scholarly journals Deficiency of the Endocytic ProteinHip1Leads to DecreasedGdpd3expression, Low Phosphocholine, and Kypholordosis

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.

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.


2015 ◽  
Vol 112 (35) ◽  
pp. 11007-11012 ◽  
Author(s):  
Lingjun Meng ◽  
Wei Jin ◽  
Xiaodong Wang

Systematic inflammation contributes to the development of many diseases, including cardiovascular disease, which is the leading cause of mortality worldwide. How such inflammation is initiated and maintained throughout the course of disease remains unclear. In the current study, we report the observation of specific phosphorylation of the receptor-interacting protein 3 (RIP3) kinase that marks the activation of programmed necrosis (also called the “necroptosis pathway”) in the atherosclerotic plaques in apolipoprotein E (ApoE)-knockout mice. The mRNA expression levels of 10 inflammatory cytokines, including IL-1α, were decreased significantly in the plaque regions of mice lacking RIP3. Lymphocyte infiltrations in the adipocyte tissue and in skin lesions of ApoE single-knockout mice were significantly mitigated in ApoE/RIP3 double-knockout mice. The high percentage of inflammatory monocytes with high levels of lymphocyte antigen 6C in the blood of ApoE single-knockout mice also was greatly decreased in the ApoE/RIP3 double-knockout mice. Most significantly, the double-knockout mice displayed dramatically delayed mortality compared with ApoE single-knockout mice. Our findings indicate that necrotic death in areas such as atherosclerotic plaques may release cytokines that mobilize monocytes from bone marrow to the lesion sites, exacerbating the lesions in multiple tissues and resulting in the premature death of the animals.


2018 ◽  
Vol 77 (11) ◽  
pp. 1627-1635 ◽  
Author(s):  
Teresina Laragione ◽  
Max Brenner ◽  
Amit Lahiri ◽  
Erjing Gao ◽  
Carolyn Harris ◽  
...  

ObjectivesWhile new treatments for rheumatoid arthritis (RA) have markedly improved disease control by targeting immune/inflammatory pathways, current treatments rarely induce remission, underscoring the need for therapies that target other aspects of the disease. Little is known about the regulation of disease severity and joint damage, which are major predictors of disease outcome, and might be better or complementary targets for therapy. In this study, we aimed to discover and characterise a new arthritis severity gene.MethodsAn unbiased and phenotype-driven strategy including studies of unique congenic rat strains was used to identify new arthritis severity and joint damage genes. Fibroblast-like synoviocytes (FLS) from rats and patients with RA expressing or not Huntingtin-interacting protein 1 (HIP1) were studied for invasiveness, morphology and cell signalling. HIP1 knockout mice were used in in vivo confirmatory studies. Paired t-test was used.ResultsDNA sequencing and subcongenic strains studied in pristane-induced arthritis identified a new amino acid changing functional variant in HIP1. HIP1 was required for the increased invasiveness of FLS from arthritic rats and from patients with RA. Knocking down HIP1 expression reduced receptor tyrosine kinase-mediated responses in RA FLS, including RAC1 activation, affecting actin cytoskeleton and cell morphology and interfering with the formation of lamellipodia, consistent with reduced invasiveness. HIP1 knockout mice were protected in KRN serum-induced arthritis and developed milder disease.ConclusionHIP1 is a new arthritis severity gene and a potential novel prognostic biomarker and target for therapy in RA.


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.


Author(s):  
AC Langheinrich ◽  
D Sedding ◽  
M Kampschulte ◽  
J Wilhelm ◽  
W Haberbosch ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document