Laura Monteonofrio, Ilaria Virdia, Sara Pozzi, Roberto Quadri, Alessandra Amendolare, Flaviana Marzano, Micaela Braile, Virginia Sulfaro, Moira Paroni, Apollonia Tullo, Silvia Soddu, Luisa Guerrini
{"title":"Molecular mechanisms of thalidomide effectiveness on COVID-19 patients explained: ACE2 is a new ΔNp63α target gene","authors":"Laura Monteonofrio, Ilaria Virdia, Sara Pozzi, Roberto Quadri, Alessandra Amendolare, Flaviana Marzano, Micaela Braile, Virginia Sulfaro, Moira Paroni, Apollonia Tullo, Silvia Soddu, Luisa Guerrini","doi":"10.1007/s00109-024-02485-x","DOIUrl":"https://doi.org/10.1007/s00109-024-02485-x","url":null,"abstract":"<h3 data-test=\"abstract-sub-heading\">Abstract</h3><p>COVID-19 pandemic is caused by the SARS-CoV-2 virus, whose internalization and infection are mediated by the angiotensin-converting enzyme 2 (ACE2). The identification of novel approaches to tackle this step is instrumental for the development of therapies for the management of COVID-19 and other diseases with a similar mechanism of infection. Thalidomide, a drug sadly known for its teratogenic effects, has potent immunomodulatory and anti-inflammatory properties. Treatment with this drug has been shown to improve the immune functions of COVID-19 patients and proposed for the management of COVID-19 in clinical practice through drug repositioning. Here, we investigated the molecular details linking thalidomide to ACE2 and COVID-19, showing that in conditions mimicking SARS-CoV-2-associated cytokine storm, the transcription factor ΔNp63α and ACE2 are stabilized, and IL-8 production is increased. In such conditions, we found p63 to bind to and regulate the expression of the <i>ACE2</i> gene. We previously showed that ΔNp63α is degraded upon thalidomide treatment and now found that treatment with this drug—or with its analogue lenalidomide—downregulates ACE2 in a p63-dependent manner. Finally, we found that thalidomide treatment reduces in vitro infection by pseudo-SARS-CoV-2, a baculovirus pseudotyped with the SARS-CoV-2 spike protein. Overall, we propose the dual effect of thalidomide in reducing SARS-CoV-2 viral re-entry and inflammation through p63 degradation to weaken SARS-CoV-2 entry into host cells and mitigate lung inflammation, making it a valuable option in clinical management of COVID-19.</p><h3 data-test=\"abstract-sub-heading\">Key messages</h3>\u0000<ul>\u0000<li>\u0000<p>Thalidomide treatment results in p63-dependent ACE2 downregulation.</p>\u0000</li>\u0000<li>\u0000<p>ACE2 is a p63 transcriptional target.</p>\u0000</li>\u0000<li>\u0000<p>Thalidomide reduces the “cytokine storm” associated to COVID-19.</p>\u0000</li>\u0000<li>\u0000<p>Thalidomide prevents viral re-entry of SARS-CoV-2 by p63-dependent ACE2 downregulation.</p>\u0000</li>\u0000<li>\u0000<p>Thalidomide is a modulator of SARS-CoV-2 or other ACE2-dependent infections.</p>\u0000</li>\u0000<li>\u0000<p>ACE2 is modulated by a pharmacological substance.</p>\u0000</li>\u0000</ul>","PeriodicalId":16341,"journal":{"name":"Journal of Molecular Medicine","volume":"1 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142265384","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"The role of circular RNA targeting IGF2BPs in cancer—a potential target for cancer therapy","authors":"Xia Luo, Jiaxin Shi, Siyuan Wang, Xiaofeng Jin","doi":"10.1007/s00109-024-02488-8","DOIUrl":"https://doi.org/10.1007/s00109-024-02488-8","url":null,"abstract":"<p>Circular RNAs (circRNAs) are an interesting class of conserved single-stranded RNA molecules derived from exon or intron sequences produced by the reverse splicing of precursor mRNA. CircRNAs play important roles as microRNA sponges, gene splicing and transcriptional regulators, RNA-binding protein sponges, and protein/peptide translation factors. Abnormal functions of circRNAs and RBPs in tumor progression have been widely reported. Insulin-like growth factor-2 mRNA-binding proteins (IGF2BPs) are a highly conserved family of RBPs identified in humans that function as post-transcriptional fine-tuners of target transcripts. Emerging evidence suggests that IGF2BPs regulate the processing and metabolism of RNA, including its stability, translation, and localization, and participate in a variety of cellular functions and pathophysiology. In this review, we have summarized the roles and molecular mechanisms of circRNAs and IGF2BPs in cancer development and progression. In addition, we briefly introduce the role of other RNAs and IGF2BPs in cancer, discuss the current clinical applications and challenges faced by circRNAs and IGF2BPs, and propose future directions for this promising research field.</p>","PeriodicalId":16341,"journal":{"name":"Journal of Molecular Medicine","volume":"31 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142265385","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yarui Pan, Lan Xiang, Tingting Zhu, Haiyan Wang, Qi Xu, Faxue Liao, Juan He, Yongquan Wang
{"title":"Prefrontal cortex astrocytes in major depressive disorder: exploring pathogenic mechanisms and potential therapeutic targets","authors":"Yarui Pan, Lan Xiang, Tingting Zhu, Haiyan Wang, Qi Xu, Faxue Liao, Juan He, Yongquan Wang","doi":"10.1007/s00109-024-02487-9","DOIUrl":"https://doi.org/10.1007/s00109-024-02487-9","url":null,"abstract":"<h3 data-test=\"abstract-sub-heading\">Abstract</h3><p>Major depressive disorder (MDD) is a prevalent mental health condition characterized by persistent feelings of sadness and hopelessness, affecting millions globally. The precise molecular mechanisms underlying MDD remain elusive, necessitating comprehensive investigations. Our study integrates transcriptomic analysis, functional assays, and computational modeling to explore the molecular landscape of MDD, focusing on the DLPFC. We identify key genomic alterations and co-expression modules associated with MDD, highlighting potential therapeutic targets. Functional enrichment and protein–protein interaction analyses emphasize the role of astrocytes in MDD progression. Machine learning is employed to develop a predictive model for MDD risk assessment. Single-cell and spatial transcriptomic analyses provide insights into cell type–specific expression patterns, particularly regarding astrocytes. We have identified significant genomic alterations and co-expression modules associated with MDD in the DLPFC. Key genes involved in neuroactive ligand-receptor interaction pathways, notably in astrocytes, have been highlighted. Additionally, we developed a predictive model for MDD risk assessment based on selected key genes. Single-cell and spatial transcriptomic analyses underscored the role of astrocytes in MDD. Virtual screening of compounds targeting GPR37L1, KCNJ10, and PPP1R3C proteins has identified potential therapeutic candidates. In summary, our comprehensive approach enhances the understanding of MDD’s molecular underpinnings and offers promising opportunities for advancing therapeutic interventions, ultimately aiming to alleviate the burden of this debilitating mental health condition.</p><h3 data-test=\"abstract-sub-heading\">Key messages</h3>\u0000<ul>\u0000<li>\u0000<p>Our investigation furnishes insightful revelations concerning the dysregulation of astrocyte-associated processes in MDD.</p>\u0000</li>\u0000<li>\u0000<p>We have pinpointed specific genes, namely KCNJ10, PPP1R3C, and GPR37L1, as potential candidates warranting further exploration and therapeutic intervention.</p>\u0000</li>\u0000<li>\u0000<p>We incorporate a virtual screening of small molecule compounds targeting KCNJ10, PPP1R3C, and GPR37L1, presenting a promising trajectory for drug discovery in MDD.</p>\u0000</li>\u0000</ul>","PeriodicalId":16341,"journal":{"name":"Journal of Molecular Medicine","volume":"37 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142265387","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Marketa Rackova, Rafael Mattera, Michael Svaton, Filip Fencl, Veronika Kanderova, Karolina Spicakova, Sang Yoon Park, Ondrej Fabian, Miroslav Koblizek, Eva Fronkova, Juan S. Bonifacino, Karolina Skvarova Kramarzova
{"title":"Revising pathogenesis of AP1S1-related MEDNIK syndrome: a missense variant in the AP1S1 gene as a causal genetic lesion","authors":"Marketa Rackova, Rafael Mattera, Michael Svaton, Filip Fencl, Veronika Kanderova, Karolina Spicakova, Sang Yoon Park, Ondrej Fabian, Miroslav Koblizek, Eva Fronkova, Juan S. Bonifacino, Karolina Skvarova Kramarzova","doi":"10.1007/s00109-024-02482-0","DOIUrl":"https://doi.org/10.1007/s00109-024-02482-0","url":null,"abstract":"<h3 data-test=\"abstract-sub-heading\">Abstract</h3><p>MEDNIK syndrome is a rare autosomal recessive disease characterized by mental retardation, enteropathy, deafness, peripheral neuropathy, ichthyosis, and keratoderma, and caused by variants in the adaptor-related protein complex 1 subunit sigma 1 (<i>AP1S1</i>) gene. This gene encodes the σ1A protein, which is a subunit of the adaptor protein complex 1 (AP-1), a key component of the intracellular protein trafficking machinery. Previous work identified three <i>AP1S1</i> nonsense, frameshift and splice-site variants in MEDNIK patients predicted to encode truncated σ1A proteins, with consequent AP-1 dysfunction. However, two <i>AP1S1</i> missense variants (c.269 T > C and c.346G > A) were recently reported in patients who presented with severe enteropathy but no additional symptoms of MEDNIK. This condition was described as a novel non-syndromic form of congenital diarrhea caused specifically by the <i>AP1S1</i> missense variants. In this study, we report two patients with the same c.269 T > C variant, who, contrary to the previous cases, presented as complete MEDNIK syndrome. These data substantially revise the presentation of disorders associated with <i>AP1S1</i> gene variants and indicate that all the identified pathogenic <i>AP1S1</i> variants result in MEDNIK syndrome. We also provide a series of functional analyses that elucidate the impact of the c.269 T > C variant on σ1A function, contributing to a better understanding of the molecular pathogenesis of MEDNIK syndrome.</p><h3 data-test=\"abstract-sub-heading\">Key messages</h3>\u0000<ul>\u0000<li>\u0000<p>A missense <i>AP1S1</i> c.269 T > C (σ1A L90P) variant causes full MEDNIK syndrome.</p>\u0000</li>\u0000<li>\u0000<p>The σ1A L90P variant is largely unable to assemble into the AP-1 complex.</p>\u0000</li>\u0000<li>\u0000<p>The σ1A L90P variant fails to bind [DE]XXXL[LI] sorting motifs.</p>\u0000</li>\u0000<li>\u0000<p>The σ1A L90P variant results in loss-of-function of the protein.</p>\u0000</li>\u0000</ul>","PeriodicalId":16341,"journal":{"name":"Journal of Molecular Medicine","volume":"12 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142226797","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Qiumei Lan, Jie Li, Hanqing Zhang, Zijun Zhou, Yaxuan Fang, Bo Yang
{"title":"Mechanistic complement of autosomal dominant polycystic kidney disease: the role of aquaporins.","authors":"Qiumei Lan, Jie Li, Hanqing Zhang, Zijun Zhou, Yaxuan Fang, Bo Yang","doi":"10.1007/s00109-024-02446-4","DOIUrl":"https://doi.org/10.1007/s00109-024-02446-4","url":null,"abstract":"","PeriodicalId":16341,"journal":{"name":"Journal of Molecular Medicine","volume":"35 5","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-04-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140652058","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Analysis of the action mechanisms and targets of herbal anticonvulsants highlights opportunities for therapeutic engagement with refractory epilepsy.","authors":"Sobia Tabassum, Susan Shorter, S. Ovsepian","doi":"10.1007/s00109-024-02445-5","DOIUrl":"https://doi.org/10.1007/s00109-024-02445-5","url":null,"abstract":"","PeriodicalId":16341,"journal":{"name":"Journal of Molecular Medicine","volume":"3 6","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-04-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140658843","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"The role of annexins in central nervous system development and disease","authors":"Zachary B. White, Sindhu Nair, Markus Bredel","doi":"10.1007/s00109-024-02443-7","DOIUrl":"https://doi.org/10.1007/s00109-024-02443-7","url":null,"abstract":"<p>Annexins, a group of Ca<sup>2+</sup>-dependent phospholipid-binding proteins, exert diverse roles in neuronal development, normal central nervous system (CNS) functioning, neurological disorders, and CNS tumors. This paper reviews the roles of individual annexins (A1-A13) in these contexts. Annexins possess unique structural and functional features, such as Ca<sup>2+</sup>-dependent binding to phospholipids, participating in membrane organization, and modulating cell signaling. They are implicated in various CNS processes, including endocytosis, exocytosis, and stabilization of plasma membranes. Annexins exhibit dynamic roles in neuronal development, influencing differentiation, proliferation, and synaptic formation in CNS tissues. Notably, annexins such as ANXA1 and ANXA2 play roles in apoptosis and blood-brain barrier (BBB) integrity. Neurological disorders, including Alzheimer’s disease, multiple sclerosis, and depression, involve annexin dysregulation, influencing neuroinflammation, blood-brain barrier integrity, and stress responses. Moreover, annexins contribute to the pathogenesis of CNS tumors, either promoting or suppressing tumor growth, angiogenesis, and invasion. Annexin expression patterns vary across different CNS tumor types, providing potential prognostic markers and therapeutic targets. This review underscores the multifaceted roles of annexins in the CNS, highlighting their importance in normal functioning, disease progression, and potential therapeutic interventions.</p>","PeriodicalId":16341,"journal":{"name":"Journal of Molecular Medicine","volume":"163 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-04-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140629646","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"The role of the immunosuppressive PD-1/PD-L1 checkpoint pathway in the aging process and age-related diseases","authors":"Antero Salminen","doi":"10.1007/s00109-024-02444-6","DOIUrl":"https://doi.org/10.1007/s00109-024-02444-6","url":null,"abstract":"<h3 data-test=\"abstract-sub-heading\">Abstract</h3><p>The accumulation of senescent cells within tissues is a hallmark of the aging process. Senescent cells are also commonly present in many age-related diseases and in the cancer microenvironment. The escape of abnormal cells from immune surveillance indicates that there is some defect in the function of cytotoxic immune cells, e.g., CD8<sup>+</sup> T cells and natural killer (NK) cells. Recent studies have revealed that the expression of programmed death-ligand 1 (PD-L1) protein is abundantly increased in senescent cells. An increase in the amount of PD-L1 protein protects senescent cells from clearance by the PD-1 checkpoint receptor in cytotoxic immune cells. In fact, the activation of the PD-1 receptor suppresses the cytotoxic properties of CD8<sup>+</sup> T and NK cells, promoting a state of immunosenescence. The inhibitory PD-1/PD-L1 checkpoint pathway acts in cooperation with immunosuppressive cells; for example, activation of PD-1 receptor can enhance the differentiation of regulatory T cells (Treg), myeloid-derived suppressor cells (MDSC), and M2 macrophages, whereas the cytokines secreted by immunosuppressive cells stimulate the expression of the immunosuppressive PD-L1 protein. Interestingly, many signaling pathways known to promote cellular senescence and the aging process are crucial stimulators of the expression of PD-L1 protein, e.g., epigenetic regulation, inflammatory mediators, mTOR-related signaling, cGAS-STING pathway, and AhR signaling. It seems that the inhibitory PD-1/PD-L1 immune checkpoint axis has a crucial role in the accumulation of senescent cells and thus it promotes the aging process in tissues. Thus, the blockade of the PD-1/PD-L1 checkpoint signaling might be a potential anti-aging senolytic therapy.</p><h3 data-test=\"abstract-sub-heading\">Key messages</h3>\u0000<ul>\u0000<li>\u0000<p>Senescent cells accumulate within tissues during aging and age-related diseases.</p>\u0000</li>\u0000<li>\u0000<p>Senescent cells are able to escape immune surveillance by cytotoxic immune cells.</p>\u0000</li>\u0000<li>\u0000<p>Expression of programmed death-ligand 1 (PD-L1) markedly increases in senescent cells.</p>\u0000</li>\u0000<li>\u0000<p>Age-related signaling stimulates the expression of PD-L1 protein in senescent cells.</p>\u0000</li>\u0000<li>\u0000<p>Inhibitory PD-1/PD-L1 checkpoint pathway suppresses clearance of senescent cells.</p>\u0000</li>\u0000</ul>","PeriodicalId":16341,"journal":{"name":"Journal of Molecular Medicine","volume":"4 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-04-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140574869","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Metabolomic profiling of maternal plasma identifies inverse associations of acetate and urea with anti-SARS-CoV-2 antibody titers following COVID-19 vaccination during pregnancy","authors":"An-Shine Chao, Chiao-Yun Lin, Meng-Han Chiang, Kuan-Ying Lu, Cheng-Kun Tsai, Kuan-Ju Chen, Chih-Wei Chien, Ting-Shu Wu, Yao-Lung Chang, Angel Chao, Gigin Lin, Chih-Yung Chiu","doi":"10.1007/s00109-024-02438-4","DOIUrl":"https://doi.org/10.1007/s00109-024-02438-4","url":null,"abstract":"<h3 data-test=\"abstract-sub-heading\">Abstract</h3><p>We conducted a comprehensive metabolomic analysis of plasma samples obtained from pregnant women who displayed varying post-vaccination antibody titers after receiving mRNA-1273-SARS-CoV-2 vaccines. The study involved 62 pregnant women, all of whom had been vaccinated after reaching 24 weeks of gestation. To quantify post-vaccination plasma antibody titers, we employed binding antibody units (BAU) in accordance with the World Health Organization International Standard. Subsequently, we classified the study participants into three distinct BAU/mL categories: those with high titers (above 2000), medium titers (ranging from 1000 to 2000), and low titers (below 1000). Plasma metabolomic profiling was conducted using <sup>1</sup>H nuclear magnetic resonance spectroscopy, and the obtained data were correlated with the categorized antibody titers. Notably, in pregnant women exhibiting elevated anti-SARS-CoV-2 antibody titers, reduced plasma concentrations of acetate and urea were observed. A significant negative correlation between these compounds and antibody titers was also evident. An analysis of metabolomics pathways revealed significant inverse associations between antibody titers and four distinct amino acid metabolic pathways: (1) biosynthesis of phenylalanine, tyrosine, and tryptophan; (2) biosynthesis of valine, leucine, and isoleucine; (3) phenylalanine metabolism; and (4) degradation of valine, leucine, and isoleucine. Additionally, an association between the synthesis and degradation pathways of ketone bodies was evident. In conclusion, we identified different metabolic pathways that underlie the diverse humoral responses triggered by COVID-19 mRNA vaccines during pregnancy. Our data hold significant implications for refining COVID-19 vaccination approaches in expectant mothers.</p><h3 data-test=\"abstract-sub-heading\">\u0000<b>Key messages</b>\u0000</h3><ul>\u0000<li>\u0000<p>Anti-SARS-CoV-2 antibody titers decline as the number of days since COVID-19 vaccination increases.</p>\u0000</li>\u0000<li>\u0000<p>Anti-SARS-CoV-2 antibody titers are inversely associated with acetate, a microbial-derived metabolite, and urea.</p>\u0000</li>\u0000<li>\u0000<p>Amino acid metabolism is significantly associated with SARS-CoV-2 antibody titers.</p>\u0000</li>\u0000</ul>","PeriodicalId":16341,"journal":{"name":"Journal of Molecular Medicine","volume":"27 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-04-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140575243","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}