Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
2.
Transl Lung Cancer Res ; 13(4): 733-748, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38736495

ABSTRACT

Background: The identification of prognostic biomarkers is crucial for guiding treatment strategies in mesothelioma patients. The Duchenne muscular dystrophy (DMD) gene and its specific transcripts have been associated with patient survival in various tumours. In this study, we aimed to investigate the prognostic potential of DMD gene expression and its transcripts in mesothelioma patients. Methods: We analysed The Cancer Genome Atlas (TCGA) mesothelioma RNAseq, mutation, and clinical data to assess the association between DMD gene expression and its transcripts (Dp427, Dp71 splice variants) and mesothelioma survival. We also evaluated the specific Dp71 transcript as a unique prognostic biomarker across mesothelioma subtypes. Additionally, we performed differential gene expression analysis between high and low DMD gene/transcript expression groups. Results: The analysis included 57 epithelioid, 23 biphasic, two sarcomatoid, and five not otherwise specified (NOS) histological subtypes of mesothelioma samples. Univariate analysis revealed that high expression of the DMD gene and its Dp71 transcript was significantly associated with shorter survival in mesothelioma patients (P=0.003 and P<0.001, respectively). In a multivariate analysis, the association between Dp71 expression and survival remained significant [hazard ratio (HR) 2.29, 95% confidence interval (CI): 1.24-4.23, P=0.008] across all mesothelioma patients, and also among patients with mesotheliomas without deep CDKN2A deletions (HR 3.58, 95% CI: 1.31-9.80, P=0.01). Pathway analysis revealed enrichment of cell cycle (P=3.01×10-4) and homologous recombination (P=0.01) pathways in differentially expressed genes (DEGs) between high and low Dp71 groups. Furthermore, there were correlations between Dp71 transcript expression and tumour microenvironment (TME) cells, including a weak positive correlation with macrophages (R=0.32, P=0.002) specifically M2 macrophages (R=0.34, P=0.001). Conclusions: Our findings indicate that the differential expression of specific DMD transcripts is associated with poor survival in mesothelioma patients. The specific Dp71 transcript can serve as a potential biomarker for predicting patient survival in diverse histological subtypes of mesothelioma. Further studies are needed to understand the role of specific dystrophin transcripts in cancer and TME cells, and their implications in the pathogenesis and progression of mesothelioma. Identifying patients at risk of poor survival based on DMD transcript expression can guide treatment strategies in mesothelioma, informing decisions regarding treatment intensity, follow-up schedules, eligibility for clinical trials, and ultimately, end-of-life care planning.

3.
Front Pharmacol ; 15: 1367581, 2024.
Article in English | MEDLINE | ID: mdl-38681192

ABSTRACT

Introduction: Drug development is systemically inefficient. Research and development costs for novel therapeutics average hundreds of millions to billions of dollars, with the overall likelihood of approval estimated to be as low as 6.7% for oncology drugs. Over half of these failures are due to a lack of drug efficacy. This pervasive and repeated low rate of success exemplifies how preclinical models fail to adequately replicate the complexity and heterogeneity of human cancer. Therefore, new methods of evaluation, early in the development trajectory, are essential both to rule-in and rule-out novel agents with more rigor and speed, but also to spare clinical trial patients from the potentially toxic sequelae (high risk) of testing investigational agents that have a low likelihood of producing a response (low benefit). Methods: The clinical in vivo oncology (CIVO®) platform was designed to change this drug development paradigm. CIVO precisely delivers microdose quantities of up to 8 drugs or combinations directly into patient tumors 4-96 h prior to planned surgical resection. Resected tissue is then analyzed for responses at each site of intratumoral drug exposure. Results: To date, CIVO has been used safely in 6 clinical trials, including 68 subjects, with 5 investigational and 17 approved agents. Resected tissues were analyzed initially using immunohistochemistry and in situ hybridization assays (115 biomarkers). As technology advanced, the platform was paired with spatial biology analysis platforms, to successfully track anti-neoplastic and immune-modulating activity of the injected agents in the intact tumor microenvironment. Discussion: Herein we provide a report of the use of CIVO technology in patients, a depiction of the robust analysis methods enabled by this platform, and a description of the operational and regulatory mechanisms used to deploy this approach in synergistic partnership with pharmaceutical partners. We further detail how use of the CIVO platform is a clinically safe and scientifically precise alternative or complement to preclinical efficacy modeling, with outputs that inform, streamline, and de-risk drug development.

4.
Clin Cancer Res ; 29(18): 3813-3825, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37389981

ABSTRACT

PURPOSE: Cancer drug development is currently limited by a paradigm of preclinical evaluation that does not adequately recapitulate the complexity of the intact human tumor microenvironment (TME). To overcome this, we combined trackable intratumor microdosing (CIVO) with spatial biology readouts to directly assess drug effects in patient tumors in situ. EXPERIMENTAL DESIGN: In a first-of-its-kind phase 0 clinical trial, we explored the effects of an investigational stage SUMOylation-activating enzyme (SAE) inhibitor, subasumstat (TAK-981) in 12 patients with head and neck carcinoma (HNC). Patients scheduled for tumor resection received percutaneous intratumor injections of subasumstat and vehicle control 1 to 4 days before surgery, resulting in spatially localized and graded regions of drug exposure (∼1,000-2,000 µm in diameter). Drug-exposed (n = 214) and unexposed regions (n = 140) were compared by GeoMx Digital Spatial Profiler, with evaluation at single-cell resolution in a subset of these by CosMx Spatial Molecular Imager. RESULTS: Localized regions of subasumstat exposure revealed SUMO pathway inhibition, elevation of type I IFN response, and inhibition of cell cycle across all tumor samples. Single-cell analysis by CosMx demonstrated cell-cycle inhibition specific to the tumor epithelium, and IFN pathway induction commensurate with a TME shift from immune-suppressive to immune-permissive. CONCLUSIONS: Pairing CIVO with spatial profiling enabled detailed investigation of response to subasumstat across a diverse sampling of native and intact TME. We demonstrate that drug mechanism of action can be directly evaluated in a spatially precise manner in the most translationally relevant setting: an in situ human tumor.


Subject(s)
Antineoplastic Agents , Head and Neck Neoplasms , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Enzyme Inhibitors , Head and Neck Neoplasms/drug therapy , Tumor Microenvironment
5.
Sci Rep ; 11(1): 20885, 2021 10 22.
Article in English | MEDLINE | ID: mdl-34686736

ABSTRACT

SLC4A11 is a H+/NH3/water transport protein, of corneal endothelial cells. SLC4A11 mutations cause congenital hereditary endothelial dystrophy and some cases of Fuchs endothelial corneal dystrophy. To probe SLC4A11's roles, we compared gene expression in RNA from corneas of 17-week-old slc4a11-/- (n = 3) and slc4a11+/+ mice (n = 3) and subjected to RNA sequencing. mRNA levels for a subset of genes were also assessed by quantitative real-time reverse transcription PCR (qRT RT-PCR). Cornea expressed 13,173 genes, which were rank-ordered for their abundance. In slc4a11-/- corneas, 100 genes had significantly altered expression. Abundant slc14a1 expression, encoding the urea transporter UT-A, suggests a significant role in the cornea. The set of genes with altered expression was subjected to Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses, revealing that alterations clustered into extracellular region, cytoskeleton, cell adhesion and plasma membrane functions. Gene expression changes further clustered into classes (with decreasing numbers of genes): cell fate and development, extracellular matrix and cell adhesion, cytoskeleton, ion homeostasis and energy metabolism. Together these gene changes confirm earlier suggestions of a role of SLC4A11 in ion homeostasis, energy metabolism, cell adhesion, and reveal an unrecognized SLC4A11 role in cytoskeletal organization.


Subject(s)
Anion Transport Proteins/genetics , Cornea/physiology , Gene Expression/genetics , Symporters/genetics , Animals , Cell Adhesion/genetics , Cell Membrane/genetics , Endothelial Cells/physiology , Endothelium, Corneal/physiology , Epithelial Cells/physiology , Extracellular Matrix/genetics , Gene Expression Regulation/genetics , Ion Transport/genetics , Male , Mice , Mutation/genetics
6.
J Clin Invest ; 116(11): 3042-9, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17053834

ABSTRACT

Cylindromatosis (CYLD) is a deubiquitinating enzyme that is altered in patients with familial cylindromatosis, a condition characterized by numerous benign adnexal tumors. However, the regulatory function of CYLD remains unsettled. Here we show that the development of B cells, T cells, and myeloid cells was unaffected in CYLD-deficient mice, but that the activation of these cells with mediators of innate and adaptive immunity resulted in enhanced NF-kappaB and JNK activity associated with increased TNF receptor-associated factor 2 (TRAF2) and NF-kappaB essential modulator (NEMO) ubiquitination. CYLD-deficient mice were more susceptible to induced colonic inflammation and showed a dramatic increase in the incidence of tumors compared with controls in a colitis-associated cancer model. These results suggest that CYLD limits inflammation and tumorigenesis by regulating ubiquitination in vivo.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Colitis/metabolism , Colitis/pathology , Cysteine Endopeptidases/metabolism , Genetic Predisposition to Disease/genetics , NF-kappa B/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Colitis/complications , Colitis/genetics , Colonic Neoplasms/etiology , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cysteine Endopeptidases/deficiency , Cysteine Endopeptidases/genetics , Cytokines/biosynthesis , Deubiquitinating Enzyme CYLD , Enzyme Activation , Intracellular Signaling Peptides and Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Macrophages/metabolism , Mice , Mice, Knockout , Phenotype , Protein Binding , TNF Receptor-Associated Factor 2/metabolism , Ubiquitin/metabolism
7.
J Clin Invest ; 114(11): 1593-602, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15578091

ABSTRACT

Hypomorphic mutations in the zinc finger domain of NF-kappaB essential modulator (NEMO) cause X-linked hyper-IgM syndrome with ectodermal dysplasia (XHM-ED). Here we report that patient B cells are characterized by an absence of Ig somatic hypermutation (SHM) and defective class switch recombination (CSR) despite normal induction of activation-induced cytidine deaminase (AID) and Iepsilon-Cepsilon transcripts. This indicates that AID expression alone is insufficient to support neutralizing antibody responses. Furthermore, we show that patient B cells stimulated with CD40 ligand are impaired in both p65 and c-Rel activation, and whereas addition of IL-4 can enhance p65 activity, c-Rel activity remains deficient. This suggests that these NF-kappaB components have different activation requirements and that IL-4 can augment some but not all NEMO-dependent NF-kappaB signaling. Finally, using microarray analysis of patient B cells we identified downstream effects of impaired NF-kappaB activation and candidate factors that may be necessary for CSR and SHM in B cells.


Subject(s)
B-Lymphocytes/physiology , CD40 Antigens/metabolism , Carrier Proteins/genetics , Carrier Proteins/immunology , Cell Differentiation/physiology , Mutation , Proto-Oncogene Proteins c-rel/immunology , Adolescent , Adult , B-Lymphocytes/cytology , Child, Preschool , Cytidine Deaminase , Cytosine Deaminase/genetics , Cytosine Deaminase/metabolism , Ectodermal Dysplasia/genetics , Ectodermal Dysplasia/immunology , Gene Expression Regulation , Humans , Hypergammaglobulinemia/genetics , Hypergammaglobulinemia/immunology , I-kappa B Kinase , Immunoglobulin Class Switching/genetics , Immunoglobulins/blood , Interleukin-4/metabolism , Molecular Sequence Data , NF-kappa B/immunology , Proto-Oncogene Proteins c-rel/genetics , Somatic Hypermutation, Immunoglobulin/genetics , Syndrome
8.
Sci Transl Med ; 9(394)2017 06 14.
Article in English | MEDLINE | ID: mdl-28615356

ABSTRACT

A potentially useful approach for drug discovery is to connect gene expression profiles of disease-affected tissues ("disease signatures") to drug signatures, but it remains to be shown whether it can be used to identify clinically relevant treatment options. We analyzed coexpression networks and genetic data to identify a disease signature for type 2 diabetes in liver tissue. By interrogating a library of 3800 drug signatures, we identified sulforaphane as a compound that may reverse the disease signature. Sulforaphane suppressed glucose production from hepatic cells by nuclear translocation of nuclear factor erythroid 2-related factor 2 (NRF2) and decreased expression of key enzymes in gluconeogenesis. Moreover, sulforaphane reversed the disease signature in the livers from diabetic animals and attenuated exaggerated glucose production and glucose intolerance by a magnitude similar to that of metformin. Finally, sulforaphane, provided as concentrated broccoli sprout extract, reduced fasting blood glucose and glycated hemoglobin (HbA1c) in obese patients with dysregulated type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Isothiocyanates/therapeutic use , Liver/drug effects , Liver/metabolism , Animals , Blood Glucose/drug effects , Cell Line , Female , Glycated Hemoglobin/metabolism , Humans , Hypoglycemic Agents/therapeutic use , Male , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/metabolism , Obesity/drug therapy , Obesity/metabolism , Sulfoxides
9.
Nat Neurosci ; 19(11): 1442-1453, 2016 11.
Article in English | MEDLINE | ID: mdl-27668389

ABSTRACT

Over 100 genetic loci harbor schizophrenia-associated variants, yet how these variants confer liability is uncertain. The CommonMind Consortium sequenced RNA from dorsolateral prefrontal cortex of people with schizophrenia (N = 258) and control subjects (N = 279), creating a resource of gene expression and its genetic regulation. Using this resource, ∼20% of schizophrenia loci have variants that could contribute to altered gene expression and liability. In five loci, only a single gene was involved: FURIN, TSNARE1, CNTN4, CLCN3 or SNAP91. Altering expression of FURIN, TSNARE1 or CNTN4 changed neurodevelopment in zebrafish; knockdown of FURIN in human neural progenitor cells yielded abnormal migration. Of 693 genes showing significant case-versus-control differential expression, their fold changes were ≤ 1.33, and an independent cohort yielded similar results. Gene co-expression implicates a network relevant for schizophrenia. Our findings show that schizophrenia is polygenic and highlight the utility of this resource for mechanistic interpretations of genetic liability for brain diseases.


Subject(s)
Gene Expression Regulation/genetics , Genetic Predisposition to Disease , Multifactorial Inheritance/genetics , Schizophrenia/genetics , Brain/metabolism , Female , Genome-Wide Association Study , Humans , Male , Polymorphism, Single Nucleotide , Risk
10.
PLoS One ; 5(12): e14319, 2010 Dec 14.
Article in English | MEDLINE | ID: mdl-21179467

ABSTRACT

To identify the genes and pathways that underlie cardiovascular and metabolic phenotypes we performed an integrated analysis of a mouse C57BL/6JxA/J F2 (B6AF2) cross by relating genome-wide gene expression data from adipose, kidney, and liver tissues to physiological endpoints measured in the population. We have identified a large number of trait QTLs including loci driving variation in cardiac function on chromosomes 2 and 6 and a hotspot for adiposity, energy metabolism, and glucose traits on chromosome 8. Integration of adipose gene expression data identified a core set of genes that drive the chromosome 8 adiposity QTL. This chromosome 8 trans eQTL signature contains genes associated with mitochondrial function and oxidative phosphorylation and maps to a subnetwork with conserved function in humans that was previously implicated in human obesity. In addition, human eSNPs corresponding to orthologous genes from the signature show enrichment for association to type II diabetes in the DIAGRAM cohort, supporting the idea that the chromosome 8 locus perturbs a molecular network that in humans senses variations in DNA and in turn affects metabolic disease risk. We functionally validate predictions from this approach by demonstrating metabolic phenotypes in knockout mice for three genes from the trans eQTL signature, Akr1b8, Emr1, and Rgs2. In addition we show that the transcriptional signatures for knockout of two of these genes, Akr1b8 and Rgs2, map to the F2 network modules associated with the chromosome 8 trans eQTL signature and that these modules are in turn very significantly correlated with adiposity in the F2 population. Overall this study demonstrates how integrating gene expression data with QTL analysis in a network-based framework can aid in the elucidation of the molecular drivers of disease that can be translated from mice to humans.


Subject(s)
Cardiovascular Diseases/genetics , Cardiovascular System , Crosses, Genetic , Quantitative Trait Loci , Animals , Blood Pressure , Body Composition , Cholesterol/metabolism , Cohort Studies , Electrocardiography/methods , Female , Male , Mice , Mice, Inbred C57BL , Models, Genetic , Phenotype
11.
Pharmacogenomics ; 10(2): 203-12, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19207021

ABSTRACT

The pharmaceutical industry faces unprecedented pressures based largely on the inability to bring sufficient new medicines to market. The high failure rate of drug candidates in clinical development highlights a need for new approaches to the study of disease mechanisms and drug discovery. We advocate an integrated approach based on the study of the entire organism leveraging the power of detailed phenotyping, high-throughput genomic technologies and mathematical modeling. Key to this paradigm is the realization that the systematic genetic perturbations that exist in populations provide an ideal structure for uncovering the interactions that define molecular networks or states. By linking molecular states to physiological states and in turn understanding how molecular states drive disease processes, the promise of truly rational drug-design with a high probability of success in clinical development can be realized.


Subject(s)
Drug Therapy/methods , Genetic Diseases, Inborn/genetics , Genome, Human , Genomics/methods , Integrative Medicine/methods , Animals , Humans , Integrative Medicine/standards , Mammals/genetics , Models, Genetic , Models, Molecular , Molecular Biology/methods , Phenotype
12.
J Pharmacol Toxicol Methods ; 57(1): 42-51, 2008.
Article in English | MEDLINE | ID: mdl-17928241

ABSTRACT

INTRODUCTION: Coverslip hypoxia (CSH) is a recently described method for producing rapid and severe ischemia derived from the metabolic activity of synchronously contracting isolated neonatal rat ventricular myocytes (NRVMs). While the effect of acute ischemia produced by CSH is documented, the contribution of reperfusion to cell viability has not been fully studied. METHODS: We therefore used fluorescence microscopy and expression profiling by microarray to determine the morphological and genetic effects in NRVMs of both the ischemic and reperfusion events of CSH. RESULTS: Fluorescence microscopy studies in coverslipped NRVMs showed cell death at 1 h as previously reported. Matched samples coverslipped for up to 2 h and then reperfused 18 h showed myocyte recovery prior to but not beyond 1 h upon post-staining, suggesting a limited window of recovery. Expression profiling of more than 30,000 genes using total RNA collected from NRVMs subjected to varying periods of ischemia and reperfusion revealed 103 genes regulated at least 2-fold at p<10(-7). These genes fall into discrete functional groups including apoptosis, metabolism, and hypoxia/acidosis. The regulation of a subset of genes from these groups was confirmed by RT-PCR. Interestingly, the hypoxia/acidosis gene BNip3 (a Bcl-2 family member implicated in hypoxia/acidosis-associated cell death) was upregulated early during ischemia and persisted throughout reperfusion. In addition, other hypoxia/acidosis genes such as heme oxygenase 1, pyruvate dehydrogenase kinase 1, prolyl hydroxylases, and hypoxia-inducible protein 2 were upregulated. DISCUSSION: These data suggest that the ischemic and reperfusion events created by CSH induce gene regulation within distinct functional groups related to in vivo ischemia.


Subject(s)
Gene Expression Profiling , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Animals , Animals, Newborn , Cell Death , Cell Hypoxia/physiology , Cell Survival , Cells, Cultured , Cluster Analysis , Gene Expression Regulation , Heart Ventricles/cytology , Microscopy, Fluorescence , Myocardial Ischemia , Protein Array Analysis , Rats , Rats, Sprague-Dawley , Reperfusion Injury
15.
Blood ; 108(7): 2324-31, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-16794254

ABSTRACT

Ectodermal dysplasia with immune deficiency (EDI) is caused by alterations in NEMO (nuclear factor [NF]-kappaB essential modulator). Most genetic mutations are located in exon 10 and affect the C-terminal zinc finger domain. However, the biochemical mechanism by which they cause immune dysfunction remains undetermined. In this report, we investigated the effect of a cysteine-to-arginine mutation (C417R) found in the NEMO zinc finger domain on dendritic cell (DC) function. Following CD40 stimulation of DCs prepared from 2 unrelated patients with the NEMO C417R mutation, we found NEMO ubiquitination was absent, and this was associated with preserved RelA but absent c-Rel activity. As a consequence, CD40 stimulated EDI DCs failed to synthesize the c-Rel-dependent cytokine interleukin-12, had impaired up-regulation of costimulatory molecules, and failed to support allogeneic lymphocyte proliferation in vitro. In contrast, EDI DCs stimulated with the TLR4 ligand lipopolysaccharide (LPS) showed normal downstream NF-kappaB activity, DC maturation, and NEMO ubiquitination. These findings show for the first time how mutations in the zinc finger domain of NEMO can lead to pathway specific defects in NEMO ubiquitination and thus immune deficiency.


Subject(s)
Dendritic Cells/cytology , Ectodermal Dysplasia/pathology , I-kappa B Kinase/chemistry , I-kappa B Kinase/genetics , Immunologic Deficiency Syndromes/immunology , Ubiquitin/chemistry , Arginine/chemistry , CD3 Complex/biosynthesis , CD40 Antigens/biosynthesis , Cell Proliferation , Cysteine/chemistry , Dendritic Cells/metabolism , Ectodermal Dysplasia/immunology , Exons , Humans , Immunologic Deficiency Syndromes/blood , Mutation , Protein Structure, Tertiary , Zinc Fingers
SELECTION OF CITATIONS
SEARCH DETAIL