Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Nature ; 547(7661): 55-60, 2017 07 06.
Article in English | MEDLINE | ID: mdl-28658208

ABSTRACT

Genomic analysis of tumours has led to the identification of hundreds of cancer genes on the basis of the presence of mutations in protein-coding regions. By contrast, much less is known about cancer-causing mutations in non-coding regions. Here we perform deep sequencing in 360 primary breast cancers and develop computational methods to identify significantly mutated promoters. Clear signals are found in the promoters of three genes. FOXA1, a known driver of hormone-receptor positive breast cancer, harbours a mutational hotspot in its promoter leading to overexpression through increased E2F binding. RMRP and NEAT1, two non-coding RNA genes, carry mutations that affect protein binding to their promoters and alter expression levels. Our study shows that promoter regions harbour recurrent mutations in cancer with functional consequences and that the mutations occur at similar frequencies as in coding regions. Power analyses indicate that more such regions remain to be discovered through deep sequencing of adequately sized cohorts of patients.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , Mutation , Promoter Regions, Genetic/genetics , Cohort Studies , E2F Transcription Factors/metabolism , Exome/genetics , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-alpha/metabolism , High-Throughput Nucleotide Sequencing , Humans , Protein Binding/genetics , RNA, Long Noncoding/genetics , Receptors, Estrogen/antagonists & inhibitors
2.
J Biol Chem ; 292(7): 2679-2689, 2017 02 17.
Article in English | MEDLINE | ID: mdl-28007963

ABSTRACT

The negative regulator of Rho family GTPases, p190A RhoGAP, is one of six mammalian proteins harboring so-called FF motifs. To explore the function of these and other p190A segments, we identified interacting proteins by tandem mass spectrometry. Here we report that endogenous human p190A, but not its 50% identical p190B paralog, associates with all 13 eIF3 subunits and several other translational preinitiation factors. The interaction involves the first FF motif of p190A and the winged helix/PCI domain of eIF3A, is enhanced by serum stimulation and reduced by phosphatase treatment. The p190A/eIF3A interaction is unaffected by mutating phosphorylated p190A-Tyr308, but disrupted by a S296A mutation, targeting the only other known phosphorylated residue in the first FF domain. The p190A-eIF3 complex is distinct from eIF3 complexes containing S6K1 or mammalian target of rapamycin (mTOR), and appears to represent an incomplete preinitiation complex lacking several subunits. Based on these findings we propose that p190A may affect protein translation by controlling the assembly of functional preinitiation complexes. Whether such a role helps to explain why, unique among the large family of RhoGAPs, p190A exhibits a significantly increased mutation rate in cancer remains to be determined.


Subject(s)
Eukaryotic Initiation Factor-3/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Protein Biosynthesis , Repressor Proteins/metabolism , Animals , Chromatography, Affinity , Eukaryotic Initiation Factor-3/chemistry , Eukaryotic Initiation Factor-3/genetics , Gene Knockdown Techniques , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/genetics , HeLa Cells , Humans , Mice , Mutation, Missense , NIH 3T3 Cells , Protein Binding , Repressor Proteins/chemistry , Repressor Proteins/genetics , Subcellular Fractions/metabolism
3.
PLoS Genet ; 9(11): e1003958, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24278035

ABSTRACT

Neurofibromatosis type 1 (NF1), a genetic disease that affects 1 in 3,000, is caused by loss of a large evolutionary conserved protein that serves as a GTPase Activating Protein (GAP) for Ras. Among Drosophila melanogaster Nf1 (dNf1) null mutant phenotypes, learning/memory deficits and reduced overall growth resemble human NF1 symptoms. These and other dNf1 defects are relatively insensitive to manipulations that reduce Ras signaling strength but are suppressed by increasing signaling through the 3'-5' cyclic adenosine monophosphate (cAMP) dependent Protein Kinase A (PKA) pathway, or phenocopied by inhibiting this pathway. However, whether dNf1 affects cAMP/PKA signaling directly or indirectly remains controversial. To shed light on this issue we screened 486 1(st) and 2(nd) chromosome deficiencies that uncover >80% of annotated genes for dominant modifiers of the dNf1 pupal size defect, identifying responsible genes in crosses with mutant alleles or by tissue-specific RNA interference (RNAi) knockdown. Validating the screen, identified suppressors include the previously implicated dAlk tyrosine kinase, its activating ligand jelly belly (jeb), two other genes involved in Ras/ERK signal transduction and several involved in cAMP/PKA signaling. Novel modifiers that implicate synaptic defects in the dNf1 growth deficiency include the intersectin-related synaptic scaffold protein Dap160 and the cholecystokinin receptor-related CCKLR-17D1 drosulfakinin receptor. Providing mechanistic clues, we show that dAlk, jeb and CCKLR-17D1 are among mutants that also suppress a recently identified dNf1 neuromuscular junction (NMJ) overgrowth phenotype and that manipulations that increase cAMP/PKA signaling in adipokinetic hormone (AKH)-producing cells at the base of the neuroendocrine ring gland restore the dNf1 growth deficiency. Finally, supporting our previous contention that ALK might be a therapeutic target in NF1, we report that human ALK is expressed in cells that give rise to NF1 tumors and that NF1 regulated ALK/RAS/ERK signaling appears conserved in man.


Subject(s)
Drosophila melanogaster/genetics , Memory Disorders/genetics , Neurofibromatosis 1/genetics , Anaplastic Lymphoma Kinase , Animals , Cyclic AMP/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Memory Disorders/pathology , Mutation , Neurofibromatosis 1/metabolism , Neurofibromatosis 1/physiopathology , Neuromuscular Junction/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/genetics , ras GTPase-Activating Proteins/genetics , ras GTPase-Activating Proteins/metabolism
4.
Am J Med Genet A ; 164A(3): 563-78, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24443315

ABSTRACT

The neurofibromatoses (NF) are autosomal dominant genetic disorders that encompass the rare diseases NF1, NF2, and schwannomatosis. The NFs affect more people worldwide than Duchenne muscular dystrophy and Huntington's disease combined. NF1 and NF2 are caused by mutations of known tumor suppressor genes (NF1 and NF2, respectively). For schwannomatosis, although mutations in SMARCB1 were identified in a subpopulation of schwannomatosis patients, additional causative gene mutations are still to be discovered. Individuals with NF1 may demonstrate manifestations in multiple organ systems, including tumors of the nervous system, learning disabilities, and physical disfigurement. NF2 ultimately can cause deafness, cranial nerve deficits, and additional severe morbidities caused by tumors of the nervous system. Unmanageable pain is a key finding in patients with schwannomatosis. Although today there is no marketed treatment for NF-related tumors, a significant number of clinical trials have become available. In addition, significant preclinical efforts have led to a more rational selection of potential drug candidates for NF trials. An important element in fueling this progress is the sharing of knowledge. For over 20 years the Children's Tumor Foundation has convened an annual NF Conference, bringing together NF professionals to share novel findings, ideas, and build collaborations. The 2012 NF Conference held in New Orleans hosted over 350 NF researchers and clinicians. This article provides a synthesis of the highlights presented at the conference and as such, is a "state-of-the-field" for NF research in 2012.


Subject(s)
Neurilemmoma/etiology , Neurofibromatoses/etiology , Neurofibromatosis 1/etiology , Neurofibromatosis 2/etiology , Skin Neoplasms/etiology , Humans , Neurilemmoma/genetics , Neurilemmoma/therapy , Neurofibromatoses/genetics , Neurofibromatoses/therapy , Neurofibromatosis 1/genetics , Neurofibromatosis 1/therapy , Neurofibromatosis 2/genetics , Neurofibromatosis 2/therapy , Skin Neoplasms/genetics , Skin Neoplasms/therapy
5.
PLoS Genet ; 7(9): e1002281, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21949657

ABSTRACT

Anaplastic Lymphoma Kinase (Alk) is a Receptor Tyrosine Kinase (RTK) activated in several cancers, but with largely unknown physiological functions. We report two unexpected roles for the Drosophila ortholog dAlk, in body size determination and associative learning. Remarkably, reducing neuronal dAlk activity increased body size and enhanced associative learning, suggesting that its activation is inhibitory in both processes. Consistently, dAlk activation reduced body size and caused learning deficits resembling phenotypes of null mutations in dNf1, the Ras GTPase Activating Protein-encoding conserved ortholog of the Neurofibromatosis type 1 (NF1) disease gene. We show that dAlk and dNf1 co-localize extensively and interact functionally in the nervous system. Importantly, genetic or pharmacological inhibition of dAlk rescued the reduced body size, adult learning deficits, and Extracellular-Regulated-Kinase (ERK) overactivation dNf1 mutant phenotypes. These results identify dAlk as an upstream activator of dNf1-regulated Ras signaling responsible for several dNf1 defects, and they implicate human Alk as a potential therapeutic target in NF1.


Subject(s)
Association Learning , Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , Nerve Tissue Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , ras GTPase-Activating Proteins/metabolism , Anaplastic Lymphoma Kinase , Animals , Body Size/genetics , Brain/metabolism , Central Nervous System/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Humans , MAP Kinase Signaling System/genetics , Molecular Targeted Therapy , Mutation , Nerve Tissue Proteins/genetics , Neurofibromin 1/antagonists & inhibitors , Neurofibromin 1/genetics , Neurofibromin 1/metabolism , Neurons/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction , ras GTPase-Activating Proteins/genetics
6.
Am J Med Genet A ; 155A(2): 307-21, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21271647

ABSTRACT

The neurofibromatoses (NF) encompass the rare diseases NF1, NF2, and schwannomatosis. The NFs affect 100,000 Americans; over 2 million persons worldwide; and are caused by mutation of tumor suppressor genes. Individuals with NF1 in particular may develop tumors anywhere in the nervous system; additional manifestations can include learning disabilities, bone dysplasia, cardiovascular defects, unmanageable pain, and physical disfigurement. Ultimately, the NFs can cause blindness, deafness, severe morbidity, and increased mortality and NF1 includes a risk of malignant cancer. Today there is no treatment for the NFs (other than symptomatic); however, research efforts to understand these genetic conditions have made tremendous strides in the past few years. Progress is being made on all fronts, from discovery studies-understanding the molecular signaling deficits that cause the manifestations of NF-to the growth of preclinical drug screening initiatives and the emergence of a number of clinical trials. An important element in fuelling this progress is the sharing of knowledge, and to this end, for over 20 years the Children's Tumor Foundation has convened an annual NF Conference, bringing together NF professionals to share ideas and build collaborations. The 2010 NF Conference held in Baltimore, MD June 5-8, 2010 hosted over 300 NF researchers and clinicians. This paper provides a synthesis of the highlights presented at the Conference and as such, is a "state-of-the-field" for NF research in 2010.


Subject(s)
Genes, Tumor Suppressor , Neurofibromatoses/diagnosis , Neurofibromatoses/drug therapy , Neurofibromatoses/pathology , Signal Transduction/physiology , Animals , Disease Models, Animal , Genes, ras/genetics , Humans , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Neurofibromatoses/genetics
8.
Trends Cell Biol ; 14(7): 377-85, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15246431

ABSTRACT

The small GTPases of the Ras superfamily mediate numerous biological processes through their ability to cycle between an inactive GDP-bound and an active GTP-bound form. Among the key regulators of GTPase cycling are the GTPase-activating proteins (GAPs), which stimulate the weak intrinsic GTP-hydrolysis activity of the GTPases, thereby inactivating them. Despite the abundance of GAPs and the fact that mutations in GAP-encoding genes underlie several human diseases, these proteins have received relatively little attention. Recent studies have addressed the regulatory mechanisms that influence GAP activity. So far, findings suggest that GAP activity is regulated by several mechanisms, including protein-protein interactions, phospholipid interactions, phosphorylation, subcellular translocation and proteolytic degradation.


Subject(s)
GTP Phosphohydrolases/metabolism , GTPase-Activating Proteins/metabolism , Humans , Lipid Metabolism , Models, Biological , Phosphorylation , Protein Binding/physiology , Protein Structure, Tertiary/physiology
9.
Cancer Cell ; 36(3): 288-301.e14, 2019 09 16.
Article in English | MEDLINE | ID: mdl-31526759

ABSTRACT

Current statistical models for assessing hotspot significance do not properly account for variation in site-specific mutability, thereby yielding many false-positives. We thus (i) detail a Log-normal-Poisson (LNP) background model that accounts for this variability in a manner consistent with models of mutagenesis; (ii) use it to show that passenger hotspots arise from all common mutational processes; and (iii) apply it to a ∼10,000-patient cohort to nominate driver hotspots with far fewer false-positives compared with conventional methods. Overall, we show that many cancer hotspot mutations recurring at the same genomic site across multiple tumors are actually passenger events, recurring at inherently mutable genomic sites under no positive selection.


Subject(s)
Carcinogenesis/genetics , Genomics/methods , Models, Genetic , Mutagenesis , Neoplasms/genetics , DNA Mutational Analysis , Datasets as Topic , Genes, Tumor Suppressor , Humans , Poisson Distribution , ROC Curve , Selection, Genetic , Exome Sequencing
10.
J Neurosci ; 27(17): 4716-24, 2007 Apr 25.
Article in English | MEDLINE | ID: mdl-17460084

ABSTRACT

In the germinative zone of the adult rodent brain, neural progenitors migrate into niches delimited by astrocyte processes and differentiate into neuronal precursors. In the present study, we report a modulating role for the scaffolding protein IQGAP1 in neural progenitor migration. We have identified IQGAP1 as a new marker of amplifying neural progenitor and neuronal precursor cells of the subventricular zone (SVZ) and the rostral migratory stream (RMS) in the adult mouse brain. To determine functions for IQGAP1 in neural progenitors, we compared the properties of neural progenitor cells from wild-type and Iqgap1-null mutant mice in vivo and in vitro. The in vivo studies reveal a delay in the transition of de novo neural progenitors into neuronal precursor cells in Iqgap1-null mice. In vitro, we demonstrated that IQGAP1 acts as a downstream effector in the vascular endothelial growth factor (VEGF)-dependent migratory response of neural progenitors that also impacts on their neuronal differentiation. The Rho-family GTPases cdc42/Rac1 and Lis1 are major partners of IQGAP1 in this migratory process. Finally, astrocytes of the neurogenic SVZ and RMS are shown to express VEGF. We propose that VEGF synthesized by astrocytes could be involved in the guidance of neural progenitors to neurogenic niches and that IQGAP1 is an effector of the VEGF-dependent migratory signal.


Subject(s)
Adult Stem Cells/cytology , Adult Stem Cells/physiology , Cell Movement/physiology , Neurons/cytology , ras GTPase-Activating Proteins/metabolism , Animals , Astrocytes/cytology , Astrocytes/metabolism , Cell Communication/physiology , Cell Differentiation/physiology , Cell Movement/drug effects , Cerebral Ventricles/cytology , In Vitro Techniques , Mice , Mice, Knockout , Neuropeptides/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology , cdc42 GTP-Binding Protein/metabolism , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein , ras GTPase-Activating Proteins/genetics
11.
Nat Neurosci ; 5(2): 95-6, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11788835

ABSTRACT

Neurofibromatosis type 1 (NF1) is a dominant genetic disorder characterized by multiple benign and malignant nervous system tumors, and by learning defects in 45% of children with NF1 mutations. Studies of neurofibromin, the protein encoded by NF1, have focused on its functions in tumorigenesis and regulation of Ras activity; however, Drosophila NF1 regulates both Ras and cyclic AMP (cAMP) pathways. Expression of a human NF1 transgene rescued cAMP-related phenotypes in NF1 mutant flies (small body size and G protein-stimulated adenylyl cyclase (AC) activity defects), and neuropeptide- and G protein-stimulated AC activity were lower in Nf1-/- as compared to Nf1+/- mouse brains, demonstrating that neurofibromin regulates AC activity in both mammals and flies.


Subject(s)
Adenylyl Cyclases/metabolism , GTP-Binding Proteins/physiology , Neurofibromin 1/physiology , Animals , Drosophila , GTP-Binding Proteins/genetics , Genes, Neurofibromatosis 1/physiology , Humans , Mice , Mice, Knockout/genetics , Mutation/physiology , Neurofibromin 1/genetics , Transgenes/physiology
12.
J Cell Biol ; 217(9): 3183-3201, 2018 09 03.
Article in English | MEDLINE | ID: mdl-29934311

ABSTRACT

ARHGAP35 encoding p190A RhoGAP is a cancer-associated gene with a mutation spectrum suggestive of a tumor-suppressor function. In this study, we demonstrate that loss of heterozygosity for ARHGAP35 occurs in human tumors. We sought to identify tumor-suppressor capacities for p190A RhoGAP (p190A) and its paralog p190B in epithelial cells. We reveal an essential role for p190A and p190B to promote contact inhibition of cell proliferation (CIP), a function that relies on RhoGAP activity. Unbiased mRNA sequencing analyses establish that p190A and p190B modulate expression of genes associated with the Hippo pathway. Accordingly, we determine that p190A and p190B induce CIP by repressing YAP-TEAD-regulated gene transcription through activation of LATS kinases and inhibition of the Rho-ROCK pathway. Finally, we demonstrate that loss of a single p190 paralog is sufficient to elicit nuclear translocation of YAP and perturb CIP in epithelial cells cultured in Matrigel. Collectively, our data reveal a novel mechanism consistent with a tumor-suppressor function for ARHGAP35.


Subject(s)
Cell Proliferation/physiology , Contact Inhibition/physiology , Epithelial Cells/metabolism , GTPase-Activating Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Neoplasms/pathology , Repressor Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Line , DNA-Binding Proteins/genetics , Dogs , GTPase-Activating Proteins/genetics , Gene Expression Regulation, Neoplastic/genetics , Guanine Nucleotide Exchange Factors/genetics , Hippo Signaling Pathway , Humans , Madin Darby Canine Kidney Cells , Neoplasms/genetics , Nuclear Proteins/genetics , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Small Interfering/genetics , Repressor Proteins/genetics , TEA Domain Transcription Factors , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , YAP-Signaling Proteins , rho-Associated Kinases/metabolism
13.
J Exp Med ; 214(10): 2875-2887, 2017 Oct 02.
Article in English | MEDLINE | ID: mdl-28878000

ABSTRACT

Recent advances in single-cell, transcriptomic profiling have provided unprecedented access to investigate cell heterogeneity during tissue and organ development. In this study, we used massively parallel, single-cell RNA sequencing to define cell heterogeneity within the zebrafish kidney marrow, constructing a comprehensive molecular atlas of definitive hematopoiesis and functionally distinct renal cells found in adult zebrafish. Because our method analyzed blood and kidney cells in an unbiased manner, our approach was useful in characterizing immune-cell deficiencies within DNA-protein kinase catalytic subunit (prkdc), interleukin-2 receptor γ a (il2rga), and double-homozygous-mutant fish, identifying blood cell losses in T, B, and natural killer cells within specific genetic mutants. Our analysis also uncovered novel cell types, including two classes of natural killer immune cells, classically defined and erythroid-primed hematopoietic stem and progenitor cells, mucin-secreting kidney cells, and kidney stem/progenitor cells. In total, our work provides the first, comprehensive, single-cell, transcriptomic analysis of kidney and marrow cells in the adult zebrafish.


Subject(s)
Hematopoiesis, Extramedullary/genetics , Kidney/cytology , RNA/genetics , Zebrafish/anatomy & histology , Animals , Animals, Genetically Modified , Cell Lineage/genetics , Cell Lineage/physiology , Gene Expression Profiling , Hematopoiesis, Extramedullary/physiology , Hematopoietic Stem Cells , Kidney/metabolism , Sequence Analysis, RNA , Zebrafish/genetics , Zebrafish/metabolism
14.
Nat Biotechnol ; 35(10): 951-959, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28892075

ABSTRACT

Microsatellites (MSs) are tracts of variable-length repeats of short DNA motifs that exhibit high rates of mutation in the form of insertions or deletions (indels) of the repeated motif. Despite their prevalence, the contribution of somatic MS indels to cancer has been largely unexplored, owing to difficulties in detecting them in short-read sequencing data. Here we present two tools: MSMuTect, for accurate detection of somatic MS indels, and MSMutSig, for identification of genes containing MS indels at a higher frequency than expected by chance. Applying MSMuTect to whole-exome data from 6,747 human tumors representing 20 tumor types, we identified >1,000 previously undescribed MS indels in cancer genes. Additionally, we demonstrate that the number and pattern of MS indels can accurately distinguish microsatellite-stable tumors from tumors with microsatellite instability, thus potentially improving classification of clinically relevant subgroups. Finally, we identified seven MS indel driver hotspots: four in known cancer genes (ACVR2A, RNF43, JAK1, and MSH3) and three in genes not previously implicated as cancer drivers (ESRP1, PRDM2, and DOCK3).


Subject(s)
INDEL Mutation/genetics , Microsatellite Repeats/genetics , Neoplasms/genetics , Exome/genetics , Genes, Neoplasm , High-Throughput Nucleotide Sequencing , Humans , Microsatellite Instability , Mutation/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
15.
Methods Enzymol ; 407: 1-9, 2006.
Article in English | MEDLINE | ID: mdl-16757309

ABSTRACT

For geneticists and other researchers alike it is often useful to know how many related proteins might perform similar functions. With this in mind, a survey was performed to determine what proportion of human and Drosophila genes code for Ras superfamily members and their positive or negative regulators. Results indicate that just < 2% of genes in both genomes predict such proteins. A database was compiled to provide easy access to this information. This database also includes information on approximately 360 putative Ras superfamily effector proteins and may be a useful tool for those interested in GTPase biology.


Subject(s)
Databases, Protein , ras GTPase-Activating Proteins/genetics , ras Proteins/genetics , Animals , Humans , Monomeric GTP-Binding Proteins/genetics
16.
Biochim Biophys Acta ; 1603(2): 47-82, 2003 Mar 17.
Article in English | MEDLINE | ID: mdl-12618308

ABSTRACT

Typical members of the Ras superfamily of small monomeric GTP-binding proteins function as regulators of diverse processes by cycling between biologically active GTP- and inactive GDP-bound conformations. Proteins that control this cycling include guanine nucleotide exchange factors or GEFs, which activate Ras superfamily members by catalyzing GTP for GDP exchange, and GTPase activating proteins or GAPs, which accelerate the low intrinsic GTP hydrolysis rate of typical Ras superfamily members, thus causing their inactivation. Two among the latter class of proteins have been implicated in common genetic disorders associated with an increased cancer risk, neurofibromatosis-1, and tuberous sclerosis. To facilitate genetic analysis, I surveyed Drosophila and human sequence databases for genes predicting proteins related to GAPs for Ras superfamily members. Remarkably, close to 0.5% of genes in both species (173 human and 64 Drosophila genes) predict proteins related to GAPs for Arf, Rab, Ran, Rap, Ras, Rho, and Sar family GTPases. Information on these genes has been entered into a pair of relational databases, which can be used to identify evolutionary conserved proteins that are likely to serve basic biological functions, and which can be updated when definitive information on the coding potential of both genomes becomes available.


Subject(s)
ras GTPase-Activating Proteins/genetics , ADP-Ribosylation Factors/genetics , Animals , Databases, Genetic , Drosophila , GTPase-Activating Proteins/genetics , Humans , Monomeric GTP-Binding Proteins/genetics , Protein Structure, Tertiary , Saccharomyces cerevisiae Proteins/genetics , Substrate Specificity , Vesicular Transport Proteins , rab GTP-Binding Proteins/genetics , ran GTP-Binding Protein/genetics , rap GTP-Binding Proteins/genetics , ras GTPase-Activating Proteins/chemistry , rho GTP-Binding Proteins/genetics
18.
Mol Immunol ; 38(9): 701-8, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11858825

ABSTRACT

Nf1(-/-) fetal liver cells were used to reconstitute B and T cells in Rag-1(-/-) mice. Lymphocyte development was largely unimpaired in the absence of neurofibromin. However antigen-receptor induced proliferation was defective in neurofibromin deficient peripheral B cells and CD4(+) single positive thymocytes. In contrast to its role as a negative regulator of proliferation in many other cell types, neurofibromin may be a positive regulator of lymphocyte proliferation. Peripheral B cells exhibited circumscribed defects in anti-IgM induced protein tyrosine phosphorylation, which may contribute to the unexpected proliferative defect seen in these cells.


Subject(s)
B-Lymphocytes/physiology , CD4-Positive T-Lymphocytes/physiology , Neurofibromin 1/physiology , Animals , B-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/pathology , Cell Division/physiology , Gene Deletion , Lymphocyte Activation/physiology , Mice , Mice, Knockout , Neurofibromin 1/genetics , Signal Transduction/immunology
19.
Nat Med ; 21(11): 1290-7, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26501192

ABSTRACT

Genome-wide association studies (GWASs) have linked genes to various pathological traits. However, the potential contribution of regulatory noncoding RNAs, such as microRNAs (miRNAs), to a genetic predisposition to pathological conditions has remained unclear. We leveraged GWAS meta-analysis data from >188,000 individuals to identify 69 miRNAs in physical proximity to single-nucleotide polymorphisms (SNPs) associated with abnormal levels of circulating lipids. Several of these miRNAs (miR-128-1, miR-148a, miR-130b, and miR-301b) control the expression of key proteins involved in cholesterol-lipoprotein trafficking, such as the low-density lipoprotein (LDL) receptor (LDLR) and the ATP-binding cassette A1 (ABCA1) cholesterol transporter. Consistent with human liver expression data and genetic links to abnormal blood lipid levels, overexpression and antisense targeting of miR-128-1 or miR-148a in high-fat diet-fed C57BL/6J and Apoe-null mice resulted in altered hepatic expression of proteins involved in lipid trafficking and metabolism, and in modulated levels of circulating lipoprotein-cholesterol and triglycerides. Taken together, these findings support the notion that altered expression of miRNAs may contribute to abnormal blood lipid levels, predisposing individuals to human cardiometabolic disorders.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Cholesterol, HDL/metabolism , Cholesterol, LDL/metabolism , Diet, High-Fat , Dyslipidemias/genetics , MicroRNAs/genetics , Receptors, LDL/metabolism , Triglycerides/metabolism , Animals , Apolipoproteins E/genetics , Cholesterol/metabolism , Genome-Wide Association Study , Homeostasis/genetics , Humans , Lipoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Polymorphism, Single Nucleotide
20.
Rare Dis ; 2: e28341, 2014.
Article in English | MEDLINE | ID: mdl-25054093

ABSTRACT

Neurofibromatosis type 1 (NF1) is caused by loss of a negative regulator of Ras oncoproteins. Unknown genetic modifiers have been implicated in NF1's characteristic variability. Drosophila melanogaster dNf1 phenotypes include cognitive deficits and reduced growth, both of which resemble human symptoms. We recently reported results of a screen for dominant modifiers of dNf1 growth. Suppressors include the dAlk tyrosine kinase and its activating ligand, two other genes involved in Ras/ERK signal transduction, the synaptic scaffold Dap160 and the CCKLR-17D1 drosulfakinin receptor. Additional modifiers include several genes involved in cAMP/PKA signaling. Providing mechanistic insights, dAlk, jeb, and CCKLR-17D1 also suppress a dNf1 synaptic overgrowth defect, and increasing cAMP/PKA signaling in the neuroendocrine ring gland rescued the dNf1 growth deficiency. Finally, among the several suppressors identified in our screen, we specifically implicate ALK as a potential therapeutic target by showing that NF1-regulated ALK/RAS/ERK signaling is conserved in human cells.

SELECTION OF CITATIONS
SEARCH DETAIL