Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Nucleic Acids Res ; 51(21): 11453-11465, 2023 Nov 27.
Article in English | MEDLINE | ID: mdl-37823611

ABSTRACT

SINE-VNTR-Alu (SVA) retrotransposons are evolutionarily young and still-active transposable elements (TEs) in the human genome. Several pathogenic SVA insertions have been identified that directly mutate host genes to cause neurodegenerative and other types of diseases. However, due to their sequence heterogeneity and complex structures as well as limitations in sequencing techniques and analysis, SVA insertions have been less well studied compared to other mobile element insertions. Here, we identified polymorphic SVA insertions from 3646 whole-genome sequencing (WGS) samples of >150 diverse populations and constructed a polymorphic SVA insertion reference catalog. Using 20 long-read samples, we also assembled reference and polymorphic SVA sequences and characterized the internal hexamer/variable-number-tandem-repeat (VNTR) expansions as well as differing SVA activity for SVA subfamilies and human populations. In addition, we developed a module to annotate both reference and polymorphic SVA copies. By characterizing the landscape of both reference and polymorphic SVA retrotransposons, our study enables more accurate genotyping of these elements and facilitate the discovery of pathogenic SVA insertions.


Subject(s)
Genome, Human , Retroelements , Humans , Alu Elements , Genome, Human/genetics , Minisatellite Repeats/genetics , Retroelements/genetics , Short Interspersed Nucleotide Elements
2.
FASEB J ; 34(7): 9051-9073, 2020 07.
Article in English | MEDLINE | ID: mdl-32413188

ABSTRACT

Laminopathies are rare diseases associated with mutations in LMNA, which encodes nuclear lamin A/C. LMNA variants lead to diverse tissue-specific phenotypes including cardiomyopathy, lipodystrophy, myopathy, neuropathy, progeria, bone/skin disorders, and overlap syndromes. The mechanisms underlying these heterogeneous phenotypes remain poorly understood, although post-translational modifications, including phosphorylation, are postulated as regulators of lamin function. We catalogued all known lamin A/C human mutations and their associated phenotypes, and systematically examined the putative role of phosphorylation in laminopathies. In silico prediction of specific LMNA mutant-driven changes to lamin A phosphorylation and protein structure was performed using machine learning methods. Some of the predictions we generated were validated via assessment of ectopically expressed wild-type and mutant LMNA. Our findings indicate phenotype- and mutant-specific alterations in lamin phosphorylation, and that some changes in phosphorylation may occur independently of predicted changes in lamin protein structure. Therefore, therapeutic targeting of phosphorylation in the context of laminopathies will likely require mutant- and kinase-specific approaches.


Subject(s)
Genetic Association Studies , Genotype , Lamin Type A/genetics , Laminopathies/pathology , Mutation , Phenotype , Female , Humans , Lamin Type A/metabolism , Laminopathies/classification , Laminopathies/genetics , Laminopathies/metabolism , Male , Phosphorylation
3.
PLoS Genet ; 13(8): e1006938, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28787442

ABSTRACT

Advances in genomics in recent years have provided key insights into defining cancer subtypes "within-a-tissue"-that is, respecting traditional anatomically driven divisions of medicine. However, there remains a dearth of data regarding molecular profiles that are shared across tissues, an understanding of which could lead to the development of highly versatile, broadly applicable therapies. Using data acquired from The Cancer Genome Atlas (TCGA), we performed a transcriptomics-centered analysis on 1494 patient samples, comparing the two major histological subtypes of solid tumors (adenocarcinomas and squamous cell carcinomas) across organs, with a focus on tissues in which both subtypes arise: esophagus, lung, and uterine cervix. Via principal component and hierarchical clustering analysis, we discovered that histology-driven differences accounted for a greater degree of inherent molecular variation in the tumors than did tissue of origin. We then analyzed differential gene expression, DNA methylation, and non-coding RNA expression between adenocarcinomas and squamous cell carcinomas and found 1733 genes, 346 CpG sites, and 42 microRNAs in common between organ sites, indicating specific adenocarcinoma-associated and squamous cell carcinoma-associated molecular patterns that were conserved across tissues. We then identified specific pathways that may be critical to the development of adenocarcinomas and squamous cell carcinomas, including Liver X receptor activation, which was upregulated in adenocarcinomas but downregulated in squamous cell carcinomas, possibly indicating important differences in cancer cell metabolism between these two histological subtypes of cancer. In addition, we highlighted genes that may be common drivers of adenocarcinomas specifically, such as IGF2BP1, which suggests a possible link between embryonic development and tumor subtype. Altogether, we demonstrate the need to consider biological similarities that transcend anatomical boundaries to inform the development of novel therapeutic strategies. All data sets from our analysis are available as a resource for further investigation.


Subject(s)
Adenocarcinoma/genetics , Carcinoma, Squamous Cell/genetics , Gene Expression Regulation, Neoplastic , Transcriptome , Adenocarcinoma/diagnosis , Carcinoma, Squamous Cell/diagnosis , Cervix Uteri/pathology , DNA Methylation , Down-Regulation , Epigenomics , Esophagus/pathology , Female , Genetic Markers , Genetic Variation , Humans , Liver X Receptors/genetics , Liver X Receptors/metabolism , Lung/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , Principal Component Analysis , Prognosis , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Reproducibility of Results , Up-Regulation
4.
Proc Natl Acad Sci U S A ; 109 Suppl 2: 17213-20, 2012 Oct 16.
Article in English | MEDLINE | ID: mdl-23045690

ABSTRACT

Preference behaviors are often established during early life, but the underlying neural circuit mechanisms remain unknown. Adapting a unique nesting behavior assay, we confirmed a "critical period" for developing music preference in C57BL/6 mice. Early music exposure between postnatal days 15 and 24 reversed their innate bias for silent shelter, which typically could not be altered in adulthood. Instead, exposing adult mice treated acutely with valproic acid or carrying a targeted deletion of the Nogo receptor (NgR(-/-)) unmasked a strong plasticity of preference consistent with a reopening of the critical period as seen in other systems. Imaging of cFos expression revealed a prominent neuronal activation in response to the exposed music in the prelimbic and infralimbic medial prefrontal cortex only under conditions of open plasticity. Neither behavioral changes nor selective medial prefrontal cortex activation was observed in response to pure tone exposure, indicating a music-specific effect. Open-field center crossings were increased concomitant with shifts in music preference, suggesting a potential anxiolytic effect. Thus, music may offer both a unique window into the emotional state of mice and a potentially efficient assay for molecular "brakes" on critical period plasticity common to sensory and higher order brain areas.


Subject(s)
Auditory Cortex/growth & development , Auditory Cortex/physiology , Behavior, Animal/physiology , Acoustic Stimulation , Animals , Anxiety/physiopathology , Anxiety/psychology , Auditory Cortex/drug effects , Behavior, Animal/drug effects , Female , GPI-Linked Proteins/deficiency , GPI-Linked Proteins/genetics , GPI-Linked Proteins/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Music/psychology , Myelin Proteins/deficiency , Myelin Proteins/genetics , Myelin Proteins/physiology , Neuronal Plasticity/physiology , Nogo Receptor 1 , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Receptors, Cell Surface/physiology , Valproic Acid/pharmacology
5.
Mol Cancer Res ; 21(7): 741-752, 2023 07 05.
Article in English | MEDLINE | ID: mdl-37027010

ABSTRACT

Cancer-associated fibroblasts (CAF) can promote tumor growth, metastasis, and therapeutic resistance in esophageal squamous cell carcinoma (ESCC), but the mechanisms of action remain elusive. Our objective was to identify secreted factor(s) that mediate the communication between CAFs and ESCC tumor cells with the aim of identifying potential druggable targets. Through unbiased cytokine arrays, we have identified CC motif chemokine ligand 5 (CCL5) as a secreted factor that is increased upon co-culture of ESCC cells and CAFs, which we replicated in esophageal adenocarcinoma (EAC) with CAFs. Loss of tumor-cell-derived CCL5 reduces ESCC cell proliferation in vitro and in vivo and we propose this is mediated, in part, by a reduction in ERK1/2 signaling. Loss of tumor-derived CCL5 reduces the percentage of CAFs recruited to xenograft tumors in vivo. CCL5 is a ligand for the CC motif receptor 5 (CCR5), for which a clinically approved inhibitor exists, namely Maraviroc. Maraviroc treatment reduced tumor volume, CAF recruitment, and ERK1/2 signaling in vivo, thus, mimicking the effects observed with genetic loss of CCL5. High CCL5 or CCR5 expression is associated with worse prognosis in low-grade esophageal carcinomas. IMPLICATIONS: These data highlight the role of CCL5 in tumorigenesis and the therapeutic potential of targeting the CCL5-CCR5 axis in ESCC.


Subject(s)
Cancer-Associated Fibroblasts , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Humans , Cancer-Associated Fibroblasts/metabolism , Cell Line, Tumor , Cell Proliferation , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Chemokine CCL5/pharmacology , Chemokines/metabolism , Chemokines/pharmacology , Chemokines/therapeutic use , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/drug therapy , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/metabolism , Fibroblasts/metabolism , Ligands , Maraviroc/metabolism , Maraviroc/pharmacology , Maraviroc/therapeutic use , Animals
6.
J Invest Dermatol ; 142(12): 3282-3293, 2022 12.
Article in English | MEDLINE | ID: mdl-35691363

ABSTRACT

Epidermolysis bullosa simplex (EBS) is a severe and potentially life-threatening disorder for which no adequate therapy exists. Most cases are caused by dominant sequence variations in keratin genes K5 or K14, leading to the formation of cytoplasmic keratin aggregates, profound keratinocyte fragility, and cytolysis. We hypothesized that pharmacological reduction of keratin aggregates, which compromise keratinocyte integrity, represents a viable strategy for the treatment of EBS. In this study, we show that the multikinase inhibitor PKC412, which is currently in clinical use for acute myeloid leukemia and advanced systemic mastocytosis, reduced keratin aggregation by 40% in patient-derived K14.R125C EBS-associated keratinocytes. Using a combination of epithelial shear stress assay and real-time impedance spectroscopy, we show that PKC412 restored intercellular adhesion. Molecularly, global phosphoproteomic analysis together with immunoblots using phosphoepitope-specific antibodies revealed that PKC412 treatment altered phosphorylated sites on keratins and desmoplakin. Thus, our data provide a proof of concept to repurpose existing drugs for the targeted treatment of EBS and showcase how one broad-range kinase inhibitor reduced keratin filament aggregation in patient-derived EBS keratinocytes and the fragility of EBS cell monolayers. Our study paves the way for a clinical trial using PKC412 for systemic or local application in patients with EBS.


Subject(s)
Epidermolysis Bullosa Simplex , Humans , Epidermolysis Bullosa Simplex/genetics , Epidermolysis Bullosa Simplex/metabolism , Keratins/metabolism , Staurosporine/metabolism , Cytoskeleton/metabolism , Cytoskeletal Proteins/genetics , Keratin-14/genetics , Keratin-14/metabolism , Keratin-5/genetics , Keratin-5/metabolism , Mutation
7.
Sci Rep ; 11(1): 21304, 2021 10 29.
Article in English | MEDLINE | ID: mdl-34716381

ABSTRACT

3D patient-derived organoids (PDOs) have been utilized to evaluate potential therapies for patients with different cancers. However, the use of PDOs created from treatment-naive patient biopsies for prediction of clinical outcomes in patients with esophageal cancer has not yet been reported. Herein we describe a pilot prospective observational study with the goal of determining whether esophageal cancer PDOs created from treatment naive patients can model or predict clinical outcomes. Endoscopic biopsies of treatment-naive patients at a single tertiary care center were used to generate esophageal cancer PDOs, which were treated with standard-of-care chemotherapy, gamma-irradiation, and newer non-standard approaches, such as proton beam therapy or two small molecule inhibitors. Clinical outcomes of patients following neoadjuvant treatment were compared to their in vitro PDO responses, demonstrating the PDO's ability to mirror clinical response, suggesting the value of PDOs in prediction of clinical response to new therapeutic approaches. Future prospective clinical trials should test the use of pre-treatment PDOs to identify specific, targeted therapies for individual patients with esophageal adenocarcinoma.


Subject(s)
Adenocarcinoma/therapy , Antineoplastic Agents/pharmacology , Chemoradiotherapy/methods , Esophageal Neoplasms/therapy , Neoadjuvant Therapy , Organoids/drug effects , Aged , Drug Resistance, Neoplasm/drug effects , Humans , Male , Middle Aged , Pilot Projects , Precision Medicine , Prospective Studies
8.
Prostate ; 68(15): 1615-23, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18663729

ABSTRACT

Molecular targeting of apoptotic signaling pathways has been extensively studied in recent years and directed towards the development of effective therapeutic modalities for treating advanced androgen-independent prostate tumors. The majority of therapeutic agents act through intrinsic or mitochondrial pathways to induce programmed cell death. The induction of apoptosis through endoplasmic reticulum (ER) stress pathways may provide an alternative to treat patients. The functional interaction between the BCL-2 family members and regulation of calcium homeostasis in the ER provides a critical link to the life or death outcome of the cell. Apoptosis induction mediated by ER stress-inducing agents is just beginning to be exploited for therapeutic targeting of prostate tumors. Insightful dissection of recently discovered apoptotic signaling pathways that function through the endoplasmic reticulum may identify novel molecules that could effectively target both androgen-dependent and androgen-independent prostate tumors. In this review, we focus on linking ER stress-induced apoptosis to therapeutic targeting of prostate tumors and dissect its cross-talk with the intrinsic and extrinsic apoptotic pathways.


Subject(s)
Apoptosis , Endoplasmic Reticulum/metabolism , Prostatic Neoplasms/drug therapy , Signal Transduction , Stress, Physiological/physiopathology , Calcium/metabolism , Homeostasis , Humans , Male , Proto-Oncogene Proteins c-bcl-2/metabolism
9.
JCI Insight ; 3(6)2018 03 22.
Article in English | MEDLINE | ID: mdl-29563330

ABSTRACT

BACKGROUND: Myeloid-derived suppressor cells (MDSCs) are a population of immature immune cells with several protumorigenic functions. CD38 is a transmembrane receptor-ectoenzyme expressed by MDSCs in murine models of esophageal cancer. We hypothesized that CD38 could be expressed on MDSCs in human colorectal cancer (CRC), which might allow for a new perspective on therapeutic targeting of human MDSCs with anti-CD38 monoclonal antibodies in this cancer. METHODS: Blood samples were collected from 41 CRC patients and 8 healthy donors, followed by peripheral blood mononuclear cell (PBMC) separation. Polymorphonuclear (PMN-) and monocytic (M-) MDSCs and CD38 expression levels were quantified by flow cytometry. The immunosuppressive capacity of M-MDSCs from 10 CRC patients was validated in a mixed lymphocyte reaction (MLR) assay. RESULTS: A significant expansion of CD38+ M-MDSCs and a trend of expansion of CD38+ PMN-MDSCs (accompanied by a trend of increased CD38 expression on both M- and PMN-MDSCs) were observed in PBMCs of CRC patients when compared with healthy donors. The CD38+ M-MDSCs from CRC patients were found to be immunosuppressive when compared with mature monocytes. CD38+ M- and PMN-MDSC frequencies were significantly higher in CRC patients who previously received treatment when compared with treatment-naive patients. CONCLUSIONS: This study provides a rationale for an attempt to target M-MDSCs with an anti-CD38 monoclonal antibody in metastatic CRC patients. FUNDING: NCI P01-CA14305603, the American Cancer Society, Scott and Suzi Lustgarten Family Colon Cancer Research Fund, Hansen Foundation, and Janssen Research and Development.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , Colorectal Neoplasms/metabolism , Esophageal Neoplasms/metabolism , Leukocytes, Mononuclear/metabolism , Membrane Glycoproteins/metabolism , Myeloid-Derived Suppressor Cells/metabolism , Neutrophils/metabolism , ADP-ribosyl Cyclase 1/immunology , Adult , Aged , Animals , Antibodies, Monoclonal , Colorectal Neoplasms/immunology , Esophageal Neoplasms/immunology , Female , Humans , Immunosuppressive Agents/pharmacology , Lymphocytes , Male , Membrane Glycoproteins/immunology , Mice , Middle Aged , Monocytes , Pennsylvania
10.
Cancer Res ; 78(17): 4957-4970, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29976575

ABSTRACT

The tumor microenvironment (TME) plays a major role in the pathogenesis of multiple cancer types, including upper-gastrointestinal (GI) cancers that currently lack effective therapeutic options. Cancer-associated fibroblasts (CAF) are an essential component of the TME, contributing to tumorigenesis by secreting growth factors, modifying the extracellular matrix, supporting angiogenesis, and suppressing antitumor immune responses. Through an unbiased approach, we have established that IL-6 mediates cross-talk between tumor cells and CAF not only by supporting tumor cell growth, but also by promoting fibroblast activation. As a result, IL-6 receptor (IL6Rα) and downstream effectors offer opportunities for targeted therapy in upper-GI cancers. IL-6 loss suppressed tumorigenesis in physiologically relevant three-dimensional (3D) organotypic and 3D tumoroid models and murine models of esophageal cancer. Tocilizumab, an anti-IL6Rα antibody, suppressed tumor growth in vivo in part via inhibition of STAT3 and MEK/ERK signaling. Analysis of a pan-cancer TCGA dataset revealed an inverse correlation between IL-6 and IL6Rα overexpression and patient survival. Therefore, we expanded evaluation of tocilizumab to head and neck squamous cell carcinoma patient-derived xenografts and gastric adenocarcinoma xenografts, demonstrating suppression of tumor growth and altered STAT3 and ERK1/2 gene signatures. We used small-molecule inhibitors of STAT3 and MEK1/2 signaling to suppress tumorigenesis in the 3D organotypic model of esophageal cancer. We demonstrate that IL6 is a major contributor to the dynamic cross-talk between tumor cells and CAF in the TME. Our findings provide a translational rationale for inhibition of IL6Rα and downstream signaling pathways as a novel targeted therapy in oral-upper-GI cancers.Significance: These findings demonstrate the interaction of esophageal cancer and cancer-associated fibroblasts through IL-6 signaling, providing rationale for a novel therapeutic approach to target these cancers. Cancer Res; 78(17); 4957-70. ©2018 AACR.


Subject(s)
Esophageal Neoplasms/genetics , Gastrointestinal Neoplasms/genetics , Interleukin-6/genetics , Receptors, Interleukin-6/genetics , Animals , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Carcinogenesis/genetics , Cell Line, Tumor , Esophageal Neoplasms/pathology , Gastrointestinal Neoplasms/pathology , Humans , MAP Kinase Signaling System/genetics , Mice , STAT3 Transcription Factor/genetics , Signal Transduction , Tumor Microenvironment/genetics , Xenograft Model Antitumor Assays
11.
Virology ; 417(2): 268-80, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21784501

ABSTRACT

The human cytomegalovirus DNA polymerase subunit UL44 is a phosphoprotein, but its sites and roles of phosphorylation have not been investigated. We compared sites of phosphorylation of UL44 in vitro by the viral protein kinase UL97 and cyclin-dependent kinase 1 with those in infected cells. Transient treatment of infected cells with a UL97 inhibitor greatly reduced labeling of two minor UL44 phosphopeptides. Viruses containing alanine substitutions of most UL44 residues that are phosphorylated in infected cells exhibited at most modest effects on viral DNA synthesis and yield. However, substitution of highly phosphorylated sites adjacent to the nuclear localization signal abolished viral replication. The results taken together are consistent with UL44 being phosphorylated directly by UL97 during infection, and a crucial role for phosphorylation-mediated nuclear localization of UL44 for viral replication, but lend little support to the widely held hypothesis that UL97-mediated phosphorylation of UL44 is crucial for viral DNA synthesis.


Subject(s)
Cell Nucleus/metabolism , Cytomegalovirus/physiology , DNA-Binding Proteins/metabolism , Host-Pathogen Interactions , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Viral Proteins/metabolism , Virus Replication , Active Transport, Cell Nucleus , Amino Acid Substitution/genetics , Humans , Phosphorylation
SELECTION OF CITATIONS
SEARCH DETAIL