{"title":"Unveiling the P2X7 receptor: Exploring its mechanisms, pathogenic role in ocular diseases, and emerging therapeutic potential","authors":"Kai-Yang Chen , Hoi-Chun Chan , Chi-Ming Chan","doi":"10.1016/j.mam.2025.101389","DOIUrl":"10.1016/j.mam.2025.101389","url":null,"abstract":"<div><div>Purinergic signaling, mediated by extracellular ATP (eATP) and P2 receptors, plays a vital role in physiological and pathological processes. The P2X7 receptor (P2X7R), a ligand-gated cation channel, is crucial in inflammation, cell death, and immune responses. Widely expressed in retinal cells, P2X7R contributes to visual function regulation and retinal degeneration. This review explores P2X7R involvement in retinal diseases, including age-related macular degeneration (AMD), Behçet's disease (BD), diabetic retinopathy (DR), glaucoma, retinitis pigmentosa (RP), uveitis, Stargardt's disease (STGD), and toxoplasmosis.</div><div>P2X7R activation drives inflammation, oxidative stress, apoptosis, and immune dysregulation. For instance, it contributes to RPE degeneration in AMD, vascular proliferation in DR, neuroinflammation in glaucoma, and photoreceptor loss in RP. In uveitis, P2X7R enhances Th1 and Th17 responses. Targeting P2X7R with antagonists or modulators holds therapeutic potential, offering strategies to preserve retinal function and prevent vision loss in these debilitating diseases.</div></div>","PeriodicalId":49798,"journal":{"name":"Molecular Aspects of Medicine","volume":"105 ","pages":"Article 101389"},"PeriodicalIF":10.3,"publicationDate":"2025-07-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144720951","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jing Xia , Jing Jin , Shuang Dai , HaoHan Fan , KeLiang Chen , JianMei Li , Feng Luo , Xingchen Peng
{"title":"Mitophagy: A key regulator of radiotherapy resistance in the tumor immune microenvironment","authors":"Jing Xia , Jing Jin , Shuang Dai , HaoHan Fan , KeLiang Chen , JianMei Li , Feng Luo , Xingchen Peng","doi":"10.1016/j.mam.2025.101385","DOIUrl":"10.1016/j.mam.2025.101385","url":null,"abstract":"<div><div>Cancer remains a leading global cause of mortality, with radiation therapy (RT) as a cornerstone of treatment despite frequent radioresistance. Emerging evidence indicates that mitophagy activation contributes to adaptive radioresistance of cancer cells within the tumor microenvironment (TME). In this review, we highlight the dual role of mitophagy in modulating RT resistance and shaping the immune landscape of the TME. Mitophagy enhances cancer cell resilience by clearing radiation-damaged mitochondria, preserving metabolic homeostasis and reducing oxidative stress, while simultaneously altering the balance between immune activation and suppression within the TME. To provide mechanistic insight, we summarize key mitophagy-regulating pathways—including the PINK1/Parkin axis, BNIP3/NIX, and FUNDC1-mediated mechanisms—that respond to RT-induced mitochondrial stress and represent potential therapeutic targets. Furthermore, we explore how the interplay between mitophagy, metabolic reprogramming, and immune modulation shapes resistance not only to RT but also to immunotherapies such as immune checkpoint inhibitors (ICIs) and chimeric antigen receptor T (CAR-T) cell therapy. Additionally, we examine how Type 2 diabetes(T2DM) mellitus impacts this process, as its associated metabolic disturbances exacerbate mitochondrial vulnerability to radiation and create an immunosuppressive milieu that compromises the tumor immune landscape. Understanding these interactions may support development of personalized therapeutic strategies for diabetic cancer patients.</div></div>","PeriodicalId":49798,"journal":{"name":"Molecular Aspects of Medicine","volume":"105 ","pages":"Article 101385"},"PeriodicalIF":8.7,"publicationDate":"2025-07-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144665892","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Qiang Wu , Wen Li , Hongli Liu , Yinyin Xue , Kaili Huang , Hossein Pourghadamyari , Zhenkun Liu
{"title":"Respiratory emerging viruses and drug resistance in lung cancer: challenges and new perspectives in treatment","authors":"Qiang Wu , Wen Li , Hongli Liu , Yinyin Xue , Kaili Huang , Hossein Pourghadamyari , Zhenkun Liu","doi":"10.1016/j.mam.2025.101383","DOIUrl":"10.1016/j.mam.2025.101383","url":null,"abstract":"<div><div>Emerging viral pathogens, particularly those targeting the respiratory system such as SARS-CoV-2 and influenza virus, pose significant challenges for patients with lung cancer. These patients show increased susceptibility to serious infections caused by these viruses as a result of immunosuppression from various therapies such as immunotherapy, chemotherapy, and targeted agents. Respiratory viral infections can induce tumor progression and drug resistance through affecting the tumor microenvironment (TME), modulating immune checkpoints, or interfering with therapeutic efficacy. The present review discusses the reciprocal interactions between these viral pathogens and lung cancer, underscoring mechanisms by which SARS-CoV-2 and influenza viruses affect the lung cancer TME promoting tumor progression, immune evasion, and ultimately, resistance to anti-cancer therapies such as tyrosine kinase inhibitors (TKIs) and immune checkpoint inhibitors (ICIs). Sections discussed in this study include: (i) increased susceptibility to viral infections, (ii) virus-induced modulation of the tumor microenvironment, and (iii) mechanisms of therapy resistance. This review also addresses emerging strategies, such as oncolytic virotherapy, as potential candidates for lung cancer treatment. It aims to provide new perspectives on therapeutic optimization for patients with lung cancer in the context of SARS-CoV-2 and influenza viruses, by reviewing evidence from virology, oncology, and immunology.</div></div>","PeriodicalId":49798,"journal":{"name":"Molecular Aspects of Medicine","volume":"104 ","pages":"Article 101383"},"PeriodicalIF":8.7,"publicationDate":"2025-07-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144632758","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Protein and RNA chaperones","authors":"Bikash R. Sahoo, James CA. Bardwell","doi":"10.1016/j.mam.2025.101384","DOIUrl":"10.1016/j.mam.2025.101384","url":null,"abstract":"<div><div>Cells preserve macromolecular homeostasis by utilizing molecular chaperones that prevent aggregation or promote correct folding of protein and RNA. Here we discuss non-traditional proteinaceous chaperones like RNA-binding chaperones that work by modulating RNA structure, preventing aberrant interactions, and regulating intracellular granule dynamics. We also discuss the chaperone functions of other macromolecules such as nucleic acids, and in particular G-quadruplexes, which are very effective at preventing protein aggregation and accelerating protein folding. These chaperones are particularly important in G-quadruplex linked amyloid aggregation and repeat-expansion diseases such as Parkinson's disease and amyotrophic lateral sclerosis, where RNA aggregation and misfolded protein accumulation co-occur. By comparing protein and non-protein chaperone systems, we highlight the principles that underlie chaperone action across molecular classes.</div></div>","PeriodicalId":49798,"journal":{"name":"Molecular Aspects of Medicine","volume":"104 ","pages":"Article 101384"},"PeriodicalIF":8.7,"publicationDate":"2025-07-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144614681","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Julia Lis, Jakub Fichna, Aleksandra Tarasiuk-Zawadzka
{"title":"The role of free fatty acid receptors activation in pancreatic disorders","authors":"Julia Lis, Jakub Fichna, Aleksandra Tarasiuk-Zawadzka","doi":"10.1016/j.mam.2025.101386","DOIUrl":"10.1016/j.mam.2025.101386","url":null,"abstract":"<div><div>Free fatty acid receptors (FFARs), a subset of G protein-coupled receptors, play a pivotal role in metabolic and immune homeostasis by modulating signaling pathways in response to free fatty acids. The four main FFARs (FFAR1, FFAR2, FFAR3, and FFAR4) are especially significant in pancreatic function, regulating insulin secretion, inflammation, and glucose metabolism. These receptors are involved in key pancreatic disorders, including acute pancreatitis (AP), pancreatic cancer (PC), type 1 diabetes (T1D), and type 2 diabetes (T2D).</div><div>FFAR1, FFAR3, and FFAR4 exhibit protective effects against AP due to their anti-inflammatory properties. In PC, FFAR1 inhibits tumor cell motility, while FFAR2 downregulation may contribute to tumor progression. FFAR3 plays a role in limiting tumor proliferation, whereas FFAR4 has a dual effect, promoting metastasis while also triggering tumor cell apoptosis. In T1D, FFAR2 and FFAR4 help regulate glycemia without directly stimulating insulin secretion. In T2D, all four FFARs contribute to glycemic control and may protect pancreatic β-cells.</div><div>Despite their therapeutic potential, the precise mechanisms underlying FFAR function in pancreatic disorders remain incompletely understood. Ongoing research aims to clarify these pathways, identify optimal ligands, and assess the safety and efficacy of FFAR-targeted therapies. This growing body of evidence underscores the importance of FFARs as potential targets for innovative treatments in pancreatic diseases.</div></div>","PeriodicalId":49798,"journal":{"name":"Molecular Aspects of Medicine","volume":"104 ","pages":"Article 101386"},"PeriodicalIF":8.7,"publicationDate":"2025-07-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144605148","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Wei Li , Yan Liao , Jie Chen , Weichao Kang , Xintao Wang , Xiaozhu Zhai , Ying Xue , Wangzheqi Zhang , Yiyang Xia , Derong Cui
{"title":"Ischemia - Reperfusion injury: A roadmap to precision therapies","authors":"Wei Li , Yan Liao , Jie Chen , Weichao Kang , Xintao Wang , Xiaozhu Zhai , Ying Xue , Wangzheqi Zhang , Yiyang Xia , Derong Cui","doi":"10.1016/j.mam.2025.101382","DOIUrl":"10.1016/j.mam.2025.101382","url":null,"abstract":"<div><div>Ischemia-reperfusion (I/R) injury is prevalent in the medical field and significantly limits the therapeutic outcomes of various ischemic diseases, adversely affecting patient prognosis. The pathogenesis of I/R injury is highly complex, involving intricate interactions among oxidative stress, inflammatory responses, mitochondrial dysfunction, and multiple cell death pathways. Once the mitochondrial respiratory chain is impaired, it triggers oxidative stress responses, leading to the excessive production of reactive oxygen species (ROS). Excessive ROS not only directly damage cells but also activate inflammatory responses and initiate multiple cell death signalling pathways, such as necroptosis, pyroptosis, and ferroptosis, thereby exacerbating tissue damage. Moreover, the clinical manifestations of I/R injury vary significantly across different organs, such as the heart, brain, kidneys, liver, and lungs, and are further influenced by patients' underlying conditions, posing challenges for clinical diagnosis and treatment. Therefore, constructing a comprehensive assessment system based on individual patient characteristics (such as genetic polymorphisms and comorbidities) to accurately predict the risk of I/R injury is particularly important. Currently, there are diverse strategies for the prevention and treatment of I/R injury, but translating basic research into clinical application remains challenging. Developing personalized treatment plans tailored to different cell types holds promise for overcoming existing therapeutic bottlenecks, significantly improving patient outcomes, and providing new directions for addressing the challenges of I/R injury.</div></div>","PeriodicalId":49798,"journal":{"name":"Molecular Aspects of Medicine","volume":"104 ","pages":"Article 101382"},"PeriodicalIF":8.7,"publicationDate":"2025-07-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144572362","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Type 2 Diabetes Mellitus and bladder cancer: A narrative review on associated signaling pathways","authors":"Lívia da Cunha Agostini, Glenda Nicioli da Silva","doi":"10.1016/j.mam.2025.101381","DOIUrl":"10.1016/j.mam.2025.101381","url":null,"abstract":"<div><div>Bladder cancer is one of the most common cancers globally. The risk factors for urothelial bladder cancer can be broadly divided into genetic predispositions and external environmental exposures. Type 2 Diabetes Mellitus (T2DM) is a chronic, non-communicable metabolic disorder, and the interaction between genetic, behavioral, and environmental factors plays a significant role in its development. The management of T2DM includes lifestyle modifications and medication. Several studies suggest that T2DM is associated with an increased risk of bladder cancer. This review highlights the key signaling mechanisms involved in this association and explores the impact of T2DM medications on bladder cancer. In conclusion, the literature suggests that metabolic abnormalities associated with T2DM —such as hyperglycemia, insulin resistance and elevated levels of insulin, insulin-like growth factor 1 (IGF-1), inflammatory cytokines, iNOS/eNOS activity, hypoxia, dyslipidemia, matrix metalloproteinase (MMPs), leptin, vimentin, N-cadherin, fibronectin, advanced glycation end products (AGEs), endoplasmic reticulum stress (ERS), and <em>Arntl2</em> gene expression; in addition to reduced E-cadherin, adiponectin, autophagy, and <em>IGF-1</em> and <em>Usp2</em> gene expression—significantly influence signaling pathways essential for bladder tumor development. Additionally, the choice of hypoglycemic treatment should be carefully considered, taking into account potential effects on carcinogenesis.</div></div>","PeriodicalId":49798,"journal":{"name":"Molecular Aspects of Medicine","volume":"104 ","pages":"Article 101381"},"PeriodicalIF":8.7,"publicationDate":"2025-06-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144322188","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Li Li , Xu Shi , Ruiming Wang , Yuxi Fan , Zhihan Xu , Habibollah Mirzaei , Wuran Wei
{"title":"Cardiovascular impact of emerging and Re-emerging Viruses: Pathophysiological mechanisms, diagnosis, and management with a pediatric focus","authors":"Li Li , Xu Shi , Ruiming Wang , Yuxi Fan , Zhihan Xu , Habibollah Mirzaei , Wuran Wei","doi":"10.1016/j.mam.2025.101371","DOIUrl":"10.1016/j.mam.2025.101371","url":null,"abstract":"<div><div>Emerging and re-emerging viruses are currently known as a major public health issue. These viruses can cause various human complications such as cardiovascular diseases (CVDs), both in adults and pediatric populations. Although various CVDs have been previously reported for emerging and re-emerging viruses, the mechanisms underlying these complications remain relatively unknown. Children and infants, while commonly developing less severe symptoms, may experience notable cardiovascular manifestations during infections caused by emerging and re-emerging viral infections, which can result in both acute and long-term complications. The present review aims to discuss various cardiovascular complications linked to emerging and re-emerging viral pathogens (including severe acute respiratory syndrome <em>coronavirus 2</em> (<em>SARS</em>-<em>CoV</em>-<em>2</em>), dengue virus (DENV), Zika virus (ZIKV), and chikungunya virus (CHIKV)) such as arrhythmias, myocarditis, vascular disorders, and thromboembolic conditions, particularly among the pediatric population. This review also addresses the potential mechanisms by which <em>SARS</em>-<em>CoV</em>-<em>2</em>, DENV, ZIKV, and CHIKV may impact the cardiovascular system and their clinical implications. Moreover, it discusses the diagnostic challenges for viral-caused cardiovascular disorders in children, owing to their common subtle or atypical manifestations. Finally, it addresses the present therapeutic specifically used for pediatric cases.</div></div>","PeriodicalId":49798,"journal":{"name":"Molecular Aspects of Medicine","volume":"104 ","pages":"Article 101371"},"PeriodicalIF":8.7,"publicationDate":"2025-05-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144139220","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Adiponectin: its role in diabetic and pancreatic cancer","authors":"Seema Kumari , Sujatha Peela , Mundla Srilatha , Bala Prabhakar Girish , Ganji Purnachandra Nagaraju","doi":"10.1016/j.mam.2025.101370","DOIUrl":"10.1016/j.mam.2025.101370","url":null,"abstract":"<div><div>Adiponectin (ApN) is an antidiabetic and anti-inflammatory protein synthesized by adipose tissue. It is essential in regulating insulin sensitivity, glucose, and lipid metabolism by controlling AMPK, PPARα, and MAPK signals. It is an anti-inflammatory property that protects pancreatic β-cells. Often, low levels of ApN are linked to obesity, type II diabetes and the development of PDAC. However, changes in lifestyle and the use of certain drugs can improve ApN function and insulin sensitivity. PDAC is a highly aggressive cancer linked to obesity, type II diabetes, and insulin resistance. ApN plays a complex role in PDAC progression and can suppress PDAC development by weakening β-catenin signaling. Decreases in ApN levels are associated with increased PDAC risk in diabetic patients. PDAC and diabetes are interconnected through the development of insulin resistance, islet dysfunction, change in immunological response, inflammation, oxidative stress, and altered hormone secretion. Genetic studies highlight specific genes like HNF4G and PDX1 that influence both conditions and miRNAs such as miR-19a promote tumor progression through the PI3K/AKT pathway. This review discusses the role of ApN in diabetes and PDAC and the interrelation between diabetes and PDAC.</div></div>","PeriodicalId":49798,"journal":{"name":"Molecular Aspects of Medicine","volume":"103 ","pages":"Article 101370"},"PeriodicalIF":8.7,"publicationDate":"2025-05-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144099281","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Virus-like particles of retroviral origin in protein aggregation and neurodegenerative diseases","authors":"Serena Carra , Balazs Fabian , Hamed Taghavi , Edoardo Milanetti , Valeria Giliberti , Giancarlo Ruocco , Jason Shepherd , Michele Vendruscolo , Monika Fuxreiter","doi":"10.1016/j.mam.2025.101369","DOIUrl":"10.1016/j.mam.2025.101369","url":null,"abstract":"<div><div>A wide range of human diseases are associated with protein misfolding and amyloid aggregates. Recent studies suggest that in certain neurological disorders, including Amyotrophic Lateral Sclerosis (ALS), Frontotemporal Dementia (FTD) and various tauopathies, protein aggregation may be promoted by virus-like particles (VLPs) formed by endogenous retroviruses (ERVs). The molecular mechanisms by which these VLPs contribute to protein aggregation, however, remain enigmatic. Here, we discuss possible molecular mechanisms of ERV-derived VLPs in the formation and spread of protein aggregates. An intriguing possibility is that liquid-like condensates may facilitate the formation of both protein aggregates and ERV-derived VLPs. We also describe how RNA chaperoning, and the encapsulation and trafficking of misfolded proteins, may contribute to protein homeostasis through the elimination of protein aggregates from cells. Based on these insights, we discuss future potential therapeutic opportunities.</div></div>","PeriodicalId":49798,"journal":{"name":"Molecular Aspects of Medicine","volume":"103 ","pages":"Article 101369"},"PeriodicalIF":8.7,"publicationDate":"2025-05-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144089163","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}