Romina Nassini, Martina Chieca, Matilde Marini, Lorenzo Landini, Francesco De Logu
{"title":"AAV vectors: an emerging strategy for chronic pain management.","authors":"Romina Nassini, Martina Chieca, Matilde Marini, Lorenzo Landini, Francesco De Logu","doi":"10.1016/j.molmed.2025.08.008","DOIUrl":"https://doi.org/10.1016/j.molmed.2025.08.008","url":null,"abstract":"<p><p>Chronic pain affects millions of people worldwide and represents a crucial public health issue. A growing body of preclinical evidence suggests that adeno-associated virus (AAV) vectors are a powerful gene therapy tool for chronic pain management. We systematically summarize how AAV vectors, by targeting molecular pain pathways in neuronal and non-neuronal cells, may offer precise and sustained pain relief. We provide an overview of the latest findings and emerging concepts in this area, and discuss preclinical innovations in neuropathic, inflammatory and cancer-related pain. We also present clinical data on pain modulation by AAV in osteoarthritis (OA) patients. Finally, we provide a thorough analysis of key concerns about AAV therapy, including potential immune responses, toxicity, and adverse effects.</p>","PeriodicalId":23263,"journal":{"name":"Trends in molecular medicine","volume":" ","pages":""},"PeriodicalIF":13.8,"publicationDate":"2025-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145091537","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}
{"title":"MAFLD: a ferroptotic disease.","authors":"Shaojie Cui, Jin Ye","doi":"10.1016/j.molmed.2025.08.006","DOIUrl":"10.1016/j.molmed.2025.08.006","url":null,"abstract":"<p><p>Ferroptosis, a regulated cell death pathway driven by iron-catalyzed lipid peroxidation, has recently been implicated as a major cause of hepatic injury in metabolic dysfunction-associated fatty liver disease (MAFLD). This review highlights how the identification of hyperoxidized peroxiredoxin 3 (PRDX3) as a ferroptosis-specific marker has led to the discovery that ferroptosis contributes to liver injury in MAFLD, and summarizes other emerging evidence connecting ferroptosis to MAFLD pathogenesis. These new findings suggest that dietary fat composition and genetic variants such as PNPLA3(I148M) may affect the progression of MAFLD by regulating cellular sensitivity to ferroptosis. Recognizing MAFLD as a ferroptotic disease provides novel insights into the pathogenesis of the disease, and supports the exploration of ferroptosis as a potential target for therapeutic intervention.</p>","PeriodicalId":23263,"journal":{"name":"Trends in molecular medicine","volume":" ","pages":""},"PeriodicalIF":13.8,"publicationDate":"2025-09-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12431636/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145034306","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Sean Escopete, Madelyn Arzt, Maedeh Mozneb, Jemima Moses, Arun Sharma
{"title":"Human cardiac organoids for disease modeling and drug discovery.","authors":"Sean Escopete, Madelyn Arzt, Maedeh Mozneb, Jemima Moses, Arun Sharma","doi":"10.1016/j.molmed.2025.08.004","DOIUrl":"https://doi.org/10.1016/j.molmed.2025.08.004","url":null,"abstract":"<p><p>Cardiac organoids are 3D self-assembling structures that recapitulate some of the functional, structural, and cellular aspects of the developing heart. Cardiac organoid modeling has overcome many of the limitations of current cardiac modeling systems by providing a human-relevant, multicellular, spatially advanced model that can replicate early key developmental stages of human cardiogenesis. Recent advancements in cardiac organoid modeling have enabled further understanding of cardiogenesis, cardiovascular disease, and drug-induced cardiotoxicity. Emerging tools to effectively characterize cardiac organoid models to understand their morphology, function, and cellular phenotype will enable further understanding of cardiac development, cardiovascular disease, and preclinical drug discovery.</p>","PeriodicalId":23263,"journal":{"name":"Trends in molecular medicine","volume":" ","pages":""},"PeriodicalIF":13.8,"publicationDate":"2025-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145008499","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}
Constanze Schanbacher, Maria-Elisabeth Goebeler, Brenda Gerull, Kristina Lorenz
{"title":"Targeting pathological ERK1/2 signaling in cancer and beyond.","authors":"Constanze Schanbacher, Maria-Elisabeth Goebeler, Brenda Gerull, Kristina Lorenz","doi":"10.1016/j.molmed.2025.08.001","DOIUrl":"https://doi.org/10.1016/j.molmed.2025.08.001","url":null,"abstract":"<p><p>Dysregulation of the RAF-MEK-ERK1/2 pathway is involved in the pathoetiology of many diseases. Its central role in cancer has led to the development of drugs targeting upstream receptors, RAS, and kinases in the extracellular signal-regulated kinase 1 (ERK1) and 2 (ERK2) signaling cascade. The use of these drugs in cancer therapy - together with ongoing monitoring of their effectiveness, evolving side-effects, and resistance mechanisms - has expanded our knowledge of both the physiological and pathological functions of ERK1/2 and could thus provide potential alternative therapeutic strategies. In this review we discuss the latest insights into targeting of MEK1/2 and ERK1/2 and the transfer of the lessons learned from cancer treatment to further indications involving ERK1/2 dysregulation such as genetic disorders (RASopathies) and beyond.</p>","PeriodicalId":23263,"journal":{"name":"Trends in molecular medicine","volume":" ","pages":""},"PeriodicalIF":13.8,"publicationDate":"2025-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145008455","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}
Stephen D Robinson, Chrysa Filippopoulou, Simoni Besta, Mark Samuels, Andrea Lauer Betrán, Maha Abu Ajamieh, Viviana Vella, William Jones, Georgios Giamas
{"title":"Spatial biology - unravelling complexity within the glioblastoma microenvironment.","authors":"Stephen D Robinson, Chrysa Filippopoulou, Simoni Besta, Mark Samuels, Andrea Lauer Betrán, Maha Abu Ajamieh, Viviana Vella, William Jones, Georgios Giamas","doi":"10.1016/j.molmed.2025.01.014","DOIUrl":"10.1016/j.molmed.2025.01.014","url":null,"abstract":"<p><p>The advent and refinement of state-of-the-art spatial biology technologies have facilitated analysis that combines the advantages of high-throughput single cell analysis with techniques that preserve tissue architecture. This combination of cellular phenotyping with retained spatial context provides a much greater understanding of cellular interactions within the tumour microenvironment (TME). For glioblastoma, with its significant intra-tumoural heterogeneity, cellular plasticity, and complex TME, appreciating and understanding these spatial patterns may prove key to improving patient outcomes. This review examines the advances in spatial biology techniques, discusses how these methodologies are being applied to study glioblastoma, and explores how spatial information improves understanding of the TME. Ultimately, it is this spatial context that will accelerate the identification of more effective treatments for glioblastoma.</p>","PeriodicalId":23263,"journal":{"name":"Trends in molecular medicine","volume":" ","pages":"846-859"},"PeriodicalIF":13.8,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143400179","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}
Luca Perico, Federica Casiraghi, Ariela Benigni, Giuseppe Remuzzi
{"title":"Is there a place for engineered immune cell therapies in autoimmune diseases?","authors":"Luca Perico, Federica Casiraghi, Ariela Benigni, Giuseppe Remuzzi","doi":"10.1016/j.molmed.2025.01.011","DOIUrl":"10.1016/j.molmed.2025.01.011","url":null,"abstract":"<p><p>The ability to engineer immune cells yielded a transformative era in oncology. Early clinical trials demonstrated the efficacy of chimeric antigen receptor (CAR) T cells in resetting the immune system, motivating the expansion of this treatment beyond cancer, including autoimmune conditions. In this review, we discuss the current state of CAR T cell research in autoimmune diseases, examining the main challenges that limit widespread adoption of this therapy, such as complex isolation protocols, stringent immunosuppression, risk of secondary malignancies, and variable efficacy. We also review the studies addressing these limitations by development of off-the-shelf allogeneic CAR T cells, tunable safety systems, and antigen-specific therapies, which hold the potential to improve safety and accessibility of this treatment in clinical practice.</p>","PeriodicalId":23263,"journal":{"name":"Trends in molecular medicine","volume":" ","pages":"827-845"},"PeriodicalIF":13.8,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143473004","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}
William M McFadden, Mia Faerch, Karen A Kirby, Robert A Dick, Bruce E Torbett, Stefan G Sarafianos
{"title":"Considerations for capsid-targeting antiretrovirals in pre-exposure prophylaxis.","authors":"William M McFadden, Mia Faerch, Karen A Kirby, Robert A Dick, Bruce E Torbett, Stefan G Sarafianos","doi":"10.1016/j.molmed.2025.01.013","DOIUrl":"10.1016/j.molmed.2025.01.013","url":null,"abstract":"<p><p>Antiretroviral therapy (ART) impairs viral replication in people living with HIV (PLWH) by suppressing infection or spread. However, not all treatment strategies apply to preventive applications like pre-exposure prophylaxis (PrEP) for uninfected individuals. To prevent the establishment of HIV infection, PrEP must block viral replication either before, or at the stage of integration into the host genome. A promising PrEP approach under investigation utilizes lenacapavir (LEN), which targets the HIV-1 capsid protein (CA) potently before integration. LEN, a first-in-class antiretroviral, has shown high protective efficacy in the ongoing PURPOSE trials thus far. Here, we discuss clinical investigations of LEN, theoretical suitability of preclinical CA-binding antivirals in PrEP, and other key considerations for preventing HIV-1 infection by targeting the capsid.</p>","PeriodicalId":23263,"journal":{"name":"Trends in molecular medicine","volume":" ","pages":"801-813"},"PeriodicalIF":13.8,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12270248/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143531891","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Gasdermins in pyroptosis, inflammation, and cancer.","authors":"Rui Min, Yang Bai, Ning-Rui Wang, Xing Liu","doi":"10.1016/j.molmed.2025.04.003","DOIUrl":"10.1016/j.molmed.2025.04.003","url":null,"abstract":"<p><p>Pyroptosis is a type of programmed inflammatory cell death characterized by balloon-like swelling, membrane rupture, and the release of inflammatory cytokines and danger signals. Pyroptosis is directly triggered by activated gasdermins (GSDMs) which bind to membrane phospholipids, oligomerize, and form pores in cell membranes. GSDM activation is mediated by various effector proteases via cleavage of the linker region or post-translational modification to release the active N-terminal fragment in response to a variety of pathogenic or intrinsic danger signals. GSDM-mediated pyroptosis is involved in the pathogenesis of an array of infectious and inflammatory diseases and cancers. This review discusses recent advances related to the physiological and pathological functions of GSDM-mediated pyroptosis, as well as therapeutic strategies targeting pyroptosis.</p>","PeriodicalId":23263,"journal":{"name":"Trends in molecular medicine","volume":" ","pages":"860-875"},"PeriodicalIF":13.8,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144033847","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}
Orrin Devinsky, Jeff Coller, Rebecca Ahrens-Nicklas, X Shawn Liu, Nadav Ahituv, Beverly L Davidson, Kathie M Bishop, Yael Weiss, Ana Mingorance
{"title":"Gene therapies for neurogenetic disorders.","authors":"Orrin Devinsky, Jeff Coller, Rebecca Ahrens-Nicklas, X Shawn Liu, Nadav Ahituv, Beverly L Davidson, Kathie M Bishop, Yael Weiss, Ana Mingorance","doi":"10.1016/j.molmed.2025.01.015","DOIUrl":"10.1016/j.molmed.2025.01.015","url":null,"abstract":"<p><p>Pathogenic variants in over 1700 genes can cause neurogenetic disorders. Monogenetic diseases are ideal targets for genetic therapies; however, the blood-brain barrier (BBB), post-mitotic neurons, and inefficient delivery platforms make gene therapies for neurogenetic diseases challenging. Following nusinersen's 2016 approval, the development of gene therapies for neurogenetic disorders has advanced rapidly, with new delivery vehicles [e.g., BBB-crossing capsids, engineered viral-like proteins, lipid nanoparticles (LNPs)] and novel therapeutic strategies (e.g., regulatory elements, novel RNA therapeutics, tRNA therapies, epigenetic and gene editing). Patient-led disease foundations have accelerated treatment development by addressing trial readiness and supporting translational research. We review the current landscape and future directions in developing gene therapies for neurogenetic disorders.</p>","PeriodicalId":23263,"journal":{"name":"Trends in molecular medicine","volume":" ","pages":"814-826"},"PeriodicalIF":13.8,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143450316","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}