Stéphanie A. Eid , Sarah E. Elzinga , Kai Guo , Lucy M. Hinder , John M. Hayes , Crystal M. Pacut , Emily J. Koubek , Junguk Hur , Eva L. Feldman
{"title":"Transcriptomic profiling of sciatic nerves and dorsal root ganglia reveals site-specific effects of prediabetic neuropathy","authors":"Stéphanie A. Eid , Sarah E. Elzinga , Kai Guo , Lucy M. Hinder , John M. Hayes , Crystal M. Pacut , Emily J. Koubek , Junguk Hur , Eva L. Feldman","doi":"10.1016/j.trsl.2024.03.009","DOIUrl":"10.1016/j.trsl.2024.03.009","url":null,"abstract":"<div><p>Peripheral neuropathy (PN) is a severe and frequent complication of obesity, prediabetes, and type 2 diabetes characterized by progressive distal-to-proximal peripheral nerve degeneration. However, a comprehensive understanding of the mechanisms underlying PN, and whether these mechanisms change during PN progression, is currently lacking. Here, gene expression data were obtained from distal (sciatic nerve; SCN) and proximal (dorsal root ganglia; DRG) injury sites of a high-fat diet (HFD)-induced mouse model of obesity/prediabetes at early and late disease stages. Self-organizing map and differentially expressed gene analyses followed by pathway enrichment analysis identified genes and pathways altered across disease stage and injury site. Pathways related to immune response, inflammation, and glucose and lipid metabolism were consistently dysregulated with HFD-induced PN, irrespective of injury site. However, regulation of oxidative stress was unique to the SCN while dysregulated Hippo and Notch signaling were only observed in the DRG. The role of the immune system and inflammation in disease progression was supported by an increase in the percentage of immune cells in the SCN with PN progression. Finally, when comparing these data to transcriptomic signatures from human patients with PN, we observed conserved pathways related to metabolic dysregulation across species, highlighting the translational relevance of our mouse data. Our findings demonstrate that PN is associated with distinct site-specific molecular re-programming in the peripheral nervous system, identifying novel, clinically relevant therapeutic targets.</p></div>","PeriodicalId":23226,"journal":{"name":"Translational Research","volume":"270 ","pages":"Pages 24-41"},"PeriodicalIF":7.8,"publicationDate":"2024-03-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140332407","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Hanlin Yao , Hongchao Zhao , Yang Du , Ye Zhang , Yanze Li , Hengcheng Zhu
{"title":"Sex-related differences in SIRT3-mediated mitochondrial dynamics in renal ischemia/reperfusion injury","authors":"Hanlin Yao , Hongchao Zhao , Yang Du , Ye Zhang , Yanze Li , Hengcheng Zhu","doi":"10.1016/j.trsl.2024.03.005","DOIUrl":"10.1016/j.trsl.2024.03.005","url":null,"abstract":"<div><p>The prevalence of renal ischemia/reperfusion injury (IRI) in premenopausal women is considerably lower than that in age-matched men. This suggests that sex-related differences in mitochondrial function and homeostasis may contribute to sexual dimorphism in renal injury, though the mechanism remains unclear. Mouse model of unilateral left renal IRI with contralateral kidney enucleation, Ovariectomy in female mice, and a human embryonic kidney (HEK) cell model of hypoxia-reoxygenation were used to study how estrogen affects the sexual dimorphism of renal IRI through SIRT3 in vitro and in vivo, respectively. Here, we demonstrate differential expression of renal SIRT3 may induce sexual dimorphism in IRI using the renal IRI model. Higher SIRT3 level in female mice was associated with E2-induced protection of renal tubular epithelium, reduced mitochondrial reactive oxygen species (ROS), and IRI resistance. In hypoxia-reoxygenated HEK cells, SIRT3 knockdown increased oxidative stress, shifted the interconnected mitochondrial network toward fission, exacerbated hypoxia/reoxygenation-induced endoplasmic reticulum stress (ERS), and abolished the protective effects of E2 on IRI. Mechanistically, the SIRT3 level is E2-dependent and that E2 increases the SIRT3 protein level via estrogen receptor. SIRT3 targeted an i-AAA protease, yeast mitochondrial AAA metalloprotease (YME1L1), and hydrolyzed long optic atrophy 1 (L-OPA) to short-OPA1 (S-OPA1) by deacetylating YME1L1, regulating mitochondrial dynamics toward fusion to reduce oxidative stress and ERS. These findings explored the mechanism by how estrogen alleviates renal IRI and providing a basis for potential therapeutic interventions targeting SIRT3.</p></div>","PeriodicalId":23226,"journal":{"name":"Translational Research","volume":"270 ","pages":"Pages 1-12"},"PeriodicalIF":7.8,"publicationDate":"2024-03-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140332406","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Sixian Chen , Qingyun Zhao , Ruirui Zhang , Jungang Liu , Wenyi Peng , Haotian Xu , Xiaofei Li , Xin Wang , Shuilian Wu , Gang Li , Aruo Nan
{"title":"A transcribed ultraconserved noncoding RNA, uc.285+, promotes colorectal cancer proliferation through dual targeting of CDC42 by directly binding mRNA and protein","authors":"Sixian Chen , Qingyun Zhao , Ruirui Zhang , Jungang Liu , Wenyi Peng , Haotian Xu , Xiaofei Li , Xin Wang , Shuilian Wu , Gang Li , Aruo Nan","doi":"10.1016/j.trsl.2024.03.008","DOIUrl":"10.1016/j.trsl.2024.03.008","url":null,"abstract":"<div><p>The transcribed ultraconserved region (T-UCR) belongs to a new type of lncRNAs that are conserved in homologous regions of the rat, mouse and human genomes. A lot of research has reported that differential expression of T-UCRs can influence the development of various cancers, revealing the ability of T-UCRs as new therapeutic targets or potential cancer biomarkers. Most studies on the molecular mechanisms of T-UCRs in cancer have focused on ceRNA regulatory networks and interactions with target proteins, but the present study reveals an innovative dual-targeted regulatory approach in which T-UCRs bind directly to mRNAs and directly to proteins. We screened T-UCRs that may be related to colorectal cancer (CRC) by performing a whole-genome T-UCR gene microarray and further studied the functional mechanism of T-UCR uc.285+ in the development of CRC. Modulation of uc.285+ affected the proliferation of CRC cell lines and influenced the expression of the CDC42 gene. We also found that uc.285+ promoted the proliferation of CRC cells by directly binding to CDC42 mRNA and enhancing its stability while directly binding to CDC42 protein and affecting its stability. In short, our research on the characteristics of cell proliferation found that uc.285+ has a biological function in promoting CRC proliferation. uc.285+ may have considerable potential as a new diagnostic biomarker for CRC.</p></div>","PeriodicalId":23226,"journal":{"name":"Translational Research","volume":"270 ","pages":"Pages 52-65"},"PeriodicalIF":7.8,"publicationDate":"2024-03-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1931524424000616/pdfft?md5=0602f01e7f9f9c2a59b4b10da28d1974&pid=1-s2.0-S1931524424000616-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140327590","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jia-Yu Peng , Xiao Fu , Xue-Yang Luo , Fang Liu , Bing Zhang , Bin Zhou , Kun Sun , Alex F. Chen
{"title":"Endothelial ELABELA improves post-ischemic angiogenesis by upregulating VEGFR2 expression","authors":"Jia-Yu Peng , Xiao Fu , Xue-Yang Luo , Fang Liu , Bing Zhang , Bin Zhou , Kun Sun , Alex F. Chen","doi":"10.1016/j.trsl.2024.03.011","DOIUrl":"10.1016/j.trsl.2024.03.011","url":null,"abstract":"<div><h3>Background</h3><p>Post-ischemic angiogenesis is critical for perfusion recovery and tissue repair. ELABELA (ELA) plays an essential role in embryonic heart development and vasculogenesis. However, the mechanism of ELA on post-ischemic angiogenesis is poorly characterized.</p></div><div><h3>Methods</h3><p>We first assessed ELA expression after hind limb ischemia (HLI) in mice. We then established a HLI model in tamoxifen-inducible endothelial-ELA-specific knockout mice (ELA<sup>ECKO</sup>) and assessed the rate of perfusion recovery, capillary density, and VEGFR2 pathway. Knockdown of ELA with lentivirus or siRNA and exogenous addition of ELA peptides were employed to analyze the effects of ELA on angiogenic capacity and VEGFR2 pathway in endothelial cells <em>in vitro</em>. The serum levels of ELA in healthy people and patients with type 2 diabetes mellitus (T2DM) and diabetic foot ulcer (DFU) were detected by a commercial ELISA kit.</p></div><div><h3>Results</h3><p>In murine HLI models, ELA was significantly up-regulated in the ischemic hindlimb. Endothelial-specific deletion of ELA impaired perfusion recovery and angiogenesis. In physiologic conditions, no significant difference in VEGFR2 expression was found between ELA<sup>ECKO</sup> mice and ELA<sup>WT</sup> mice. After ischemia, the expression of VEGFR2, p-VEGFR2, and p-AKT was significantly lower in ELA<sup>ECKO</sup> mice than in ELA<sup>WT</sup> mice. In cellular experiments, the knockdown of ELA inhibited endothelial cell proliferation and tube formation, and the addition of ELA peptides promoted proliferation and tube formation. Mechanistically, ELA upregulated the expression of VEGFR2, p-VEGFR2, and p-AKT in endothelial cells under hypoxic conditions. In clinical investigations, DFU patients had significantly lower serum levels of ELA compared to T2DM patients.</p></div><div><h3>Conclusion</h3><p>Our results indicated that endothelial ELA is a positive regulator of post-ischemic angiogenesis via upregulating VEGFR2 expression. Targeting ELA may be a potential therapeutic option for peripheral arterial diseases.</p></div>","PeriodicalId":23226,"journal":{"name":"Translational Research","volume":"270 ","pages":"Pages 13-23"},"PeriodicalIF":7.8,"publicationDate":"2024-03-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140320347","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yuanbo Zhu , Yue Hu , Zhongwang Liu , Luping Chang , Xue Geng , Xuhui Yin , Bing-Qiao Zhao , Wenying Fan
{"title":"The LPS-inactivating enzyme acyloxyacyl hydrolase protects the brain from experimental stroke","authors":"Yuanbo Zhu , Yue Hu , Zhongwang Liu , Luping Chang , Xue Geng , Xuhui Yin , Bing-Qiao Zhao , Wenying Fan","doi":"10.1016/j.trsl.2024.03.007","DOIUrl":"10.1016/j.trsl.2024.03.007","url":null,"abstract":"<div><p>Blood-brain-barrier (BBB) disruption is a pathological hallmark of ischemic stroke, and inflammation occurring at the BBB contributes to the pathogenesis of ischemic brain injury. Lipopolysaccharide (LPS), a cell wall component of Gram-negative bacteria, is elevated in patients with acute stroke. The activity of LPS is controlled by acyloxyacyl hydrolase (AOAH), a host enzyme that deacylates LPS to inactivated forms. However, whether AOAH influences the pathogenesis of ischemic stroke remain elusive. We performed in vivo experiments to explore the role and mechanism of AOAH on neutrophil extravasation, BBB disruption, and brain infarction. We found that AOAH was upregulated in neutrophils in peri-infarct areas from mice with transient focal cerebral ischemia. AOAH deficiency increased neutrophil extravasation into the brain parenchyma and proinflammatory cytokine production, broke down the BBB and worsened stroke outcomes in mice. These effects require Toll-like receptor 4 (TLR4) because absence of TLR4 or pharmacologic inhibition of TLR4 signaling prevented the exacerbated inflammation and BBB damage in <em>Aoah</em><sup>−/−</sup> mice after ischemic stroke. Importantly, neutrophil depletion or inhibition of neutrophil trafficking by blocking LFA-1 integrin dramatically reduced stroke-induced BBB breakdown in <em>Aoah</em><sup>−/−</sup> mice. Furthermore, virus-mediated overexpression of AOAH induced a substantial decrease in neutrophil recruitment that was accompanied by reducing BBB damage and stroke volumes. Our findings show the importance of AOAH in regulating neutrophil-dependent BBB breakdown and cerebral infarction. Consequently, strategies that modulate AOAH may be a new therapeutic approach for treatment of ischemic stroke.</p></div>","PeriodicalId":23226,"journal":{"name":"Translational Research","volume":"270 ","pages":"Pages 42-51"},"PeriodicalIF":7.8,"publicationDate":"2024-03-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140208766","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pratikshya Sa , Priya Singh , Sudhakar Panda , Rajeeb K. Swain , Rupesh Dash , Sanjeeb Kumar Sahoo
{"title":"Reversal of cisplatin resistance in oral squamous cell carcinoma by piperlongumine loaded smart nanoparticles through inhibition of Hippo-YAP signaling pathway","authors":"Pratikshya Sa , Priya Singh , Sudhakar Panda , Rajeeb K. Swain , Rupesh Dash , Sanjeeb Kumar Sahoo","doi":"10.1016/j.trsl.2024.03.004","DOIUrl":"10.1016/j.trsl.2024.03.004","url":null,"abstract":"<div><p>Cisplatin alone or in combination with 5FU and docetaxel is the preferred chemotherapy regimen for advanced-stage OSCC patients. However, its use has been linked to recurrence and metastasis due to the development of drug resistance. Therefore, sensitization of cancer cells to conventional chemotherapeutics can be an effective strategy to overcome drug resistance. Piperlongumine (PL), an alkaloid, have shown anticancer properties and sensitizes numerous neoplasms, but its effect on OSCC has not been explored. However, low aqueous solubility and poor pharmacokinetics limit its clinical application. Therefore, to improve its therapeutic efficacy, we developed piperlongumine-loaded PLGA-based smart nanoparticles (smart PL-NPs) that can rapidly release PL in an acidic environment of cancer cells and provide optimum drug concentrations to overcome chemoresistance. Our results revealed that smart PL-NPs has high cellular uptake in acidic environment, facilitating the intracellular delivery of PL and sensitizing cancer cells to cisplatin, resulting in synergistic anticancer activity <em>in vitro</em> by increasing DNA damage, apoptosis, and inhibiting drug efflux. Further, we have mechanistically explored the Hippo-YAP signaling pathway, which is the critical mediator of chemoresistance, and investigated the chemosensitizing effect of PL in OSCC. We observed that PL alone and in combination with cisplatin significantly inhibits the activation of YAP and its downstream target genes and proteins. In addition, the combination of cisplatin with smart PL-NPs significantly inhibited tumor growth in two preclinical models (patient-derived cell based nude mice and zebrafish xenograft). Taken together, our findings suggest that smart PL-NPs with cisplatin will be a novel formulation to reverse cisplatin resistance in patients with advanced OSCC.</p></div>","PeriodicalId":23226,"journal":{"name":"Translational Research","volume":"268 ","pages":"Pages 63-78"},"PeriodicalIF":7.8,"publicationDate":"2024-03-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140159844","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Baicheng Kuang , Mengqin Wang , Hao Yan , Qin Jiang , Zhiheng Wang , Haiqiang Ni , Shuaiheng Hou , Xuan Peng , Shiqi Gu , Yuanyuan Zhao , Tongwen Ou , Nianqiao Gong
{"title":"Genetic and pharmacological targeting of XBP1 alleviates hepatic ischemia reperfusion injury by enhancing FoxO1-dependent mitophagy","authors":"Baicheng Kuang , Mengqin Wang , Hao Yan , Qin Jiang , Zhiheng Wang , Haiqiang Ni , Shuaiheng Hou , Xuan Peng , Shiqi Gu , Yuanyuan Zhao , Tongwen Ou , Nianqiao Gong","doi":"10.1016/j.trsl.2024.03.006","DOIUrl":"10.1016/j.trsl.2024.03.006","url":null,"abstract":"<div><p>Hepatic ischemia reperfusion (I/R) injury is a common clinical complication. X-box binding protein 1 (XBP1), as a critical regulator of the endoplasmic reticulum stress, has been implicated in a variety of diseases. In this study, we aimed to investigate the effects and the underlying mechanism of XBP1 in the progression of hepatic I/R injury. Hepatocyte-specific XBP1 knockout mice, multiple viral delivery systems and specific pharmacological inhibitors were applied <em>in vivo</em> in a partial hepatic I/R injury mouse model and <em>in vitro</em> in a cell model of hypoxia-reoxygenation (H/R) injury. Mitophagy and autophagic flux were evaluated and fluorescence resonance energy transfer (FRET) as well as immunoprecipitation were performed. The results demonstrated that reperfusion for 6 h represented a critical timepoint in hepatic I/R injury and resulted in significant intracellular mitochondrial dysfunction; led to the breakdown of hepatocytes accompanied by the highest expression levels of XBP1. Hepatocyte-specific <em>XBP1</em> knockout alleviated hepatic I/R injury <em>via</em> enhanced mitophagy, as demonstrated by the reduction in hepatocellular damage/necrosis and increased expression of mitophagy markers. Mechanistically, XBP1 interacted with FoxO1 directly and catalyzed the ubiquitination of FoxO1 for proteasomal degradation. Targeting XBP1 by genetic or pharmacological techniques potentiated the protein levels of FoxO1, further promoting the activity of the PINK1/Parkin signaling pathway, thus augmenting mitophagy and exerting hepatoprotective effects upon I/R injury. In conclusion, the inhibition of XBP1 potentiated FoxO1-mediated mitophagy in hepatic I/R injury. Specific genetic and pharmacological treatment targeting XBP1 in the perioperative 6 h prior to reperfusion exerted beneficial effects, thus providing a novel therapeutic approach.</p></div>","PeriodicalId":23226,"journal":{"name":"Translational Research","volume":"272 ","pages":"Pages 162-176"},"PeriodicalIF":6.4,"publicationDate":"2024-03-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140144978","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Sarah Richtmann , Sebastian Marwitz , Thomas Muley , Hannu Koistinen , Petros Christopoulos , Michael Thomas , Daniel Kazdal , Michael Allgäuer , Hauke Winter , Torsten Goldmann , Michael Meister , Ursula Klingmüller , Marc A. Schneider
{"title":"The pregnancy-associated protein glycodelin as a potential sex-specific target for resistance to immunotherapy in non-small cell lung cancer","authors":"Sarah Richtmann , Sebastian Marwitz , Thomas Muley , Hannu Koistinen , Petros Christopoulos , Michael Thomas , Daniel Kazdal , Michael Allgäuer , Hauke Winter , Torsten Goldmann , Michael Meister , Ursula Klingmüller , Marc A. Schneider","doi":"10.1016/j.trsl.2024.02.007","DOIUrl":"10.1016/j.trsl.2024.02.007","url":null,"abstract":"<div><p>Lung cancer has been shown to be targetable by novel immunotherapies which reactivate the immune system and enable tumor cell killing. However, treatment failure and resistance to these therapies is common. Consideration of sex as a factor influencing therapy resistance is still rare. We hypothesize that the success of the treatment is impaired by the presence of the immunosuppressive pregnancy-associated glycoprotein glycodelin that is expressed in patients with non-small-cell lung cancer (NSCLC). We demonstrate that the glycan pattern of NSCLC-derived glycodelin detected by a lectin-based enrichment assay highly resembles amniotic fluid-derived glycodelin A, which is known to have immunosuppressive properties. NSCLC-derived glycodelin interacts with immune cells <em>in vitro</em> and regulates the expression of genes associated with inflammatory and tumor microenvironment pathways. In tumor microarray samples of patients, high glycodelin staining in tumor areas results in an impaired overall survival of female patients. Moreover, glycodelin colocalizes to tumor infiltrating CD8+ T cells and pro-tumorigenic M2 macrophages. High serum concentrations of glycodelin prior to immunotherapy are associated with a poor progression-free survival (<em>p</em> < 0.001) of female patients receiving PD-(L)1 inhibitors. In summary, our findings suggest that glycodelin not only is a promising immunological biomarker for early identification of female patients that do not benefit from the costly immunotherapy, but also represents a promising immunotherapeutic target in NSCLC to improve therapeutic options in lung cancer.</p></div>","PeriodicalId":23226,"journal":{"name":"Translational Research","volume":"272 ","pages":"Pages 177-189"},"PeriodicalIF":6.4,"publicationDate":"2024-03-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1931524424000367/pdfft?md5=f879972cbb0aed4e7506c65c67ce17eb&pid=1-s2.0-S1931524424000367-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140137649","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Victoria Lorant , Martin Klein , Damien Garçon , Thibaud Sotin , Samuel Frey , Marie-Aude Cheminant , Audrey Ayer , Mikaël Croyal , Laurent Flet , Antoine Rimbert , Luc Colas , Bertrand Cariou , Grégory Bouchaud , Cédric Le May
{"title":"PCSK9 inhibition protects mice from food allergy","authors":"Victoria Lorant , Martin Klein , Damien Garçon , Thibaud Sotin , Samuel Frey , Marie-Aude Cheminant , Audrey Ayer , Mikaël Croyal , Laurent Flet , Antoine Rimbert , Luc Colas , Bertrand Cariou , Grégory Bouchaud , Cédric Le May","doi":"10.1016/j.trsl.2024.03.001","DOIUrl":"10.1016/j.trsl.2024.03.001","url":null,"abstract":"<div><p>The Proprotein Convertase Subtilisin Kexin of type 9 (PCSK9) has been identified in 2003 as the third gene involved in familial hypercholesterolemia. PCSK9 binds to the membrane low-density lipoprotein receptor (LDLR) and promotes its cellular internalization and lysosomal degradation. Beyond this canonical role, PCSK9 was recently described to be involved in several immune responses. However, to date, the contribution of PCSK9 in food allergy remains unknown. Here, we showed that <em>Pcsk9</em> deficiency or pharmacological inhibition of circulating PCSK9 with a specific monoclonal antibody (m-Ab) protected mice against symptoms of gliadin-induced-food allergy, such as increased intestinal transit time and ear oedema. Furthermore, specific PCSK9 inhibition during the elicitation steps of allergic process was sufficient to ensure anti-allergic effects in mice. Interestingly, the protective effect of PCSK9 inhibition against food allergy symptoms was independent of the LDLR as PCSK9 inhibitors remained effective in <em>Ldlr</em> deficient mice. <em>In vitro</em>, we showed that recombinant gain of function PCSK9 (PCSK9 D374Y) increased the percentage of mature bone marrow derived dendritic cells (BMDCs), promoted naïve T cell proliferation and potentiated the gliadin induced basophils degranulation. Altogether, our data demonstrate that PCSK9 inhibition is protective against gliadin induced food allergy in a LDLR-independent manner.</p></div>","PeriodicalId":23226,"journal":{"name":"Translational Research","volume":"272 ","pages":"Pages 151-161"},"PeriodicalIF":6.4,"publicationDate":"2024-03-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140112565","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}