Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Science ; 384(6701): 1196-1202, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38870301

ABSTRACT

In vivo genome correction holds promise for generating durable disease cures; yet, effective stem cell editing remains challenging. In this work, we demonstrate that optimized lung-targeting lipid nanoparticles (LNPs) enable high levels of genome editing in stem cells, yielding durable responses. Intravenously administered gene-editing LNPs in activatable tdTomato mice achieved >70% lung stem cell editing, sustaining tdTomato expression in >80% of lung epithelial cells for 660 days. Addressing cystic fibrosis (CF), NG-ABE8e messenger RNA (mRNA)-sgR553X LNPs mediated >95% cystic fibrosis transmembrane conductance regulator (CFTR) DNA correction, restored CFTR function in primary patient-derived bronchial epithelial cells equivalent to Trikafta for F508del, corrected intestinal organoids and corrected R553X nonsense mutations in 50% of lung stem cells in CF mice. These findings introduce LNP-enabled tissue stem cell editing for disease-modifying genome correction.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator , Cystic Fibrosis , Gene Editing , Lung , Nanoparticles , Stem Cells , Animals , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Mice , Cystic Fibrosis/therapy , Cystic Fibrosis/genetics , Lung/metabolism , Stem Cells/metabolism , Humans , Lipids , Organoids , Epithelial Cells/metabolism , CRISPR-Cas Systems , Genetic Therapy/methods , Liposomes
2.
FASEB J ; 38(8): e23606, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38648465

ABSTRACT

Rhodopsin mislocalization encompasses various blind conditions. Rhodopsin mislocalization is the primary factor leading to rod photoreceptor dysfunction and degeneration in autosomal dominant retinitis pigmentosa (adRP) caused by class I mutations. In this study, we report a new knock-in mouse model that harbors a class I Q344X mutation in the endogenous rhodopsin gene, which causes rod photoreceptor degeneration in an autosomal dominant pattern. In RhoQ344X/+ mice, mRNA transcripts from the wild-type (Rho) and RhoQ344X mutant rhodopsin alleles are expressed at equal levels. However, the amount of RHOQ344X mutant protein is 2.7 times lower than that of wild-type rhodopsin, a finding consistent with the rapid degradation of the mutant protein. Immunofluorescence microscopy indicates that RHOQ344X is mislocalized to the inner segment and outer nuclear layers of rod photoreceptors in both RhoQ344X/+ and RhoQ344X/Q344X mice, confirming the essential role of the C-terminal VxPx motif in promoting OS delivery of rhodopsin. The mislocalization of RHOQ344X is associated with the concurrent mislocalization of wild-type rhodopsin in RhoQ344X/+ mice. To understand the global changes in proteostasis, we conducted quantitative proteomics analysis and found attenuated expression of rod-specific OS membrane proteins accompanying reduced expression of ciliopathy causative gene products, including constituents of BBSome and axonemal dynein subunit. Those studies unveil a novel negative feedback regulation involving ciliopathy-associated proteins. In this process, a defect in the trafficking signal leads to a reduced quantity of the trafficking apparatus, culminating in a widespread reduction in the transport of ciliary proteins.


Subject(s)
Disease Models, Animal , Gene Knock-In Techniques , Retinal Rod Photoreceptor Cells , Retinitis Pigmentosa , Rhodopsin , Animals , Rhodopsin/metabolism , Rhodopsin/genetics , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Mice , Retinal Rod Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/pathology , Cilia/metabolism , Cilia/pathology
3.
J Thromb Haemost ; 22(6): 1715-1726, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38508397

ABSTRACT

BACKGROUND: Protease-activated receptor 4 (PAR4) mediates thrombin signaling on platelets and other cells. Our recent structural studies demonstrated that a single nucleotide polymorphism in extracellular loop 3 and PAR4-P310L (rs2227376) leads to a hyporeactive receptor. OBJECTIVES: The goal of this study was to determine how the hyporeactive PAR4 variant in extracellular loop 3 impacts platelet function in vivo using a novel knock-in mouse model (PAR4-322L). METHODS: A point mutation was introduced into the PAR4 gene F2rl3 via CRISPR/Cas9 to create PAR4-P322L, the mouse homolog to human PAR4-P310L. Platelet response to PAR4 activation peptide (AYPGKF), thrombin, ADP, and convulxin was monitored by αIIbß3 integrin activation and P-selectin translocation using flow cytometry or platelet aggregation. In vivo responses were determined by the tail bleeding assay and the ferric chloride-induced carotid artery injury model. RESULTS: PAR4-P/L and PAR4-L/L platelets had a reduced response to AYPGKF and thrombin measured by P-selectin translocation or αIIbß3 activation. The response to ADP and convulxin was unchanged among genotypes. In addition, both PAR4-P/L and PAR4-L/L platelets showed a reduced response to thrombin in aggregation studies. There was an increase in the tail bleeding time for PAR4-L/L mice. The PAR4-P/L and PAR4-L/L mice both showed an extended time to arterial thrombosis. CONCLUSION: PAR4-322L significantly reduced platelet responsiveness to AYPGKF and thrombin, which is in agreement with our previous structural and cell signaling studies. In addition, PAR4-322L had prolonged arterial thrombosis time. Our mouse model provides a foundation to further evaluate the role of PAR4 in other pathophysiological contexts.


Subject(s)
Blood Platelets , Mice, Inbred C57BL , Platelet Aggregation , Platelet Glycoprotein GPIIb-IIIa Complex , Receptors, Thrombin , Thrombin , Animals , Blood Platelets/metabolism , Receptors, Thrombin/genetics , Receptors, Thrombin/metabolism , Thrombin/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Disease Models, Animal , Crotalid Venoms/pharmacology , Crotalid Venoms/toxicity , Adenosine Diphosphate/metabolism , Adenosine Diphosphate/pharmacology , P-Selectin/metabolism , P-Selectin/genetics , Point Mutation , Gene Knock-In Techniques , Signal Transduction , Thrombosis/genetics , Thrombosis/blood , Male , Chlorides , Mice , Platelet Activation , CRISPR-Cas Systems , Humans , Phenotype , Ferric Compounds , Oligopeptides , Lectins, C-Type , Receptors, Proteinase-Activated
4.
bioRxiv ; 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-38077081

ABSTRACT

Background: Protease activated receptor 4 (PAR4) mediates thrombin signaling on platelets and other cells. Our recent structural studies demonstrated a single nucleotide polymorphism in extracellular loop 3 (ECL3), PAR4-P310L (rs2227376) leads to a hypo-reactive receptor. Objectives: The goal of this study was to determine how the hypo-reactive PAR4 variant in ECL3 impacts platelet function in vivo using a novel knock-in mouse model (PAR4-322L). Methods: A point mutation was introduced into the PAR4 gene, F2rl3, via CRISPR/Cas9 to create PAR4-P322L, the mouse homolog to human PAR4-P310L. Platelet response to PAR4 activation peptide (AYPGKF), thrombin, ADP, and convulxin was monitored by αIIbß3 integrin activation and P-selectin translocation using flow cytometry or platelet aggregation. In vivo responses were determined by the tail bleeding assay and the ferric chloride-induced carotid artery injury model. Results: PAR4-P/L and PAR4-L/L platelets had a reduced response to AYPGKF and thrombin measured by P-selectin translocation or αIIbß3 activation. The response to ADP and convulxin was unchanged among genotypes. In addition, both PAR4-P/L and PAR4-L/L platelets showed a reduced response to thrombin in aggregation studies. There was an increase in the tail bleeding time for PAR4-L/L mice. The PAR4-P/L and PAR4-L/L mice both showed an extended time to arterial thrombosis. Conclusions: PAR4-322L significantly reduced platelet responsiveness to AYPGKF and thrombin, which is in agreement with our previous structural and cell signaling studies. In addition, PAR4-322L had prolonged arterial thrombosis time. Our mouse model provides a foundation to further evaluate the role of PAR4 in other pathophysiological contexts.

5.
Genes Dis ; 10(4): 1702-1713, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37397533

ABSTRACT

How PD-L1 expression is regulated in cancer is poorly understood. Here, we report that the ATP-binding activity of ERBB3 pseudokinase regulates PD-L1 gene expression in colorectal cancers (CRCs). ERBB3 is one of the four members of the EGF receptor family, all with protein tyrosine kinase domains. ERBB3 is a pseudokinase with a high binding affinity to ATP. We showed that ERBB3 ATP-binding inactivation mutant reduces tumorigenicity in genetically engineered mouse models and impairs xenograft tumor growth of CRC cell lines. The ERBB3 ATP-binding mutant cells dramatically reduce IFN-γ-induced PD-L1 expression. Mechanistically, ERBB3 regulates IFN-γ-induced PD-L1 expression through the IRS1-PI3K-PDK1-RSK-CREB signaling axis. CREB is the transcription factor that regulates PD-L1 gene expression in CRC cells. Knockin of a tumor-derived ERBB3 mutation located in the kinase domain sensitizes mouse colon cancers to anti-PD1 antibody therapy, suggesting that ERBB3 mutations could be predictive biomarkers for tumors amenable to immune checkpoint therapy.

6.
J Law Biosci ; 10(1): lsad003, 2023.
Article in English | MEDLINE | ID: mdl-36910719

ABSTRACT

A large and highly heterogeneous group of individuals conducts genetic and genomic research outside of traditional corporate and academic settings. They can be an important source of innovation, but their activities largely take place beyond the purview of existing regulatory systems for promoting safe and ethical practices. Historically the gene-targeting technology available for non-traditional genomic research has been limited, and therefore these activities have attracted little regulatory attention. New technologies such as CRISPR/Cas9, however, give nonconventional experimenters more extensive gene editing abilities at an unprecedented level of accessibility. The affordability and accessibility of these powerful technologies are raising questions about whether the current largely laissez-faire governance approach is adequate. This article recommends steps to enhance self-governance, including establishing umbrella organizations to represent community interests, creating a community IRB modelled on the DIYBio Ask a Safety Expert Service, and adopting an ethical obligation to report rogue experiments.

7.
Nat Commun ; 13(1): 1974, 2022 04 13.
Article in English | MEDLINE | ID: mdl-35418124

ABSTRACT

PI3Ks consist of p110 catalytic subunits and p85 regulatory subunits. PIK3CA, encoding p110α, is frequently mutated in human cancers. Most PIK3CA mutations are clustered in the helical domain or the kinase domain. Here, we report that p85ß disassociates from p110α helical domain mutant protein and translocates into the nucleus through a nuclear localization sequence (NLS). Nuclear p85ß recruits deubiquitinase USP7 to stabilize EZH1 and EZH2 and enhances H3K27 trimethylation. Knockout of p85ß or p85ß NLS mutant reduces the growth of tumors harboring a PIK3CA helical domain mutation. Our studies illuminate a novel mechanism by which PIK3CA helical domain mutations exert their oncogenic function. Finally, a combination of Alpelisib, a p110α-specific inhibitor, and an EZH inhibitor, Tazemetostat, induces regression of xenograft tumors harboring a PIK3CA helical domain mutation, but not tumors with either a WT PIK3CA or a PIK3CA kinase domain mutation, suggesting that the drug combination could be an effective therapeutic approach for PIK3CA helical domain mutant tumors.


Subject(s)
Neoplasms , Phosphatidylinositol 3-Kinases , Carcinogenesis/genetics , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Humans , Mutation , Neoplasms/drug therapy , Neoplasms/genetics , Oncogenes , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Ubiquitin-Specific Peptidase 7/genetics
8.
Cancer Res ; 80(21): 4815-4827, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32907836

ABSTRACT

PIK3CA encodes the p110α catalytic subunit of PI3K and is frequently mutated in human cancers, including ∼30% of colorectal cancer. Oncogenic mutations in PIK3CA render colorectal cancers more dependent on glutamine. Here we report that the glutaminase inhibitor CB-839 preferentially inhibits xenograft growth of PIK3CA-mutant, but not wild-type (WT), colorectal cancers. Moreover, the combination of CB-839 and 5-fluorouracil (5-FU) induces PIK3CA-mutant tumor regression in xenograft models. CB-839 treatment increased reactive oxygen species and caused nuclear translocation of Nrf2, which in turn upregulated mRNA expression of uridine phosphorylase 1 (UPP1). UPP1 facilitated the conversion of 5-FU to its active compound, thereby enhancing the inhibition of thymidylate synthase. Consistently, knockout of UPP1 abrogated the tumor inhibitory effect of combined CB-839 and 5-FU administration. A phase I clinical trial showed that the combination of CB-839 and capecitabine, a prodrug of 5-FU, was well tolerated at biologically-active doses. Although not designed to test efficacy, an exploratory analysis of the phase I data showed a trend that PIK3CA-mutant patients with colorectal cancer might derive greater benefit from this treatment strategy as compared with PIK3CA WT patients with colorectal cancer. These results effectively demonstrate that targeting glutamine metabolism may be an effective approach for treating patients with PIK3CA-mutant colorectal cancers and warrants further clinical evaluation. SIGNIFICANCE: Preclinical and clinical trial data suggest that the combination of CB-839 with capecitabine could serve as an effective treatment for PIK3CA-mutant colorectal cancers.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzeneacetamides/administration & dosage , Class I Phosphatidylinositol 3-Kinases/genetics , Colorectal Neoplasms/drug therapy , Fluorouracil/administration & dosage , Thiadiazoles/administration & dosage , Adult , Animals , Benzeneacetamides/adverse effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Dose-Response Relationship, Drug , Female , Fluorouracil/adverse effects , Humans , Male , Maximum Tolerated Dose , Mice , Middle Aged , Thiadiazoles/adverse effects , Xenograft Model Antitumor Assays
9.
Genome Biol ; 21(1): 202, 2020 08 10.
Article in English | MEDLINE | ID: mdl-32778141

ABSTRACT

BACKGROUND: The complex interspersed pattern of segmental duplications in humans is responsible for rearrangements associated with neurodevelopmental disease, including the emergence of novel genes important in human brain evolution. We investigate the evolution of LCR16a, a putative driver of this phenomenon that encodes one of the most rapidly evolving human-ape gene families, nuclear pore interacting protein (NPIP). RESULTS: Comparative analysis shows that LCR16a has independently expanded in five primate lineages over the last 35 million years of primate evolution. The expansions are associated with independent lineage-specific segmental duplications flanking LCR16a leading to the emergence of large interspersed duplication blocks at non-orthologous chromosomal locations in each primate lineage. The intron-exon structure of the NPIP gene family has changed dramatically throughout primate evolution with different branches showing characteristic gene models yet maintaining an open reading frame. In the African ape lineage, we detect signatures of positive selection that occurred after a transition to more ubiquitous expression among great ape tissues when compared to Old World and New World monkeys. Mouse transgenic experiments from baboon and human genomic loci confirm these expression differences and suggest that the broader ape expression pattern arose due to mutational changes that emerged in cis. CONCLUSIONS: LCR16a promotes serial interspersed duplications and creates hotspots of genomic instability that appear to be an ancient property of primate genomes. Dramatic changes to NPIP gene structure and altered tissue expression preceded major bouts of positive selection in the African ape lineage, suggestive of a gene undergoing strong adaptive evolution.


Subject(s)
Evolution, Molecular , Gene Duplication , Primates/genetics , Segmental Duplications, Genomic , Animals , Biodiversity , Brain , Chromosome Mapping , Chromosomes , Exons , Gene Fusion , Genome, Human , Genomic Instability , Hominidae , Humans , Phylogeny
10.
Nature ; 585(7825): 397-403, 2020 09.
Article in English | MEDLINE | ID: mdl-32610343

ABSTRACT

Mutations in PLP1, the gene that encodes proteolipid protein (PLP), result in failure of myelination and neurological dysfunction in the X-chromosome-linked leukodystrophy Pelizaeus-Merzbacher disease (PMD)1,2. Most PLP1 mutations, including point mutations and supernumerary copy variants, lead to severe and fatal disease. Patients who lack PLP1 expression, and Plp1-null mice, can display comparatively mild phenotypes, suggesting that PLP1 suppression might provide a general therapeutic strategy for PMD1,3-5. Here we show, using CRISPR-Cas9 to suppress Plp1 expression in the jimpy (Plp1jp) point-mutation mouse model of severe PMD, increased myelination and restored nerve conduction velocity, motor function and lifespan of the mice to wild-type levels. To evaluate the translational potential of this strategy, we identified antisense oligonucleotides that stably decrease the levels of Plp1 mRNA and PLP protein throughout the neuraxis in vivo. Administration of a single dose of Plp1-targeting antisense oligonucleotides in postnatal jimpy mice fully restored oligodendrocyte numbers, increased myelination, improved motor performance, normalized respiratory function and extended lifespan up to an eight-month end point. These results suggest that PLP1 suppression could be developed as a treatment for PMD in humans. More broadly, we demonstrate that oligonucleotide-based therapeutic agents can be delivered to oligodendrocytes in vivo to modulate neurological function and lifespan, establishing a new pharmaceutical modality for myelin disorders.


Subject(s)
Disease Models, Animal , Myelin Proteolipid Protein/deficiency , Pelizaeus-Merzbacher Disease/genetics , Pelizaeus-Merzbacher Disease/therapy , Animals , CRISPR-Cas Systems , Female , Gene Editing , Hypoxia/metabolism , Male , Mice , Mice, Mutant Strains , Motor Activity/genetics , Myelin Proteolipid Protein/genetics , Myelin Proteolipid Protein/metabolism , Myelin Sheath/metabolism , Oligodendroglia/metabolism , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/genetics , Pelizaeus-Merzbacher Disease/metabolism , Point Mutation , Respiratory Function Tests , Survival Analysis
11.
Sci Rep ; 9(1): 19180, 2019 12 16.
Article in English | MEDLINE | ID: mdl-31844152

ABSTRACT

Cancer cells in culture rely on glutamine as an anaplerotic substrate to replenish tricarboxylic acid (TCA) cycle intermediates that have been consumed. but it is uncertain whether cancers in vivo depend on glutamine for anaplerosis. Here, following in vivo infusions of [13C5]-glutamine in mice bearing subcutaneous colon cancer xenografts, we showed substantial amounts of infused [13C5]-glutamine enters the TCA cycle in the tumors. Consistent with our prior observation that colorectal cancers (CRCs) with oncogenic mutations in the phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic (PIK3CA) subunit are more dependent on glutamine than CRCs with wild type PIK3CA, labeling from glutamine to most TCA cycle intermediates was higher in PIK3CA-mutant subcutaneous xenograft tumors than in wild type PIK3CA tumors. Moreover, using orthotopic mouse colon tumors estalished from human CRC cells or patient-derived xenografts, we demonstrated substantial amounts of infused [13C5]-glutamine enters the TCA cycle in the tumors and tumors utilize anaplerotic glutamine to a greater extent than adjacent normal colon tissues. Similar results were seen in spontaneous colon tumors arising in genetically engineered mice. Our studies provide compelling evidence CRCs utilizes glutamine to replenish the TCA cycle in vivo, suggesting that targeting glutamine metabolism could be a therapeutic approach for CRCs, especially for PIK3CA-mutant CRCs.


Subject(s)
Citric Acid Cycle , Colorectal Neoplasms/metabolism , Glutamine/metabolism , Animals , Carbon Isotopes/blood , Class I Phosphatidylinositol 3-Kinases/genetics , Colorectal Neoplasms/blood , Female , Glutamine/blood , HCT116 Cells , Humans , Kinetics , Mice, Nude , Mutation/genetics , Subcutaneous Tissue/pathology , Substrate Specificity , Xenograft Model Antitumor Assays
12.
Genes Dis ; 6(2): 97-108, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31193992

ABSTRACT

In this review, we describe a path for translation of gene editing into therapy for cystic fibrosis (CF). Cystic fibrosis results from mutations in the CFTR gene, with one allele predominant in patient populations. This simple, genetic etiology makes gene editing appealing for treatment of this disease. There already have been success in applying this approach to cystic fibrosis in cell and animal models, although these advances have been modest in comparison to advances for other disease. Less than six years after its first demonstration in animals, CRISPR/Cas gene editing is in early clinical trials for several disorders. Most clinical trials, thus far, attempt to edit genes in cells of the blood lineages. The advantage of the blood is that the stem cells are known, can be isolated, edited, selected, expanded, and returned to the body. The likely next trials will be in the liver, which is accessible to many delivery methods. For cystic fibrosis, the biggest hurdle is to deliver editors to other, less accessible organs. We outline a path by which delivery can be improved. The translation of new therapies doesn't occur in isolation, and the development of gene editors is occurring as advances in gene therapy and small molecule therapeutics are being made. The advances made in gene therapy may help develop delivery vehicles for gene editing, although major improvements are needed. Conversely, the approval of effective small molecule therapies for many patients with cystic fibrosis will raise the bar for translation of gene editing.

13.
PLoS One ; 13(6): e0199573, 2018.
Article in English | MEDLINE | ID: mdl-29924856

ABSTRACT

Nonsense mutations are present in 10% of patients with CF, produce a premature termination codon in CFTR mRNA causing early termination of translation, and lead to lack of CFTR function. There are no currently available animal models which contain a nonsense mutation in the endogenous Cftr locus that can be utilized to test nonsense mutation therapies. In this study, we create a CF mouse model carrying the G542X nonsense mutation in Cftr using CRISPR/Cas9 gene editing. The G542X mouse model has reduced Cftr mRNA levels, demonstrates absence of CFTR function, and displays characteristic manifestations of CF mice such as reduced growth and intestinal obstruction. Importantly, CFTR restoration is observed in G542X intestinal organoids treated with G418, an aminoglycoside with translational readthrough capabilities. The G542X mouse model provides an invaluable resource for the identification of potential therapies of CF nonsense mutations as well as the assessment of in vivo effectiveness of these potential therapies targeting nonsense mutations.


Subject(s)
Codon, Nonsense , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/therapy , Disease Models, Animal , Genetic Therapy/methods , Mice, Transgenic , Animals , CRISPR-Cas Systems , Cells, Cultured , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Female , Intestines , Male , Mice, Inbred C57BL , Organoids/drug effects , Organoids/metabolism , RNA, Messenger/metabolism , Tissue Culture Techniques
14.
Nat Commun ; 7: 11971, 2016 06 20.
Article in English | MEDLINE | ID: mdl-27321283

ABSTRACT

Cancer cells often require glutamine for growth, thereby distinguishing them from most normal cells. Here we show that PIK3CA mutations reprogram glutamine metabolism by upregulating glutamate pyruvate transaminase 2 (GPT2) in colorectal cancer (CRC) cells, making them more dependent on glutamine. Compared with isogenic wild-type (WT) cells, PIK3CA mutant CRCs convert substantially more glutamine to α-ketoglutarate to replenish the tricarboxylic acid cycle and generate ATP. Mutant p110α upregulates GPT2 gene expression through an AKT-independent, PDK1-RSK2-ATF4 signalling axis. Moreover, aminooxyacetate, which inhibits the enzymatic activity of aminotransferases including GPT2, suppresses xenograft tumour growth of CRCs with PIK3CA mutations, but not with WT PIK3CA. Together, these data establish oncogenic PIK3CA mutations as a cause of glutamine dependency in CRCs and suggest that targeting glutamine metabolism may be an effective approach to treat CRC patients harbouring PIK3CA mutations.


Subject(s)
Adenocarcinoma/genetics , Class I Phosphatidylinositol 3-Kinases/genetics , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Glutamine/metabolism , Mutation , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Adenocarcinoma/drug therapy , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Adenosine Triphosphate/antagonists & inhibitors , Adenosine Triphosphate/biosynthesis , Aminooxyacetic Acid/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Citric Acid Cycle/drug effects , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Class I Phosphatidylinositol 3-Kinases/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , Enzyme Inhibitors/pharmacology , Female , Glutamine/antagonists & inhibitors , HCT116 Cells , HT29 Cells , Humans , Ketoglutaric Acids/antagonists & inhibitors , Ketoglutaric Acids/metabolism , Mice , Mice, Nude , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Signal Transduction , Transaminases/genetics , Transaminases/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
16.
Nat Med ; 17(6): 684-91, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21602802

ABSTRACT

The human skeleton is affected by mutations in low-density lipoprotein receptor-related protein 5 (LRP5). To understand how LRP5 influences bone properties, we generated mice with osteocyte-specific expression of inducible Lrp5 mutations that cause high and low bone mass phenotypes in humans. We found that bone properties in these mice were comparable to bone properties in mice with inherited mutations. We also induced an Lrp5 mutation in cells that form the appendicular skeleton but not in cells that form the axial skeleton; we observed that bone properties were altered in the limb but not in the spine. These data indicate that Lrp5 signaling functions locally, and they suggest that increasing LRP5 signaling in mature bone cells may be a strategy for treating human disorders associated with low bone mass, such as osteoporosis.


Subject(s)
Bone Density/genetics , LDL-Receptor Related Proteins/physiology , Alleles , Animals , Bone Density/physiology , Bone and Bones/metabolism , Bone and Bones/physiology , Female , Gene Knock-In Techniques , Gene Knockout Techniques , Genotype , LDL-Receptor Related Proteins/genetics , Low Density Lipoprotein Receptor-Related Protein-5 , Male , Mice , Mice, Knockout , Mice, Mutant Strains , Osteocytes/metabolism , Osteocytes/physiology , Serotonin/biosynthesis , Spine/metabolism , Spine/physiology , Tryptophan Hydroxylase/physiology
17.
Hum Mol Genet ; 19(4): 573-83, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-19933700

ABSTRACT

Huntington's disease (HD) is caused by expansion of the polymorphic polyglutamine segment in the huntingtin protein. Full-length huntingtin is thought to be a predominant HEAT repeat alpha-solenoid, implying a role as a facilitator of macromolecular complexes. Here we have investigated huntingtin's domain structure and potential intersection with epigenetic silencer polycomb repressive complex 2 (PRC2), suggested by shared embryonic deficiency phenotypes. Analysis of a set of full-length recombinant huntingtins, with different polyglutamine regions, demonstrated dramatic conformational flexibility, with an accessible hinge separating two large alpha-helical domains. Moreover, embryos lacking huntingtin exhibited impaired PRC2 regulation of Hox gene expression, trophoblast giant cell differentiation, paternal X chromosome inactivation and histone H3K27 tri-methylation, while full-length endogenous nuclear huntingtin in wild-type embryoid bodies (EBs) was associated with PRC2 subunits and was detected with trimethylated histone H3K27 at Hoxb9. Supporting a direct stimulatory role, full-length recombinant huntingtin significantly increased the histone H3K27 tri-methylase activity of reconstituted PRC2 in vitro, and structure-function analysis demonstrated that the polyglutamine region augmented full-length huntingtin PRC2 stimulation, both in Hdh(Q111) EBs and in vitro, with reconstituted PRC2. Knowledge of full-length huntingtin's alpha-helical organization and role as a facilitator of the multi-subunit PRC2 complex provides a novel starting point for studying PRC2 regulation, implicates this chromatin repressive complex in a neurodegenerative disorder and sets the stage for further study of huntingtin's molecular function and the impact of its modulatory polyglutamine region.


Subject(s)
Huntington Disease/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Animals , Disease Models, Animal , Female , Histones/genetics , Histones/metabolism , Humans , Huntingtin Protein , Huntington Disease/embryology , Huntington Disease/genetics , Male , Mice , Mice, Knockout , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Polycomb-Group Proteins , Protein Binding , Repressor Proteins/genetics , Sequence Homology, Amino Acid
18.
Methods Mol Med ; 129: 41-67, 2006.
Article in English | MEDLINE | ID: mdl-17085804

ABSTRACT

Diseases with a genetic basis can be modeled with knockout, knock-in, and conditional mutant gene-targeted mice. In the following, we provide detailed protocols for gene targeting. Gene targeting of embryonic stem cells can be accomplished by laboratories equipped for tissue culture. Alternatively, many gene-targeting services divide the work of targeting with a customer lab. In this collaborative situation, knowledge of the entire process helps ensure a successful outcome. The construction of chimeras for germ-line transmission is not described here, because this procedure is beyond the means of most laboratories, typically is provided by transgenic core facilities, and is best learned through hands-on demonstration.


Subject(s)
Disease Models, Animal , Gene Targeting/methods , Mice/genetics , Animals , Blotting, Southern , Cardiovascular Diseases/genetics , Chromosomes, Mammalian , DNA/isolation & purification , Embryonic Stem Cells , Genetic Vectors , Integrases , Mice, Knockout , Transfection/methods
19.
Dev Biol ; 296(1): 164-76, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16730693

ABSTRACT

Dorsal dermis and epaxial muscle have been shown to arise from the central dermomyotome in the chick. En1 is a homeobox transcription factor gene expressed in the central dermomyotome. We show by genetic fate mapping in the mouse that En1-expressing cells of the central dermomyotome give rise to dorsal dermis and epaxial muscle and, unexpectedly, to interscapular brown fat. Thus, the En1-expressing central dermomyotome normally gives rise to three distinct fates in mice. Wnt signals are important in early stages of dermomyotome development, but the signal that acts to specify the dermal fate has not been identified. Using a reporter transgene for Wnt signal transduction, we show that the En1-expressing cells directly underneath the surface ectoderm transduce Wnt signals. When the essential Wnt transducer beta-catenin is mutated in En1 cells, it results in the loss of Dermo1-expressing dorsal dermal progenitors and dermis. Conversely, when beta-catenin was activated in En1 cells, it induces Dermo1 expression in all cells of the En1 domain and disrupts muscle gene expression. Our results indicate that the mouse central dermomyotome gives rise to dermis, muscle, and brown fat, and that Wnt signalling normally instructs cells to select the dorsal dermal fate.


Subject(s)
Dermis/embryology , beta Catenin/metabolism , Adipose Tissue, Brown/embryology , Animals , Back , Homeodomain Proteins/physiology , Mice , Mice, Transgenic , Muscle, Skeletal/embryology , beta Catenin/physiology
20.
Nutr Metab (Lond) ; 2: 33, 2005 Nov 21.
Article in English | MEDLINE | ID: mdl-16300682

ABSTRACT

BACKGROUND: The metabolic function of PEPCK-C is not fully understood; deletion of the gene for the enzyme in mice provides an opportunity to fully assess its function. METHODS: The gene for the cytosolic form of phosphoenolpyruvate carboxykinase (GTP) (EC 4.1.1.32) (PEPCK-C) was deleted in mice by homologous recombination (PEPCK-C-/- mice) and the metabolic consequences assessed. RESULTS: PEPCK-C-/- mice became severely hypoglycemic by day two after birth and then died with profound hypoglycemia (12 mg/dl). The mice had milk in their stomachs at day two after birth and the administration of glucose raised the concentration of blood glucose in the mice but did not result in an increased survival. PEPCK-C-/- mice have two to three times the hepatic triglyceride content as control littermates on the second day after birth. These mice also had an elevation of lactate (2.5 times), beta-hydroxybutyrate (3 times) and triglyceride (50%) in their blood, as compared to control animals. On day two after birth, alanine, glycine, glutamine, glutamate, aspartate and asparagine were elevated in the blood of the PEPCK-C-/- mice and the blood urea nitrogen concentration was increased by 2-fold. The rate of oxidation of [2-14C]-acetate, and [5-14C]-glutamate to 14CO2 by liver slices from PEPCK-C-/- mice at two days of age was greatly reduced, as was the rate of fatty acid synthesis from acetate and glucose. As predicted by the lack of PEPCK-C, the concentration of malate in the livers of the PEPCK-C-/- mice was 10 times that of controls. CONCLUSION: We conclude that PEPCK-C is required not only for gluconeogenesis and glyceroneogenesis but also for cataplerosis (i.e. the removal of citric acid cycle anions) and that the failure of this process in the livers of PEPCK-C-/- mice results in a marked reduction in citric acid cycle flux and the shunting of hepatic lipid into triglyceride, resulting in a fatty liver.

SELECTION OF CITATIONS
SEARCH DETAIL
...