Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Cell Rep ; 17(1): 221-232, 2016 09 27.
Article in English | MEDLINE | ID: mdl-27681433

ABSTRACT

Microvascular endothelial cells maintain a tight barrier to prevent passage of plasma and circulating immune cells into the extravascular tissue compartment, yet endothelial cells respond rapidly to vasoactive substances, including thrombin, allowing transient paracellular permeability. This response is a cornerstone of acute inflammation, but the mechanisms responsible are still incompletely understood. Here, we demonstrate that thrombin triggers MALT1 to proteolytically cleave cylindromatosis (CYLD). Fragmentation of CYLD results in microtubule disruption and a cascade of events leading to endothelial cell retraction and an acute permeability response. This finding reveals an unexpected role for the MALT1 protease, which previously has been viewed mostly as a driver of pro-inflammatory NF-κB signaling in lymphocytes. Thus, MALT1 not only promotes immune cell activation but also acutely regulates endothelial cell biology, actions that together facilitate tissue inflammation. Pharmacologic inhibition of MALT1 may therefore have synergistic impact by targeting multiple disparate steps in the overall inflammatory response.


Subject(s)
Caspases/immunology , Cysteine Endopeptidases/immunology , Endothelial Cells/drug effects , Microtubules/drug effects , Neoplasm Proteins/immunology , Thrombin/pharmacology , Animals , Biological Transport , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/immunology , Caspases/genetics , Cell Line , Cysteine Endopeptidases/genetics , Deubiquitinating Enzyme CYLD , Endothelial Cells/cytology , Endothelial Cells/immunology , Gene Expression Regulation , I-kappa B Kinase/genetics , I-kappa B Kinase/immunology , Mice , Mice, Transgenic , Microtubules/ultrastructure , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , NF-kappa B/genetics , NF-kappa B/immunology , Neoplasm Proteins/genetics , Permeability/drug effects , Primary Cell Culture , Receptor, PAR-1/genetics , Receptor, PAR-1/immunology , Signal Transduction , Thrombin/metabolism
2.
Hypertension ; 64(5): 1032-9, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25185127

ABSTRACT

Angiotensin (Ang) II is a potent mediator of both hypertension and cardiac damage; however, the mechanisms by which this occur remain unclear. B-cell lymphoma/leukemia 10 (Bcl10) is a member of the CBM signalosome, which links Ang II and nuclear factor-κB signaling. We hypothesized that Bcl10 is pivotal in the pathogenesis of Ang II-induced cardiac damage. Ang II infusion in mice lacking Bcl10 resulted in reduced cardiac fibrosis, less cellular infiltration, and improved arrhythmogenic electric remodeling, despite a similar degree of hypertension or cardiac hypertrophy. Adoptive transfer of bone marrow (BM), whereby Bcl10 knockout or wildtype BM was transferred to their opposite genotype recipients, revealed the dual importance of Bcl10 within both cardiac and immune cells. Loss of Bcl10 in cardiac cells resulted in reduced expression of genes important for the adhesion and recruitment of immune cells. In vitro experiments demonstrated that adhesion of monocytes to Ang II-treated endothelial cells also required Bcl10. Additionally, Bcl10 deficiency in macrophages reduced their intrinsic migratory ability. To address the role of BM-derived fibroblasts in the formation of cardiac fibrosis, we explored whether Bcl10 is also important for the infiltration of BM-derived (myo)fibroblasts into the heart. The transfer of green fluorescent protein positive wildtype BM into Bcl10 knockout recipient mice revealed a reduced number of noncardiac (myo)fibroblasts compared with those wildtype recipients. Our results demonstrate the significant role of Bcl10 in multiple cell types important for the generation of Ang II-induced cardiac damage and electric remodeling and may provide a new avenue for therapeutic intervention.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Angiotensin II/adverse effects , Atrial Remodeling/physiology , Heart Diseases/chemically induced , Heart Diseases/physiopathology , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , B-Cell CLL-Lymphoma 10 Protein , Cell Adhesion/physiology , Cell Movement/physiology , Cells, Cultured , Disease Models, Animal , Endothelium, Vascular/pathology , Fibrosis , Heart Diseases/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/pathology , Myocardium/metabolism , Myocardium/pathology , NF-kappa B/metabolism
3.
Cell Rep ; 5(1): 51-60, 2013 Oct 17.
Article in English | MEDLINE | ID: mdl-24095735

ABSTRACT

Chronic myeloid leukemia (CML) and some acute lymphoblastic leukemias are characterized by the t(9;22) chromosome, which encodes the BCR/ABL oncogene. Multiple mouse models of CML express BCR/ABL at high levels from non-Bcr promoters, resulting in the development of leukemias. In contrast, a significant fraction of healthy humans have been found to have BCR/ABL-positive hematopoietic cells. To bridge the gap between the information derived from current mouse models and nonleukemic humans with the BCR/ABL oncogene, we generated a knockin model with BCR/ABL p210 expressed from the Bcr locus. Unlike previous models, expression of BCR/ABL from the knockin allele did not induce leukemia. BCR/ABL mutant cells did exhibit favorable bone marrow engraftment compared to control cells. These data suggest that BCR/ABL expression alone is insufficient to induce disease. This model allows for inducible spatial and temporal control of BCR/ABL expression for analysis of early steps in the pathogenesis of BCR/ABL-expressing leukemias.


Subject(s)
Bone Marrow Transplantation/methods , Fusion Proteins, bcr-abl/biosynthesis , Fusion Proteins, bcr-abl/genetics , Alleles , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Fusion Proteins, bcr-abl/analysis , Gene Expression Regulation , Gene Knock-In Techniques , Humans , Leukemia, Myeloid/genetics , Leukemia, Myeloid/metabolism , Mice , Mice, Transgenic
4.
Cell Rep ; 1(5): 444-52, 2012 May 31.
Article in English | MEDLINE | ID: mdl-22708078

ABSTRACT

Excess serum free fatty acids (FFAs) are fundamental to the pathogenesis of insulin resistance. With high-fat feeding, FFAs activate NF-kB in target tissues, initiating negative crosstalk with insulin signaling. However, the mechanisms underlying FFA-dependent NF-kB activation remain unclear. Here, we demonstrate that the saturated FA, palmitate, requires Bcl10 for NF-kB activation in hepatocytes. Uptake of palmitate, metabolism to diacylglycerol, and subsequent activation of protein kinase C (PKC) appear to mechanistically link palmitate with Bcl10, known as a central component of a signaling complex that, along with CARMA3 and MALT1, activates NF-kB downstream of selected cell surface receptors. Consequently, Bcl10-deficient mice are protected from hepatic NF-kB activation and insulin resistance following brief high-fat diet, suggesting that Bcl10 plays a major role in the metabolic consequences of acute overnutrition. Surprisingly, while CARMA3 also participates in the palmitate response, MALT1 is completely dispensable, thereby revealing an apparent nonclassical role for Bcl10 in NF-kB signaling.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carcinoma, Hepatocellular/metabolism , Fatty Acids/pharmacology , Hepatocytes/metabolism , Insulin Resistance/physiology , Liver Neoplasms/metabolism , NF-kappa B/metabolism , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , B-Cell CLL-Lymphoma 10 Protein , CARD Signaling Adaptor Proteins/metabolism , Carcinoma, Hepatocellular/pathology , Caspases/metabolism , Cell Line, Tumor , Cells, Cultured , Diet, High-Fat , Hepatocytes/drug effects , Hepatocytes/pathology , Humans , Liver Neoplasms/pathology , Male , Mice , Mice, Knockout , Models, Animal , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , Neoplasm Proteins/metabolism , Overnutrition/metabolism , Palmitates/pharmacology , Rats
5.
J Invest Dermatol ; 131(10): 2113-20, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21697888

ABSTRACT

Merkel cell carcinoma (MCC) is a neoplasm thought to originate from the neuroendocrine Merkel cells of the skin. Although the prevalence of MCC has been increasing, treatments for this disease remain limited because of a paucity of information regarding MCC biology. We have found that the endocytic oncoprotein Huntingtin-interacting protein 1 (HIP1) is expressed at high levels in ∼90% of MCC tumors and serves as a more reliable histological cytoplasmic stain than the gold standard, cytokeratin 20. Furthermore, high anti-HIP1 antibody reactivity in the sera of a cohort of MCC patients predicts the presence of metastases. Another protein that is frequently expressed at high levels in MCC tumors is the stem cell factor (SCF) receptor tyrosine kinase, c-Kit. In working toward an understanding of how HIP1 might contribute to MCC tumorigenesis, we have discovered that HIP1 interacts with SCF-activated c-Kit. These data not only identify HIP1 as a molecular marker for management of MCC patients but also show that HIP1 interacts with and slows the degradation of c-Kit.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Merkel Cell/metabolism , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Aged , Animals , Cytoplasm/metabolism , Endocytosis , Female , Humans , Male , Mice , Middle Aged , Neoplasm Metastasis , Prevalence , Stem Cell Factor/metabolism
6.
Cancer Cell ; 16(2): 137-48, 2009 Aug 04.
Article in English | MEDLINE | ID: mdl-19647224

ABSTRACT

Despite remarkable responses to the tyrosine kinase inhibitor imatinib, CML patients are rarely cured by this therapy perhaps due to imatinib refractoriness of leukemia-initiating cells (LICs). Evidence for this is limited because of poor engraftment of human CML-LICs in NOD-SCID mice and nonphysiologic expression of oncogenes in retroviral transduction mouse models. To address these challenges, we generated mice bearing conditional knockin alleles of two human oncogenes: HIP1/PDGFbetaR (H/P) and AML1-ETO (A/E). Unlike retroviral transduction, physiologic expression of H/P or A/E individually failed to induce disease, but coexpression of both H/P and A/E led to rapid onset of a fully penetrant, myeloproliferative disorder, indicating cooperativity between these two alleles. Although imatinib dramatically decreased disease burden, LICs persisted, demonstrating imatinib refractoriness of LICs.


Subject(s)
Antineoplastic Agents/therapeutic use , Core Binding Factor Alpha 2 Subunit/genetics , DNA-Binding Proteins/genetics , Leukemia, Myelomonocytic, Chronic/drug therapy , Myeloproliferative Disorders/drug therapy , Oncogene Proteins, Fusion/genetics , Piperazines/therapeutic use , Pyrimidines/therapeutic use , Animals , Benzamides , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Gene Knock-In Techniques , Genotype , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Imatinib Mesylate , Leukemia, Myelomonocytic, Chronic/genetics , Leukemia, Myelomonocytic, Chronic/pathology , Mice , Mice, Transgenic , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , RUNX1 Translocation Partner 1 Protein , Spleen/metabolism , Spleen/pathology
7.
Cancer Res ; 68(4): 1064-73, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18281481

ABSTRACT

Huntingtin interacting protein 1 (HIP1) is a 116-kDa endocytic protein, which is necessary for the maintenance of several tissues in vivo as its deficiency leads to degenerative adult phenotypes. HIP1 deficiency also inhibits prostate tumor progression in mice. To better understand how deficiency of HIP1 leads to such phenotypes, we analyzed tumorigenic potential in mice homozygous for a Hip1 mutant allele, designated Hip1(Delta 3-5), which is predicted to result in a frame-shifted, nonsense mutation in the NH(2) terminus of HIP1. In contrast to our previous studies using the Hip1 null allele, an inhibition of tumorigenesis was not observed as a result of the homozygosity of the nonsense Delta 3-5 allele. To further examine the contrasting results from the prior Hip1 mutant mice, we cultured tumor cells from homozygous Delta 3-5 allele-bearing mice and discovered the presence of a 110-kDa form of HIP1 in tumor cells. Upon sequencing of Hip1 DNA and message from these tumors, we determined that this 110-kDa form of HIP1 is the product of splicing of a cryptic U12-type AT-AC intron. This event results in the insertion of an AG dinucleotide between exons 2 and 6 and restoration of the reading frame. Remarkably, this mutant protein retains its capacity to bind lipids, clathrin, AP2, and epidermal growth factor receptor providing a possible explanation for why tumorigenesis was not altered after this knockout mutation. Our data show how knowledge of the transcript that is produced by a knockout allele can lead to discovery of novel types of molecular compensation at the level of splicing.


Subject(s)
DNA-Binding Proteins/genetics , Mammary Neoplasms, Experimental/genetics , RNA Splice Sites , Alleles , Amino Acid Sequence , Animals , Breast Neoplasms/genetics , DNA-Binding Proteins/biosynthesis , Exons , Female , Gene Deletion , Humans , Male , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Prostatic Neoplasms/genetics
8.
Hum Mol Genet ; 16(11): 1279-92, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17452370

ABSTRACT

The members of the huntingtin-interacting protein-1 (HIP1) family, HIP1 and HIP1-related (HIP1r), are multi-domain proteins that interact with inositol lipids, clathrin and actin. HIP1 is over-expressed in a variety of cancers and both HIP1 and HIP1r prolong the half-life of multiple growth factor receptors. To better understand the physiological importance of the HIP1 family in vivo, we have analyzed a large cohort of double Hip1/Hip1r knockout (DKO) mice. All DKO mice were dwarfed, afflicted with severe vertebral defects and died in early adulthood. These phenotypes were not observed during early adulthood in the single Hip1 or Hip1r knockouts, indicating that HIP1 and HIP1r compensate for one another. Despite the ability of HIP1 and HIP1r to modulate growth factor receptor levels when over-expressed, studies herein using DKO fibroblasts indicate that the HIP1 family is not necessary for endocytosis but is necessary for the maintenance of diverse adult tissues in vivo. To test if human HIP1 can function similar to mouse HIP1, transgenic mice with 'ubiquitous' expression of the human HIP1 cDNA were generated and crossed with DKO mice. Strikingly, the compound human HIP1 transgenic DKO mice were completely free from dwarfism and spinal defects. This successful rescue demonstrates that the human HIP1 protein shares some interchangeable functions with both HIP1 and HIP1r in vivo. In addition, we conclude that the degenerative phenotypes seen in the DKO mice are due mainly to HIP1 and HIP1r protein deficiency rather than altered expression of neighboring genes or disrupted intronic elements.


Subject(s)
DNA-Binding Proteins/genetics , Lordosis/genetics , Weight Loss/genetics , Adaptor Proteins, Signal Transducing , Animals , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/physiology , Genes, Lethal , Humans , Introns , Kyphosis/genetics , Mice , Mice, Knockout , Microfilament Proteins , Phenotype
9.
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
10.
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
11.
Hum Mol Genet ; 13(8): 851-67, 2004 Apr 15.
Article in English | MEDLINE | ID: mdl-14998932

ABSTRACT

Huntingtin Interacting Protein 1 (HIP1) binds clathrin and AP2, is overexpressed in multiple human tumors, and transforms fibroblasts. The function of HIP1 is unknown although it is thought to play a fundamental role in clathrin trafficking. Gene-targeted Hip1-/- mice develop premature testicular degeneration and severe spinal deformities. Yet, although HIP1 is expressed in many tissues including the spleen and bone marrow and was part of a leukemogenic translocation, its role in hematopoiesis has not been examined. In this study we report that three different mutations of murine Hip1 lead to hematopoietic abnormalities reflected by diminished early progenitor frequencies and resistance to 5-FU-induced bone marrow toxicity. Two of the Hip1 mutant lines also display the previously described spinal defects. These observations indicate that, in addition to being required for the survival/proliferation of cancer cells and germline progenitors, HIP1 is also required for the survival/proliferation of diverse types of somatic cells, including hematopoietic progenitors.


Subject(s)
Cataract/genetics , DNA-Binding Proteins/genetics , Hematopoiesis/genetics , Spine/abnormalities , Animals , Chimera/genetics , Chimera/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/metabolism , Gene Targeting , Hematopoiesis/physiology , Hematopoietic Stem Cells , Infertility, Male/etiology , Infertility, Male/genetics , Male , Mice , Mutation , Sequence Analysis, DNA
12.
J Biol Chem ; 279(14): 14294-306, 2004 Apr 02.
Article in English | MEDLINE | ID: mdl-14732715

ABSTRACT

Huntingtin-interacting protein 1-related (HIP1r) is the only known mammalian relative of huntingtin-interacting protein 1 (HIP1), a protein that transforms fibroblasts via undefined mechanisms. Here we demonstrate that both HIP1r and HIP1 bind inositol lipids via their epsin N-terminal homology (ENTH) domains. In contrast to other ENTH domain-containing proteins, lipid binding is preferential to the 3-phosphate-containing inositol lipids, phosphatidylinositol 3,4-bisphosphate and phosphatidylinositol 3,5-bisphosphate. Furthermore, the HIP1r ENTH domain, like that of HIP1, is necessary for lipid binding, and expression of an ENTH domain-deletion mutant, HIP1r/deltaE, induces apoptosis. Consistent with the ability of HIP1r and HIP1 to affect cell survival, full-length HIP1 and HIP1r stabilize pools of growth factor receptors by prolonging their half-life following ligand-induced endocytosis. Although HIP1r and HIP1 display only a partially overlapping pattern of protein interactions, these data suggest that both proteins share a functional homology by binding 3-phosphorylated inositol lipids and stabilizing receptor tyrosine kinases in a fashion that may contribute to their ability to alter cell growth and survival.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , DNA-Binding Proteins , Endocytosis/physiology , Neuropeptides/genetics , Phosphatidylinositols/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Vesicular Transport Proteins , Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport , Animals , Antibodies, Monoclonal , Binding Sites , COS Cells , Carrier Proteins/chemistry , Carrier Proteins/immunology , Cell Death/physiology , Clathrin/metabolism , ErbB Receptors/metabolism , HeLa Cells , Humans , Inositol/metabolism , Kinetics , Microfilament Proteins , Mutagenesis , Protein Structure, Tertiary , Protein Transport/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...