A comprehensive understanding of human systemic immune responses to mosquito-borne dengue virus (DENV) infection is vital for addressing challenges posed by viral heterologous serotypes and potential adverse memory immune responses. Asymptomatic DENV infection offers an opportunity to explore protective immunity because infected individuals effectively clear the virus without symptomatic manifestations. However, data on asymptomatic dengue are scarce because of limited sample availability during silent viremia. Here, we conducted single-cell RNA and immune receptor sequencing of peripheral blood mononuclear cells (PBMCs) from donors with varying disease severities including asymptomatic dengue and performed longitudinal analysis in a symptomatic dengue cohort, enabling identification of distinct immune responses. In asymptomatic dengue, we observed potential indications of enhanced viral antigen processing via MHC-I, correlating with increased CD8 effector T cell activities, distinct NK cell profiles, and enriched IGHA1+ plasmablasts. In contrast, symptomatic dengue cases exhibited indications toward antibody-mediated viral entry, elevated type I interferon responses, and IL-10–associated expansion of IGHG1+ plasmablasts with biased V(D)J gene usage and a shared B cell receptor clonotype network. Our study reports a gene expression and immune receptor repertoire resource for systemic immune responses to DENV infection and suggests distinct mechanisms for potential protection and pathogenicity in individuals with asymptomatic compared with symptomatic dengue.
{"title":"Distinct systemic immune responses in asymptomatic and symptomatic dengue virus infection","authors":"Waradon Sungnak, Natnicha Jiravejchakul, Tiraput Poonpanichakul, Chawinya Trakoolsoontorn, Sirawit Srikor, Anunya Opasawatchai, Jantarika Arora, Damita Jevapatarakul, Narita Thungsatianpun, Sarintip Nguantad, Juthamard Chantaraamporn, Pattarakul Pakchotanon, Nuntaya Punyadee, Thaneeya Duangchinda, Panisadee Avirutnan, DENFREE Thailand, Juthathip Mongkolsapaya, Kerstin B. Meyer, Oranart Matangkasombut, Varodom Charoensawan, Sarah A. Teichmann, Ponpan Matangkasombut","doi":"10.1126/scitranslmed.ads5932","DOIUrl":"10.1126/scitranslmed.ads5932","url":null,"abstract":"<div >A comprehensive understanding of human systemic immune responses to mosquito-borne dengue virus (DENV) infection is vital for addressing challenges posed by viral heterologous serotypes and potential adverse memory immune responses. Asymptomatic DENV infection offers an opportunity to explore protective immunity because infected individuals effectively clear the virus without symptomatic manifestations. However, data on asymptomatic dengue are scarce because of limited sample availability during silent viremia. Here, we conducted single-cell RNA and immune receptor sequencing of peripheral blood mononuclear cells (PBMCs) from donors with varying disease severities including asymptomatic dengue and performed longitudinal analysis in a symptomatic dengue cohort, enabling identification of distinct immune responses. In asymptomatic dengue, we observed potential indications of enhanced viral antigen processing via MHC-I, correlating with increased CD8 effector T cell activities, distinct NK cell profiles, and enriched <i>IGHA1<sup>+</sup></i> plasmablasts. In contrast, symptomatic dengue cases exhibited indications toward antibody-mediated viral entry, elevated type I interferon responses, and IL-10–associated expansion of <i>IGHG1</i><sup>+</sup> plasmablasts with biased V(D)J gene usage and a shared B cell receptor clonotype network. Our study reports a gene expression and immune receptor repertoire resource for systemic immune responses to DENV infection and suggests distinct mechanisms for potential protection and pathogenicity in individuals with asymptomatic compared with symptomatic dengue.</div>","PeriodicalId":21580,"journal":{"name":"Science Translational Medicine","volume":"17 829","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145766427","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-17DOI: 10.1126/scitranslmed.adp5088
Carla L. Alves, Leena Karimi, Mikkel G. Terp, Mie K. Jakobsen, Fiona H. Zhou, Benedetta Policastro, Nikoline Nissen, Lene E. Johansen, Tina Ravnsborg, Leila Eshraghi, Sana Tamboowala, Ole N. Jensen, Elgene Lim, Henrik J. Ditzel
Combined CDK4/6 inhibitor (CDK4/6i) and endocrine therapy (ET) improves outcomes in advanced estrogen receptor–positive (ER+) breast cancer, but emergence of resistance to this combination underscores the pressing need for alternative therapeutic strategies. A promising approach involves adding an inhibitor of the PI3K/AKT/mTOR pathway to the standard combined CDK4/6i and ET, but selecting the most effective inhibitors and their optimal combinations has proven to be challenging. Here, we compared the efficacy of various triple combinations using single- or dual-point PI3K/AKT/mTOR pathway inhibitors in breast cancer cell lines, cell line xenografts, patient-derived xenografts, and organoids resistant to CDK4/6i and ET and exhibiting PIK3CA, PTEN, or AKT1 mutations. PIK3CA-mutant, PTEN–wild type, CDK4/6i-resistant, and ET-resistant models required the addition of the dual PI3K/mTOR inhibitor gedatolisib to effectively impede tumor growth by blocking the HIF-1α pathway through both mTORC1 inhibition and PI3K/AKT-mediated modulation of GSK3α/β activity. Conversely, PIK3CA–wild type, PTEN-null cells benefited from triple combinations incorporating either the AKT inhibitor capivasertib or the dual mTORC1/2 inhibitor sapanisertib to block tumor growth. In addition, gedatolisib reduced viability of PIK3CA- or AKT1-mutant and PTEN–wild type CDK4/6i-resistant patient-derived organoids compared with the α-specific PI3K inhibitor alpelisib. Our data support the higher efficacy of gedatolisib over alpelisib in ER+ breast tumors harboring alterations of the PI3K/AKT/mTOR pathway including PIK3CA or AKT1 mutations.
{"title":"Dual PI3K/mTOR inhibition is required to combat resistance to CDK4/6 inhibitor and endocrine therapy in PIK3CA-mutant breast cancer","authors":"Carla L. Alves, Leena Karimi, Mikkel G. Terp, Mie K. Jakobsen, Fiona H. Zhou, Benedetta Policastro, Nikoline Nissen, Lene E. Johansen, Tina Ravnsborg, Leila Eshraghi, Sana Tamboowala, Ole N. Jensen, Elgene Lim, Henrik J. Ditzel","doi":"10.1126/scitranslmed.adp5088","DOIUrl":"10.1126/scitranslmed.adp5088","url":null,"abstract":"<div >Combined CDK4/6 inhibitor (CDK4/6i) and endocrine therapy (ET) improves outcomes in advanced estrogen receptor–positive (ER<sup>+</sup>) breast cancer, but emergence of resistance to this combination underscores the pressing need for alternative therapeutic strategies. A promising approach involves adding an inhibitor of the PI3K/AKT/mTOR pathway to the standard combined CDK4/6i and ET, but selecting the most effective inhibitors and their optimal combinations has proven to be challenging. Here, we compared the efficacy of various triple combinations using single- or dual-point PI3K/AKT/mTOR pathway inhibitors in breast cancer cell lines, cell line xenografts, patient-derived xenografts, and organoids resistant to CDK4/6i and ET and exhibiting <i>PIK3CA</i>, <i>PTEN</i>, or <i>AKT1</i> mutations. <i>PIK3CA</i>-mutant, <i>PTEN</i>–wild type, CDK4/6i-resistant, and ET-resistant models required the addition of the dual PI3K/mTOR inhibitor gedatolisib to effectively impede tumor growth by blocking the HIF-1α pathway through both mTORC1 inhibition and PI3K/AKT-mediated modulation of GSK3α/β activity. Conversely, <i>PIK3CA</i>–wild type, <i>PTEN</i>-null cells benefited from triple combinations incorporating either the AKT inhibitor capivasertib or the dual mTORC1/2 inhibitor sapanisertib to block tumor growth. In addition, gedatolisib reduced viability of <i>PIK3CA</i>- or <i>AKT1</i>-mutant and <i>PTEN</i>–wild type CDK4/6i-resistant patient-derived organoids compared with the α-specific PI3K inhibitor alpelisib. Our data support the higher efficacy of gedatolisib over alpelisib in ER<sup>+</sup> breast tumors harboring alterations of the PI3K/AKT/mTOR pathway including <i>PIK3CA</i> or <i>AKT1</i> mutations.</div>","PeriodicalId":21580,"journal":{"name":"Science Translational Medicine","volume":"17 829","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145765538","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Monoclonal antibodies have transformed the therapeutic landscape across oncology, immunology, and infectious diseases by enabling high-affinity, antigen-specific targeting to neutralize soluble molecules, block cellular interactions, or deplete cells. Although high specificity is critical for safety, it may confer an inherent susceptibility to even minor variations in the target epitope. Here, we investigated the impact of natural single-nucleotide variants on antigen recognition by therapeutic monoclonal antibodies, both approved and in clinical development. For almost every antibody analyzed, we identified protein variants in or near the antibody-antigen interface, a subset of which were predicted to disrupt antigen recognition. Experimental studies corroborated the impact of select variants for four different antigens, revealing complete loss of antibody binding in some cases. For example, a human breast cancer cell line overexpressing human epidermal growth factor receptor 2 (HER-2), but engineered to carry the HER-2P594H variant, was completely resistant to killing even by highly potent clinical antibody-drug conjugates. These findings suggest that natural variants can confer primary resistance to antibody-based therapies, with critical implications for treatment outcomes, patient management, and safety, particularly in the context of potent modalities such as antibody-drug conjugates. Individual resistance-associated variants, although globally rare, are enriched in specific populations, underscoring the importance of accounting for genetic diversity in both drug development and clinical decision-making.
{"title":"Single–amino acid variants in target epitopes can confer resistance to antibody-based therapies","authors":"Romina Marone, Erblin Asllanaj, Giuseppina Capoferri, Torsten Schwede, Lukas T. Jeker, Rosalba Lepore","doi":"10.1126/scitranslmed.ady4877","DOIUrl":"10.1126/scitranslmed.ady4877","url":null,"abstract":"<div >Monoclonal antibodies have transformed the therapeutic landscape across oncology, immunology, and infectious diseases by enabling high-affinity, antigen-specific targeting to neutralize soluble molecules, block cellular interactions, or deplete cells. Although high specificity is critical for safety, it may confer an inherent susceptibility to even minor variations in the target epitope. Here, we investigated the impact of natural single-nucleotide variants on antigen recognition by therapeutic monoclonal antibodies, both approved and in clinical development. For almost every antibody analyzed, we identified protein variants in or near the antibody-antigen interface, a subset of which were predicted to disrupt antigen recognition. Experimental studies corroborated the impact of select variants for four different antigens, revealing complete loss of antibody binding in some cases. For example, a human breast cancer cell line overexpressing human epidermal growth factor receptor 2 (HER-2), but engineered to carry the HER-2<sup>P594H</sup> variant, was completely resistant to killing even by highly potent clinical antibody-drug conjugates. These findings suggest that natural variants can confer primary resistance to antibody-based therapies, with critical implications for treatment outcomes, patient management, and safety, particularly in the context of potent modalities such as antibody-drug conjugates. Individual resistance-associated variants, although globally rare, are enriched in specific populations, underscoring the importance of accounting for genetic diversity in both drug development and clinical decision-making.</div>","PeriodicalId":21580,"journal":{"name":"Science Translational Medicine","volume":"17 829","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145765542","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-17DOI: 10.1126/scitranslmed.adw9446
Ankit Uniyal, Niyada Hin, Chi Zhang, Qian Xu, Qian Huang, Ilyas Berhane, Ajit G. Thomas, John Maragakis, Sadakatali Gori, Jing Liu, Qin Zheng, Xiang Cui, Qi Peng, Jieru Wan, Rana Rais, Barbara S. Slusher, Srinivasa. N. Raja, Xinzhong Dong, Takashi Tsukamoto, Yun Guan
The human Mas-related G protein–coupled receptor X1 (MrgprX1) represents a promising nonopioid analgesic target because of its selective expression in primary nociceptive sensory neurons. Positive allosteric modulators (PAMs) promote receptor signaling, depending on the availability of endogenous ligands, offering physiological selectivity over orthosteric agonists. We developed an orally bioavailable MrgprX1 PAM, 6-tert-butyl-5-(4-chlorophenyl)-4-(2-fluoro-6-(trifluoromethoxy)phenoxy)thieno[2,3-d]pyrimidine (BCFTP). BCFTP selectively potentiated the functional response of MrgprX1 in HEK293 cells, was metabolically stable, and demonstrated a favorable in vitro safety profile. BCFTP was orally bioavailable and distributed into the spinal cords of wild-type mice. BAM22, an endogenous ligand for MrgprX1, was up-regulated in the spinal cord after nerve injury in both wild-type and humanized MrgprX1 mice and was expressed in peptidergic and nonpeptidergic dorsal root ganglion neurons. Oral administration of BCFTP dose-dependently inhibited heat hyperalgesia and spontaneous pain-like behavior but not mechanical hypersensitivity after sciatic chronic constrictive injury (CCI) in MrgprX1 mice. BCFTP did not have analgesic effects in Mrgpr cluster knockout (Mrgpr−/−) mice, indicating that the analgesic effects in MrgprX1 mice were MrgprX1 dependent. BCFTP enhanced BAM8-22–induced, MrgprX1-mediated reduction of C-fiber eEPSC amplitudes in spinal lamina II neurons, indicating inhibition of spinal nociceptive synaptic transmission. BCFTP did not induce tolerance or side effects, such as itch, sedation, and motor incoordination, and had no rewarding properties. The mRNAs encoding MrgprX1 and μ-opioid receptors were colocalized in human DRG neurons, and BCFTP synergistically enhanced morphine analgesia in CCI MrgprX1 mice. Our research suggests an approach for developing safer, orally bioavailable MrgprX1 PAM as a nonopioid therapy for neuropathic pain.
{"title":"An orally bioavailable MrgprX1-positive allosteric modulator alleviates certain neuropathic pain–related behaviors in humanized mice","authors":"Ankit Uniyal, Niyada Hin, Chi Zhang, Qian Xu, Qian Huang, Ilyas Berhane, Ajit G. Thomas, John Maragakis, Sadakatali Gori, Jing Liu, Qin Zheng, Xiang Cui, Qi Peng, Jieru Wan, Rana Rais, Barbara S. Slusher, Srinivasa. N. Raja, Xinzhong Dong, Takashi Tsukamoto, Yun Guan","doi":"10.1126/scitranslmed.adw9446","DOIUrl":"10.1126/scitranslmed.adw9446","url":null,"abstract":"<div >The human <i>Mas</i>-related G protein–coupled receptor X1 (MrgprX1) represents a promising nonopioid analgesic target because of its selective expression in primary nociceptive sensory neurons. Positive allosteric modulators (PAMs) promote receptor signaling, depending on the availability of endogenous ligands, offering physiological selectivity over orthosteric agonists. We developed an orally bioavailable MrgprX1 PAM, 6-tert-butyl-5-(4-chlorophenyl)-4-(2-fluoro-6-(trifluoromethoxy)phenoxy)thieno[2,3-d]pyrimidine (BCFTP). BCFTP selectively potentiated the functional response of MrgprX1 in HEK293 cells, was metabolically stable, and demonstrated a favorable in vitro safety profile. BCFTP was orally bioavailable and distributed into the spinal cords of wild-type mice. BAM22, an endogenous ligand for MrgprX1, was up-regulated in the spinal cord after nerve injury in both wild-type and humanized MrgprX1 mice and was expressed in peptidergic and nonpeptidergic dorsal root ganglion neurons. Oral administration of BCFTP dose-dependently inhibited heat hyperalgesia and spontaneous pain-like behavior but not mechanical hypersensitivity after sciatic chronic constrictive injury (CCI) in MrgprX1 mice. BCFTP did not have analgesic effects in Mrgpr cluster knockout (Mrgpr<sup>−/−</sup>) mice, indicating that the analgesic effects in MrgprX1 mice were MrgprX1 dependent. BCFTP enhanced BAM8-22–induced, MrgprX1-mediated reduction of C-fiber eEPSC amplitudes in spinal lamina II neurons, indicating inhibition of spinal nociceptive synaptic transmission. BCFTP did not induce tolerance or side effects, such as itch, sedation, and motor incoordination, and had no rewarding properties. The mRNAs encoding MrgprX1 and μ-opioid receptors were colocalized in human DRG neurons, and BCFTP synergistically enhanced morphine analgesia in CCI MrgprX1 mice. Our research suggests an approach for developing safer, orally bioavailable MrgprX1 PAM as a nonopioid therapy for neuropathic pain.</div>","PeriodicalId":21580,"journal":{"name":"Science Translational Medicine","volume":"17 829","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145765548","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-17DOI: 10.1126/scitranslmed.adq4060
Yahya Mohammadzadeh, Vojislav Gligorovski, Olga Egorova, Gabriele Casagrande Raffi, Jort J. van der Schans, Ali Ghasemi, Katharina Jonas, Bruno Torchia, Alan Guichard, Rachel Marcone, Amaia Martinez-Usatorre, Anna Köck, Raphael Genolet, Nadine Fournier, Tatiana V. Petrova, Daniel E. Speiser, Sahand Jamal Rahi, Nahal Mansouri, Michele De Palma
Effective antitumor immunity requires dendritic cells (DCs) to internalize, process, and present tumor antigens to T cells. Adoptive transfer of DCs that were loaded ex vivo with tumor antigens has been shown to stimulate antitumor immunity in patients with cancer, but clinical responses have been mixed. To address the limitations of traditional DC-based therapies, we constructed and functionally screened a panel of chimeric antigen receptors (CARs) optimized for expression and activity in DCs. Through this screening, we identified key functional components that guided the development of an inducible platform centered on an instructive chimeric antigen receptor (iCAR). This iCAR enabled DCs to (i) recognize a surface molecule present on cancer cells or their extracellular vesicles (EVs), such as disialoganglioside GD2 (expressed in melanoma and other tumors of neuroectodermal origin) or HER2 (expressed in some epithelial cancers), thereby promoting the acquisition of tumor-derived material containing putative tumor antigens; (ii) undergo immunostimulatory activation to prime antigen-specific T cells via both cross-dressing and cross-presentation; and (iii) transactivate the expression of the therapeutic cytokine interleukin-12 (IL-12) in response to antigen uptake. The iCAR converted melanoma-derived EVs from immune-suppressive to stimulatory cues for DCs in cell culture assays. Moreover, systemic administration of iCAR-DCs enhanced antigen-specific T cells, expanded low-frequency T cell clonotypes, and delayed tumor growth in immunotherapy-resistant melanoma models without the need for ex vivo antigen loading or cell maturation. iCAR-DCs may therefore provide a platform for antigen-agnostic cancer immunotherapy that integrates antigen uptake with programmable DC activation.
{"title":"Coordinate tumor-antigen uptake and dendritic cell activation by chimeric antigen receptors","authors":"Yahya Mohammadzadeh, Vojislav Gligorovski, Olga Egorova, Gabriele Casagrande Raffi, Jort J. van der Schans, Ali Ghasemi, Katharina Jonas, Bruno Torchia, Alan Guichard, Rachel Marcone, Amaia Martinez-Usatorre, Anna Köck, Raphael Genolet, Nadine Fournier, Tatiana V. Petrova, Daniel E. Speiser, Sahand Jamal Rahi, Nahal Mansouri, Michele De Palma","doi":"10.1126/scitranslmed.adq4060","DOIUrl":"10.1126/scitranslmed.adq4060","url":null,"abstract":"<div >Effective antitumor immunity requires dendritic cells (DCs) to internalize, process, and present tumor antigens to T cells. Adoptive transfer of DCs that were loaded ex vivo with tumor antigens has been shown to stimulate antitumor immunity in patients with cancer, but clinical responses have been mixed. To address the limitations of traditional DC-based therapies, we constructed and functionally screened a panel of chimeric antigen receptors (CARs) optimized for expression and activity in DCs. Through this screening, we identified key functional components that guided the development of an inducible platform centered on an instructive chimeric antigen receptor (iCAR). This iCAR enabled DCs to (i) recognize a surface molecule present on cancer cells or their extracellular vesicles (EVs), such as disialoganglioside GD2 (expressed in melanoma and other tumors of neuroectodermal origin) or HER2 (expressed in some epithelial cancers), thereby promoting the acquisition of tumor-derived material containing putative tumor antigens; (ii) undergo immunostimulatory activation to prime antigen-specific T cells via both cross-dressing and cross-presentation; and (iii) transactivate the expression of the therapeutic cytokine interleukin-12 (IL-12) in response to antigen uptake. The iCAR converted melanoma-derived EVs from immune-suppressive to stimulatory cues for DCs in cell culture assays. Moreover, systemic administration of iCAR-DCs enhanced antigen-specific T cells, expanded low-frequency T cell clonotypes, and delayed tumor growth in immunotherapy-resistant melanoma models without the need for ex vivo antigen loading or cell maturation. iCAR-DCs may therefore provide a platform for antigen-agnostic cancer immunotherapy that integrates antigen uptake with programmable DC activation.</div>","PeriodicalId":21580,"journal":{"name":"Science Translational Medicine","volume":"17 829","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145765535","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-17DOI: 10.1126/scitranslmed.adv9403
Amit Manhas, Yu Liu, Chikage Noishiki, David Wu, Dipti Tripathi, Sarah Mirza, Dilip Thomas, Lu Liu, Avirup Guha, Patricia K. Nguyen, Ian Y. Chen, Vipul Chitalia, Paul Cheng, Danish Sayed, Melinda L. Telli, Karim Sallam, Joseph C. Wu, Nazish Sayed
Tyrosine kinase inhibitors (TKIs) have improved cancer outcomes but are limited by cardiovascular toxicity, most notably hypertension and heart failure. The underlying mechanisms remain poorly understood, hindering the development of protective strategies. Here, we investigated the role of endothelial mechanotransduction in mediating vascular and cardiac injury caused by the vascular endothelial growth factor receptor–targeting TKI sunitinib. Using patient-specific induced pluripotent stem cell–derived endothelial cells (iPSC-ECs) and a mouse model of TKI-induced hypertension, we identified down-regulation of piezo-type mechanosensitive ion channel component 1 (PIEZO1), a mechanically activated ion channel, as a driver of endothelial dysfunction. Restoring PIEZO1 expression, either pharmacologically with Yoda1, a selective agonist, or through inducible overexpression in iPSC-ECs, reversed sunitinib-induced endothelial dysfunction and mitigated its hypertensive effects, providing both mechanistic and genetic validation of PIEZO1’s protective role against vascular toxicity. In mice, cotreatment with sunitinib and Yoda1 prevented the long-term cardiac dysfunction observed after sunitinib exposure and normalized elevations in circulating cardiac stress biomarkers. Single-nucleus multiomic profiling of mouse hearts revealed that sunitinib exposure activated chromatin remodeling and fibrogenic programs, which were reversed with PIEZO1 activation. Human engineered cardiac organoids further demonstrated that sunitinib impaired cardiomyocyte function only in the presence of endothelial cells, confirming a role for disrupted endothelial-cardiomyocyte cross-talk in TKI cardiotoxicity. Together, these findings identify endothelial PIEZO1 as a mediator of TKI-induced hypertension and cardiac dysfunction and highlight PIEZO1 activation as a potential therapeutic strategy for protecting cardiovascular health during cancer therapy.
{"title":"Multiscale profiling of tyrosine kinase inhibitor cardiotoxicity reveals mechanosensitive ion channel PIEZO1 as cardioprotective","authors":"Amit Manhas, Yu Liu, Chikage Noishiki, David Wu, Dipti Tripathi, Sarah Mirza, Dilip Thomas, Lu Liu, Avirup Guha, Patricia K. Nguyen, Ian Y. Chen, Vipul Chitalia, Paul Cheng, Danish Sayed, Melinda L. Telli, Karim Sallam, Joseph C. Wu, Nazish Sayed","doi":"10.1126/scitranslmed.adv9403","DOIUrl":"10.1126/scitranslmed.adv9403","url":null,"abstract":"<div >Tyrosine kinase inhibitors (TKIs) have improved cancer outcomes but are limited by cardiovascular toxicity, most notably hypertension and heart failure. The underlying mechanisms remain poorly understood, hindering the development of protective strategies. Here, we investigated the role of endothelial mechanotransduction in mediating vascular and cardiac injury caused by the vascular endothelial growth factor receptor–targeting TKI sunitinib. Using patient-specific induced pluripotent stem cell–derived endothelial cells (iPSC-ECs) and a mouse model of TKI-induced hypertension, we identified down-regulation of <i>piezo-type mechanosensitive ion channel component 1</i> (<i>PIEZO1</i>), a mechanically activated ion channel, as a driver of endothelial dysfunction. Restoring <i>PIEZO1</i> expression, either pharmacologically with Yoda1, a selective agonist, or through inducible overexpression in iPSC-ECs, reversed sunitinib-induced endothelial dysfunction and mitigated its hypertensive effects, providing both mechanistic and genetic validation of PIEZO1’s protective role against vascular toxicity. In mice, cotreatment with sunitinib and Yoda1 prevented the long-term cardiac dysfunction observed after sunitinib exposure and normalized elevations in circulating cardiac stress biomarkers. Single-nucleus multiomic profiling of mouse hearts revealed that sunitinib exposure activated chromatin remodeling and fibrogenic programs, which were reversed with PIEZO1 activation. Human engineered cardiac organoids further demonstrated that sunitinib impaired cardiomyocyte function only in the presence of endothelial cells, confirming a role for disrupted endothelial-cardiomyocyte cross-talk in TKI cardiotoxicity. Together, these findings identify endothelial PIEZO1 as a mediator of TKI-induced hypertension and cardiac dysfunction and highlight PIEZO1 activation as a potential therapeutic strategy for protecting cardiovascular health during cancer therapy.</div>","PeriodicalId":21580,"journal":{"name":"Science Translational Medicine","volume":"17 829","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145765533","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-10DOI: 10.1126/scitranslmed.adq0143
Xu Cao, Amit Manhas, Yi-Ing Chen, Arianne Caudal, Gema Mondejar-Parreño, Wenjuan Zhu, Wenqiang Liu, Xiaohui Kong, Wenshu Zeng, Lichao Liu, Shane R. Zhao, James W. S. Jahng, Paul J. Utz, Kari C. Nadeau, Masataka Nishiga, Joseph C. Wu
Messenger RNA (mRNA) vaccines against SARS-CoV-2 are highly effective and were instrumental in curbing the COVID-19 pandemic. However, rare cases of noninfective myocarditis, particularly in young males and typically after the second dose, have been observed. Here, we explore the mediators of this myocarditis to better understand and to enhance the safety of future mRNA vaccines. Through analysis of human plasma data and in vitro experiments with human macrophages and T cells, we identified increased C-X-C motif chemokine ligand 10 (CXCL10) and interferon-γ (IFN-γ) after exposure to BNT162b2 (Pfizer) or mRNA-1273 (Moderna). Neutralization of CXCL10 and IFN-γ during the second dose (21 days after the first dose) reduced vaccine-induced cardiac injury in mice. Neutralization also reduced cardiac stress markers such as the release of N-terminal pro-B-type natriuretic peptide (NT-proBNP) and expression of inflammatory genes in human induced pluripotent stem cell (iPSC)–derived cardiac spheroids. When exposed to these cytokines in vitro, human iPSC-derived cardiomyocytes (iPSC-CMs) exhibited impaired contractility, arrhythmogenicity, and proinflammatory gene expression patterns. Genistein, a phytoestrogen implicated in reducing cardiovascular inflammation, mitigated these effects in iPSC-CMs. In mice exposed to these cytokines or receiving BNT162b2 vaccination, genistein treatment reduced cardiac injury markers and attenuated infiltration of neutrophils and macrophages into the heart. These findings implicate CXCL10–IFN-γ signaling as a contributor to myocardial injury in experimental models of mRNA vaccination and indicate that pharmacologic modulation, such as with genistein, may mitigate cytokine-driven injury.
{"title":"Inhibition of CXCL10 and IFN-γ ameliorates myocarditis in preclinical models of SARS-CoV-2 mRNA vaccination","authors":"Xu Cao, Amit Manhas, Yi-Ing Chen, Arianne Caudal, Gema Mondejar-Parreño, Wenjuan Zhu, Wenqiang Liu, Xiaohui Kong, Wenshu Zeng, Lichao Liu, Shane R. Zhao, James W. S. Jahng, Paul J. Utz, Kari C. Nadeau, Masataka Nishiga, Joseph C. Wu","doi":"10.1126/scitranslmed.adq0143","DOIUrl":"10.1126/scitranslmed.adq0143","url":null,"abstract":"<div >Messenger RNA (mRNA) vaccines against SARS-CoV-2 are highly effective and were instrumental in curbing the COVID-19 pandemic. However, rare cases of noninfective myocarditis, particularly in young males and typically after the second dose, have been observed. Here, we explore the mediators of this myocarditis to better understand and to enhance the safety of future mRNA vaccines. Through analysis of human plasma data and in vitro experiments with human macrophages and T cells, we identified increased C-X-C motif chemokine ligand 10 (CXCL10) and interferon-γ (IFN-γ) after exposure to BNT162b2 (Pfizer) or mRNA-1273 (Moderna). Neutralization of CXCL10 and IFN-γ during the second dose (21 days after the first dose) reduced vaccine-induced cardiac injury in mice. Neutralization also reduced cardiac stress markers such as the release of N-terminal pro-B-type natriuretic peptide (NT-proBNP) and expression of inflammatory genes in human induced pluripotent stem cell (iPSC)–derived cardiac spheroids. When exposed to these cytokines in vitro, human iPSC-derived cardiomyocytes (iPSC-CMs) exhibited impaired contractility, arrhythmogenicity, and proinflammatory gene expression patterns. Genistein, a phytoestrogen implicated in reducing cardiovascular inflammation, mitigated these effects in iPSC-CMs. In mice exposed to these cytokines or receiving BNT162b2 vaccination, genistein treatment reduced cardiac injury markers and attenuated infiltration of neutrophils and macrophages into the heart. These findings implicate CXCL10–IFN-γ signaling as a contributor to myocardial injury in experimental models of mRNA vaccination and indicate that pharmacologic modulation, such as with genistein, may mitigate cytokine-driven injury.</div>","PeriodicalId":21580,"journal":{"name":"Science Translational Medicine","volume":"17 828","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-12-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145711322","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-10DOI: 10.1126/scitranslmed.ado6948
Tao Zhang, Tingyun Lei, Jie Han, Yangwu Chen, Yuepeng Nie, Ru Zhang, Honglu Cai, Zijin Guo, Ruojin Yan, Zan Li, Yongqiang Xu, Xianzhu Zhang, Hongxia Xu, Jiahe Chen, Youguo Liao, Qiulin He, Hong Zhang, Jianquan Chen, Shouan Zhu, Jiansheng Guo, Yi Liu, Haihua Pan, Jingyao Chen, Wei Yin, Hongwei Ouyang, Weiliang Shen, Zi Yin, Ruikang Tang, Chen Xiao
The enthesis, a fibrocartilaginous tissue connecting tendon or ligament to bone, is critical for joint movement but lacks regenerative capacity after injury. Current clinical treatments for enthesis healing remain limited. Here, with a resolution of 2 to 3 nanometers, we found that mineral particles form a continuous cross-fibrillar phase with a discontinuous distribution in the fibrocartilage layer. Building on this finding, we developed a series of bioinspired mineralized collagen matrices, characterized by both intra- and extrafibrillar localization of crystallites, with a tunable mass percentage of inorganic content as scaffolds for enthesis repair. Our results revealed that mineralized collagen with controlled inorganic content (33% mineral content) facilitated fibrocartilage healing across multiple animal enthesis injury models, including mice, rats, rabbits, and goats. In direct comparisons with other biomaterials in a rabbit model, the bioinspired mineralized collagen resulted in 82% fibrocartilage width recovery, more than two times the healing observed with other materials. Treatment with the bioinspired mineralized collagen scaffold produced joint healing with an ability to sustain a higher maximum load in both rat and rabbit models, with the animals able to walk normally. The goat model exhibited an improvement in jumping ability. Mechanistically, we found that the bioinspired mineralized collagen modulated Hedgehog signaling intensity in a mineralization-dependent manner, which in turn up-regulated Gli1 expression. This modulation regulated the differentiation of mesenchymal progenitor cells and promoted fibrocartilage healing. Overall, we demonstrate that a bioinspired mineralized collagen scaffold effectively promotes enthesis injury repair, demonstrating potential for clinical translation.
{"title":"A bioinspired mineralized collagen scaffold promotes enthesis healing and activates Gli1 expression in preclinical models","authors":"Tao Zhang, Tingyun Lei, Jie Han, Yangwu Chen, Yuepeng Nie, Ru Zhang, Honglu Cai, Zijin Guo, Ruojin Yan, Zan Li, Yongqiang Xu, Xianzhu Zhang, Hongxia Xu, Jiahe Chen, Youguo Liao, Qiulin He, Hong Zhang, Jianquan Chen, Shouan Zhu, Jiansheng Guo, Yi Liu, Haihua Pan, Jingyao Chen, Wei Yin, Hongwei Ouyang, Weiliang Shen, Zi Yin, Ruikang Tang, Chen Xiao","doi":"10.1126/scitranslmed.ado6948","DOIUrl":"10.1126/scitranslmed.ado6948","url":null,"abstract":"<div >The enthesis, a fibrocartilaginous tissue connecting tendon or ligament to bone, is critical for joint movement but lacks regenerative capacity after injury. Current clinical treatments for enthesis healing remain limited. Here, with a resolution of 2 to 3 nanometers, we found that mineral particles form a continuous cross-fibrillar phase with a discontinuous distribution in the fibrocartilage layer. Building on this finding, we developed a series of bioinspired mineralized collagen matrices, characterized by both intra- and extrafibrillar localization of crystallites, with a tunable mass percentage of inorganic content as scaffolds for enthesis repair. Our results revealed that mineralized collagen with controlled inorganic content (33% mineral content) facilitated fibrocartilage healing across multiple animal enthesis injury models, including mice, rats, rabbits, and goats. In direct comparisons with other biomaterials in a rabbit model, the bioinspired mineralized collagen resulted in 82% fibrocartilage width recovery, more than two times the healing observed with other materials. Treatment with the bioinspired mineralized collagen scaffold produced joint healing with an ability to sustain a higher maximum load in both rat and rabbit models, with the animals able to walk normally. The goat model exhibited an improvement in jumping ability. Mechanistically, we found that the bioinspired mineralized collagen modulated Hedgehog signaling intensity in a mineralization-dependent manner, which in turn up-regulated <i>Gli1</i> expression. This modulation regulated the differentiation of mesenchymal progenitor cells and promoted fibrocartilage healing. Overall, we demonstrate that a bioinspired mineralized collagen scaffold effectively promotes enthesis injury repair, demonstrating potential for clinical translation.</div>","PeriodicalId":21580,"journal":{"name":"Science Translational Medicine","volume":"17 828","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-12-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145711357","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-10DOI: 10.1126/scitranslmed.aed9347
{"title":"Erratum for the Research Article “Impairment of stromal-epithelial regenerative cross-talk in Hirschsprung disease primes for the progression to enterocolitis” by Z. Zhang et al.","authors":"","doi":"10.1126/scitranslmed.aed9347","DOIUrl":"10.1126/scitranslmed.aed9347","url":null,"abstract":"","PeriodicalId":21580,"journal":{"name":"Science Translational Medicine","volume":"17 828","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-12-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145717556","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-10DOI: 10.1126/scitranslmed.adr0024
Erkin Kurganov, Lei Cui, Nikita Budnik, Siwei Chen, Erick Olivares, David Baez-Nieto, Ahmet S. Asan, Laina Lusk, Lacey Smith, Sooyeon Jo, Diogo Marques, Ralda Nehme, Sureni V. Mullegama, Amanda Lindy, Alfred L. George Jr., Annapurna Poduri, Ingo Helbig, Mark Daly, Jen Q. Pan
CACNA1A encodes the α1A subunit of the CaV2.1 voltage–gated calcium channel essential for neuronal excitability, neurotransmitter release, and calcium signaling in neurons. Pathogenic CACNA1A variants are linked to diverse neurological and developmental disorders, including hemiplegic migraine, epilepsy, developmental delay, and ataxia. However, the contribution of CACNA1A variants to neurodevelopmental disorders has been less well established. Clinical heterogeneity and the complex impact of missense variants on channel function have made genotype-phenotype relationships difficult to resolve. Here, we characterized the biophysical properties of 42 de novo CACNA1A missense variants identified in a neurodevelopmental disorder cohort of over 31,000 individuals, alongside eight common variants from the general population. All but one de novo variant altered biophysical properties of the CaV2.1 channel, with 50% abolishing measurable currents. Among variants with detectable currents, nearly half disrupted voltage-dependent gating, whereas common variants had no measurable effect. Similar results were obtained using representative variants in the presence of β3 or β4 auxiliary subunits. The expression of two selected variants in CACNA1A-knockdown hiPSC-derived neurons confirmed similar functional alterations in human neurons. Simulations using the NEURON model of a Purkinje neuron demonstrated that these biophysical changes profoundly affected excitability. Coupling functional data with AlphaMissense predictions, we found that CaV2.1 missense variants contribute to the risk for developmental epileptic encephalopathy. Finally, correlating clinical features with molecular consequences of each variant revealed meaningful associations between specific channel dysfunctions and distinct clinical outcomes. These findings provide insights into CACNA1A molecular pathology and highlight the potential for precision medicine treatment of CACNA1A-associated disorders.
CACNA1A编码Ca V 2.1电压门控钙通道的α1A亚基,该通道对神经元的兴奋性、神经递质释放和钙信号传导至关重要。致病性CACNA1A变异与多种神经和发育障碍有关,包括偏瘫性偏头痛、癫痫、发育迟缓和共济失调。然而,CACNA1A变异对神经发育障碍的贡献尚未得到很好的证实。临床异质性和错义变异对通道功能的复杂影响使得基因型-表型关系难以解决。在这里,我们描述了在一个超过31000人的神经发育障碍队列中发现的42个新生CACNA1A错义变异的生物物理特性,以及来自普通人群的8个常见变异。除了一个全新的变体外,其他所有变体都改变了Ca V 2.1通道的生物物理特性,其中50%的变体消除了可测量电流。在具有可检测电流的变体中,近一半破坏了电压依赖性门控,而普通变体没有可测量的影响。在β3或β4辅助亚基存在的情况下,使用具有代表性的变体获得了类似的结果。在CACNA1A敲低hipsc衍生的神经元中,两种选择的变体的表达证实了人类神经元中类似的功能改变。使用浦肯野神经元模型的模拟表明,这些生物物理变化深刻地影响了兴奋性。将功能数据与AlphaMissense预测相结合,我们发现Ca V 2.1错义变异增加了发育性癫痫性脑病的风险。最后,将临床特征与每种变异的分子后果相关联,揭示了特定通道功能障碍与不同临床结果之间有意义的关联。这些发现提供了对CACNA1A分子病理学的见解,并强调了CACNA1A相关疾病的精准医学治疗的潜力。
{"title":"Characterization of the functional and clinical impacts of CACNA1A missense variants found in neurodevelopmental disorders","authors":"Erkin Kurganov, Lei Cui, Nikita Budnik, Siwei Chen, Erick Olivares, David Baez-Nieto, Ahmet S. Asan, Laina Lusk, Lacey Smith, Sooyeon Jo, Diogo Marques, Ralda Nehme, Sureni V. Mullegama, Amanda Lindy, Alfred L. George Jr., Annapurna Poduri, Ingo Helbig, Mark Daly, Jen Q. Pan","doi":"10.1126/scitranslmed.adr0024","DOIUrl":"10.1126/scitranslmed.adr0024","url":null,"abstract":"<div ><i>CACNA1A</i> encodes the α1A subunit of the Ca<sub>V</sub>2.1 voltage–gated calcium channel essential for neuronal excitability, neurotransmitter release, and calcium signaling in neurons. Pathogenic <i>CACNA1A</i> variants are linked to diverse neurological and developmental disorders, including hemiplegic migraine, epilepsy, developmental delay, and ataxia. However, the contribution of <i>CACNA1A</i> variants to neurodevelopmental disorders has been less well established. Clinical heterogeneity and the complex impact of missense variants on channel function have made genotype-phenotype relationships difficult to resolve. Here, we characterized the biophysical properties of 42 de novo <i>CACNA1A</i> missense variants identified in a neurodevelopmental disorder cohort of over 31,000 individuals, alongside eight common variants from the general population. All but one de novo variant altered biophysical properties of the Ca<sub>V</sub>2.1 channel, with 50% abolishing measurable currents. Among variants with detectable currents, nearly half disrupted voltage-dependent gating, whereas common variants had no measurable effect. Similar results were obtained using representative variants in the presence of β3 or β4 auxiliary subunits. The expression of two selected variants in <i>CACNA1A</i>-knockdown hiPSC-derived neurons confirmed similar functional alterations in human neurons. Simulations using the <i>NEURON</i> model of a Purkinje neuron demonstrated that these biophysical changes profoundly affected excitability. Coupling functional data with AlphaMissense predictions, we found that Ca<sub>V</sub>2.1 missense variants contribute to the risk for developmental epileptic encephalopathy. Finally, correlating clinical features with molecular consequences of each variant revealed meaningful associations between specific channel dysfunctions and distinct clinical outcomes. These findings provide insights into <i>CACNA1A</i> molecular pathology and highlight the potential for precision medicine treatment of <i>CACNA1A</i>-associated disorders.</div>","PeriodicalId":21580,"journal":{"name":"Science Translational Medicine","volume":"17 828","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-12-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145711356","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}