ID: mdl-32759276

ABSTRACT

Blockade of epidermal growth factor receptor (EGFR) causes tumor regression in some patients with metastatic colorectal cancer (mCRC). However, residual disease reservoirs typically remain even after maximal response to therapy, leading to relapse. Using patient-derived xenografts (PDXs), we observed that mCRC cells surviving EGFR inhibition exhibited gene expression patterns similar to those of a quiescent subpopulation of normal intestinal secretory precursors with Paneth cell characteristics. Compared with untreated tumors, these pseudodifferentiated tumor remnants had reduced expression of genes encoding EGFR-activating ligands, enhanced activity of human epidermal growth factor receptor 2 (HER2) and HER3, and persistent signaling along the phosphatidylinositol 3-kinase (PI3K) pathway. Clinically, properties of residual disease cells from the PDX models were detected in lingering tumors of responsive patients and in tumors of individuals who had experienced early recurrence. Mechanistically, residual tumor reprogramming after EGFR neutralization was mediated by inactivation of Yes-associated protein (YAP), a master regulator of intestinal epithelium recovery from injury. In preclinical trials, Pan-HER antibodies minimized residual disease, blunted PI3K signaling, and induced long-term tumor control after treatment discontinuation. We found that tolerance to EGFR inhibition is characterized by inactivation of an intrinsic lineage program that drives both regenerative signaling during intestinal repair and EGFR-dependent tumorigenesis. Thus, our results shed light on CRC lineage plasticity as an adaptive escape mechanism from EGFR-targeted therapy and suggest opportunities to preemptively target residual disease.


Subject(s)
Colorectal Neoplasms , Phosphatidylinositol 3-Kinases , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , ErbB Receptors , Humans , Neoplasm Recurrence, Local , Neoplasm, Residual , Paneth Cells , Phenotype
14.
Sci Adv ; 6(26): eaba4353, 2020 06.
Article in English | MEDLINE | ID: mdl-32637608

ABSTRACT

Fibroblast-like synoviocytes (FLS) are joint-lining cells that promote rheumatoid arthritis (RA) pathology. Current disease-modifying antirheumatic agents (DMARDs) operate through systemic immunosuppression. FLS-targeted approaches could potentially be combined with DMARDs to improve control of RA without increasing immunosuppression. Here, we assessed the potential of immunoglobulin-like domains 1 and 2 (Ig1&2), a decoy protein that activates the receptor tyrosine phosphatase sigma (PTPRS) on FLS, for RA therapy. We report that PTPRS expression is enriched in synovial lining RA FLS and that Ig1&2 reduces migration of RA but not osteoarthritis FLS. Administration of an Fc-fusion Ig1&2 attenuated arthritis in mice without affecting innate or adaptive immunity. Furthermore, PTPRS was down-regulated in FLS by tumor necrosis factor (TNF) via a phosphatidylinositol 3-kinase-mediated pathway, and TNF inhibition enhanced PTPRS expression in arthritic joints. Combination of ineffective doses of TNF inhibitor and Fc-Ig1&2 reversed arthritis in mice, providing an example of synergy between FLS-targeted and immunosuppressive DMARD therapies.


Subject(s)
Antirheumatic Agents , Arthritis, Rheumatoid , Synoviocytes , Animals , Antirheumatic Agents/therapeutic use , Cells, Cultured , Fibroblasts/metabolism , Mice , Synoviocytes/metabolism , Synoviocytes/pathology , Tumor Necrosis Factor-alpha/metabolism
15.
J Am Soc Nephrol ; 19(10): 1904-18, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18614774

ABSTRACT

Macrophage-stimulating protein (MSP) exerts proliferative and antiapoptotic effects, suggesting that it may play a role in tubular regeneration after acute kidney injury. In this study, elevated plasma levels of MSP were found both in critically ill patients with acute renal failure and in recipients of renal allografts during the first week after transplantation. In addition, MSP and its receptor, RON, were markedly upregulated in the regenerative phase after glycerol-induced tubular injury in mice. In vitro, MSP stimulated tubular epithelial cell proliferation and conferred resistance to cisplatin-induced apoptosis by inhibiting caspase activation and modulating Fas, mitochondrial proteins, Akt, and extracellular signal-regulated kinase. MSP also enhanced migration, scattering, branching morphogenesis, tubulogenesis, and mesenchymal de-differentiation of surviving tubular cells. In addition, MSP induced an embryonic phenotype characterized by Pax-2 expression. In conclusion, MSP is upregulated during the regeneration of injured tubular cells, and it exerts multiple biologic effects that may aid recovery from acute kidney injury.


Subject(s)
Acute Kidney Injury/blood , Hepatocyte Growth Factor/blood , Kidney Transplantation , Kidney Tubules/physiology , Proto-Oncogene Proteins/blood , Receptor Protein-Tyrosine Kinases/blood , Regeneration/physiology , Aged , Animals , Case-Control Studies , Cell Culture Techniques , Cell Survival , Critical Illness , Humans , Mice , Mice, Inbred C57BL , Middle Aged
16.
Clin Cancer Res ; 24(17): 4297-4308, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29764854

ABSTRACT

Purpose: c-KIT overexpression is well recognized in cancers such as gastrointestinal stromal tumors (GIST), small cell lung cancer (SCLC), melanoma, non-small cell lung cancer (NSCLC), and acute myelogenous leukemia (AML). Treatment with the small-molecule inhibitors imatinib, sunitinib, and regorafenib resulted in resistance (c-KIT mutant tumors) or limited activity (c-KIT wild-type tumors). We selected an anti-c-KIT ADC approach to evaluate the anticancer activity in multiple disease models.Experimental Design: A humanized anti-c-KIT antibody LMJ729 was conjugated to the microtubule destabilizing maytansinoid, DM1, via a noncleavable linker (SMCC). The activity of the resulting ADC, LOP628, was evaluated in vitro against GIST, SCLC, and AML models and in vivo against GIST and SCLC models.Results: LOP628 exhibited potent antiproliferative activity on c-KIT-positive cell lines, whereas LMJ729 displayed little to no effect. At exposures predicted to be clinically achievable, LOP628 demonstrated single administration regressions or stasis in GIST and SCLC xenograft models in mice. LOP628 also displayed superior efficacy in an imatinib-resistant GIST model. Further, LOP628 was well tolerated in monkeys with an adequate therapeutic index several fold above efficacious exposures. Safety findings were consistent with the pharmacodynamic effect of neutropenia due to c-KIT-directed targeting. Additional toxicities were considered off-target and were consistent with DM1, such as effects in the liver and hematopoietic/lymphatic system.Conclusions: The preclinical findings suggest that the c-KIT-directed ADC may be a promising therapeutic for the treatment of mutant and wild-type c-KIT-positive cancers and supported the clinical evaluation of LOP628 in GIST, AML, and SCLC patients. Clin Cancer Res; 24(17); 4297-308. ©2018 AACR.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Immunoconjugates/pharmacology , Neoplasms/drug therapy , Proto-Oncogene Proteins c-kit/genetics , Animals , Antibodies, Anti-Idiotypic/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/immunology , Heterografts , Humans , Imatinib Mesylate/pharmacology , Immunoconjugates/immunology , Mice , Mutation , Neoplasms/classification , Neoplasms/immunology , Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-kit/immunology
17.
Nat Commun ; 8: 15107, 2017 05 31.
Article in English | MEDLINE | ID: mdl-28561063

ABSTRACT

Stromal content heavily impacts the transcriptional classification of colorectal cancer (CRC), with clinical and biological implications. Lineage-dependent stromal transcriptional components could therefore dominate over more subtle expression traits inherent to cancer cells. Since in patient-derived xenografts (PDXs) stromal cells of the human tumour are substituted by murine counterparts, here we deploy human-specific expression profiling of CRC PDXs to assess cancer-cell intrinsic transcriptional features. Through this approach, we identify five CRC intrinsic subtypes (CRIS) endowed with distinctive molecular, functional and phenotypic peculiarities: (i) CRIS-A: mucinous, glycolytic, enriched for microsatellite instability or KRAS mutations; (ii) CRIS-B: TGF-ß pathway activity, epithelial-mesenchymal transition, poor prognosis; (iii) CRIS-C: elevated EGFR signalling, sensitivity to EGFR inhibitors; (iv) CRIS-D: WNT activation, IGF2 gene overexpression and amplification; and (v) CRIS-E: Paneth cell-like phenotype, TP53 mutations. CRIS subtypes successfully categorize independent sets of primary and metastatic CRCs, with limited overlap on existing transcriptional classes and unprecedented predictive and prognostic performances.


Subject(s)
Colorectal Neoplasms/classification , Colorectal Neoplasms/genetics , Stromal Cells/metabolism , Transcriptome , Animals , Antineoplastic Agents, Immunological/pharmacology , Cell Lineage , Cetuximab/pharmacology , Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/immunology , ErbB Receptors/metabolism , Female , Gene Expression Profiling , Genes, p53 , Glycolysis , Heterografts , Humans , Insulin-Like Growth Factor II/genetics , Male , Mice , Microsatellite Instability , Mutation , Prognosis , Signal Transduction , Stromal Cells/pathology , Transforming Growth Factor beta/metabolism
18.
J Comp Neurol ; 498(4): 525-38, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-16874803

ABSTRACT

The adult mammalian spinal cord contains neural stem and/or progenitor cells that slowly multiply throughout life and differentiate exclusively into glia. The contribution of adult progenitors to repair has been highlighted in recent studies, demonstrating extensive cell proliferation and gliogenesis following central nervous system (CNS) trauma. The present experiments aimed to determine the relative roles of endogenously dividing progenitor cells versus quiescent progenitor cells in posttraumatic gliogenesis. Using the mitotic indicator bromodeoxyuridine (BrdU) and a retroviral vector, we found that, in the adult female Fisher 344 rat, endogenously dividing neural progenitors are acutely vulnerable in response to T8 dorsal hemisection spinal cord injury. We then studied the population of cells that divide postinjury in the injury epicenter by delivering BrdU or retrovirus at 24 hours after spinal cord injury. Animals were euthanized at five timepoints postinjury, ranging from 6 hours to 9 weeks after BrdU delivery. At all timepoints, we observed extensive proliferation of ependymal and periependymal cells that immunohistochemically resembled stem/progenitor cells. BrdU+ incorporation was noted to be prominent in NG2-immunoreactive progenitors that matured into oligodendrocytes, and in a transient population of microglia. Using a green fluorescence protein (GFP) hematopoietic chimeric mouse, we determined that 90% of the dividing cells in this early proliferation event originate from the spinal cord, whereas only 10% originate from the bone marrow. Our results suggest that dividing, NG2-expressing progenitor cells are vulnerable to injury, but a separate, immature population of neural stem and/or progenitor cells is activated by injury and rapidly divides to replace this vulnerable population.


Subject(s)
Cell Lineage/physiology , Nerve Regeneration/physiology , Neuroglia/cytology , Spinal Cord Injuries/pathology , Stem Cells/cytology , Wound Healing/physiology , Animals , Cell Differentiation , Cell Proliferation , Female , Immunohistochemistry , Mice , Mice, Inbred C57BL , Rats , Rats, Inbred F344 , Spinal Cord/cytology , Spinal Cord/physiology , Stem Cells/classification , Time Factors
19.
J Hematol Oncol ; 9(1): 63, 2016 07 29.
Article in English | MEDLINE | ID: mdl-27473052

ABSTRACT

We studied telomere length in 32 CML patients who discontinued imatinib after achieving complete molecular remission and 32 age-sex-matched controls. The relative telomere length (RTL) was determined by q-PCR as the telomere to single copy gene (36B4) ratio normalized to a reference sample (K-562 DNA). Age-corrected RTL (acRTL) was also obtained. The 36-month probability of treatment-free remission (TFR) was 59.4 %. TFR patients showed shorter acRTL compared to relapsed (mean ± SD = 0.01 ± 0.14 vs 0.20 ± 0.21; p = 0.01). TFR was significantly higher in CML patients with acRTL ≤0.09 (78.9 vs 30.8 %, p = 0.002). CML stem cells harboring longer telomeres possibly maintain a proliferative potential after treatment discontinuation.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Telomere Shortening , Withholding Treatment , Adult , Age Factors , Aged , Aged, 80 and over , Case-Control Studies , Female , Humans , Imatinib Mesylate/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Male , Middle Aged , Polymerase Chain Reaction , Remission Induction , Telomere/ultrastructure , Telomere Shortening/genetics
SELECTION OF CITATIONS
SEARCH DETAIL