Moritz Pernecker, Miriam Dibos, Sophie Götz, Rouvier Al-Monajjed, Vivien Barz, Christian Albiker, Rita Schröter, Ute Neugebauer, Lena Ludwig-Radtke, R Verena Taudte, Thomas Vogl, Giuliano Ciarimboli
{"title":"Guilty by association: direct interaction with the tetraspanin CD63 suggests a role for organic cation transporter 3 in histamine release from granulocytes.","authors":"Moritz Pernecker, Miriam Dibos, Sophie Götz, Rouvier Al-Monajjed, Vivien Barz, Christian Albiker, Rita Schröter, Ute Neugebauer, Lena Ludwig-Radtke, R Verena Taudte, Thomas Vogl, Giuliano Ciarimboli","doi":"10.1186/s12929-025-01158-2","DOIUrl":"10.1186/s12929-025-01158-2","url":null,"abstract":"<p><strong>Background: </strong>The organic cation transporter 3 (OCT3) is a ubiquitous transporter that carries both endogenous and exogenous substrates, such as histamine and cisplatin. Our investigations have shown that OCT3 directly interacts with the tetraspanin CD63. CD63 is a marker for activated basophils and mast cells, which are granulocytes capable of rapidly releasing large amounts of histamine. This makes them key players in the development of allergic reactions.</p><p><strong>Methods and results: </strong>In this work, we demonstrated that OCT3 is present in murine and human basophils and is strongly colocalized with CD63 in a specific region of the plasma membrane, particularly after cell activation leading to histamine release. Furthermore, we confirmed that part of the histamine release from basophils is mediated by OCT3. In a mouse model of contact dermatitis, the presence of OCT3 is crucial for determining the severity of the allergic reaction. The presence of CD63 also seems to be important for regulating the allergic response, although it does not directly affect histamine secretion. RNA-Seq and metabolome analyses revealed that wild-type mice and mice with genetic deletion of OCT3 (OCT3<sup>-/-</sup>) are phenotypically very similar, and that the observed effects in OCT3<sup>-/-</sup> organisms can be attributed mainly to the genetic deletion of the OCT3 transporter.</p><p><strong>Conclusions: </strong>In conclusion, OCT3 is a transporter for histamine in granulocytes, which plays a crucial role in determining the intensity of allergic reactions and may be a target for interventions aimed at reducing their severity.</p>","PeriodicalId":15365,"journal":{"name":"Journal of Biomedical Science","volume":"32 1","pages":"68"},"PeriodicalIF":9.0,"publicationDate":"2025-07-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12255121/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144618124","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}
Sanaa Dekkar, Kamilia Mahloul, Amandine Falco, Karidia Konate, Romane Pisteur, Sarah Maurel, Laurent Maïmoun, Norbert Chauvet, Prisca Boisguérin, David Nocca, Ariane Sultan, Florian Pallot, Guillaume Walther, Nicolas Cenac, Cyril Breuker, Sandrine Faure, Pascal de Santa Barbara
{"title":"Obesity induces phenotypic switching of gastric smooth muscle cells through the activation of the PPARD/PDK4/ANGPTL4 pathway.","authors":"Sanaa Dekkar, Kamilia Mahloul, Amandine Falco, Karidia Konate, Romane Pisteur, Sarah Maurel, Laurent Maïmoun, Norbert Chauvet, Prisca Boisguérin, David Nocca, Ariane Sultan, Florian Pallot, Guillaume Walther, Nicolas Cenac, Cyril Breuker, Sandrine Faure, Pascal de Santa Barbara","doi":"10.1186/s12929-025-01163-5","DOIUrl":"10.1186/s12929-025-01163-5","url":null,"abstract":"<p><strong>Background: </strong>Clinical research has identified stomach dysmotility as a common feature of obesity. However, the specific mechanisms driving gastric emptying dysfunction in patients with obesity remain largely unknown. In this study, we investigated potential mechanisms by focusing on the homeostasis of gastric smooth muscle.</p><p><strong>Methods: </strong>An obese mouse model was established using a high-fat diet (HFD). Immunofluorescence analysis and Western blotting were employed to assess smooth muscle status using stage-specific markers. An in vitro culture model of differentiated human gastric smooth muscle cells (SMCs) was treated with lipids, siRNA-peptide-based nanoparticles and pharmaceutical compounds. Global lipidomic and RNA sequencing analyses were performed. The findings were evaluated in patients with obesity, using gastric samples from individuals who underwent sleeve gastrectomy, to evaluate their clinical relevance.</p><p><strong>Results: </strong>The smooth muscle layers in gastric tissue from both mice fed on a HFD as well as patients with obesity exhibited altered differentiation status. Treatment of differentiated human gastric SMCs with lipids phenocopies these alterations and is associated with increased expression of PDK4 and ANGPTL4. Inhibition of PDK4 or ANGPTL4 upregulation prevents these lipid-induced modifications. PPARD activation stimulates PDK4 and ANGPTL4 upregulation, leading to SMC dedifferentiation. Notably, PDK4 and ANGPTL4 levels correlate with immaturity and alteration of gastric smooth muscle in patients with obesity.</p><p><strong>Conclusions: </strong>Obesity triggers a phenotypic change in gastric SMCs, driven by the activation of the PPARD/PDK4/ANGPTL4 pathway. These mechanistic insights offer potential biomarkers for diagnosing stomach dysmotility in patients with obesity.</p>","PeriodicalId":15365,"journal":{"name":"Journal of Biomedical Science","volume":"32 1","pages":"67"},"PeriodicalIF":9.0,"publicationDate":"2025-07-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12254972/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144618125","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}
Katia Fettucciari, Luigi Cari, Andrea Spaterna, Rachele Del Sordo, Filippo Tavanti, Pierfrancesco Marconi, Gabrio Bassotti
{"title":"Clostridioides difficile meets the adenosine system: the art of manipulating host homeostasis.","authors":"Katia Fettucciari, Luigi Cari, Andrea Spaterna, Rachele Del Sordo, Filippo Tavanti, Pierfrancesco Marconi, Gabrio Bassotti","doi":"10.1186/s12929-025-01160-8","DOIUrl":"10.1186/s12929-025-01160-8","url":null,"abstract":"<p><strong>Background: </strong>Adenosine is a ubiquitous endogenous molecule capable of influencing several pathophysiological aspects. The adenosine system is extremely complex, starting from the generation of intracellular and extracellular adenosine, the regulation of its levels, and its action on four different receptors that vary in affinity and distribution in the different cell types and tissues. The most relevant effects of adenosine during infections and inflammation are documented on all types of immune cells, including those of adaptive immunity (T lymphocytes, B lymphocytes, regulatory cells) and of natural immunity (macrophages, polymorphonuclear cells, dendritic cells, natural killer). Of interest, the adenosine system is also strongly involved in the pathophysiology of colonic cells. Clostridioides difficile (C. difficile), responsible for 15-20% of all cases of antibiotic-associated diarrhea, is an infection that has been evolving over the past two decades due to the unstoppable spread of C. difficile in the anthropized environment and the progressive human colonization. The pathological activity of C. difficile is due to toxin A (TcdA) and B (TcdB) which profoundly alter the homeostasis of the adenosine system, acting both at the level of its generation and on the expression and regulation of adenosine receptors. The final effect consists in an attenuation of the inflammatory response to favor the persistence of the C. difficile infection.</p><p><strong>Conclusion: </strong>This review highlights a new ability of C. difficile, through its Tcds, of manipulating the host to its advantage.</p>","PeriodicalId":15365,"journal":{"name":"Journal of Biomedical Science","volume":"32 1","pages":"66"},"PeriodicalIF":9.0,"publicationDate":"2025-07-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12255077/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144618123","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}
Ángel J García-Yagüe, Lucía Cañizares-Moscato, José Antonio Encinar, Eduardo Cazalla, Raquel Fernández-Ginés, Maribel Escoll, Ana I Rojo, Antonio Cuadrado
{"title":"A novel β-TrCP1/NRF2 interaction inhibitor for effective anti-inflammatory therapy.","authors":"Ángel J García-Yagüe, Lucía Cañizares-Moscato, José Antonio Encinar, Eduardo Cazalla, Raquel Fernández-Ginés, Maribel Escoll, Ana I Rojo, Antonio Cuadrado","doi":"10.1186/s12929-025-01157-3","DOIUrl":"10.1186/s12929-025-01157-3","url":null,"abstract":"<p><strong>Background: </strong>Non-communicable chronic diseases are characterized by low-grade inflammation and oxidative stress. Extensive research has identified the transcription factor NRF2 as a potential therapeutic target. Current NRF2 activators, designed to inhibit its repressor KEAP1, often exhibit undesirable side effects. As an alternative approach, we previously developed PHAR, a protein-protein interaction inhibitor of β-TrCP1/NRF2, which promotes NRF2 activation. Using the same in silico screening platform, we have now identified a novel compound, P10. This small molecule selectively interferes with the β-TrCP1/NRF2 interaction, leading to NRF2 stabilization and transcriptional activation of its target genes in a β-TrCP1-dependent manner, demonstrating promising effects in a liver model of acute inflammation.</p><p><strong>Methods: </strong>After an in silico screening of ∼1 million compounds, including molecular docking analysis, ADMET evaluation, and molecular dynamics simulations, we identified and characterized a novel small molecule, P10, which inhibits β-TrCP1/NRF2 interaction. The compound was validated using luciferase reporter assays, co-immunoprecipitation, and ubiquitination experiments. The specificity of P10 was assessed by comparing NRF2 signatures in wild-type and Nrf2-null cells. The impact of NRF2 activation induced by P10 was investigated by evaluating its antioxidant and anti-inflammatory responses against tert-butyl hydroperoxide and lipopolysaccharide, respectively. Finally, wild-type and Nrf2-null mice were administered P10 intraperitoneally at a dose of 20 mg/kg daily for five consecutive days. Four hours before sacrifice, all animals received a lipopolysaccharide (LPS) injection at 10 mg/kg.</p><p><strong>Results: </strong>P10 selectively disrupts the interaction between β-TrCP1 and NRF2, thereby inhibiting β-TrCP1-mediated ubiquitination of NRF2 and leading to the upregulation of NRF2 target genes. Additionally, P10 mitigates oxidative stress induced by tert-butyl hydroperoxide and reduces pro-inflammatory markers in an NRF2-dependent manner in macrophages treated with lipopolysaccharide. In a preclinical model of liver inflammation, P10 specifically targets the liver, significantly attenuating lipopolysaccharide-induced inflammation through the activation of NRF2. This is demonstrated by decreased expression of inflammatory cytokine genes and a reduction in F4/80-stained liver macrophages. Notably, this anti-inflammatory effect is absent in Nrf2-knockout mice, confirming its NRF2-dependent mechanism of action.</p><p><strong>Conclusions: </strong>P10 emerges as a promising NRF2 activator by selectively disrupting the β-TrCP1/NRF2 interaction, highlighting its potential as a therapeutic agent for diseases presenting acute liver inflammation.</p>","PeriodicalId":15365,"journal":{"name":"Journal of Biomedical Science","volume":"32 1","pages":"65"},"PeriodicalIF":9.0,"publicationDate":"2025-07-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12247323/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144618122","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}
Daniela Scribano, Claudia Tito, Astri D Tagueha, Martina Pasqua, Luciana De Angelis, Francesco Fazi, Dolores Limongi, Marta De Angelis, Lucia Nencioni, Anna Teresa Palamara, Cecilia Ambrosi
{"title":"Goblet cell breakdown: transcriptomics reveals Acinetobacter baumannii early and robust inflammatory response in differentiated human bronchial epithelial cells.","authors":"Daniela Scribano, Claudia Tito, Astri D Tagueha, Martina Pasqua, Luciana De Angelis, Francesco Fazi, Dolores Limongi, Marta De Angelis, Lucia Nencioni, Anna Teresa Palamara, Cecilia Ambrosi","doi":"10.1186/s12929-025-01159-1","DOIUrl":"10.1186/s12929-025-01159-1","url":null,"abstract":"<p><strong>Background: </strong>The airway epithelium represents the first line of defense of the lungs, functioning both as a physical barrier as well as an active immune modulator. However, in the last years, pneumonia caused by the opportunistic pathogen Acinetobacter baumannii have become difficult to treat due to the increase of the number of extensively drug resistant strains. In this study, we report for the first time the use of an ex vivo air-liquid interface (ALI) model of differentiated human bronchial epithelial cells to unravel the early response to A. baumannii infection.</p><p><strong>Methods: </strong>Epithelial integrity, tissue architecture, and goblet cell function were assessed through FITC-dextran permeability assays, hematoxylin and eosin staining, and indirect immunofluorescence. Transcriptomic profiling was performed to characterize host gene expression changes.</p><p><strong>Results: </strong>Initial tissue damage began as early as at 4 h post-infection (hpi); at 24 hpi, goblet cell hypertrophy, reduced mucin secretion, and compromised epithelial integrity were highly evident. Transcriptomic data at 4 hpi revealed 668 differentially expressed genes (441 upregulated, 227 downregulated), mainly involved in a strong pro-inflammatory response and characterized by IL-8/CCL20-driven neutrophil recruitment and type 2 cytokine activation (IL-4, IL-13). Noteworthy, genes related to cytoskeletal organization, adhesion, and extracellular matrix remodeling were significantly altered, suggesting a bacterial mechanism to enhanced tissue dissemination. The PI3K-Akt survival pathway was inhibited, with downregulation of PIK3R1 and PIK3R2 genes, implying the induction of apoptosis/cell death and epithelial damage. Our findings are in agreement with previous in vivo studies, further strengthening the value of our ALI model in mimicking the early infection response of bronchial cells to A. baumannii infection.</p><p><strong>Conclusion: </strong>Our data highlight the early molecular mechanisms underlying A. baumannii pathogenesis and open new avenues for future investigations for therapeutic interventions.</p>","PeriodicalId":15365,"journal":{"name":"Journal of Biomedical Science","volume":"32 1","pages":"63"},"PeriodicalIF":9.0,"publicationDate":"2025-07-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12239265/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144600558","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}
Weronika Gonciarz, Ewa Balcerczak, Marek Brzeziński, Agnieszka Jeleń, Agnieszka J Pietrzyk-Brzezińska, Vedha Hari B Narayanan, Magdalena Chmiela
{"title":"Chitosan-based formulations for therapeutic applications. A recent overview.","authors":"Weronika Gonciarz, Ewa Balcerczak, Marek Brzeziński, Agnieszka Jeleń, Agnieszka J Pietrzyk-Brzezińska, Vedha Hari B Narayanan, Magdalena Chmiela","doi":"10.1186/s12929-025-01161-7","DOIUrl":"10.1186/s12929-025-01161-7","url":null,"abstract":"<p><p>Chitosan is a cationic natural polymer composed of glucosamine and N-acetylglucosamine residues that are held together by a glycosidic bond. Chitosan has many excellent properties, including physicochemical properties, i.e., stability in the natural environment, chelation of metal ions, high sorption properties, biological properties such as biocompatibility and biological activity, ecological properties resulting from biodegradability, and physiological properties, which include non-toxicity, and economic affordability, and is used in various biomedical and industrial applications. The presented article highlights recent developments in chitosan-based formulations for the treatment of bacteria, viruses, cancer, or gastroesophageal reflux disease. Moreover, chitosan-derived biomaterials can also be used in regenerative medicine or food packaging to prevent contamination by pathogenic microorganisms. In summary, this is a valuable compilation in this emerging field that focuses on the biomedical application of chitosan-based biomaterials.</p>","PeriodicalId":15365,"journal":{"name":"Journal of Biomedical Science","volume":"32 1","pages":"62"},"PeriodicalIF":9.0,"publicationDate":"2025-07-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12239286/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144591301","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}
{"title":"Cardiac-specific overexpression of PRMT5 exacerbates pressure overload-induced hypertrophy and heart failure.","authors":"Yasufumi Katanasaka, Yoichi Sunagawa, Ryoga Sakurai, Mikuto Tojima, Ryuya Naruta, Yuya Hojo, Yuto Kawase, Toshihide Hamabe-Horiike, Kiyoshi Mori, Koji Hasegawa, Tatsuya Morimoto","doi":"10.1186/s12929-025-01162-6","DOIUrl":"10.1186/s12929-025-01162-6","url":null,"abstract":"<p><strong>Background: </strong>Various epigenetic modifiers are involved in the regulation of gene expression during pathological cardiac hypertrophy-a critical event in the development of heart failure. Our previous research has demonstrated that protein arginine methyltransferase 5 (PRMT5) in cardiac fibroblasts is a crucial epigenetic writer implicated in pathological cardiac fibrosis. Moreover, treatment with a PRMT5 inhibitor also suppressed cardiac hypertrophy in mice after transverse aortic constriction (TAC) surgery. However, as the functional role of PRMT5 in cardiomyocytes remains to be fully elucidated in pathological cardiac hypertrophy and systolic dysfunction, this study aimed to clarify the gain-of-function of PRMT5 in cardiomyocytes.</p><p><strong>Methods: </strong>Cardiac-specific PRMT5 transgenic (PRMT5-TG) mice were generated to evaluate the gain-of-function of PRMT5 in cardiac hypertrophy and dysfunction in male mice undergoing TAC surgery. Cardiac function and myocardial cell hypertrophy were evaluated in wild-type (WT) and PRMT5-TG mice after TAC surgery. To elucidate the molecular mechanistic basis through which PRMT5 induces cardiomyocyte hypertrophy, we examined epigenetic modifications of histones in cardiomyocytes.</p><p><strong>Results: </strong>Echocardiography revealed that fractional shortening was reduced in PRMT5-TG mice compared to WT mice after TAC surgery. Both heart weight/BW and lung weight/BW ratios increased significantly more in PRMT5-TG than in WT mice. Histological analyses showed that cardiomyocyte diameter and perivascular fibrosis were elevated in PRMT5-TG mice in comparison to WT mice. Hypertrophic gene expression significantly increased in PRMT5-TG mice after TAC surgery. In primary cultured neonatal rat cardiac myocytes, EPZ015666, a specific inhibitor of PRMT5, and PRMT5 knockdown significantly inhibited phenylephrine (PE)-induced cell hypertrophy. Cardiac overexpression of PRMT5 promoted the acetylation of H3K9, a histone marker associated with cardiomyocyte hypertrophy, and the histone acetyltransferase activity of p300. Conversely, treatment with EPZ015666 reduced the acetylation of H3K9 induced by TAC surgery and PE treatment. Finally, we found that PRMT5 interacts with and methylates p300 at R200. The R200 point mutation in p300 abolished PRMT5-mediated enhancement of its histone acetyltransferase activity.</p><p><strong>Conclusions: </strong>The gain-of-function of PRMT5 in cardiomyocytes exacerbates pressure overload-induced cardiac hypertrophy and left ventricular systolic dysfunction, at least partially, through p300 methylation and histone acetyltransferase activation.</p>","PeriodicalId":15365,"journal":{"name":"Journal of Biomedical Science","volume":"32 1","pages":"61"},"PeriodicalIF":9.0,"publicationDate":"2025-07-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12229037/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144575570","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}
Antonella Campanale, Dario Siniscalco, Vincenzo Di Marzo
{"title":"The endocannabinoidome-gut microbiome-brain axis as a novel therapeutic target for autism spectrum disorder.","authors":"Antonella Campanale, Dario Siniscalco, Vincenzo Di Marzo","doi":"10.1186/s12929-025-01145-7","DOIUrl":"10.1186/s12929-025-01145-7","url":null,"abstract":"<p><strong>Introduction: </strong>Autism spectrum disorder (ASD) is characterized by disruption of the gut-brain axis, which leads to behavioral, psychiatric, metabolic and gastrointestinal symptoms. Effective ASD treatments are limited. Research highlights the roles of the endocannabinoidome (eCBome) and gut microbiome (GM), both crucial for brain and gut function. This review summarizes research on therapeutic targets within the eCBome-GM-brain axis for ASD and related comorbidities.</p><p><strong>Discussion: </strong>Evidence suggests that reduced levels of eCBome mediators, like oleoylethanolamide and anandamide, and altered cannabinoid type 1 and type 2 (CB1 and CB2) receptors activity may contribute to ASD symptoms, making them promising targets. Modulating the eCBome-GM-brain axis with inhibitors of fatty acid amide hydrolase (FAAH), transient receptor potential vanilloid 1, and monoacylglycerol lipase (MAGL) may improve repetitive, stereotypical, and sensory behaviors, and alleviate sociability impairments, depression and anxiety. However, inhibition of FAAH and MAGL may also induce ADHD-like behaviors, which can be reversed by CB1 inverse agonists. Targeting metabotropic glutamate receptor 5 to increase levels of the eCBome mediator 2-arachidonoylglycerol (2-AG) may benefit ASD-related behaviors. eCBome mediators such as 2-AG, 1/2-palmitoylglycerol and palmitoylethanolamide may also help manage ASD- and GI-related symptoms, and systemic inflammation. Other potential therapeutic targets that deserve further investigation are eCBome-related receptors G-protein-coupled receptor 55 and peroxisome proliferator-activated receptors-alpha and -gamma, and the cyclooxygenase-2/prostaglandin E2 pathway, which may address hyperactivity and repetitive behaviors. Additionally, mucin-degrading genera like Akkermansia and Ruminococcus may improve ASD-related GI symptoms such as hypersensitivity and inflammation. Selective antibiotics against specific Clostridium strains may improve irritability and aggression. In ASD with ADHD and OCD, treatments may involve modulating the CB1 and CB2 receptor, and bacterial families like Ruminococcaceae and Lachnospiraceae. Lastly, modulating the abundance of anti-inflammatory genera like Prevotella and Anaeroplasma, and taxa associated with gut health such as Roseburia may also offer therapeutic value.</p><p><strong>Conclusion: </strong>The eCBome-GM-brain axis is a promising target for ASD treatment, meriting further clinical and preclinical research.</p>","PeriodicalId":15365,"journal":{"name":"Journal of Biomedical Science","volume":"32 1","pages":"60"},"PeriodicalIF":9.0,"publicationDate":"2025-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12220735/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144553598","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}
Ming-Shiu Lin, Tai-Ling Chao, Yu-Chi Chou, Yao Yi, Ci-Ling Chen, Kuo-Yen Huang, Sui-Yuan Chang, Pan-Chyr Yang
{"title":"The ACE2 decoy receptor can overcome immune escape by rapid mutating SARS-CoV-2 variants and reduce cytokine induction and clot formation.","authors":"Ming-Shiu Lin, Tai-Ling Chao, Yu-Chi Chou, Yao Yi, Ci-Ling Chen, Kuo-Yen Huang, Sui-Yuan Chang, Pan-Chyr Yang","doi":"10.1186/s12929-025-01156-4","DOIUrl":"10.1186/s12929-025-01156-4","url":null,"abstract":"<p><strong>Background: </strong>The COVID-19 pandemic continues to affect the world in 2025. The rapid mutation of SARS-CoV-2 results in breakthrough infections and diminishes the efficacy of vaccines and anti-viral drugs. The severity of the disease varies across different variants, and the underlying mechanisms driving these differences remain unclear. This study explores the relationship between different Spike variants and cytotoxicity, aiming to determine whether the humanized decoy receptor ACE2-Fc can neutralize spikes from diverse variants, offering a solution to overcome rapid mutating SARS-CoV-2 induced immune escape.</p><p><strong>Methods: </strong>We co-cultured 293 T-ACE2 cells with 293 T cells transfected with various Spike protein variants or used H1650-ACE2 cells transfected with these Spike variants. This allowed us to observe the effects of different Spike mutations, specifically focusing on cell fusion, cytotoxicity, and cytokine release from human peripheral blood mononuclear cells. Flow cytometry is employed to determine if ACE2-Fc can recognize different Spike variants. We also assess the ability of ACE2-Fc to inhibit infection, cell fusion, cytotoxicity, and cytokine release through pseudovirus infections or Spike protein transfections. Additionally, we use actual viruses from SARS-CoV-2 patients to validate the impacts of Spike mutations and the effectiveness of ACE2-Fc. Furthermore, human plasma is utilized to evaluate ACE2-Fc's capability to inhibit Spike-induced clot formation.</p><p><strong>Results: </strong>We found that different Spike variants, particularly those with enhancements at the S2' site, increased cell-cell fusion capability, which correlated positively with cytotoxicity and cytokine IL-6 and TNF-α released from PBMCs. ACE2-Fc recognized spikes from wide-range of variants, including wild type, Alpha, Delta, Delta plus, Lambda, BA.2, BA.2.75, BA.5, BF.7, BQ.1, XBB.1, JN.1, KP.2, and KP.3, and effectively prevented these spike-expressing pseudo-viruses from entering host cells. Crucially, ACE2-Fc can prevent spike-induced cell fusion, thereby reducing subsequent cytotoxicity and the release of IL-6 and TNF-α from PBMCs. ACE2-Fc also effectively reduces plasma clot formation induced by trimeric spike proteins.</p><p><strong>Conclusions: </strong>These findings demonstrated that ACE2-Fc could effectively combat the infection of rapidly mutating SARS-CoV-2, providing a potential solution to overcome immune evasion.</p>","PeriodicalId":15365,"journal":{"name":"Journal of Biomedical Science","volume":"32 1","pages":"59"},"PeriodicalIF":9.0,"publicationDate":"2025-06-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12199494/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144505828","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}