scholarly journals Deficiency of the Endocytic Protein Hip1 Leads to DecreasedGdpd3Expression, Low Phosphocholine, and Kypholordosis

2018 ◽  
Vol 38 (23) ◽  
Author(s):  
Ranjula Wijayatunge ◽  
Sam R. Holmstrom ◽  
Samantha B. Foley ◽  
Victoria E. Mgbemena ◽  
Varsha Bhargava ◽  
...  

ABSTRACTDeficiency of huntingtin-interacting protein 1 (Hip1) results in degenerative phenotypes. Here we generated aHip1deficiency allele where a floxed transcriptional stop cassette and a humanHIP1cDNA were knocked into intron 1 of the mouseHip1locus.CMV-Cre-mediated germ line excision of the stop cassette resulted in expression of HIP1 and rescue of theHip1knockout 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 severalHip1knockout 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,Gdpd3downregulation could lead to the low PC levels. To test whetherGdpd3contributes to theHip1deficiency phenotype, we generatedGdpd3knockout mice. Double knockout ofGdpd3andHip1worsened the Hip1 phenotype. This suggests that Gdpd3 compensates for Hip1 loss. More-detailed knowledge of howHip1deficiency leads to low PC will improve our understanding of HIP1 in choline metabolism in normal and disease states.

2018 ◽  
Author(s):  
Ranjula Wijayatunge ◽  
Sam R. Holmstrom ◽  
Samantha B. Foley ◽  
Victoria E. Mgbemena ◽  
Varsha Bhargava ◽  
...  

ABSTRACTDeficiency of huntingtin interacting protein 1 (Hip1) results in degenerative phenotypes. Here we generated aHip1deficiency allele where a floxed transcriptional stop-cassette and a humanHIP1cDNA were knocked-in to intron 1 of mouseHip1locus.CMV-Cre-mediated germline excision of the stop-cassette resulted in expression of HIP1 and rescue of theHip1knockout phenotype.Mxl-Cre--mediated excision led to HIP1 expression in spleen, kidney and liver, and also rescued the phenotype. In contrast,GFAP-Cre-mediatedHIP1expression in brain did not rescue the phenotype. Metabolomics and microarrays of severalHip1knockout tissues identified low phosphocholine (PC) levels and lowGlycerophosphodiester Phosphodiesterase Domain Containing 3 (Gdpd3) expression. Since Gdpd3 has lysophospholipase D activity that results in the formation of choline, a precursor of PC,Gdpd3downregulation could lead to the low PC levels. To test ifGdpd3contributes to the Hip1 deficiency phenotype, we generatedGdpd3knockout mice. Double knockout ofGdpd3andHip1worsened 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.


2005 ◽  
Vol 170 (2) ◽  
pp. 169-171 ◽  
Author(s):  
Manuela Vecchi ◽  
Pier Paolo Di Fiore

Many endocytic proteins shuttle between the nucleus and the cytoplasm; however, their putative function in the nucleus is unclear. Now, new data demonstrate that huntingtin interacting protein 1 (HIP1), an endocytic protein, modulates the transcriptional activity of nuclear hormone receptors. In network theory, therefore, HIP1 can be regarded as a hub connecting heterogeneous functional “territories:” a possibility with important physiological and pathological implications.


2004 ◽  
Vol 24 (10) ◽  
pp. 4329-4340 ◽  
Author(s):  
Teresa S. Hyun ◽  
Lina Li ◽  
Katherine I. Oravecz-Wilson ◽  
Sarah V. Bradley ◽  
Melissa M. Provot ◽  
...  

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.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3757-3757
Author(s):  
Kah-Keng Wong ◽  
Karen Pulford ◽  
Hong Chen ◽  
Derek Murphy ◽  
Alison Banham

Abstract We screened lymphoma patients’ serum samples against protein/antigen arrays, containing over 10,000 different human proteins, to identify disease-associated auto-IgG antibody interactions. This approach identified a humoral immune response to the HIP1R antigen in 40% of patients’ sera (4/6 follicular lymphomas, 3/5 germinal centre [GC]-derived diffuse large B-cell lymphomas [DLBCL], 2/4 transformed DLBCL, 1/5 mantle cell lymphomas, 1/5 non-GC DLBCL and 1/5 peripheral T-cell lymphomas). HIP1R has also previously been identified as an autoantigen in colon cancer using the SEREX technique. HIP1R is one of two members of the Huntingtin interacting protein 1 family. Both HIP1R and HIP1 share significant sequence homology and are cytoplasmic proteins that interact with inositol lipids, clathrin and actin. Interestingly, both proteins can stabilize pools of receptor tyrosine kinases by inhibiting their trafficking to the lysosome for degradation, which may mediate affects on cell growth and transformation. HIP1 has been more extensively characterized in cancer and was originally implicated in hematological malignancy when it was identified as a platelet-derived growth factor β receptor fusion partner in chronic myelomonocytic leukemia. HIP1 expression is elevated in lymphomas and can be induced by activated NF-κB while transgenic overexpression of HIP1 is associated with the development of lymphoid neoplasms. Interestingly, the increased severity of a murine Hip1/Hip1r double-knockout phenotype indicates that Hip1r can partially functionally compensate for the loss of Hip1, suggesting they have overlapping roles in vivo. Autoantibodies to HIP1 have been reported in 59% of human lymphoma sera, compared to 31% of healthy controls and showed higher titre in patients who had undergone remission than those that relapsed (Bradley et al. 2007. Cancer Res 67:8923). However, the region of HIP1 tested for this serum reactivity shares high sequence homology with HIP1R, and thus this previous study is likely to have identified autoantibodies to both HIP1 and HIP1R. In contrast the region of HIP1R on our antigen array does not share homology with HIP1, suggesting that HIP1R expression is also altered in lymphomas. Quantitative RT-PCR was performed for both HIP1 and HIP1R on a panel of lymphoma cell lines, normal tissues and purified B- and T-cell populations. Overall HIP1R was more widely expressed and at higher levels than HIP1 in both normal lymphocytes and lymphoma cell lines. The particularly high level expression of HIP1R in Burkitt lymphoma cell lines and increased expression in GC- compared to non-GC DLBCL is consistent with the high frequency of autoantibodies in the sera from patients with GC-derived lymphomas. Two commercial monoclonal antibodies were successfully validated for the detection of HIP1R, although only the BD Transduction Laboratories’ antibody is raised to a region that lacks any significant stretches of homology with HIP1. Western blotting experiments have confirmed the expression of the HIP1R protein in lymphoma cell lines and showed a good correlation with mRNA expression levels. The BD Transduction Laboratories anti-HIP1R antibody recognises a formalin resistant epitope and studies are already underway to elucidate HIP1R expression patterns in lymphoma biopsies.


2007 ◽  
Vol 28 (1) ◽  
pp. 386-396 ◽  
Author(s):  
Trevelyan R. Menheniott ◽  
Kathryn Woodfine ◽  
Reiner Schulz ◽  
Andrew J. Wood ◽  
David Monk ◽  
...  

ABSTRACT By combining a tissue-specific microarray screen with mouse uniparental duplications, we have identified a novel imprinted gene, Dopa decarboxylase (Ddc), on chromosome 11. Ddc_exon1a is a 2-kb transcript variant that initiates from an alternative first exon in intron 1 of the canonical Ddc transcript and is paternally expressed in trabecular cardiomyocytes of the embryonic and neonatal heart. Ddc displays tight conserved linkage with the maternally expressed and methylated Grb10 gene, suggesting that these reciprocally imprinted genes may be coordinately regulated. In Dnmt3L mutant embryos that lack maternal germ line methylation imprints, we show that Ddc is overexpressed and Grb10 is silenced. Their imprinting is therefore dependent on maternal germ line methylation, but the mechanism at Ddc does not appear to involve differential methylation of the Ddc_exon1a promoter region and may instead be provided by the oocyte mark at Grb10. Our analysis of Ddc redefines the imprinted Grb10 domain on mouse proximal chromosome 11 and identifies Ddc_exon1a as the first example of a heart-specific imprinted gene.


2010 ◽  
Vol 38 (1) ◽  
pp. 187-191 ◽  
Author(s):  
Irit Gottfried ◽  
Marcelo Ehrlich ◽  
Uri Ashery

HIP1 (huntingtin interacting protein 1) has two close relatives: HIP1R (HIP1-related) and yeast Sla2p. All three members of the family have a conserved domain structure, suggesting a common function. Over the past decade, a number of studies have characterized these proteins using a combination of biochemical, imaging, structural and genetic techniques. These studies provide valuable information on binding partners, structure and dynamics of HIP1/HIP1R/Sla2p. In general, all suggest a role in CME (clathrin-mediated endocytosis) for the three proteins, though some differences have emerged. In this mini-review we summarize the current views on the roles of these proteins, while emphasizing the unique attributes of each family member.


2019 ◽  
Author(s):  
Debasish Kumar Ghosh ◽  
Ajit Roy ◽  
Akash Ranjan

ABSTRACTSelective autophagy of protein aggregates is necessary for maintaining the cellular proteostasis. Several regulatory proteins play critical roles in this process. Here, we report that the huntingtin interacting protein K (HYPK) modulates the autophagic degradation of poly-neddylated huntingtin exon1 aggregates. HYPK functions as a scaffolding protein that binds to the Nedd8 and LC3 proteins. The C-terminal ubiquitin-associated (UBA) domain of HYPK binds to the Nedd8, whereas an N-terminal tyrosine-type (Y-type) LC3 interacting region (LIR) of HYPK binds to the LC3. Several conserved amino acids in the UBA domain of HYPK are necessary to mediate the efficient binding of HYPK to Nedd8. The autophagy inducing properties of HYPK are manifested by the increased lipidation of LC3 protein, increased expression of beclin-1 and ATG-5 proteins, and generation of puncta-like granules of LC3 in the HYPK overexpressing cells. Association of the ‘H-granules’ of HYPK with the poly-neddylated huntingtin exon1 aggregates results in the formation of autophagosome around the huntingtin exon1 aggregates, thereby clearing the aggregates by aggrephagy. Poly-neddylation of huntingtin exon1 is required for its autophagic degradation by HYPK. Thus, overexpression of Nedd8 also increases the basal level of cellular autophagy, other than maintaining the autophagy flux. The poly-neddylation dependent autophagic clearance of huntingtin exon1 by HYPK leads to better cell physiology and survival. Taken together, our study describes a novel mechanism of HYPK mediated autophagy of poly-neddylated huntingtin exon1 aggregates.


2007 ◽  
Vol 71 (4) ◽  
pp. 1686-1698 ◽  
Author(s):  
Swasti Raychaudhuri ◽  
Pritha Majumder ◽  
Somosree Sarkar ◽  
Kalyan Giri ◽  
Debashis Mukhopadhyay ◽  
...  

2001 ◽  
Vol 18 (1) ◽  
pp. 68-79 ◽  
Author(s):  
Susanne Rega ◽  
Thorsten Stiewe ◽  
Dae-In Chang ◽  
Barbara Pollmeier ◽  
Helmut Esche ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document