Pub Date : 2024-12-31DOI: 10.1016/j.ymthe.2024.12.038
Vishnu Raman, Christopher L Hall, Victoria E Wetherby, Samantha A Witney, Nele Van Dessel, Neil S Forbes
Effectively targeting intracellular pathways in cancers requires a system that specifically delivers to tumors and internalizes into cancer cells. To achieve this goal, we developed intracellular-delivering (ID) Salmonella with controllable expression of flhDC, to regulate flagella production and cell invasion. We hypothesized that controlling flhDC would overcome the poor colonization seen in prior clinical trials. To test this hypothesis, we incorporated the aspirin-responsive Psal promoter and tuned flhDC expression with ssra degradation tags. In tumor-bearing mice, controlling flhDC increased protein release, tissue dispersion and tumor colonization more than ten million times. We discovered that inducing flhDC increases escape from intracellular vacuoles; however, deleting sseJ prevented escape and further increased protein delivery. Delivering constitutively active caspase-3 with ID-f-s Salmonella (ΔsseJ and induced PSal-flhDC) induced cell death in pancreatic, breast and liver cancer cells and reduced the growth of breast tumors. This clinically ready strain preferentially colonized metastatic breast tissue 280 and 800 times more than surrounding healthy tissue in the lung and liver, respectively. By precisely controlling tumor colonization and cell invasion, this strain overcomes critical limitations of bacterial therapy and will enable treatment of many hard-to-treat cancers444.
{"title":"Controlling intracellular protein delivery, tumor colonization and tissue distribution using the master regulator flhDC in a clinically relevant ΔsseJ Salmonella strain.","authors":"Vishnu Raman, Christopher L Hall, Victoria E Wetherby, Samantha A Witney, Nele Van Dessel, Neil S Forbes","doi":"10.1016/j.ymthe.2024.12.038","DOIUrl":"https://doi.org/10.1016/j.ymthe.2024.12.038","url":null,"abstract":"<p><p>Effectively targeting intracellular pathways in cancers requires a system that specifically delivers to tumors and internalizes into cancer cells. To achieve this goal, we developed intracellular-delivering (ID) Salmonella with controllable expression of flhDC, to regulate flagella production and cell invasion. We hypothesized that controlling flhDC would overcome the poor colonization seen in prior clinical trials. To test this hypothesis, we incorporated the aspirin-responsive Psal promoter and tuned flhDC expression with ssra degradation tags. In tumor-bearing mice, controlling flhDC increased protein release, tissue dispersion and tumor colonization more than ten million times. We discovered that inducing flhDC increases escape from intracellular vacuoles; however, deleting sseJ prevented escape and further increased protein delivery. Delivering constitutively active caspase-3 with ID-f-s Salmonella (ΔsseJ and induced PSal-flhDC) induced cell death in pancreatic, breast and liver cancer cells and reduced the growth of breast tumors. This clinically ready strain preferentially colonized metastatic breast tissue 280 and 800 times more than surrounding healthy tissue in the lung and liver, respectively. By precisely controlling tumor colonization and cell invasion, this strain overcomes critical limitations of bacterial therapy and will enable treatment of many hard-to-treat cancers<sup>444</sup>.</p>","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":" ","pages":""},"PeriodicalIF":12.1,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142915308","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}
Rabies is a lethal zoonotic infectious disease. Vaccines against the rabies virus have significantly reduced the number of deaths from the disease. However, all the licensed rabies vaccines are inactivated vaccines, which have limited immunogenicity and complicated immunization procedures. A novel vaccine that provides sustained and comprehensive protection is urgently needed. Here, we developed a novel rabies mRNA vaccine candidate containing sequence-optimized mRNAs encoding full-length glycoprotein encapsulated in ionizable lipid nanoparticles. In mice and rhesus macaques, the rabies mRNA exhibited superior immunogenicity over licensed vaccines, especially in inducing long-lasting neutralizing antibodies and memory B cells. A single administration of 1.5 μg mRNA vaccine could provide complete protection against a lethal rabies virus challenge in mice. Additionally, the mRNA vaccine could robustly activate cellular immune responses with moderate release of several cytokines. In summary, our data demonstrated that the rabies mRNA vaccine outperformed approved inactivated vaccines in both mice and rhesus macaques. This highlights the potential of the mRNA platform in developing next-generation rabies vaccines.
{"title":"A nucleoside-modified rabies mRNA vaccine induces long-lasting and comprehensive immune responses in mice and non-human primates.","authors":"Yu Wang, Shen Wang, Lulu Haung, Wenhao Mao, Fangmeng Li, Ang Lin, Weijun Zhao, Xianhuan Zeng, Yue Zhang, Dingcao Yang, Yuhong Han, Yidan Li, Leyuan Ren, Ying Li, Liang Zhang, Feihu Yan, Yong Yang, Xinying Tang","doi":"10.1016/j.ymthe.2024.12.041","DOIUrl":"https://doi.org/10.1016/j.ymthe.2024.12.041","url":null,"abstract":"<p><p>Rabies is a lethal zoonotic infectious disease. Vaccines against the rabies virus have significantly reduced the number of deaths from the disease. However, all the licensed rabies vaccines are inactivated vaccines, which have limited immunogenicity and complicated immunization procedures. A novel vaccine that provides sustained and comprehensive protection is urgently needed. Here, we developed a novel rabies mRNA vaccine candidate containing sequence-optimized mRNAs encoding full-length glycoprotein encapsulated in ionizable lipid nanoparticles. In mice and rhesus macaques, the rabies mRNA exhibited superior immunogenicity over licensed vaccines, especially in inducing long-lasting neutralizing antibodies and memory B cells. A single administration of 1.5 μg mRNA vaccine could provide complete protection against a lethal rabies virus challenge in mice. Additionally, the mRNA vaccine could robustly activate cellular immune responses with moderate release of several cytokines. In summary, our data demonstrated that the rabies mRNA vaccine outperformed approved inactivated vaccines in both mice and rhesus macaques. This highlights the potential of the mRNA platform in developing next-generation rabies vaccines.</p>","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":" ","pages":""},"PeriodicalIF":12.1,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142915303","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 : 2024-12-30DOI: 10.1016/j.ymthe.2024.12.047
Gang Zhong, Wei Liu, Jagadeesh K Venkatesan, Dan Wang, Henning Madry, Magali Cucchiarini
Despite various available treatments, highly prevalent osteoarthritis cannot be cured in patients. In light of evidence showing mitochondria dysfunction during the disease progression, our goal was to develop a novel therapeutic concept based on the transplantation of mitochondria as platforms to deliver recombinant adeno-associated viral (rAAV) gene vectors with a potency for osteoarthritis. For the first time to our best knowledge, we report the successful creation of a safe mitochondria/rAAV system effectively promoting the overexpression of a candidate insulin-like growth factor I (IGF-I) by administration to autologous human osteoarthritic articular chondrocytes versus control conditions (reporter mitochondria/rAAV lacZ system, rAAV-free system, absence of mitochondria transplantation) (up to 8.4-fold difference). The candidate mitochondria/rAAV IGF-I system significantly improved key activities in the transplanted cells (proliferation/survival, extracellular matrix production, mitochondria functions) relative to the control conditions (up to 9.5-fold difference), including when provided in a PF127 hydrogel for reinforced delivery (up to 5.9-fold difference). Such effects were accompanied with increased levels of cartilage-specific SOX9 and Mfn-1 (mitochondria fusion) and with decreased levels of Drp-1 (mitochondria fission) and proinflammatory TNF-α (up to 4.5-fold difference). This study shows the potential of combining the use of mitochondria with rAAV as a promising approach for human OA.
{"title":"Autologous transplantation of mitochondria/rAAV IGF-I platforms in human osteoarthritic articular chondrocytes as a novel therapeutic concept for human osteoarthritis.","authors":"Gang Zhong, Wei Liu, Jagadeesh K Venkatesan, Dan Wang, Henning Madry, Magali Cucchiarini","doi":"10.1016/j.ymthe.2024.12.047","DOIUrl":"https://doi.org/10.1016/j.ymthe.2024.12.047","url":null,"abstract":"<p><p>Despite various available treatments, highly prevalent osteoarthritis cannot be cured in patients. In light of evidence showing mitochondria dysfunction during the disease progression, our goal was to develop a novel therapeutic concept based on the transplantation of mitochondria as platforms to deliver recombinant adeno-associated viral (rAAV) gene vectors with a potency for osteoarthritis. For the first time to our best knowledge, we report the successful creation of a safe mitochondria/rAAV system effectively promoting the overexpression of a candidate insulin-like growth factor I (IGF-I) by administration to autologous human osteoarthritic articular chondrocytes versus control conditions (reporter mitochondria/rAAV lacZ system, rAAV-free system, absence of mitochondria transplantation) (up to 8.4-fold difference). The candidate mitochondria/rAAV IGF-I system significantly improved key activities in the transplanted cells (proliferation/survival, extracellular matrix production, mitochondria functions) relative to the control conditions (up to 9.5-fold difference), including when provided in a PF127 hydrogel for reinforced delivery (up to 5.9-fold difference). Such effects were accompanied with increased levels of cartilage-specific SOX9 and Mfn-1 (mitochondria fusion) and with decreased levels of Drp-1 (mitochondria fission) and proinflammatory TNF-α (up to 4.5-fold difference). This study shows the potential of combining the use of mitochondria with rAAV as a promising approach for human OA.</p>","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":" ","pages":""},"PeriodicalIF":12.1,"publicationDate":"2024-12-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142915307","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 : 2024-12-30DOI: 10.1016/j.ymthe.2024.12.049
Isabel L Day, Mikayla Tamboline, Gerald S Lipshutz, Shili Xu
Gene therapy achieves therapeutic benefits by delivering genetic materials, packaged within a delivery vehicle, to target cells with defective genes. This approach has shown promise in treating various conditions, including cancer, metabolic disorders, and tissue degenerative diseases. Over the past five years, molecular imaging has increasingly supported gene therapy development in both preclinical and clinical studies. High-quality images from positron emission tomography (PET), single photon emission computed tomography (SPECT), magnetic resonance imaging (MRI), and computed tomography (CT) enable quantitative and reliable monitoring of gene therapy. Most reported studies have applied imaging biomarkers to non-invasively evaluate the outcomes of gene therapy. This review aims to inform researchers in molecular imaging and gene therapy about the integration of these two disciplines. We highlight recent developments in using imaging biomarkers to monitor the outcome of in vivo gene therapy, where the therapeutic delivery vehicle is systemically administered. In addition, we discuss prospects for further incorporating imaging biomarkers to support the development and application of gene therapy.
{"title":"Recent developments in translational imaging of in vivo gene therapy outcomes.","authors":"Isabel L Day, Mikayla Tamboline, Gerald S Lipshutz, Shili Xu","doi":"10.1016/j.ymthe.2024.12.049","DOIUrl":"https://doi.org/10.1016/j.ymthe.2024.12.049","url":null,"abstract":"<p><p>Gene therapy achieves therapeutic benefits by delivering genetic materials, packaged within a delivery vehicle, to target cells with defective genes. This approach has shown promise in treating various conditions, including cancer, metabolic disorders, and tissue degenerative diseases. Over the past five years, molecular imaging has increasingly supported gene therapy development in both preclinical and clinical studies. High-quality images from positron emission tomography (PET), single photon emission computed tomography (SPECT), magnetic resonance imaging (MRI), and computed tomography (CT) enable quantitative and reliable monitoring of gene therapy. Most reported studies have applied imaging biomarkers to non-invasively evaluate the outcomes of gene therapy. This review aims to inform researchers in molecular imaging and gene therapy about the integration of these two disciplines. We highlight recent developments in using imaging biomarkers to monitor the outcome of in vivo gene therapy, where the therapeutic delivery vehicle is systemically administered. In addition, we discuss prospects for further incorporating imaging biomarkers to support the development and application of gene therapy.</p>","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":" ","pages":""},"PeriodicalIF":12.1,"publicationDate":"2024-12-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142915323","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}
Sepsis-induced thrombocytopenia (SIT) is a widely accepted predictor of poor prognosis during sepsis, while the mechanism of SIT remains elusive. In this study, we revealed that SIT patients and septic mice exhibited higher levels of pro-inflammatory macrophages and phosphorylated BTK (p-BTK) expression in macrophages, which were closely correlated with platelet counts. Treatment with the BTK inhibitor, BGB-3111 in SIT mice resulted in enhanced production of megakaryocytes and platelets. Depletion of macrophages in SIT mice and coculture experiments further confirmed the critical role of macrophages in the improvement of platelet count induced by BGB-3111. By performing single-cell RNA sequencing on bone marrow-derived cells from SIT mice, we not only confirmed the connection between macrophages and megakaryocytes influenced by BTK but also identified a potential mediation through the Rap1 signaling pathway in macrophages. Subsequent experiments in macrophages demonstrated that inhibition of BTK signaling impeded the pro-inflammatory polarization of macrophages by targeting the Rap1/NF-κB signaling pathway. In conclusion, our study highlights the crucial role of macrophages in SIT, and inhibiting phosphorylation of BTK in macrophages may alleviate SIT through the Rap1/NF-κB signaling pathway.
{"title":"Impaired Megakaryopoiesis due to Aberrant Macrophage Polarization via BTK/Rap1/NF-κB Pathway in Sepsis-induced Thrombocytopenia.","authors":"Ziyan Zhang, Meng Zhou, Yaqiong Tang, Jiaqian Qi, Xiaoyan Xu, Peng Wang, Haohao Han, Tingting Pan, Xiaofei Song, Shuhui Jiang, Xueqian Li, Chengyuan Gu, Zhenzhen Yao, Qixiu Hou, Mengting Guo, Siyi Lu, Depei Wu, Yue Han","doi":"10.1016/j.ymthe.2024.12.048","DOIUrl":"https://doi.org/10.1016/j.ymthe.2024.12.048","url":null,"abstract":"<p><p>Sepsis-induced thrombocytopenia (SIT) is a widely accepted predictor of poor prognosis during sepsis, while the mechanism of SIT remains elusive. In this study, we revealed that SIT patients and septic mice exhibited higher levels of pro-inflammatory macrophages and phosphorylated BTK (p-BTK) expression in macrophages, which were closely correlated with platelet counts. Treatment with the BTK inhibitor, BGB-3111 in SIT mice resulted in enhanced production of megakaryocytes and platelets. Depletion of macrophages in SIT mice and coculture experiments further confirmed the critical role of macrophages in the improvement of platelet count induced by BGB-3111. By performing single-cell RNA sequencing on bone marrow-derived cells from SIT mice, we not only confirmed the connection between macrophages and megakaryocytes influenced by BTK but also identified a potential mediation through the Rap1 signaling pathway in macrophages. Subsequent experiments in macrophages demonstrated that inhibition of BTK signaling impeded the pro-inflammatory polarization of macrophages by targeting the Rap1/NF-κB signaling pathway. In conclusion, our study highlights the crucial role of macrophages in SIT, and inhibiting phosphorylation of BTK in macrophages may alleviate SIT through the Rap1/NF-κB signaling pathway.</p>","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":" ","pages":""},"PeriodicalIF":12.1,"publicationDate":"2024-12-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142915312","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 : 2024-12-30DOI: 10.1016/j.ymthe.2024.12.054
Kalkidan Ayele, Hiroaki Wakimoto, Hans J Nauwynck, Howard L Kaufman, Samuel D Rabkin, Dipongkor Saha
Oncolytic herpes simplex viruses (oHSV) preferentially replicate in cancer cells while inducing antitumor immunity, and thus, they are often referred to as in situ cancer vaccines. OHSV infection of tumors elicits diverse host immune responses comprising both innate and adaptive components. Although the innate/adaptive immune responses primarily target the tumor, they also contribute to antiviral immunity, limiting viral replication/oncolysis. OHSV-encoded proteins use various mechanisms to evade host antiviral pathways and immune recognition, favoring oHSV replication, oncolysis and spread. In general, oHSV infection and replication within tumors results in a series of sequential events, such as oncolysis and release of tumor and viral antigens, dendritic cell (DC)-mediated antigen presentation, T cell priming and activation, T cell trafficking and infiltration to tumors, and T cell recognition of cancer cells, leading to tumor (and viral) clearance. These sequential events align with all steps of the cancer-immunity cycle. However, a comprehensive understanding of the interplay between oHSV and host immune responses is crucial to optimize oHSV-induced antitumor immunity and efficacy. Therefore, this review aims to elucidate oHSV's communication with innate and adaptive immune systems and utilize such interactions to improve oHSV's potential as a potent immunovirotherapeutic agent against cancer.
{"title":"Understanding the Interplay between oHSV and the Host Immune System: Implications for Therapeutic Oncolytic Virus Development.","authors":"Kalkidan Ayele, Hiroaki Wakimoto, Hans J Nauwynck, Howard L Kaufman, Samuel D Rabkin, Dipongkor Saha","doi":"10.1016/j.ymthe.2024.12.054","DOIUrl":"https://doi.org/10.1016/j.ymthe.2024.12.054","url":null,"abstract":"<p><p>Oncolytic herpes simplex viruses (oHSV) preferentially replicate in cancer cells while inducing antitumor immunity, and thus, they are often referred to as in situ cancer vaccines. OHSV infection of tumors elicits diverse host immune responses comprising both innate and adaptive components. Although the innate/adaptive immune responses primarily target the tumor, they also contribute to antiviral immunity, limiting viral replication/oncolysis. OHSV-encoded proteins use various mechanisms to evade host antiviral pathways and immune recognition, favoring oHSV replication, oncolysis and spread. In general, oHSV infection and replication within tumors results in a series of sequential events, such as oncolysis and release of tumor and viral antigens, dendritic cell (DC)-mediated antigen presentation, T cell priming and activation, T cell trafficking and infiltration to tumors, and T cell recognition of cancer cells, leading to tumor (and viral) clearance. These sequential events align with all steps of the cancer-immunity cycle. However, a comprehensive understanding of the interplay between oHSV and host immune responses is crucial to optimize oHSV-induced antitumor immunity and efficacy. Therefore, this review aims to elucidate oHSV's communication with innate and adaptive immune systems and utilize such interactions to improve oHSV's potential as a potent immunovirotherapeutic agent against cancer.</p>","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":" ","pages":""},"PeriodicalIF":12.1,"publicationDate":"2024-12-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142915331","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 : 2024-12-30DOI: 10.1016/j.ymthe.2024.12.042
David J Young, Abigail J Edwards, Kevin G Quiroz Caceda, Ella Liberzon, Johana Barrientos, Sogun Hong, Jacob Turner, Peter L Choyke, Sean Arlauckas, Adam S Lazorchak, Richard A Morgan, Noriko Sato, Cynthia E Dunbar
B cells are an attractive platform for engineering to produce protein-based biologics absent in genetic disorders, and potentially for the treatment of metabolic diseases and cancer. As part of pre-clinical development of B cell medicines, we demonstrate a method to collect, ex vivo expand, differentiate, radioactively label, and track adoptively transferred non-human primate (NHP) B cells. These cells underwent 10- to 15-fold expansion, initiated IgG class switching, and differentiated into antibody secreting cells. Zirconium-89-oxine labeled cells were infused into autologous donors without any preconditioning and tracked by PET/CT imaging. Within 24 hours of infusion, 20% of the initial dose homed to the bone marrow and spleen and distributed stably and equally between the two. Interestingly, approximately half of the dose homed to the liver. Image analysis of the bone marrow demonstrated inhomogeneous distribution of the cells. The subjects experienced no clinically significant side effects or laboratory abnormalities. A second infusion of B cells into one of the subjects resulted in an almost identical distribution of cells, suggesting possibly a non-limiting engraftment niche and feasibility of repeated infusions. This work supports the NHP as a valuable model to assess the potential of B cell medicines as potential treatment for human diseases.
{"title":"In vivo tracking of ex vivo generated <sup>89</sup>Zr-oxine labeled plasma cells by PET in a non-human primate model.","authors":"David J Young, Abigail J Edwards, Kevin G Quiroz Caceda, Ella Liberzon, Johana Barrientos, Sogun Hong, Jacob Turner, Peter L Choyke, Sean Arlauckas, Adam S Lazorchak, Richard A Morgan, Noriko Sato, Cynthia E Dunbar","doi":"10.1016/j.ymthe.2024.12.042","DOIUrl":"https://doi.org/10.1016/j.ymthe.2024.12.042","url":null,"abstract":"<p><p>B cells are an attractive platform for engineering to produce protein-based biologics absent in genetic disorders, and potentially for the treatment of metabolic diseases and cancer. As part of pre-clinical development of B cell medicines, we demonstrate a method to collect, ex vivo expand, differentiate, radioactively label, and track adoptively transferred non-human primate (NHP) B cells. These cells underwent 10- to 15-fold expansion, initiated IgG class switching, and differentiated into antibody secreting cells. Zirconium-89-oxine labeled cells were infused into autologous donors without any preconditioning and tracked by PET/CT imaging. Within 24 hours of infusion, 20% of the initial dose homed to the bone marrow and spleen and distributed stably and equally between the two. Interestingly, approximately half of the dose homed to the liver. Image analysis of the bone marrow demonstrated inhomogeneous distribution of the cells. The subjects experienced no clinically significant side effects or laboratory abnormalities. A second infusion of B cells into one of the subjects resulted in an almost identical distribution of cells, suggesting possibly a non-limiting engraftment niche and feasibility of repeated infusions. This work supports the NHP as a valuable model to assess the potential of B cell medicines as potential treatment for human diseases.</p>","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":" ","pages":""},"PeriodicalIF":12.1,"publicationDate":"2024-12-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142915315","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 : 2024-12-30DOI: 10.1016/j.ymthe.2024.12.052
Wandi Zhu, Chunhong Dong, Lai Wei, Joo Kyung Kim, Bao-Zhong Wang
Enhancing protective immunity in the respiratory tract is crucial to combat influenza infection and transmission. Developing mucosal universal influenza vaccines requires effective delivery platforms to overcome the respiratory mucosal barrier and stimulate appropriate innate immune reactions, thereby bridging adaptive immune responses with minimal necessary inflammation. Meanwhile, the vaccine platforms must be biocompatible. This study employed cell-derived extracellular vesicles (EVs) as a mucosal universal influenza vaccine platform. By conjugating influenza hemagglutinin (HA) onto EV surfaces through HA-receptor interaction, we achieved an upside-down (inverted) influenza HA configuration that exposed the conserved HA stalk region while partially hiding the globular head domain. Intranasal immunization with the resulting EVs induced robust HA stalk- and virus-specific serum antibody and mucosal immune responses in mice, protecting against heterologous virus infection. Notably, EVs derived from the lung epithelial cell line A549 induced superior cross-reactive antibodies and enhanced protection upon intranasal immunization. EVs conjugating multivalent HA elicited broadly cross-reactive antibody and cellular responses against different influenza strains. Our results demonstrated that EVs conjugating multiple inverted HAs represented an effective strategy for developing a mucosal universal influenza vaccine.
{"title":"Immunization with extracellular vesicles conjugating inverted influenza HA elicits HA stalk-specific immunity and cross-protection in mice.","authors":"Wandi Zhu, Chunhong Dong, Lai Wei, Joo Kyung Kim, Bao-Zhong Wang","doi":"10.1016/j.ymthe.2024.12.052","DOIUrl":"https://doi.org/10.1016/j.ymthe.2024.12.052","url":null,"abstract":"<p><p>Enhancing protective immunity in the respiratory tract is crucial to combat influenza infection and transmission. Developing mucosal universal influenza vaccines requires effective delivery platforms to overcome the respiratory mucosal barrier and stimulate appropriate innate immune reactions, thereby bridging adaptive immune responses with minimal necessary inflammation. Meanwhile, the vaccine platforms must be biocompatible. This study employed cell-derived extracellular vesicles (EVs) as a mucosal universal influenza vaccine platform. By conjugating influenza hemagglutinin (HA) onto EV surfaces through HA-receptor interaction, we achieved an upside-down (inverted) influenza HA configuration that exposed the conserved HA stalk region while partially hiding the globular head domain. Intranasal immunization with the resulting EVs induced robust HA stalk- and virus-specific serum antibody and mucosal immune responses in mice, protecting against heterologous virus infection. Notably, EVs derived from the lung epithelial cell line A549 induced superior cross-reactive antibodies and enhanced protection upon intranasal immunization. EVs conjugating multivalent HA elicited broadly cross-reactive antibody and cellular responses against different influenza strains. Our results demonstrated that EVs conjugating multiple inverted HAs represented an effective strategy for developing a mucosal universal influenza vaccine.</p>","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":" ","pages":""},"PeriodicalIF":12.1,"publicationDate":"2024-12-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142915310","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 : 2024-12-30DOI: 10.1016/j.ymthe.2024.12.037
Julie Rodor, Eftychia Klimi, Simon D Brown, Georgios Krilis, Luca Braga, Nadja A R Ring, Margaret D Ballantyne, Despoina Kesidou, Aurelie Nguyen Dinh Cat, Vladislav Miscianinov, Francesca Vacante, Katarina Miteva, Matthew Bennett, Abdelaziz Beqqali, Mauro Giacca, Serena Zacchigna, Andrew H Baker
Proliferation of vascular smooth muscle cells (vSMCs) is a crucial contributor to pathological vascular remodeling. MicroRNAs (miRNAs) are powerful gene regulators and attractive therapeutic agents. Here, we aimed to systematically identify and characterize miRNAs with therapeutic potential in targeting vSMC proliferation. Using high-throughput screening, we assessed the impact of 2,042 human miRNA mimics on vSMC proliferation and identified seven miRNAs with novel vSMC anti-proliferative function: miR-323a-3p, miR-449b-5p, miR-491-3p, miR-892b, miR-1827, miR-4774-3p, and miR-5681b. miRNA-mimic treatment affects proliferation of vSMCs from different vascular beds. Focusing on vein graft failure, where miRNA-based therapeutics can be applied to the graft ex vivo, we showed that these miRNAs reduced human saphenous vein smooth muscle cell (HSVSMC) proliferation without toxic effect. HSVSMC transcriptomics revealed a distinct set of targets for each miRNA, leading to the common downregulation of a cell-cycle gene network for all miRNAs. For miR-449b-5p, we showed that its candidate target, CCND1, contributes to HSVSMC proliferation. In contrast to HSVSMCs, miRNA overexpression in endothelial cells led to a limited response in terms of proliferation and transcriptomics. In an ex vivo vein organ model, overexpression of miR-323a-3p and miR-449b-5p reduced medial proliferation. Collectively, the results of our study show the therapeutic potential of seven miRNAs to target pathological vascular remodeling.
{"title":"Functional screening identifies miRNAs with a novel function inhibiting vascular smooth muscle cell proliferation.","authors":"Julie Rodor, Eftychia Klimi, Simon D Brown, Georgios Krilis, Luca Braga, Nadja A R Ring, Margaret D Ballantyne, Despoina Kesidou, Aurelie Nguyen Dinh Cat, Vladislav Miscianinov, Francesca Vacante, Katarina Miteva, Matthew Bennett, Abdelaziz Beqqali, Mauro Giacca, Serena Zacchigna, Andrew H Baker","doi":"10.1016/j.ymthe.2024.12.037","DOIUrl":"10.1016/j.ymthe.2024.12.037","url":null,"abstract":"<p><p>Proliferation of vascular smooth muscle cells (vSMCs) is a crucial contributor to pathological vascular remodeling. MicroRNAs (miRNAs) are powerful gene regulators and attractive therapeutic agents. Here, we aimed to systematically identify and characterize miRNAs with therapeutic potential in targeting vSMC proliferation. Using high-throughput screening, we assessed the impact of 2,042 human miRNA mimics on vSMC proliferation and identified seven miRNAs with novel vSMC anti-proliferative function: miR-323a-3p, miR-449b-5p, miR-491-3p, miR-892b, miR-1827, miR-4774-3p, and miR-5681b. miRNA-mimic treatment affects proliferation of vSMCs from different vascular beds. Focusing on vein graft failure, where miRNA-based therapeutics can be applied to the graft ex vivo, we showed that these miRNAs reduced human saphenous vein smooth muscle cell (HSVSMC) proliferation without toxic effect. HSVSMC transcriptomics revealed a distinct set of targets for each miRNA, leading to the common downregulation of a cell-cycle gene network for all miRNAs. For miR-449b-5p, we showed that its candidate target, CCND1, contributes to HSVSMC proliferation. In contrast to HSVSMCs, miRNA overexpression in endothelial cells led to a limited response in terms of proliferation and transcriptomics. In an ex vivo vein organ model, overexpression of miR-323a-3p and miR-449b-5p reduced medial proliferation. Collectively, the results of our study show the therapeutic potential of seven miRNAs to target pathological vascular remodeling.</p>","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":" ","pages":""},"PeriodicalIF":12.1,"publicationDate":"2024-12-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142907285","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 : 2024-12-30DOI: 10.1016/j.ymthe.2024.12.043
Francesca Di Leva, Michele Arnoldi, Stefania Santarelli, Mathieu Massonot, Marianne Victoria Lemée, Carlotta Bon, Miguel Pellegrini, Maria Elena Castellini, Giulia Zarantonello, Andrea Messina, Yuri Bozzi, Raphael Bernier, Silvia Zucchelli, Simona Casarosa, Erik Dassi, Giuseppe Ronzitti, Christelle Golzio, Jasmin Morandell, Stefano Gustincich, Stefano Espinoza, Marta Biagioli
Loss-of-function mutations in the chromodomain helicase DNA-binding 8 (CHD8) gene are strongly associated with Autism Spectrum Disorders (ASD). Indeed, the reduction of CHD8 causes transcriptional, epigenetic and cellular phenotypic changes correlated to disease, that can be monitored in assessing new therapeutic approaches. SINEUPs are a functional class of natural and synthetic antisense long non-coding RNAs able to stimulate the translation of sense target mRNA, with no effect on transcription. Here we employed synthetic SINEUP-CHD8 targeting the first and third AUG of the CHD8 coding sequence to efficiently stimulate endogenous CHD8 protein production. SINEUP-CHD8 were effective in cells with reduced levels of the target protein and in patients'-derived fibroblasts with CHD8 mutations. Functionally, SINEUP-CHD8 were able to revert molecular phenotypes associated with CHD8-suppression, i.e. genome-wide transcriptional dysregulation, and the reduction of H3K36me3 levels. Strikingly, in chd8-morpholino-treated and ENU mutant zebrafish embryos, SINEUP-chd8 injection confirmed the ability of SINEUP RNA to rescue the chd8-suppression-induced macrocephaly phenotype and neuronal hyperproliferation. Thus, SINEUP-CHD8 molecule(s) represent a proof-of-concept towards the development of a RNA-based therapy for neurodevelopmental syndromes with implications for, and beyond ASD, and relevant to genetic disorders caused by protein haploinsufficiency.
{"title":"SINEUP RNA rescues molecular phenotypes associated with CHD8 suppression in Autism Spectrum Disorder model systems.","authors":"Francesca Di Leva, Michele Arnoldi, Stefania Santarelli, Mathieu Massonot, Marianne Victoria Lemée, Carlotta Bon, Miguel Pellegrini, Maria Elena Castellini, Giulia Zarantonello, Andrea Messina, Yuri Bozzi, Raphael Bernier, Silvia Zucchelli, Simona Casarosa, Erik Dassi, Giuseppe Ronzitti, Christelle Golzio, Jasmin Morandell, Stefano Gustincich, Stefano Espinoza, Marta Biagioli","doi":"10.1016/j.ymthe.2024.12.043","DOIUrl":"https://doi.org/10.1016/j.ymthe.2024.12.043","url":null,"abstract":"<p><p>Loss-of-function mutations in the chromodomain helicase DNA-binding 8 (CHD8) gene are strongly associated with Autism Spectrum Disorders (ASD). Indeed, the reduction of CHD8 causes transcriptional, epigenetic and cellular phenotypic changes correlated to disease, that can be monitored in assessing new therapeutic approaches. SINEUPs are a functional class of natural and synthetic antisense long non-coding RNAs able to stimulate the translation of sense target mRNA, with no effect on transcription. Here we employed synthetic SINEUP-CHD8 targeting the first and third AUG of the CHD8 coding sequence to efficiently stimulate endogenous CHD8 protein production. SINEUP-CHD8 were effective in cells with reduced levels of the target protein and in patients'-derived fibroblasts with CHD8 mutations. Functionally, SINEUP-CHD8 were able to revert molecular phenotypes associated with CHD8-suppression, i.e. genome-wide transcriptional dysregulation, and the reduction of H3K36me3 levels. Strikingly, in chd8-morpholino-treated and ENU mutant zebrafish embryos, SINEUP-chd8 injection confirmed the ability of SINEUP RNA to rescue the chd8-suppression-induced macrocephaly phenotype and neuronal hyperproliferation. Thus, SINEUP-CHD8 molecule(s) represent a proof-of-concept towards the development of a RNA-based therapy for neurodevelopmental syndromes with implications for, and beyond ASD, and relevant to genetic disorders caused by protein haploinsufficiency.</p>","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":" ","pages":""},"PeriodicalIF":12.1,"publicationDate":"2024-12-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142915326","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}