Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Nature ; 558(7711): E5, 2018 06.
Article in English | MEDLINE | ID: mdl-29849140

ABSTRACT

In Fig. 3c of this Letter, the the effects of CRISPR-Cas9-mediated deletion of NR3C1, TXNIP and CNR2 in patient-derived B-lineage leukaemia cells were shown. For curves depicting NR3C1 (left graph), data s for TXNIP (middle graph) were inadvertently plotted. This figure has been corrected online, and the original Fig. 3c is shown as Supplementary Information to this Amendment for transparency. The error does not affect the conclusions of the Letter. In addition, Source Data files have been added for the Figs. 1-4 and Extended Data Figs. 1-10 of the original Letter.

2.
Nature ; 542(7642): 479-483, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28192788

ABSTRACT

B-lymphoid transcription factors, such as PAX5 and IKZF1, are critical for early B-cell development, yet lesions of the genes encoding these transcription factors occur in over 80% of cases of pre-B-cell acute lymphoblastic leukaemia (ALL). The importance of these lesions in ALL has, until now, remained unclear. Here, by combining studies using chromatin immunoprecipitation with sequencing and RNA sequencing, we identify a novel B-lymphoid program for transcriptional repression of glucose and energy supply. Our metabolic analyses revealed that PAX5 and IKZF1 enforce a state of chronic energy deprivation, resulting in constitutive activation of the energy-stress sensor AMPK. Dominant-negative mutants of PAX5 and IKZF1, however, relieved this glucose and energy restriction. In a transgenic pre-B ALL mouse model, the heterozygous deletion of Pax5 increased glucose uptake and ATP levels by more than 25-fold. Reconstitution of PAX5 and IKZF1 in samples from patients with pre-B ALL restored a non-permissive state and induced energy crisis and cell death. A CRISPR/Cas9-based screen of PAX5 and IKZF1 transcriptional targets identified the products of NR3C1 (encoding the glucocorticoid receptor), TXNIP (encoding a glucose-feedback sensor) and CNR2 (encoding a cannabinoid receptor) as central effectors of B-lymphoid restriction of glucose and energy supply. Notably, transport-independent lipophilic methyl-conjugates of pyruvate and tricarboxylic acid cycle metabolites bypassed the gatekeeper function of PAX5 and IKZF1 and readily enabled leukaemic transformation. Conversely, pharmacological TXNIP and CNR2 agonists and a small-molecule AMPK inhibitor strongly synergized with glucocorticoids, identifying TXNIP, CNR2 and AMPK as potential therapeutic targets. Furthermore, our results provide a mechanistic explanation for the empirical finding that glucocorticoids are effective in the treatment of B-lymphoid but not myeloid malignancies. Thus, B-lymphoid transcription factors function as metabolic gatekeepers by limiting the amount of cellular ATP to levels that are insufficient for malignant transformation.


Subject(s)
B-Lymphocytes/metabolism , Energy Metabolism/genetics , Gene Expression Regulation, Neoplastic , Glucose/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Transcription Factors/metabolism , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Animals , B-Lymphocytes/drug effects , Carcinogenesis/genetics , Carrier Proteins/agonists , Carrier Proteins/metabolism , Cell Death , Chromatin Immunoprecipitation , Citric Acid Cycle , Disease Models, Animal , Female , Glucocorticoids/pharmacology , Glucocorticoids/therapeutic use , Humans , Ikaros Transcription Factor/metabolism , Mice , Mice, Transgenic , PAX5 Transcription Factor/deficiency , PAX5 Transcription Factor/genetics , PAX5 Transcription Factor/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Serine-Threonine Kinases/metabolism , Pyruvic Acid/metabolism , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/metabolism , Receptors, Glucocorticoid/metabolism , Sequence Analysis, RNA
3.
J Genet Couns ; 26(4): 716-727, 2017 Aug.
Article in English | MEDLINE | ID: mdl-27832509

ABSTRACT

This study explores the potential impact of the genetic counseling assistant (GCA) position on the efficiency of the genetic counseling field, evaluates attitudes regarding expansion of the genetic counseling field to include the GCA, and presents data on GCA endeavors and GCA job tasks as reported by GCAs, certified genetic counselors (CGCs), and program directors (PDs). Data on GCA roles and attitudes toward different aspects of the GCA position were collected via surveys of CGCs who have worked with GCAs, PDs who have and have not had experience with GCAs in their programs, and GCAs. We analyzed responses from 63 individuals: 27 PDs, 22 CGCs, and 14 GCAs. GCAs' impact on efficiency was calculated via internal analysis of genetic patient volume per genetic counselor within the University of Texas Southwestern (UTSW) patient database prior to, and since the addition of, a GCA to the practice. The response rates for PDs, CGCs, and GCAs were 27 %, 79 %, and 61 %, respectively. Every CGC stated the GCA increased their efficiency. CGCs with a GCA reported a 60 % average increase in patient volume. This figure was congruent with internal data from the UTSW cancer genetics program (58.5 % increase). Appropriate responsibilities for GCAs as reported by CGCs and PDs (>90 %) include: data entry, shipping tests, administrative tasks, research, and ordering supplies. Regarding GCAs delivering test results, there was response variation whether this should be a job duty: 42 % of CGCs agreed to GCAs delivering negative results to patients, compared to 22 % of program directors. Twenty-two percent of PDs expressed concern about the job title "Genetic Counseling Assistant." Ninety percent of CGCs felt that GCA was a career path to becoming a CGC, compared to 42 % of PDs. Eighty-three percent of GCAs who decided to apply to CGC graduate programs were accepted. We conclude the addition of a GCA to a genetic counseling practice contributes to increased efficiency and is one way to expand the reach of the profession.


Subject(s)
Allied Health Personnel/organization & administration , Attitude of Health Personnel , Delivery of Health Care/organization & administration , Genetic Counseling/organization & administration , Adult , Humans , Workforce
5.
J Genet Couns ; 23(6): 1034-44, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24916850

ABSTRACT

CancerGene Connect (CGC) is a web-based program that combines the collection of family and medical history, cancer risk assessment, psychosocial assessment, report templates, a result tracking system, and a patient follow up system. The performance of CGC was assessed in several ways: pre-appointment completion data analyzed for demographic and health variables; a time study to assess overall time per case and to compare the data entry by the genetic counselor compared to the patient, and a measured quality assessment of the program via observation and interview of patients. Prior to their appointment, 52.3% of 2,414 patients completed the online patient questionnaire section of CGC. There were significant differences in completion rates among racial and ethnic groups. County hospital patients were less likely to complete the questionnaire than insured patients (p < 0.0001); and likewise uninsured patients and patients with Medicare/Medicaid were less likely to complete the questionnaire than private patients (p < 0.0001). The average genetic counseling time per case was 82 min, with no significant differences whether the counselor or the patient completed CGC. CGC reduces genetic counselor time by approximately 14-46% compared to average time per case using traditional risk assessment and documentation methods previously reported. All surveyed users felt the questionnaire was easy to understand. CGC is an effective tool that streamlines workflow, and provides a standardized data collection tool that can be used to evaluate and improve the genetic counseling process.


Subject(s)
Genetic Counseling/methods , Genetic Testing/methods , Neoplasms/diagnosis , Neoplasms/genetics , Aged , Female , Humans , Internet , Male , Middle Aged , Professional-Patient Relations , Research Design , Risk Assessment , Surveys and Questionnaires , United States
6.
Cancer Cell ; 3(5): 471-82, 2003 May.
Article in English | MEDLINE | ID: mdl-12781365

ABSTRACT

The clathrin-associated protein, Huntingtin Interacting Protein 1 (HIP1), is overexpressed in multiple human epithelial tumors. Here, we report that HIP1 is a novel oncoprotein that transforms cells. HIP1-transformed cells, in contrast to RasV12-transformed cells, have dysregulation of multiple receptors involved in clathrin trafficking. Examples include upregulation of the epidermal growth factor receptor (EGFR) and the transferrin receptor. Furthermore, accumulation of transferrin and EGF in the HIP1-transformed cells was increased, and breast tumors that had EGFR expressed also had HIP1 upregulated. Thus, HIP1 overexpression promotes tumor formation and is associated with a general alteration in receptor trafficking. HIP1 is the first endocytic protein to be directly implicated in tumor formation.


Subject(s)
Carrier Proteins/genetics , DNA-Binding Proteins , Animals , Apoptosis , Blotting, Western , Cell Cycle , Cell Division , Cell Line, Transformed , Cell Transformation, Neoplastic , DNA, Complementary/metabolism , Endocytosis , ErbB Receptors/metabolism , Flow Cytometry , Humans , Mice , Mice, Nude , Microscopy, Confocal , Models, Biological , NIH 3T3 Cells , Phosphorylation , Promoter Regions, Genetic , Protein Transport , Time Factors , Transferrin/metabolism
7.
JCI Insight ; 7(24)2022 12 22.
Article in English | MEDLINE | ID: mdl-36346676

ABSTRACT

Mutations in the BRCA1 tumor suppressor gene, such as 5382insC (BRCA1insC), give carriers an increased risk for breast, ovarian, prostate, and pancreatic cancers. We have previously reported that, in mice, Brca1 deficiency in the hematopoietic system leads to pancytopenia and, as a result, early lethality. We explored the cellular consequences of Brca1-null and BRCA1insC alleles in combination with Trp53 deficiency in the murine hematopoietic system. We found that Brca1 and Trp53 codeficiency led to a highly penetrant erythroproliferative disorder that is characterized by hepatosplenomegaly and by expanded megakaryocyte erythroid progenitor (MEP) and immature erythroid blast populations. The expanded erythroid progenitor populations in both BM and spleen had the capacity to transmit the disease into secondary mouse recipients, suggesting that Brca1 and Trp53 codeficiency provides a murine model of hematopoietic neoplasia. This Brca1/Trp53 model replicated Poly (ADP-ribose) polymerase (PARP) inhibitor olaparib sensitivity seen in existing Brca1/Trp53 breast cancer models and had the benefits of monitoring disease progression and drug responses via peripheral blood analyses without sacrificing experimental animals. In addition, this erythroid neoplasia developed much faster than murine breast cancer, allowing for increased efficiency of future preclinical studies.


Subject(s)
Antineoplastic Agents , Neoplasms , Animals , Mice , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Poly(ADP-ribose) Polymerases/genetics , Mutation
8.
J Clin Invest ; 118(7): 2459-70, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18535670

ABSTRACT

Huntingtin interacting protein 1 related (Hip1r) is an F-actin- and clathrin-binding protein involved in vesicular trafficking. In this study, we demonstrate that Hip1r is abundantly expressed in the gastric parietal cell, predominantly localizing with F-actin to canalicular membranes. Hip1r may provide a critical function in vivo, as demonstrated by extensive changes to parietal cells and the gastric epithelium in Hip1r-deficient mice. Electron microscopy revealed abnormal apical canalicular membranes and loss of tubulovesicles in mutant parietal cells, suggesting that Hip1r is necessary for the normal trafficking of these secretory membranes. Accordingly, acid secretory dynamics were altered in mutant parietal cells, with enhanced activation and acid trapping, as measured in isolated gastric glands. At the whole-organ level, gastric acidity was reduced in Hip1r-deficient mice, and the gastric mucosa was grossly transformed, with fewer parietal cells due to enhanced apoptotic cell death and glandular hypertrophy associated with cellular transformation. Hip1r-deficient mice had increased expression of the gastric growth factor gastrin, and mice mutant for both gastrin and Hip1r exhibited normalization of both proliferation and gland height. Taken together, these studies demonstrate that Hip1r plays a significant role in gastric physiology, mucosal architecture, and secretory membrane dynamics in parietal cells.


Subject(s)
DNA-Binding Proteins/physiology , Parietal Cells, Gastric/physiology , Secretory Vesicles/physiology , Adaptor Proteins, Signal Transducing , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Chief Cells, Gastric/metabolism , Chief Cells, Gastric/pathology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gastric Acid/metabolism , Gastric Acidity Determination , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Gastrins/blood , Gastrins/genetics , Gene Expression/drug effects , H(+)-K(+)-Exchanging ATPase/genetics , H(+)-K(+)-Exchanging ATPase/metabolism , Histamine/pharmacology , Intrinsic Factor/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Microfilament Proteins , Microscopy, Electron , Parathyroid Hormone-Related Protein/metabolism , Parietal Cells, Gastric/drug effects , Rabbits , Ranitidine/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Secretory Vesicles/ultrastructure
9.
Article in English | MEDLINE | ID: mdl-32923914

ABSTRACT

PURPOSE: Hereditary cancer genetic testing can inform personalized medical management for individuals at increased cancer risk. However, many variants in cancer predisposition genes are individually rare, and traditional tools may be insufficient to evaluate pathogenicity. This analysis presents data on variant classification and reclassification over a 20-year period. PATIENTS AND METHODS: This is a retrospective analysis of > 1.9 million individuals who received hereditary cancer genetic testing from a single clinical laboratory (March 1997 to December 2017). Variant classification included review of evidence from traditional tools (eg, population frequency databases, literature) and laboratory-developed tools (eg, novel statistical methods, in-house RNA analysis) by a multidisciplinary expert committee. Variants may have been reclassified more than once and with more than one line of evidence. RESULTS: In this time period, 62,842 unique variants were observed across 25 cancer predisposition genes, and 2,976 variants were reclassified. Overall, 82.1% of reclassification events were downgrades (eg, variant of uncertain significance [VUS] to benign), and 17.9% were upgrades (eg, VUS to pathogenic). Among reclassified variants, 82.8% were initially classified as VUS, and 47.5% were identified in ≤ 20 individuals (allele frequency ≤ 0.001%). Laboratory-developed tools were used in 72.3% of variant reclassification events, which affected > 600,000 individuals. More than 1.3 million patients were identified as carrying a variant that was reclassified within this 20-year time period. CONCLUSION: The variant classification program used by the laboratory evaluated here enabled the reclassification of variants that were individually rare. Laboratory-developed tools were a key component of this program and were used in the majority of reclassifications. This demonstrates the importance of using robust and novel tools to reclassify rare variants to appropriately inform personalized medical management.

10.
Cancer Res ; 67(8): 3609-15, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17440072

ABSTRACT

Huntingtin interacting protein 1 (HIP1) is a multidomain oncoprotein whose expression correlates with increased epidermal growth factor receptor (EGFR) levels in certain tumors. For example, HIP1-transformed fibroblasts and HIP1-positive breast cancers have elevated EGFR protein levels. The combined association of HIP1 with huntingtin, the protein that is mutated in Huntington's disease, and the known overexpression of EGFR in glial brain tumors prompted us to explore HIP1 expression in a group of patients with different types of brain cancer. We report here that HIP1 is overexpressed with high frequency in brain cancers and that this overexpression correlates with EGFR and platelet-derived growth factor beta receptor expression. Furthermore, serum samples from patients with brain cancer contained anti-HIP1 antibodies more frequently than age-matched brain cancer-free controls. Finally, we report that HIP1 physically associates with EGFR and that this association is independent of the lipid, clathrin, and actin interacting domains of HIP1. These findings suggest that HIP1 may up-regulate or maintain EGFR overexpression in primary brain tumors by directly interacting with the receptor. This novel HIP1-EGFR interaction may work with or independent of HIP1 modulation of EGFR degradation via clathrin-mediated membrane trafficking pathways. Further investigation of HIP1 function in brain cancer biology and validation of its use as a prognostic or predictive brain tumor marker are now warranted.


Subject(s)
Brain Neoplasms/metabolism , DNA-Binding Proteins/metabolism , ErbB Receptors/metabolism , Actins/metabolism , Antibodies, Neoplasm/blood , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/metabolism , Brain/metabolism , Brain Neoplasms/immunology , Clathrin/metabolism , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/immunology , ErbB Receptors/biosynthesis , Humans , Immunoprecipitation , Lipid Metabolism , Protein Binding
11.
Cancer Res ; 67(18): 8923-31, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17875735

ABSTRACT

Huntingtin interacting protein 1 (HIP1) is an inositol lipid, clathrin, and actin binding protein that is overexpressed in a variety of epithelial malignancies. Here, we report for the first time that HIP1 is elevated in non-Hodgkin's and Hodgkin's lymphomas and that patients with lymphoid malignancies frequently had anti-HIP1 antibodies in their serum. Moreover, p53-deficient mice with B-cell lymphomas were 13 times more likely to have anti-HIP1 antibodies in their serum than control mice. Furthermore, transgenic overexpression of HIP1 was associated with the development of lymphoid neoplasms. The HIP1 protein was induced by activation of the nuclear factor-kappaB pathway, which is frequently activated in lymphoid malignancies. These data identify HIP1 as a new marker of lymphoid malignancies that contributes to the transformation of lymphoid cells in vivo.


Subject(s)
DNA-Binding Proteins/biosynthesis , Lymphoma/metabolism , Animals , Antibodies, Neoplasm/blood , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Disease Models, Animal , Hodgkin Disease/genetics , Hodgkin Disease/metabolism , Humans , Lymphoma/genetics , Lymphoma/pathology , Lymphoma, Mantle-Cell/genetics , Lymphoma, Mantle-Cell/metabolism , Lymphoma, Mantle-Cell/pathology , Lymphoma, Non-Hodgkin/genetics , Lymphoma, Non-Hodgkin/immunology , Lymphoma, Non-Hodgkin/metabolism , Lymphoma, Non-Hodgkin/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/metabolism , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
12.
iScience ; 19: 809-820, 2019 Sep 27.
Article in English | MEDLINE | ID: mdl-31499338

ABSTRACT

Both BRCA1 and CREBBP are tumor suppressor genes that are important for hematopoiesis. We have previously shown that mouse Brca1 is essential for hematopoietic stem cell (HSC) viability. In contrast to Brca1 deficiency, which results in pancytopenia, we report here that Crebbp deficiency results in myeloproliferation associated with an increase of splenic HSCs as well as a lethal systemic inflammatory disorder (LD50 = 86 days). To investigate the interaction of these two proteins in hematopoiesis, we generated double Crebbp/Brca1 knockout mice (DKOs). To our surprise, DKOs had accelerated bone marrow failure compared with Brca1-deficient mice and this was associated with an even shorter lifespan (LD50 = 88.5 versus 33 days). Furthermore, Crebbp or Brca1 heterozygosity influenced the hematopoietic phenotype associated with complete deficiency of Brca1 or Crebbp, respectively. We also observed lower BRCA1 protein levels in hematopoietic tissues when CREBBP is absent. Collectively, these data suggest Crebbp and Brca1 functionally interact to maintain normal hematopoiesis.

13.
Mol Genet Genomic Med ; 7(9): e886, 2019 09.
Article in English | MEDLINE | ID: mdl-31347304

ABSTRACT

BACKGROUND: There are no national guidelines for the management of patients with a family history consistent with Lynch syndrome (LS) but a negative genetic test. To determine current management practices, genetic counselors' (GCs) recommendations were assessed. METHODS: A survey of GCs using five hypothetical pedigrees was posted to National Society of Genetic Counselors (NSGC) discussion forums. Descriptive statistics were used. RESULTS: One-hundred and fifteen surveys were completed. A pedigree with a first-degree relative (FDR) with early-onset colorectal cancer (CRC) and a family history of CRC and endometrial cancer (EC) prompted 83% (n = 95) of respondents to recommend early and frequent colonoscopies, based on family history. When the CRCs and ECs occurred in family members removed from the proband, 96% (n = 110) of GCs said they would screen based on family history. However, only 52% (n = 60) suggested CRC screening should begin earlier and occur more often, and 43% (n = 50) suggested CRC screening should follow standard age and frequency guidelines. CONCLUSION: Concordance of opinion among GCs for the management of patients with negative genetic test results exists when FDRs are affected. However, when affected relatives are more distant, GCs disagreed on screening recommendations. These data suggest a need for guidelines for patients with a family history of cancer and a negative genetic test.


Subject(s)
Colonic Neoplasms/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Endometrial Neoplasms/genetics , Family , Genetic Counseling , Pedigree , Adult , Female , Humans , Male
14.
Mol Cell Biol ; 38(23)2018 12 01.
Article in English | MEDLINE | ID: mdl-30224518

ABSTRACT

Deficiency of huntingtin-interacting protein 1 (Hip1) results in degenerative phenotypes. Here we generated a Hip1 deficiency allele where a floxed transcriptional stop cassette and a human HIP1 cDNA were knocked into intron 1 of the mouse Hip1 locus. CMV-Cre-mediated germ line excision of the stop cassette resulted in expression of HIP1 and rescue of the Hip1 knockout phenotype. Mx1-Cre-mediated excision led to HIP1 expression in spleen, kidney and liver, and also rescued the phenotype. In contrast, hGFAP-Cre-mediated, brain-specific HIP1 expression did not rescue the phenotype. Metabolomics and microarrays of several Hip1 knockout tissues identified low phosphocholine (PC) levels and low glycerophosphodiester phosphodiesterase domain containing 3 (Gdpd3) gene expression. Since Gdpd3 has lysophospholipase D activity that results in the formation of choline, a precursor of PC, Gdpd3 downregulation could lead to the low PC levels. To test whether Gdpd3 contributes to the Hip1 deficiency phenotype, we generated Gdpd3 knockout mice. Double knockout of Gdpd3 and Hip1 worsened the Hip1 phenotype. This suggests that Gdpd3 compensates for Hip1 loss. More-detailed knowledge of how Hip1 deficiency leads to low PC will improve our understanding of HIP1 in choline metabolism in normal and disease states.


Subject(s)
DNA-Binding Proteins/deficiency , Endocytosis/genetics , Phosphoric Diester Hydrolases/genetics , Phosphorylcholine/metabolism , Animals , DNA, Complementary/genetics , Down-Regulation/genetics , Gene Expression/genetics , Humans , Introns/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype
15.
J Clin Invest ; 110(3): 351-60, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12163454

ABSTRACT

Huntingtin-interacting protein 1 (HIP1) is a cofactor in clathrin-mediated vesicle trafficking. It was first implicated in cancer biology as part of a chromosomal translocation in leukemia. Here we report that HIP1 is expressed in prostate and colon tumor cells, but not in corresponding benign epithelia. The relationship between HIP1 expression in primary prostate cancer and clinical outcomes was evaluated with tissue microarrays. HIP1 expression was significantly associated with prostate cancer progression and metastasis. Conversely, primary prostate cancers lacking HIP1 expression consistently showed no progression after radical prostatectomy. In addition, the expression of HIP1 was elevated in prostate tumors from the transgenic mouse model of prostate cancer (TRAMP). At the molecular level, expression of a dominant negative mutant of HIP1 led to caspase-9-dependent apoptosis, suggesting that HIP1 is a cellular survival factor. Thus, HIP1 may play a role in tumorigenesis by allowing the survival of precancerous or cancerous cells. HIP1 might accomplish this via regulation of clathrin-mediated trafficking, a fundamental cellular pathway that has not previously been associated with tumorigenesis. HIP1 represents a putative prognostic factor for prostate cancer and a potential therapy target in prostate as well as colon cancers.


Subject(s)
Carrier Proteins/genetics , Colonic Neoplasms/genetics , DNA-Binding Proteins , Gene Expression , Prostatic Neoplasms/genetics , Animals , Apoptosis , Biomarkers, Tumor/genetics , Carrier Proteins/biosynthesis , Caspase 9 , Caspases/metabolism , Cell Survival , Disease Models, Animal , Epithelial Cells/metabolism , Humans , Male , Mice , Prostatic Neoplasms/pathology , Prostatic Neoplasms/physiopathology , Tumor Cells, Cultured
16.
Mol Cell Biol ; 24(10): 4329-40, 2004 May.
Article in English | MEDLINE | ID: mdl-15121852

ABSTRACT

In mice and humans, there are two known members of the Huntingtin interacting protein 1 (HIP1) family, HIP1 and HIP1-related (HIP1r). Based on structural and functional data, these proteins participate in the clathrin trafficking network. The inactivation of Hip1 in mice leads to spinal, hematopoietic, and testicular defects. To investigate the biological function of HIP1r, we generated a Hip1r mutant allele in mice. Hip1r homozygous mutant mice are viable and fertile without obvious morphological abnormalities. In addition, embryonic fibroblasts derived from these mice do not have gross abnormalities in survival, proliferation, or clathrin trafficking pathways. Altogether, this demonstrates that HIP1r is not necessary for normal development of the embryo or for normal adulthood and suggests that HIP1 or other functionally related members of the clathrin trafficking network can compensate for HIP1r absence. To test the latter, we generated mice deficient in both HIP1 and HIP1r. These mice have accelerated development of abnormalities seen in Hip1 -deficient mice, including kypholordosis and growth defects. The severity of the Hip1r/Hip1 double-knockout phenotype compared to the Hip1 knockout indicates that HIP1r partially compensates for HIP1 function in the absence of HIP1 expression, providing strong evidence that HIP1 and HIP1r have overlapping roles in vivo.


Subject(s)
Carrier Proteins/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Dwarfism/genetics , Microfilament Proteins/genetics , Spine/abnormalities , Adaptor Proteins, Signal Transducing , Animals , Base Sequence , Chromosome Mapping , DNA/genetics , Dwarfism/pathology , Endocytosis/genetics , ErbB Receptors/metabolism , Female , Gene Expression , Humans , Male , Mice , Mice, Knockout , Phenotype , Pregnancy , Tissue Distribution , Vesicular Transport Proteins
17.
Cancer Res ; 65(10): 4126-33, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15899803

ABSTRACT

Huntingtin-interacting protein 1 (HIP1) is frequently overexpressed in prostate cancer. HIP1 is a clathrin-binding protein involved in growth factor receptor trafficking that transforms fibroblasts by prolonging the half-life of growth factor receptors. In addition to human cancers, HIP1 is also overexpressed in prostate tumors from the transgenic adenocarcinoma of the mouse prostate (TRAMP) mouse model. Here we provide evidence that HIP1 plays an important role in mouse tumor development, as tumor formation in the TRAMP mice was impaired in the Hip1null/null background. In addition, we report that autoantibodies to HIP1 developed in the sera of TRAMP mice with prostate cancer as well as in the sera from human prostate cancer patients. This led to the development of an anti-HIP1 serum test in humans that had a similar sensitivity and specificity to the anti-alpha-methylacyl CoA racemase (AMACR) and prostate-specific antigen tests for prostate cancer and when combined with the anti-AMACR test yielded a specificity of 97%. These data suggest that HIP1 plays a functional role in tumorigenesis and that a positive HIP1 autoantibody test may be an important serum marker of prostate cancer.


Subject(s)
Adenocarcinoma/immunology , Autoantibodies/blood , DNA-Binding Proteins/immunology , Prostatic Neoplasms/immunology , Adenocarcinoma/blood , Adenocarcinoma/genetics , Animals , Autoantibodies/immunology , Cohort Studies , DNA-Binding Proteins/genetics , Enzyme-Linked Immunosorbent Assay , Humans , Immunoblotting , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Prostatic Neoplasms/blood , Prostatic Neoplasms/genetics
18.
Cell Rep ; 18(4): 947-960, 2017 01 24.
Article in English | MEDLINE | ID: mdl-28122244

ABSTRACT

BRCA1 is a well-known DNA repair pathway component and a tissue-specific tumor suppressor. However, its role in hematopoiesis is uncertain. Here, we report that a cohort of patients heterozygous for BRCA1 mutations experienced more hematopoietic toxicity from chemotherapy than those with BRCA2 mutations. To test whether this reflects a requirement for BRCA1 in hematopoiesis, we generated mice with Brca1 mutations in hematopoietic cells. Mice homozygous for a null Brca1 mutation in the embryonic hematopoietic system (Vav1-iCre;Brca1F22-24/F22-24) developed hematopoietic defects in early adulthood that included reduced hematopoietic stem cells (HSCs). Although mice homozygous for a huBRCA1 knockin allele (Brca1BRCA1/BRCA1) were normal, mice with a mutant huBRCA1/5382insC allele and a null allele (Mx1-Cre;Brca1F22-24/5382insC) had severe hematopoietic defects marked by a complete loss of hematopoietic stem and progenitor cells. Our data show that Brca1 is necessary for HSC maintenance and normal hematopoiesis and that distinct mutations lead to different degrees of hematopoietic dysfunction.


Subject(s)
BRCA1 Protein/genetics , Hematopoietic Stem Cells/metabolism , Adult , Aged , Alleles , Animals , BRCA1 Protein/deficiency , BRCA1 Protein/metabolism , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Blood Cell Count , Cell Differentiation/drug effects , Cell Line , Cyclophosphamide/pharmacology , Female , Gene Knock-In Techniques , Hematopoiesis/drug effects , Hematopoietic Stem Cells/cytology , Hemoglobins/analysis , Humans , Lymphocytes/cytology , Lymphocytes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Mutagenesis , Pancytopenia/etiology , Pancytopenia/mortality , Pancytopenia/pathology , Young Adult
19.
Trends Mol Med ; 10(4): 194-9, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15059611

ABSTRACT

During recent years, alterations in proteins of the endocytic pathway have been associated with tumors. Disrupted regulation of the endocytic pathway is a relatively unstudied mechanism of tumorigenesis, which can concomitantly disrupt several different signaling pathways to affect growth, differentiation and survival. Several endocytic proteins have been identified, either as part of tumor-associated translocations or to have the ability to transform cells. Here, we summarize the information known about huntingtin interacting protein 1 (HIP1), an endocytic protein with transforming properties that is involved in a cancer-causing translocation and which is overexpressed in a variety of human cancers. We describe the known normal functions of HIP1 in endocytosis and receptor trafficking, the evidence for its role as an oncoprotein and how HIP1 might be altered to promote tumorigenesis.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation, Neoplastic , Neoplasms/metabolism , Alleles , Biological Transport , Cell Survival , Cell Transformation, Neoplastic , DNA-Binding Proteins/metabolism , Endocytosis , Humans , Models, Biological , Protein Structure, Tertiary , Protein Transport
20.
Cold Spring Harb Mol Case Stud ; 1(1): a000414, 2015 Oct.
Article in English | MEDLINE | ID: mdl-27148568

ABSTRACT

When a doctor orders a genetic test, patients assume that the test will yield a useful result to guide how their physicians take care of them. That assumption is frequently correct, but not always. Until recently, a genetic test only interrogated the sequence of one or two genes. Now, DNA-sequencing technologies are so fast and cheap that they have enabled clinicians to sequence panels of genes that may or may not be relevant to the patient's condition. The technology has outpaced our ability to interpret the results. Connecting approval of clinical tests to data sharing could help close this gap.

SELECTION OF CITATIONS
SEARCH DETAIL