Mehrnoush Safarzadeh, Nastaran Saadat, Sara Abbasi-Molaei, Mohsen Rastegari-Pouyani
{"title":"Extracellular vesicles as missiles for enhanced anti-tumor efficacy of oncolytic viruses: from disseminating oncolysis and anti-tumor immunity to targeted delivery.","authors":"Mehrnoush Safarzadeh, Nastaran Saadat, Sara Abbasi-Molaei, Mohsen Rastegari-Pouyani","doi":"10.1186/s12964-025-02283-z","DOIUrl":"https://doi.org/10.1186/s12964-025-02283-z","url":null,"abstract":"<p><p>Oncolytic viruses (OVs) have been introduced as a promising anti-cancer platform capable of selectively killing cancer cells (oncolysis) without causing damage to normal cells. These viruses also stimulate the innate and adaptive immune responses and can remodel the tumor microenvironment (TME) in favor of anti-tumor immunity. Still, successful and efficient OV application in the clinical settings encounters several challenges including immune mediated clearance and low tumor tissue targeting. Extracellular vesicles (EVs) have emerged as a useful vehicle for efficient and selective delivery of various therapeutics including chemotherapy drugs, nucleic acid-based anti-cancer agents and immunostimulatory molecules into the TME. EVs can also be employed as a proper vehicle to enhance OVs tumor-selective delivery and protect them from the host immune system mediated removal following systemic administration as a key hurdle on the way of an efficacious oncolytic virotherapy. OVs can also take advantage of EVs to spread their oncolytic activity to metastatic sites distant from the primary tumor as well as instigate robust anti-tumor immune responses. In this paper, we have tried to review the current literature regarding the roles and applications of EVs in cancer OV therapy and provide a conclusion on future directions and application of this novel technology.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"23 1","pages":"276"},"PeriodicalIF":8.2,"publicationDate":"2025-06-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144267919","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"RNA polymerase II subunit 5-mediating protein limits TLR4-induced innate immune activation in macrophages by inhibiting IKKβ/NF-κB signaling during sepsis.","authors":"Shu-Jie Pang, Tian-Yi Jiang, Nai-Guo Wang, Xiao-Wen Cui, Hui Wang, Yu-Fei Pan, Ning Yang, Li-Wei Dong","doi":"10.1186/s12964-025-02278-w","DOIUrl":"https://doi.org/10.1186/s12964-025-02278-w","url":null,"abstract":"<p><strong>Background: </strong>Nuclear factor κB (NF-κB) activity is a central component of inflammatory and innate immune responses, which plays a crucial role in sepsis. The inhibition of NF-κB signaling and the IκB kinase (IKK) complex is important for understanding the control of innate immunity and regulating the progress of sepsis.</p><p><strong>Methods: </strong>We constructed transgenic mouse strains (Rmp<sup>f/f</sup>; Lyz2-Cre<sup>+</sup>), and then established lipopolysaccharide (LPS), cecal ligation and perforation (CLP)-induced sepsis models. Hematoxylin-eosin (HE) staining, ELISA, and flow cytometry assay were employed to evaluate the sepsis-related damage and the activation of the inflammatory-related signaling pathway. In vitro, differential expression of RMP cell lines and primary macrophage isolated from transgenic mice were utilized to assess the activation of the NF-κB signaling pathway by Western blot (WB), reverse transcription-polymerase chain reaction (RT-PCR), and ELISA tests. Co‑immunoprecipitation (Co-IP), WB, GST-pulldown, phosphorylation mass spectrometry, surface plasmon resonance (SPR), and IKK activity detection assay were employed to investigate the underlying molecular mechanism by which RMP restrains IKK-NF-κB pathway.</p><p><strong>Results: </strong>We identified RNA polymerase II subunit 5 (RPB5)-mediating protein (RMP) as an inhibitor of the IKK complex, which thus inhibited NF-κB signaling in macrophages. In resting macrophages, RMP was directly bound to the kinase domain of IKKβ and inhibited its activity by recruiting protein phosphatase 2 A (PP2A) to the IKK complex. When mouse macrophages were treated with LPS, a Toll-like receptor 4 (TLR4) agonist that stimulates NF-κB signaling, RMP was phosphorylated by IKKβ at Ser<sup>439</sup> and dissociated from the IKK complex, which further activated NF-κB signaling. Macrophage-specific deletion of Rmp reduced survival in mice due to an increased inflammatory response in experimental models of sepsis.</p><p><strong>Conclusions: </strong>RMP inhibits TLR4-induced NF-κB activation and exerts homeostatic control of innate immunity, and may be promising as a therapeutic target in the limiting of NF-κB signaling and attenuating sepsis-related damage.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"23 1","pages":"274"},"PeriodicalIF":8.2,"publicationDate":"2025-06-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144267852","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Sisi Zhang, Ernest H Y Ng, William S B Yeung, Rachel W S Chan
{"title":"Myometrial extracellular vesicles promoted endometrial mesenchymal stem/stromal cells to self-renewal via jag1-mediated notch signaling.","authors":"Sisi Zhang, Ernest H Y Ng, William S B Yeung, Rachel W S Chan","doi":"10.1186/s12964-025-02282-0","DOIUrl":"https://doi.org/10.1186/s12964-025-02282-0","url":null,"abstract":"<p><strong>Background: </strong>In the human endometrium, studies show the importance of extracellular vesicles in mediating various physiological as well as pathological processes. We have demonstrated that the myometrial cells are candidate niche cells of the endometrial mesenchymal stem/stromal cells (eMSC) modulating their biological function. The Notch signaling pathway regulates the endometrial stem cell functions. Although classical Notch signaling relies on direct cell contract for actions, this pathway can also be activated at a distance by Notch ligands containing extracellular vesicles (EV). We hypothesized that certain Notch ligand(s) are packaged into the myometrial EV to mediate stem cell functions.</p><p><strong>Methods: </strong>Endometrial samples were obtained from women undergoing total abdominal hysterectomy. Endometrial MSC (CD140b<sup>+</sup>CD146<sup>+</sup> cells) were cocultured with myometrial EV and the percentage of eMSC was analysed by flow cytometry. Blockage of the secretion of EV was performed by transfection of RAB27 A siRNA. Western blot analysis and gene silencing approach were used to validate the role of Notch signaling in eMSC. The therapeutic features of transplanted eMSC/myometrial EV was determined using a mouse injured endometrium model.</p><p><strong>Results: </strong>EV released from myometrial cells could be internalized by eMSC, leading to a significant stimulatory effect on the self-renewal and clonogenic activity of eMSC. Pharmacological inhibition of Notch signaling with DAPT or silencing of NOTCH 1 nullified the stimulatory effects. Myometrial EV contains a high amount of the Notch ligand - JAG1, thus inducing a strong Notch activity in eMSC. When JAG1 was silenced in the myometrial EV, the self-renewal and clonogenic activity was reduced. Combined transplantation of eMSC with myometrial EV improves the therapeutic effect of eMSC in endometrial regeneration in vivo. The observed therapeutic feature was potentially achieved by elevating the cell proliferation and suppressing apoptosis in the injured mouse endometrium.</p><p><strong>Conclusions: </strong>This study identifies a novel EV mediated communication axis between the myometrial cells and the eMSC, providing new insights into endometrial regeneration. The findings highlight the potential of eMSC and myometrial EV as a therapeutic strategy for women with intrauterine adhesions and other endometrial disorders.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"23 1","pages":"275"},"PeriodicalIF":8.2,"publicationDate":"2025-06-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144267920","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Rosa Mistica C Ignacio, Helen Forgham, Zerong Ma, Anya Jensen, George Sharbeen, Juanfang Ruan, David S Ziegler, Maria Tsoli, Phoebe A Phillips, Chelsea Mayoh, Maria Kavallaris, Joshua McCarroll
{"title":"Chemotherapy reprograms miRNA expression profiles in apoptotic extracellular vesicles from medulloblastoma cells, regulating pro- and anti-proliferative effects on recipient drug-naïve cells.","authors":"Rosa Mistica C Ignacio, Helen Forgham, Zerong Ma, Anya Jensen, George Sharbeen, Juanfang Ruan, David S Ziegler, Maria Tsoli, Phoebe A Phillips, Chelsea Mayoh, Maria Kavallaris, Joshua McCarroll","doi":"10.1186/s12964-025-02241-9","DOIUrl":"https://doi.org/10.1186/s12964-025-02241-9","url":null,"abstract":"<p><strong>Background: </strong>Extracellular vesicles (EVs) play a crucial role in intercellular communication. While the effects of EVs released from living or non-dying cancer cells are well characterized, the impact of EVs released from chemotherapy-treated or apoptotic cancer cells is less understood. This study investigated the effects of the chemotherapy agent cisplatin on EV release and miRNA content in apoptotic medulloblastoma cells, as well as their influence on the growth of drug-naïve recipient cancer cells.</p><p><strong>Methods: </strong>EVs were isolated from cisplatin-treated and untreated SHH and group 3 medulloblastoma cells, as well as from the blood of mice with orthotopic medulloblastoma tumors. EVs were characterized using nanoparticle tracking analysis, cryo-TEM, and western blotting, and their impact on the growth of recipient medulloblastoma cells in 2D and 3D cultures was assessed. EV-miRNAs were analyzed using small RNA sequencing and qPCR, and the effects of candidate miRNA overexpression on medulloblastoma cell growth and apoptosis were evaluated.</p><p><strong>Results: </strong>We demonstrate that apoptotic SHH and group 3 medulloblastoma cells secrete increased numbers of EVs (size range 150-600 nm) both in vitro and in vivo. EVs isolated from cisplatin-treated SHH and group 3 medulloblastoma cells were internalized by recipient medulloblastoma cells and exhibited distinct effects on their growth. EVs from cisplatin-treated SHH medulloblastoma cells reduced clonogenic growth in recipient drug-naïve medulloblastoma cells, whereas EVs from cisplatin-treated group 3 medulloblastoma cells enhanced the clonogenic and sphere-forming capacity of recipient cells. These contrasting effects were associated with significant alterations in EV-miRNA expression profiles between untreated and cisplatin-treated SHH and group 3 medulloblastoma cells. Notably, miR-449a was found to be upregulated in EVs from cisplatin-treated SHH medulloblastoma cells, and its overexpression in medulloblastoma cells led to potent inhibition of growth.</p><p><strong>Conclusions: </strong>Our findings demonstrate, for the first time, that cisplatin-treated medulloblastoma cells from distinct molecular subgroups secrete EVs with altered miRNA expression profiles that either inhibit or promote the growth of recipient cancer cells. This underscores the potential of targeting EV-mediated communication as a novel therapeutic strategy in medulloblastoma.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"23 1","pages":"273"},"PeriodicalIF":8.2,"publicationDate":"2025-06-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144267918","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Behnaz Mirzaahmadi, Shahin Ahmadian, Parinaz Haddadi, Parinaz Nezhad-Mokhtari, Fereshteh Vaziri Nezamdoust, Banafsheh Yalameha, Sara Aghakhani Chegeni, Somayyeh Rashidi, Akbar Mousakhani, Emel Sokullu, Hajar Shafaei, Reza Rahbarghazi, Mohammad Karimipour
{"title":"Neuroangiogenesis potential of mesenchymal stem cell extracellular vesicles in ischemic stroke conditions.","authors":"Behnaz Mirzaahmadi, Shahin Ahmadian, Parinaz Haddadi, Parinaz Nezhad-Mokhtari, Fereshteh Vaziri Nezamdoust, Banafsheh Yalameha, Sara Aghakhani Chegeni, Somayyeh Rashidi, Akbar Mousakhani, Emel Sokullu, Hajar Shafaei, Reza Rahbarghazi, Mohammad Karimipour","doi":"10.1186/s12964-025-02286-w","DOIUrl":"10.1186/s12964-025-02286-w","url":null,"abstract":"<p><p>Ischemic stroke (IS) is a life-threatening condition in humans with high morbidity and mortality rates in developing and industrialized countries. The occlusion of blood-supporting vessels by thrombus or emboli can contribute to massive brain cell damage, neurological deficits, and long-term disability, and in more severe conditions, results in sudden death. Current therapeutic strategies, along with rehabilitation, in part, but not completely, can restore the integrity and function of the brain. These features necessitate the advent of novel therapeutic protocols for yielding better regenerative outcomes in IS patients. In past decades, the discovery of stem cells and byproducts has led to promising results in in vitro settings and pre-clinical studies. Extracellular vesicles (EVs) are nano-sized particles released from various cell types, for instance, mesenchymal stem cells (MSCs), with certain signaling biomolecules, growth factors, and cytokines involved in cell-to-cell communication. A great plethora of studies have pointed to the fact that EVs with specific cargo can distribute easily in different parts of the body, making them appropriate therapeutics under different pathological conditions. The current review articles aimed to highlight the neuroangiogenesis properties of MSC EVs in IS conditions. How and by which mechanisms MSC EVs can orchestrate the process of nervous system regeneration is at the center of debate. We think that the current article can help us better understand MSC EVs' function in the restoration of brain function under IS conditions in terms of neurogenesis and angiogenesis.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"23 1","pages":"272"},"PeriodicalIF":8.2,"publicationDate":"2025-06-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12145630/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144250918","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Combination strategies of gut microbiota in cancer therapy through metabolic reprogramming and immune remodeling.","authors":"Lixuan Wang, Weibo Jiang, Manshi Yang, Quezhu Danzeng, Shiyu Liu, Mengying Cui","doi":"10.1186/s12964-025-02275-z","DOIUrl":"10.1186/s12964-025-02275-z","url":null,"abstract":"<p><p>Over the past decade, there has been a qualitative improvement in the understanding of gut microbiota in cancer development and treatment. Gut microbiota regulates metabolic reprogramming that occurs in the competition between tumor cells and immune cells for essential nutrients in the tumor microenvironment (TME). Besides, gut microbiota dysbiosis is one of the key factors leading to the formation of aging environment, which has many similarities with the TME. Accumulating research findings have demonstrated that gut microbiota enhances the efficacy of cancer therapies by activating the immune system and facilitating the biotransformation of drugs. Other studies also have shown that specific microbial composition is the effective biomarker of drug resistance and toxicity for cancer treatment. Microbiota-directed therapies are being explored intensively for their potential in cancer prevention and treatment. In this review, we summarize the role of gut microbiota in metabolic reprogramming and immune remodeling; provide an overview of the relationship between gut microbiota and the efficacy, resistance, and toxicity of cancer treatment; propose a series of strategies to integrate gut microbiota into cancer treatment, optimizing antitumor effectiveness and reducing side effects.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"23 1","pages":"270"},"PeriodicalIF":8.2,"publicationDate":"2025-06-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12139130/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144235984","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Haixia Li, Quan Zhou, Nan Cao, Chenghao Hu, Jincheng Wang, Yu He, Shan Jiang, Qi Li, Miao Chen, Li Gong, Ming Luo, Xinzhou Deng, Zhiguo Luo
{"title":"Nanobodies and their derivatives: pioneering the future of cancer immunotherapy.","authors":"Haixia Li, Quan Zhou, Nan Cao, Chenghao Hu, Jincheng Wang, Yu He, Shan Jiang, Qi Li, Miao Chen, Li Gong, Ming Luo, Xinzhou Deng, Zhiguo Luo","doi":"10.1186/s12964-025-02270-4","DOIUrl":"10.1186/s12964-025-02270-4","url":null,"abstract":"<p><p>Cancer immunotherapy, which boosts the immune system to recognize and attack malignant cells, has revolutionized traditional cancer treatment paradigms. Approaches such as chimeric antigen receptor T cell (CAR-T) therapy and immune checkpoint inhibitors (ICIs) have demonstrated promising therapeutic outcomes, leading to the approval of numerous immuno-oncology agents by the US Food and Drug Administration (FDA) over the past few decades. Immuno-oncology agents, mainly based on conventional full-length antibodies or their derivatives, are widely used in cancer immunotherapy. However, their large size, unwanted immunogenicity, poor solubility, complex molecular architectures, and limited tumor penetration pose significant challenges that must be addressed. Nanobodies, which are single-domain antibody fragments originating from the variable regions of camelid heavy-chain immunoglobulins, represent the smallest known antigen-binding fragments. In addition to their small size (~ 15 kDa), nanobodies possess a range of advantageous properties, including high stability, strong specificity and affinity for target antigens, low immunogenicity, and cost-effective production. Nonetheless, their short serum half-life and lack of Fc-mediated functions may limit efficacy, which can be addressed by Fc fusion, albumin binding, or multivalent construct design. These properties enable nanobodies to support multifunctional constructs, such as bispecific CARs, nanobody-secreting CARs, dual ICI-containing molecules, and trispecific immune cell-engaging antibodies. In recent years, a growing number of nanobody-based immuno-oncology agents have progressed into preclinical and clinical trials, with several products approved by the US FDA and China's National Medical Products Administration for cancer therapy. In this review, we explore the unique properties of nanobodies and provide a comprehensive summary of recent preclinical and clinical advancements in nanobody-based immuno-oncology agents, with a focus on their applications in CAR-T cells, ICIs, and immune cell-engaging antibodies. Through their unique capacity to integrate innovative molecular engineering with translational clinical development, nanobody-based therapeutics are poised to revolutionize current paradigms in cancer immunotherapy.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"23 1","pages":"271"},"PeriodicalIF":8.2,"publicationDate":"2025-06-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12142954/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144235985","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Feifei Yan, Sara Göransson, Helene Olofsson, Christos Vogiatzakis, Anagha Acharekar, Staffan Strömblad
{"title":"Matrix stiffness-induced IKBKE and MAPK8 signaling drives a phenotypic switch from DCIS to invasive breast cancer.","authors":"Feifei Yan, Sara Göransson, Helene Olofsson, Christos Vogiatzakis, Anagha Acharekar, Staffan Strömblad","doi":"10.1186/s12964-025-02276-y","DOIUrl":"10.1186/s12964-025-02276-y","url":null,"abstract":"<p><p>Ductal carcinoma in situ (DCIS) is not life threatening unless it transitions into invasive breast cancer (IBC). However, although breast cancer cell exposure to matrix stiffening in vitro phenotypically mimics the DCIS to IBC switch, the molecular changes driving this switch remains unclear. Here, breast cancer cell kinome activity profiling suggested matrix stiffness-upregulation of 53 kinases, among which 16 kinases were also regulated by integrin β1. Functional validation identified matrix stiffness-activation of inhibitor of nuclear factor kappa-B kinase subunit epsilon (IKBKE) and mitogen-activated protein kinase 8 (MAPK8) signaling as critical for the stiffness-driven IBC phenotype, including for cell proliferation. The IKBKE-inhibitor Amlexanox, clinically utilized for aphthous ulcers, as well as the MAPK8 inhibitor JNK-IN-8, reinstalled the DCIS-like phenotype of breast cancer cells on high matrix stiffness. This suggests that IKBKE and/or MAPK8 inhibitors could enhance the arsenal of treatments to prevent or treat breast cancer.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"23 1","pages":"269"},"PeriodicalIF":8.2,"publicationDate":"2025-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12139146/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144227759","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Functionalized exosomes for targeted therapy in cancer and regenerative medicine: genetic, chemical, and physical modifications.","authors":"Salar Ghaffari Gabaran, Navid Ghasemzadeh, Maryam Rahnama, Erkan Karatas, Ali Akbari, Jafar Rezaie","doi":"10.1186/s12964-025-02268-y","DOIUrl":"10.1186/s12964-025-02268-y","url":null,"abstract":"<p><strong>Background: </strong>Extracellular vesicles (EVs), such as exosomes, have been extensively discovered for their function in various diseases and potential therapeutic properties. In this review, we aimed to describe the therapeutic roles of functionalized exosomes in cancer and regenerative medicine.</p><p><strong>Methods: </strong>In this review study, we studied numerous articles over the past two decades published on the application of exosomes in different diseases, as well as on perspectives and challenges in this field.</p><p><strong>Results: </strong>Recent advancements have shown that exosomes can be used as a drug delivery system. However, this approach faces challenges such as low efficiency and non-targeting effects. Different methods, including genetic, chemical, and physical modifications, are used to functionalize exosome surfaces to address these limitations. In some cases, a combination of modification methods has been used to produce smart exosomes. Different therapeutic agents have been inserted on exosome surfaces by different modification methods. These functionalized exosomes can effectively deliver therapeutic agents to target cells. A growing body of evidence shows that functionalized exosomes are promising for cancer therapy and regenerative medicine. They can not only effectively deliver therapeutic agents to cancer cells, inhibiting tumorigenesis, but also efficiently contribute to tissue repair and regeneration by increasing cell proliferation and angiogenesis. In this review, we discuss different modification methods used to functionalize exosomes and related studies. In addition, we describe the application of functionalized exosomes in cancer and regeneration, along with challenges and perspectives.</p><p><strong>Conclusions: </strong>Although functionalized exosomes show promising results, further studies are essential for the clinical translation of these exosomes.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"23 1","pages":"265"},"PeriodicalIF":8.2,"publicationDate":"2025-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12135327/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144227758","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yinghui Li, Xinzhe Chen, Shudan Xiao, Haoxuan Wang, Bo Li, MeiHua Zhang, Kun Wang
{"title":"Unlocking the life code: a review of SnoRNA functional diversity and disease relevance.","authors":"Yinghui Li, Xinzhe Chen, Shudan Xiao, Haoxuan Wang, Bo Li, MeiHua Zhang, Kun Wang","doi":"10.1186/s12964-025-02274-0","DOIUrl":"10.1186/s12964-025-02274-0","url":null,"abstract":"<p><p>Nucleolar small RNA (snoRNA), as a class of non-coding RNAs, play a crucial role in eukaryotic cells. They are widely involved in post-transcriptional modifications of ribosomal RNAs, including methylation and pseudouridylation, precisely regulating the process of ribosome biogenesis, ensuring the integrity of ribosome structure and function, and thereby guaranteeing the accuracy and efficiency of protein synthesis. snoRNAs not only maintain cell growth, proliferation, and differentiation under normal physiological conditions but also have abnormal expression closely associated with various diseases, such as cancer, cardiovascular diseases and neurodegenerative diseases. In recent years, with the innovation of research techniques, there has been a deeper exploration of the biosynthesis pathways, functional mechanisms of snoRNAs, and their potential value in disease diagnosis and treatment. This review comprehensively summarizes the structural characteristics, classification systems, biological functions, and disease associations of snoRNAs, and looks forward to future research directions, aiming to provide a systematic reference for further exploration of the mysteries of snoRNAs in related fields.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"23 1","pages":"266"},"PeriodicalIF":8.2,"publicationDate":"2025-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12135493/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144227781","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}