Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
Add more filters

Publication year range
1.
Genes Dev ; 33(3-4): 209-220, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30692207

ABSTRACT

Spatiotemporal control of Wnt signaling is essential for the development and homeostasis of many tissues. The transmembrane E3 ubiquitin ligases ZNRF3 (zinc and ring finger 3) and RNF43 (ring finger protein 43) antagonize Wnt signaling by promoting degradation of frizzled receptors. ZNRF3 and RNF43 are frequently inactivated in human cancer, but the molecular and therapeutic implications remain unclear. Here, we demonstrate that adrenocortical-specific loss of ZNRF3, but not RNF43, results in adrenal hyperplasia that depends on Porcupine-mediated Wnt ligand secretion. Furthermore, we discovered a Wnt/ß-catenin signaling gradient in the adrenal cortex that is disrupted upon loss of ZNRF3. Unlike ß-catenin gain-of-function models, which induce high Wnt/ß-catenin activation and expansion of the peripheral cortex, ZNRF3 loss triggers activation of moderate-level Wnt/ß-catenin signaling that drives proliferative expansion of only the histologically and functionally distinct inner cortex. Genetically reducing ß-catenin dosage significantly reverses the ZNRF3-deficient phenotype. Thus, homeostatic maintenance of the adrenal cortex is dependent on varying levels of Wnt/ß-catenin activation, which is regulated by ZNRF3.


Subject(s)
Adrenal Cortex/metabolism , Homeostasis/genetics , Ubiquitin-Protein Ligases/metabolism , Wnt Signaling Pathway/physiology , beta Catenin/metabolism , Adrenal Cortex/cytology , Adrenal Cortex/growth & development , Adrenal Cortex Diseases/physiopathology , Animals , Cell Proliferation/genetics , Female , Gene Knockout Techniques , Male , Mice , Models, Animal , Transcriptional Activation/genetics , Ubiquitin-Protein Ligases/genetics
2.
Gastroenterology ; 166(1): 103-116.e9, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37716376

ABSTRACT

BACKGROUND & AIMS: CXADR-like membrane protein (CLMP) is structurally related to coxsackie and adenovirus receptor. Pathogenic variants in CLMP gene have been associated with congenital short bowel syndrome, implying a role for CLMP in intestinal development. However, the contribution of CLMP to regulating gut development and homeostasis is unknown. METHODS: In this study, we investigated CLMP function in the colonic epithelium using complementary in vivo and in vitro approaches, including mice with inducible intestinal epithelial cell (IEC)-specific deletion of CLMP (ClmpΔIEC), intestinal organoids, IECs with overexpression, or loss of CLMP and RNA sequencing data from individuals with colorectal cancer. RESULTS: Loss of CLMP enhanced IEC proliferation and, conversely, CLMP overexpression reduced proliferation. Xenograft experiments revealed increased tumor growth in mice implanted with CLMP-deficient colonic tumor cells, and poor engraftment was observed with CLMP-overexpressing cells. ClmpΔIEC mice showed exacerbated tumor burden in an azoxymethane and dextran sulfate sodium-induced colonic tumorigenesis model, and CLMP expression was reduced in human colorectal cancer samples. Mechanistic studies revealed that CLMP-dependent regulation of IEC proliferation is linked to signaling through mTOR-Akt-ß-catenin pathways. CONCLUSIONS: These results reveal novel insights into CLMP function in the colonic epithelium, highlighting an important role in regulating IEC proliferation, suggesting tumor suppressive function in colon cancer.


Subject(s)
Colitis , Colonic Neoplasms , Animals , Humans , Mice , Cell Proliferation , Colitis/chemically induced , Colitis/metabolism , Colonic Neoplasms/pathology , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Epithelial Cells/pathology , Intestinal Mucosa/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism
3.
J Biol Chem ; 299(5): 104656, 2023 05.
Article in English | MEDLINE | ID: mdl-36990216

ABSTRACT

Proliferating cell nuclear antigen (PCNA) is a sliding clamp protein that coordinates DNA replication with various DNA maintenance events that are critical for human health. Recently, a hypomorphic homozygous serine to isoleucine (S228I) substitution in PCNA was described to underlie a rare DNA repair disorder known as PCNA-associated DNA repair disorder (PARD). PARD symptoms range from UV sensitivity, neurodegeneration, telangiectasia, and premature aging. We, and others, previously showed that the S228I variant changes the protein-binding pocket of PCNA to a conformation that impairs interactions with specific partners. Here, we report a second PCNA substitution (C148S) that also causes PARD. Unlike PCNA-S228I, PCNA-C148S has WT-like structure and affinity toward partners. In contrast, both disease-associated variants possess a thermostability defect. Furthermore, patient-derived cells homozygous for the C148S allele exhibit low levels of chromatin-bound PCNA and display temperature-dependent phenotypes. The stability defect of both PARD variants indicates that PCNA levels are likely an important driver of PARD disease. These results significantly advance our understanding of PARD and will likely stimulate additional work focused on clinical, diagnostic, and therapeutic aspects of this severe disease.


Subject(s)
Alleles , Ataxia Telangiectasia , DNA Repair , Proliferating Cell Nuclear Antigen , Temperature , Humans , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/metabolism , DNA Repair/genetics , DNA Replication , Proliferating Cell Nuclear Antigen/chemistry , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Protein Binding/genetics , Protein Stability , Chromatin/genetics , Chromatin/metabolism , Substrate Specificity
4.
Kidney Int ; 105(3): 593-607, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38143038

ABSTRACT

Collapsing glomerulopathy (CG) is most often associated with fast progression to kidney failure with an incidence apparently higher in Brazil than in other countries. However, the reason for this occurrence is unknown. To better understand this, we performed an integrated analysis of clinical, histological, therapeutic, causative genetic and genetic ancestry data in a highly genetically admixed cohort of 70 children and adult patients with idiopathic CG (ICG). The disease onset occurred at 23 (interquartile range: 17-31) years and approximately half of patients progressed to chronic kidney disease requiring kidney replacement therapy (CKD-KRT) 36 months after diagnosis. Causative genetic bases, assessed by targeted-gene panel or whole-exome sequencing, were identified in 58.6% of patients. Among these cases, 80.5% harbored APOL1 high-risk genotypes (HRG) and 19.5% causative Mendelian variants (MV). Self-reported non-White patients more frequently had HRG. MV was an independent risk factor for progression to CKD-KRT by 36 months and the end of follow-up, while remission was an independent protective factor. All patients with HRG manifested CG at 9-44 years of age, whereas in those with APOL1 low-risk genotype, the disease arose throughout life. HRGs were associated with higher proportion of African genetic ancestry. Novel causative MVs were identified in COL4A5, COQ2 and PLCE1 and previously described causative MVs were identified in MYH9, TRPC6, COQ2, COL4A3 and TTC21B. Three patients displayed HRG combined with a variant of uncertain significance (ITGB4, LAMA5 or PTPRO). MVs were associated with worse kidney prognosis. Thus, our data reveal that the genetic status plays a major role in ICG pathogenesis, accounting for more than half of cases in a highly admixed Brazilian population.


Subject(s)
Apolipoprotein L1 , Renal Insufficiency, Chronic , Adult , Child , Humans , Apolipoprotein L1/genetics , Genotype , Kidney/pathology , Renal Insufficiency, Chronic/epidemiology , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology , Risk Factors , Adolescent , Young Adult
5.
J Neuroinflammation ; 19(1): 111, 2022 May 14.
Article in English | MEDLINE | ID: mdl-35568856

ABSTRACT

BACKGROUND: Microglia are the tissue-resident macrophages of the CNS. They originate in the yolk sac, colonize the CNS during embryonic development and form a self-sustaining population with limited turnover. A consequence of their relative slow turnover is that microglia can serve as a long-term memory for inflammatory or neurodegenerative events. METHODS: Using ATAC-, ChIP- and RNA-sequencing, we characterized the epigenomes and transcriptomes of FACS-purified microglia from mice exposed to different stimuli. A repeated endotoxin challenge (LPS) was used to induce tolerance in microglia, while genotoxic stress (DNA repair deficiency-induced accelerated aging through Ercc1 deficiency) resulted in primed (hypersensitive) microglia. RESULTS: Whereas the enrichment of permissive epigenetic marks at enhancer regions could explain training (hyper-responsiveness) of primed microglia to an LPS challenge, the tolerized response of microglia seems to be regulated by loss of permissive epigenetic marks. We identify that inflammatory stimuli and accelerated aging as a result of genotoxic stress activate distinct gene networks. These gene networks and associated biological processes are partially overlapping, which is likely driven by specific transcription factor networks, resulting in altered epigenetic signatures and distinct functional (desensitized vs. primed) microglia phenotypes. CONCLUSION: This study provides insight into epigenetic profiles and transcription factor networks associated with transcriptional signatures of tolerized and trained microglia in vivo, leading to a better understanding of innate immune memory of microglia.


Subject(s)
Lipopolysaccharides , Microglia , Animals , Epigenesis, Genetic , Immunity, Innate , Immunologic Memory , Lipopolysaccharides/pharmacology , Mice , Transcription Factors
6.
Int J Mol Sci ; 23(9)2022 Apr 19.
Article in English | MEDLINE | ID: mdl-35562862

ABSTRACT

Astrocytoma is the most common and aggressive tumor of the central nervous system. Genetic and environmental factors, bacterial infection, and several other factors are known to be involved in gliomagenesis, although the complete underlying molecular mechanism is not fully understood. Tumorigenesis is a multistep process involving initiation, promotion, and progression. We present a human model of malignant astrocyte transformation established by subjecting primary astrocytes from healthy adults to four sequential cycles of forced anchorage impediment (deadhesion). After limiting dilution of the surviving cells obtained after the fourth deadhesion/readhesion cycle, three clones were randomly selected, and exhibited malignant characteristics, including increased proliferation rate and capacity for colony formation, migration, and anchorage-independent growth in soft agar. Functional assay results for these clonal cells, including response to temozolomide, were comparable to U87MG-a human glioblastoma-derived cell lineage-reinforcing malignant cell transformation. RNA-Seq analysis by next-generation sequencing of the transformed clones relative to the primary astrocytes revealed upregulation of genes involved in the PI3K/AKT and Wnt/ß-catenin signaling pathways, in addition to upregulation of genes related to epithelial-mesenchymal transition, and downregulation of genes related to aerobic respiration. These findings, at a molecular level, corroborate the change in cell behavior towards mesenchymal-like cell dedifferentiation. This linear progressive model of malignant human astrocyte transformation is unique in that neither genetic manipulation nor treatment with carcinogens are used, representing a promising tool for testing combined therapeutic strategies for glioblastoma patients, and furthering knowledge of astrocytoma transformation and progression.


Subject(s)
Astrocytes , Glioblastoma , Astrocytes/metabolism , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Epithelial-Mesenchymal Transition , Glioblastoma/pathology , Humans , Phosphatidylinositol 3-Kinases/metabolism
7.
J Proteome Res ; 20(10): 4693-4707, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34533964

ABSTRACT

Medulloblastomas (MBs) and glioblastomas (GBMs) are high-incidence central nervous system tumors. Different origin sites and changes in the tissue microenvironment have been associated with the onset and progression. Here, we describe differences between the extracellular matrix (ECM) signatures of these tumors. We compared the proteomic profiles of MB and GBM decellularized tumor samples between each other and their normal decellularized brain site counterparts. Our analysis revealed that 19, 28, and 11 ECM proteins were differentially expressed in MBs, GBMs, and in both MBs and GBMs, respectively. Next, we validated key findings by using a protein tissue array with 53 MB and 55 GBM cases and evaluated the clinical relevance of the identified differentially expressed proteins through their analysis on publicly available datasets, 763 MB samples from the GSE50161 and GSE85217 studies, and 115 GBM samples from RNAseq-TCGA. We report a shift toward a denser fibrillary ECM as well as a clear alteration in the glycoprotein signature, which influences the tumor pathophysiology. MS data have been submitted to the PRIDE repository, project accession: PXD023350.


Subject(s)
Brain Neoplasms , Extracellular Matrix , Glioblastoma , Medulloblastoma , Brain Neoplasms/genetics , Extracellular Matrix/pathology , Glioblastoma/genetics , Humans , Medulloblastoma/genetics , Proteome/genetics , Proteomics , Tumor Microenvironment
8.
Hum Mol Genet ; 28(10): 1620-1628, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30608580

ABSTRACT

Missense mutations in the gene, MAP3K1, are a common cause of 46,XY gonadal dysgenesis, accounting for 15-20% of cases [Ostrer, 2014, Disorders of sex development (DSDs): an update. J. Clin. Endocrinol. Metab., 99, 1503-1509]. Functional studies demonstrated that all of these mutations cause a protein gain-of-function that alters co-factor binding and increases phosphorylation of the downstream MAP kinase pathway targets, MAPK11, MAP3K and MAPK1. This dysregulation of the MAP kinase pathway results in increased CTNNB1, increased expression of WNT4 and FOXL2 and decreased expression of SRY and SOX9. Unique and recurrent pathogenic mutations cluster in three semi-contiguous domains outside the kinase region of the protein, a newly identified N-terminal domain that shares homology with the Guanine Exchange Factor (residues Met164 to Glu231), a Plant HomeoDomain (residues Met442 to Trp495) and an ARMadillo repeat domain (residues Met566 to Glu862). Despite the presence of the mutation clusters and clinical data, there exists a dearth of mechanistic insights behind the development imbalance. In this paper, we use structural modeling and functional data of these mutations to understand alterations of the MAP3K1 protein and the effects on protein folding, binding and downstream target phosphorylation. We show that these mutations have differential effects on protein binding depending on the domains in which they occur. These mutations increase the binding of the RHOA, MAP3K4 and FRAT1 proteins and generally decrease the binding of RAC1. Thus, pathologies in MAP3K1 disrupt the balance between the pro-kinase activities of the RHOA and MAP3K4 binding partners and the inhibitory activity of RAC1.


Subject(s)
Disorders of Sex Development/genetics , MAP Kinase Kinase Kinase 1/genetics , MAP Kinase Kinase Kinase 4/genetics , rac1 GTP-Binding Protein/genetics , Adaptor Proteins, Signal Transducing/genetics , Armadillo Domain Proteins/genetics , Disorder of Sex Development, 46,XY , Disorders of Sex Development/pathology , Female , Forkhead Box Protein L2/genetics , Gene Expression Regulation/genetics , Gonadal Dysgenesis, 46,XY/genetics , Gonadal Dysgenesis, 46,XY/pathology , Humans , MAP Kinase Kinase Kinase 1/chemistry , MAP Kinase Kinase Kinase 4/chemistry , MAP Kinase Signaling System/genetics , Male , Mutation, Missense/genetics , Protein Binding/genetics , Proto-Oncogene Proteins/genetics , Sex-Determining Region Y Protein/genetics , rac1 GTP-Binding Protein/chemistry , rhoA GTP-Binding Protein/chemistry , rhoA GTP-Binding Protein/genetics
9.
Int J Mol Sci ; 22(15)2021 Jul 28.
Article in English | MEDLINE | ID: mdl-34360836

ABSTRACT

Lysyl oxidase-like 3 (LOXL3), belonging to the lysyl oxidase family, is responsible for the crosslinking in collagen or elastin. The cellular localization of LOXL3 is in the extracellular space by reason of its canonical function. In tumors, the presence of LOXL3 has been associated with genomic stability, cell proliferation, and metastasis. In silico analysis has shown that glioblastoma was among tumors with the highest LOXL3 expression levels. LOXL3 silencing of U87MG cells by siRNA led to the spreading of the tumor cell surface, and the transcriptome analysis of these cells revealed an upregulation of genes coding for extracellular matrix, cell adhesion, and cytoskeleton components, convergent to an increase in cell adhesion and a decrease in cell invasion observed in functional assays. Significant correlations of LOXL3 expression with genes coding for tubulins were observed in the mesenchymal subtype in the TCGA RNA-seq dataset of glioblastoma (GBM). Conversely, genes involved in endocytosis and lysosome formation, along with MAPK-binding proteins related to focal adhesion turnover, were downregulated, which may corroborate the observed decrease in cell viability and increase in the rate of cell death. Invasiveness is a major determinant of the recurrence and poor outcome of GBM patients, and downregulation of LOXL3 may contribute to halting the tumor cell invasion.


Subject(s)
Amino Acid Oxidoreductases/metabolism , Cell Adhesion , Gene Expression Regulation, Neoplastic , Glioblastoma/enzymology , Amino Acid Oxidoreductases/genetics , Amino Acid Oxidoreductases/physiology , Cell Line, Tumor , Cell Proliferation , Computer Simulation , Cytoskeleton/metabolism , Endocytosis , Extracellular Matrix/metabolism , Gene Expression Profiling , Glioblastoma/genetics , Glioblastoma/pathology , Glioblastoma/physiopathology , Humans , Lysosomes/physiology , Neoplasm Invasiveness
10.
Horm Metab Res ; 52(8): 607-613, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32791542

ABSTRACT

Lack of routine fresh or frozen tissue is a barrier to widespread transcriptomic analysis of adrenal cortical tumors and an impediment to translational research in endocrinology and endocrine oncology. Our group has previously pioneered the use of targeted amplicon-based next-generation sequencing for archival formalin-fixed paraffin-embedded (FFPE) adrenal tissue specimens to characterize the spectrum of somatic mutations in various forms of primary aldosteronism. Herein, we developed and validated a novel 194-amplicon targeted next-generation RNA sequencing (RNAseq) assay for transcriptomic analysis of adrenal tumors using clinical-grade FFPE specimens. Targeted RNAseq-derived expression values for 27 adrenal cortical tumors, including aldosterone-producing adenomas (APA; n=8), cortisol-producing adenomas (CPA; n=11), and adrenal cortical carcinomas (ACC; n=8), highlighted known differentially-expressed genes (DEGs; i. e., CYP11B2, IGF2, etc.) and tumor type-specific transcriptional modules (i. e., high cell cycle/proliferation transcript expression in ACC, etc.), and a subset of DEGs was validated orthogonally using quantitative reverse transcription PCR (qRT-PCR). Finally, unsupervised hierarchical clustering using a subset of high-confidence DEGs revealed three discrete clusters representing APA, CPA, and ACC tumors with corresponding unique gene expression signatures, suggesting potential clinical utility for a transcriptomic-based approach to tumor classification. Overall, these data support the use of targeted amplicon-based RNAseq for comprehensive transcriptomic profiling of archival FFPE adrenal tumor material and indicate that this approach may facilitate important translational research opportunities for the study of these tumors.


Subject(s)
Adrenal Cortex Neoplasms/classification , Adrenal Cortex Neoplasms/diagnosis , Biomarkers, Tumor/genetics , Paraffin Embedding/methods , RNA-Seq/methods , Transcriptome , Adrenal Cortex Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Cohort Studies , Diagnosis, Differential , Female , Follow-Up Studies , Formaldehyde/chemistry , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Prognosis
11.
Clin Genet ; 96(3): 261-265, 2019 09.
Article in English | MEDLINE | ID: mdl-31219618

ABSTRACT

Short stature homeobox (SHOX) haploinsufficiency is a frequent cause of short stature. Despite advances in sequencing technologies, the identification of SHOX mutations continues to be performed using standard methods, including multiplex ligation-dependent probe amplification (MLPA) followed by Sanger sequencing. We designed a targeted panel of genes associated with growth impairment, including SHOX genomic and enhancer regions, to improve the resolution of next-generation sequencing for SHOX analysis. We used two software packages, CONTRA and Nexus Copy Number, in addition to visual analysis to investigate the presence of copy number variants (CNVs). We evaluated 15 patients with previously known SHOX defects, including point mutations, deletions and a duplication, and 77 patients with idiopathic short stature (ISS). The panel was able to confirm all known defects in the validation analysis. During the prospective evaluation, we identified two new partial SHOX deletions (one detected only by visual analysis), including an intragenic deletion not detected by MLPA. Additionally, we were able to determine the breakpoints in four cases. Our results show that the designed panel can be used for the molecular investigation of patients with ISS, and it may even detect CNVs in SHOX and its enhancers, which may be present in a significant fraction of patients.


Subject(s)
Genetic Association Studies , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing , Mutation , Short Stature Homeobox Protein/genetics , DNA Copy Number Variations , DNA Mutational Analysis , Female , Genotype , Humans , Male , Phenotype
12.
Clin Genet ; 95(1): 172-176, 2019 01.
Article in English | MEDLINE | ID: mdl-30294972

ABSTRACT

Molecular diagnosis is rarely established in 46,XX testicular (T) disorder of sex development (DSD) individuals with atypical genitalia. The Wilms' tumour factor-1 (WT1) gene is involved in early gonadal development in both sexes. Classically, WT1 deleterious variants are associated with 46,XY disorders of sex development (DSD) because of gonadal dysgenesis. We report a novel frameshift WT1 variant identified in an SRY-negative 46,XX testicular DSD girl born with atypical genitalia. Target massively parallel sequencing involving DSD-related genes identified a novel heterozygous WT1 c.1453_1456del; p.Arg485Glyfs*14 variant located in the fourth zinc finger of the protein which is absent in the population databases. Segregation analysis and microsatellite analysis confirmed the de novo status of the variant that is predicted to be deleterious by in silico tools and to increase WT1 target activation in crystallographic model. This novel and predicted activating frameshift WT1 variant leading to the 46,XX testicular DSD phenotype includes the fourth zinc-finger DNA-binding domain defects in the genetic aetiology of 46,XX DSD.


Subject(s)
46, XX Disorders of Sex Development/diagnosis , Pathology, Molecular , Testicular Diseases/diagnosis , WT1 Proteins/genetics , 46, XX Disorders of Sex Development/genetics , 46, XX Disorders of Sex Development/pathology , Child , DNA-Binding Proteins/genetics , Female , Heterozygote , Humans , Infant , Male , Mutation , Phenotype , Sexual Development/genetics , Testicular Diseases/genetics , Testicular Diseases/pathology , Testis/pathology
13.
Brain ; 141(8): 2299-2311, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29985992

ABSTRACT

The transcription factor BCL11B is essential for development of the nervous and the immune system, and Bcl11b deficiency results in structural brain defects, reduced learning capacity, and impaired immune cell development in mice. However, the precise role of BCL11B in humans is largely unexplored, except for a single patient with a BCL11B missense mutation, affected by multisystem anomalies and profound immune deficiency. Using massively parallel sequencing we identified 13 patients bearing heterozygous germline alterations in BCL11B. Notably, all of them are affected by global developmental delay with speech impairment and intellectual disability; however, none displayed overt clinical signs of immune deficiency. Six frameshift mutations, two nonsense mutations, one missense mutation, and two chromosomal rearrangements resulting in diminished BCL11B expression, arose de novo. A further frameshift mutation was transmitted from a similarly affected mother. Interestingly, the most severely affected patient harbours a missense mutation within a zinc-finger domain of BCL11B, probably affecting the DNA-binding structural interface, similar to the recently published patient. Furthermore, the most C-terminally located premature termination codon mutation fails to rescue the progenitor cell proliferation defect in hippocampal slice cultures from Bcl11b-deficient mice. Concerning the role of BCL11B in the immune system, extensive immune phenotyping of our patients revealed alterations in the T cell compartment and lack of peripheral type 2 innate lymphoid cells (ILC2s), consistent with the findings described in Bcl11b-deficient mice. Unsupervised analysis of 102 T lymphocyte subpopulations showed that the patients clearly cluster apart from healthy children, further supporting the common aetiology of the disorder. Taken together, we show here that mutations leading either to BCL11B haploinsufficiency or to a truncated BCL11B protein clinically cause a non-syndromic neurodevelopmental delay. In addition, we suggest that missense mutations affecting specific sites within zinc-finger domains might result in distinct and more severe clinical outcomes.


Subject(s)
Neurodevelopmental Disorders/genetics , Repressor Proteins/genetics , Repressor Proteins/physiology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology , Adolescent , Animals , Child , Child, Preschool , Female , Gene Expression Regulation/genetics , Germ-Line Mutation , Haploinsufficiency , Heterozygote , High-Throughput Nucleotide Sequencing , Humans , Infant , Lymphocytes/pathology , Lymphocytes/physiology , Male , Mice , Mutation , Repressor Proteins/metabolism , T-Lymphocytes/physiology , Transcription Factors/genetics , Tumor Suppressor Proteins/metabolism
14.
Int J Mol Sci ; 20(5)2019 Mar 06.
Article in English | MEDLINE | ID: mdl-30845661

ABSTRACT

Glioblastoma (GBM) is the most aggressive type of brain tumor, with an overall survival of 17 months under the current standard of care therapy. CD99, an over-expressed transmembrane protein in several malignancies, has been considered a potential target for immunotherapy. To further understand this potentiality, we analyzed the differential expression of its two isoforms in human astrocytoma specimens, and the CD99 involved signaling pathways in glioma model U87MG cell line. CD99 was also analyzed in GBM molecular subtypes. Whole transcriptomes by RNA-Seq of CD99-siRNA, and functional in vitro assays in CD99-shRNA, that are found in U87MG cells, were performed. Astrocytoma of different malignant grades and U87MG cells only expressed CD99 isoform 1, which was higher in mesenchymal and classical than in proneural GBM subtypes. Genes related to actin dynamics, predominantly to focal adhesion, and lamellipodia/filopodia formation were down-regulated in the transcriptome analysis, when CD99 was silenced. A decrease in tumor cell migration/invasion, and dysfunction of focal adhesion, were observed in functional assays. In addition, a striking morphological change was detected in CD99-silenced U87MG cells, further corroborating CD99 involvement in actin cytoskeleton rearrangement. Inhibiting the overexpressed CD99 may improve resectability and decrease the recurrence rate of GBM by decreasing tumor cells migration and invasion.


Subject(s)
12E7 Antigen/genetics , 12E7 Antigen/metabolism , Brain Neoplasms/genetics , Gene Expression Profiling/methods , Glioblastoma/genetics , Up-Regulation , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Focal Adhesion Kinase 1/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Genes, src/genetics , Glioblastoma/metabolism , Humans , Neoplasm Invasiveness , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Small Interfering/pharmacology , Sequence Analysis, RNA
15.
Genet Med ; 20(1): 91-97, 2018 01.
Article in English | MEDLINE | ID: mdl-28661490

ABSTRACT

PurposeC-type natriuretic peptide (CNP) and its principal receptor, natriuretic peptide receptor B (NPR-B), have been shown to be important in skeletal development. CNP and NPR-B are encoded by natriuretic peptide precursor-C (NPPC) and natriuretic peptide receptor 2 (NPR2) genes, respectively. While NPR2 mutations have been described in patients with skeletal dysplasias and idiopathic short stature (ISS), and several Npr2 and Nppc skeletal dysplasia mouse models exist, no mutations in NPPC have been described in patients to date.MethodsNPPC was screened in 668 patients (357 with disproportionate short stature and 311 with autosomal dominant ISS) and 29 additional ISS families in an ongoing whole-exome sequencing study.ResultsTwo heterozygous NPPC mutations, located in the highly conserved CNP ring, were identified. Both showed significant reductions in cyclic guanosine monophosphate synthesis, confirming their pathogenicity. Interestingly, one has been previously linked to skeletal abnormalities in the spontaneous Nppc mouse long-bone abnormality (lbab) mutant.ConclusionsOur results demonstrate, for the first time, that NPPC mutations cause autosomal dominant short stature in humans. The NPPC mutations cosegregated with a short stature and small hands phenotype. A CNP analog, which is currently in clinical trials for the treatment of achondroplasia, seems a promising therapeutic approach, since it directly replaces the defective protein.


Subject(s)
Dwarfism/diagnosis , Dwarfism/genetics , Genes, Dominant , Mutation , Natriuretic Peptide, C-Type/genetics , Adolescent , Amino Acid Sequence , Child , Computational Biology/methods , DNA Mutational Analysis , Female , Growth Charts , Heterozygote , Humans , Male , Natriuretic Peptide, C-Type/chemistry , Phenotype , Exome Sequencing
16.
Clin Genet ; 94(5): 461-466, 2018 11.
Article in English | MEDLINE | ID: mdl-30019515

ABSTRACT

SUZ12 is a core component of polycomb repressive complex 2 (PRC2) along with EZH2 and EED. Recently, germline mutations in the SUZ12, EZH2 and EED genes have been reported in Weaver syndrome (WS) or Weaver-like syndrome, suggesting a functional link between PRC2 deficits and WS. However, only one case of a SUZ12 mutation presenting with Weaver-like syndrome has been reported. Here, we report a missense and a frameshift mutation in SUZ12 (c.1797A>C; p.Gln599His and c.844_845del; p.Ala282Glnfs*7), both of which are novel, in two individuals. Their clinical features included postnatal overgrowth, increased bifrontal diameter, large ears, round face, horizontal chin crease and skeletal anomalies, but did not fulfill the WS diagnostic criteria. These data provide strong evidence that SUZ12 mutations cause Weaver-like syndrome.


Subject(s)
Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/genetics , Congenital Hypothyroidism/diagnosis , Congenital Hypothyroidism/genetics , Craniofacial Abnormalities/diagnosis , Craniofacial Abnormalities/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Hand Deformities, Congenital/diagnosis , Hand Deformities, Congenital/genetics , Mutation , Phenotype , Polycomb Repressive Complex 2/genetics , Alleles , Amino Acid Substitution , Facies , Female , Genotype , Humans , Male , Neoplasm Proteins , Pedigree , Transcription Factors
17.
Clin Endocrinol (Oxf) ; 88(3): 425-431, 2018 03.
Article in English | MEDLINE | ID: mdl-29265571

ABSTRACT

OBJECTIVES: The aetiology of congenital hypopituitarism (CH) is unknown in most patients. Rare copy number variants (CNVs) have been implicated as the cause of genetic syndromes with previously unknown aetiology. Our aim was to study the presence of CNVs and their pathogenicity in patients with idiopathic CH associated with complex phenotypes. DESIGN AND PATIENTS: We selected 39 patients with syndromic CH for array-based comparative genomic hybridization (aCGH). Patients with pathogenic CNVs were also evaluated by whole exome sequencing. RESULTS: Twenty rare CNVs were detected in 19 patients. Among the identified rare CNVs, six were classified as benign, eleven as variants of uncertain clinical significance (VUS) and four as pathogenic. The three patients with pathogenic CNVs had combined pituitary hormone deficiencies, and the associated complex phenotypes were intellectual disabilities: trichorhinophalangeal type I syndrome (TRPS1) and developmental delay/intellectual disability with cardiac malformation, respectively. Patient one has a de novo 1.6-Mb deletion located at chromosome 3q13.31q13.32, which overlaps with the region of the 3q13.31 deletion syndrome. Patient two has a 10.5-Mb de novo deletion at 8q23.1q24.11, encompassing the TRPS1 gene; his phenotype is compatible with TRPS1. Patient three carries a chromosome translocation t(2p24.3;4q35.1) resulting in two terminal alterations: a 2p25.3p24.3 duplication of 14.7 Mb and a 4-Mb deletion at 4q35.1q35.2. CONCLUSIONS: Copy number variants explained the phenotype in 8% of patients with hypopituitarism and additional complex phenotypes. This suggests that chromosomal alterations are an important contributor to syndromic hypopituitarism.


Subject(s)
DNA Copy Number Variations , Hypopituitarism/congenital , Hypopituitarism/etiology , Phenotype , Chromosome Aberrations , Comparative Genomic Hybridization , Human Growth Hormone/deficiency , Humans , Hypopituitarism/genetics , Intellectual Disability , Exome Sequencing
18.
Clin Endocrinol (Oxf) ; 82(4): 481-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25200669

ABSTRACT

OBJECTIVE: LIN28 control cells reprogramming and pluripotency mainly through miRNA regulation and has been overexpressed in many advanced cancers. In this study, we evaluated the prognostic role of LIN28 and its regulatory miRNAs in a large cohort of adrenocortical tumours (ACTs). PATIENTS AND METHODS: LIN28 protein expression was assessed in 266 adults ACTs (78 adenomas and 188 carcinomas) from Brazil and Germany. LIN28A and LIN28B gene expression was analysed in 59 ACTs (31 adenomas and 28 carcinomas) and copy number variation in 39 ACTs. In addition, we determined the expression of let-7 family, mir-9, mir-30 and mir-125 in 28 carcinomas. RESULTS: LIN28A gene was overexpressed in aggressive ACCs when compared with adenomas and nonaggressive ACCs, but no LIN28A copy number variation was found in ACTs. Unexpectedly, weak LIN28 protein expression was significantly associated with reduced disease-free survival in ACC patients (P = 0·01), but for overall survival only a trend was detectable (P = 0·117). In the multivariate analysis, only Ki67 index ≥10% (HR 4·6, P = 0·000) and weak LIN28 protein expression (HR 2·0, P = 0·03) were independent predictors of recurrence in ACC patients. Interestingly, mir-9 expression, a negative LIN28A/B regulator, was significantly higher in aggressive than in nonaggressive ACCs [2076 (from 36 to 9307) vs 133·4 (from 2·4 to 5193); P = 0·011] and was highly associated with reduced overall (P = 0·01) and disease-free survival (P = 0·01). However, mir-9 prognostic role should be further evaluated in a larger cohort. CONCLUSION: Weak LIN28 protein expression was associated with recurrence in ACCs. Additionally, overexpression of mir-9, a negative LIN28A regulator, was associated with poor outcome.


Subject(s)
Adrenal Cortex Neoplasms/genetics , Adrenal Cortex Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Adenoma/genetics , Adenoma/metabolism , Adrenocortical Carcinoma/genetics , Adrenocortical Carcinoma/metabolism , Adult , Brazil , Cohort Studies , DNA Copy Number Variations , Disease-Free Survival , Female , Gene Expression Profiling , Genome-Wide Association Study , Germany , Humans , Ki-67 Antigen/metabolism , Male , Middle Aged , Neoplasm Recurrence, Local , Prognosis , Real-Time Polymerase Chain Reaction , Treatment Outcome
19.
BMC Endocr Disord ; 14: 42, 2014 May 12.
Article in English | MEDLINE | ID: mdl-24884994

ABSTRACT

BACKGROUND: Although chronic adrenocorticotropic hormone (ACTH) and androgen hyperstimulation are assumed to be involved in the pathogenesis of adrenal myelolipomas associated with poor-compliance patients with congenital adrenal hyperplasia (CAH), the expression of their receptors has not yet been demonstrated in these tumors so far. METHODS: We analyzed Melanocortin 2 receptor (MC2R), Androgen Receptor (AR), Leptin (LEP), and Steroidogenic factor 1 (SF1) expression using real-time qRT-PCR in two giant bilateral adrenal myelolipomas from two untreated simple virilizing CAH cases and in two sporadic adrenal myelolipomas. In addition, the X-chromosome inactivation pattern and CAG repeat numbers in AR exon 1 gene were evaluated in the 4 cases. RESULTS: The MC2R gene was overexpressed in myelolipomas from 3 out of 4 patients. AR overexpression was detected in 2 tumors: a giant bilateral myelolipoma in a CAH patient and a sporadic case. Simultaneous overexpression of AR and MC2R genes was found in two of the cases. Interestingly, the bilateral giant myelolipoma associated with CAH that had high androgen and ACTH levels but lacked MC2R and AR overexpression presented a significantly shorter AR allele compared with other tumors. In addition, X-chromosome inactivation pattern analysis showed a polyclonal origin in all tumors, suggesting a stimulatory effect as the trigger for tumor development. CONCLUSION: These findings are the first evidence for MC2R or AR overexpression in giant bilateral myelolipomas from poor-compliance CAH patients.


Subject(s)
Adrenal Gland Neoplasms/complications , Adrenal Hyperplasia, Congenital/complications , Biomarkers/metabolism , Myelolipoma/diagnosis , Receptor, Melanocortin, Type 2/genetics , Receptors, Androgen/genetics , Adrenal Gland Neoplasms/genetics , Adrenal Hyperplasia, Congenital/genetics , Adult , Female , Humans , Middle Aged , Myelolipoma/etiology , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Repetitive Sequences, Nucleic Acid/genetics , Reverse Transcriptase Polymerase Chain Reaction
20.
Mol Cell Endocrinol ; 585: 112176, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38341019

ABSTRACT

Hyperaldosteronism is often associated with inappropriate aldosterone production and aldosterone synthase (Cyp11b2) expression. Normally, Cyp11b2 expression is limited to the adrenal zona glomerulosa (ZG) and regulated by angiotensin II which signals through Gq protein-coupled receptors. As cells migrate inwards, they differentiate into 11ß-hydroxylase-expressing zona fasciculata (ZF) cells lacking Cyp11b2. The mechanism causing ZG-specific aldosterone biosynthesis is still unclear. We investigated the effect of chronic Gq signaling using transgenic mice with a clozapine N-oxide (CNO)-activated human M3 muscarinic receptor (DREADD) coupled to Gq (hM3Dq) that was expressed throughout the adrenal cortex. CNO raised circulating aldosterone in the presence of a high sodium diet with greater response seen in females compared to males. Immunohistochemistry and transcriptomics indicated disrupted zonal Cyp11b2 expression while Wnt signaling remained unchanged. Chronic Gq-DREADD signaling also induced an intra-adrenal RAAS in CNO-treated mice. Chronic Gq signaling disrupted adrenal cortex zonal aldosterone production associated with ZF expression of Cyp11b2.


Subject(s)
Adrenal Cortex , Hyperaldosteronism , Male , Female , Humans , Mice , Animals , Zona Fasciculata , Aldosterone/metabolism , Adrenal Cortex/metabolism , Zona Glomerulosa/metabolism , Cytochrome P-450 CYP11B2/genetics , Wnt Signaling Pathway , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL