Background: Melanoma is recognized as a highly malignant cancer with a generally poor prognosis, underscoring the critical need for effective therapeutic strategies. Adoptive cell therapy has emerged as a promising modality to improve treatment outcomes in melanoma. For endogenous cell therapy (ECT), peripheral blood (PB) has traditionally served as the primary cell source. However, the potential of umbilical cord blood (UCB) as an alternative source for ECT remains unclear. Furthermore, the repertoire of TCRs remains limited. These deficiencies impede the optimization and broader application of ECT for melanoma, highlighting the necessity for focused investigations to resolve these issues.
Methods: To evaluate the effects of HLA-A2 restricted antigen-specific CD8+ T cells on melanoma cells, the cytotoxic activity of CD8+ T cells derived from UCB and PB were conducted in vivo and in vitro assays. Single-cell RNA sequencing combined with TCR V(D)J sequencing was employed to characterize cellular composition and quantify the frequencies of specific TCR clonotypes. The generation probability and peripheral occurrence probability of antigen-specific CD8+ TCR sequences from UCB and PB were computed using the Simple Olga Sonia algorithm. Finally, molecular docking simulations were conducted to predict the binding affinity between isolated TCRs and pMHC.
Results: No significant differences were observed in the cytotoxic effects mediated by antigen-specific CD8+ T cells derived from UCB versus PB. Phenotypic analysis revealed that PB-derived antigen-specific CD8+ T cells were predominantly effector and proliferating cells, whereas those from UCB consisted largely of memory cells. TCR sequencing identified a greater diversity of antigen-specific TCR clonotypes in PB, meanwhile UCB-derived TCRs exhibited strong pMHC binding. Molecular docking simulations confirmed high binding affinity between pMHC and TCR clones isolated from both sources.
Conclusions: Antigen-specific CD8+ T cells from UCB and PB display comparable cytotoxic efficacy against melanoma, albeit with distinct compositional profiles of antigen-specific CD8+ T cell subsets. Candidate TCRs can be effectively activated by the tumor-associated antigens MART1 and gp100. This activation promotes the expansion of the available TCR repertoires, thereby mitigating the previous constraint of a limited TCR library.
In the tumour microenvironment (TME) of renal cell carcinoma (RCC), tertiary lymphoid structures (TLS) play a crucial role in anti-tumour immune responses. Resembling secondary lymphoid organs, TLS comprises B cells, T cell zones, high endothelial venules, and antigen-presenting cells, facilitating local immune activation. While TLS has shown correlations with improved immune checkpoint inhibitors (ICIs) outcomes in other cancers, its role in RCC is still under investigation. Emerging evidence indicates that mature TLS enhances anti-tumour activity by activating T and B cells, whereas immature TLS may contribute to immune suppression. The RCC TME is highly immunosuppressive, marked by regulatory T cells, myeloid-derived suppressor cells, and elevated pro-angiogenic and immunosuppressive cytokines. In this context, TLS, particularly mature TLS, can counteract immunosuppression, boost local immune responses, and improve ICIs efficacy. However, TLS in RCC is heterogeneous, with their formation and function affected by factors like CXCL13 expression. The presence, maturity, and functionality of TLS may serve as valuable predictors of ICIs response and patient prognosis. Further research is required to understand TLS regulation and leverage their potential to enhance personalised immunotherapy for RCC.
Colorectal cancer (CRC) is one of the most common and deadliest cancers worldwide, and incidence rates are rising. However, early detection and intervention can improve the survival rates and quality of life of affected patients. Current screening tests used to streamline patients into colonoscopy either lack test adherence or sensitivity for detecting premalignant and early-stage CRC, reducing the advantages of screening measures. Cost-effective and minimally invasive diagnostic tests which can detect immune system and metabolic changes are key to lower the incidence of CRC advanced stages. We herein discuss the statistics, risk factors and unique genetic characteristics of CRC, focussing on the importance of understanding non tumour-derived information in premalignant states for developing comprehensive techniques to achieve earlier diagnosis of CRC. Moreover, the advantages and limitations of current UK and USA screening programmes and emerging detection tools are discussed, along with prospective diagnostics such as genomics, proteomics and spectroscopy.
Background: Metastatic melanoma is a highly aggressive disease with poor survival rates despite recent therapeutic advancements with immunotherapy. The proteomic landscape of advanced melanoma remains poorly understood, especially regarding proteomic heterogeneity across metastases within patients.
Methods: We collected 83 melanoma metastases from 19 different metastatic sites in 24 patients with advanced metastatic melanoma almost exclusively from the pre-immunotherapy era, using semi-rapid autopsies. The metastases were subjected to histopathological evaluation, RNA-sequencing and mass spectrometry-based proteomics for protein quantitation and non-reference peptide (NRP) sequence detection using a proteogenomic data integration approach.
Results: NRPs associated with mutations frequently occurred in proteins related to focal adhesion, vesicle-mediated transport, MAPK signalling and immune response pathways across the cohort. Intrapatient heterogeneity was negligible when considering morphology and driver gene mutation status but was substantial at the proteogenomic level. This heterogeneity was not driven by metastasis location, albeit liver metastases exhibited distinct proteogenomic patterns, including upregulation of metabolic pathways. Cluster analysis outlined four proteomic clusters (C1–4) of the metastases, characterised by the upregulation of cell cycle and RNA-splicing (C1), mitochondrial processes (C3), extracellular matrix (ECM) and immune pathways (C2) and ECM and vesicle-mediated transport pathways (C4). Around two-thirds of patients had metastases that had strongly distinct phenotypes. Patients in our cohort whose metastases were primarily assigned to clusters C1 and C3 exhibited shorter overall survival than patients whose metastases were categorised mainly into the C2 and C4 clusters.
Conclusion: Our unique multi-metastasis cohort captured the proteogenomic heterogeneity of immunotherapy-naïve melanoma distant metastases, establishing a foundation for future studies aimed at identifying novel therapeutic targets to complement current immunotherapies.
Background: Lactylation, a recently identified post-translational modification that utilizes lactic acidas a substrate, has emerged as an important regulator of gene expression andprotein function. Since its discovery in 2019, lactylation has beenincreasingly recognized for its roles in cancer biology and treatment response.
Main text: Lactylationis strongly associated with tumor progression and malignancy, underscoring itspotential as a therapeutic target. Recent studies also link lactylation tocancer treatment resistance, suggesting that modulating this modification couldenhance therapeutic efficacy. As treatment resistance remains a major clinicalchallenge in oncology, accumulating evidence indicates that dysregulatedlactylation contributes to resistance across chemotherapy, immunotherapy, targeted therapy, and radiotherapy. Preclinical and clinical research has begunto delineate the molecular pathways through which lactylation shapes theseresistance processes, and experimental approaches targeting lactylation arebeing explored to restore therapeutic sensitivity.
Conclusion: This review systematically summarizes the mechanisms of lactylation and its roles intreatment resistance, highlighting the interplay between lactylation andtherapeutic response. We discuss current and emerging strategies that targetlactylation, providing a foundation for future therapeutic development aimed atovercoming resistance and improving cancer treatment outcomes.
Radiotherapy (RT) is a cornerstone in cancer treatment, but often causes radiation-induced injury. Accumulating evidence points to the gut microbiota in modulating immune functions and maintaining intestinal integrity to impact RT efficacy. This review examines the current understanding of intestinal flora and their metabolites within the context of RT. We outlined the current research applications in how microbiota-targeted strategies such as probiotics, prebiotics, dietary interventions, and faecal microbiota transplantation could restore microbial balance, reduce toxicity, and improve patient prognosis. Microbial byproducts such as short-chain fatty acids, bile acids and tryptophan exhibit protective effects against radiation damage, supporting immune modulation and enhancing tumour radiosensitivity. These microbial products underscore the potential of gut microbiota-targeted therapies as adjunctive treatments in RT, with implications for reducing toxicity and personalizing cancer care. All these strategies targeting gut microbiota and metabolites potentially aim to develop innovative therapies that boost RT effectiveness while minimizing side effects, and finally revolutionizing personalized cancer treatment.
Background: In the realm of public health, among the primary perils menacing human well-being, the issue of pathogen infection persists as a significant concern. Precise and timely diagnosis of diseases constitutes the bedrock for effective therapeutic interventions and epidemiological monitoring. Hence, it is crucial to develop quick, sensitive, and highly effective methods for identifying pathogen and their variants.
Material and methods: This article reviews the recent research progress in the CRISPR/Cas system for detecting nucleic acids, with an emphasis on CRISPR/Cas9, CRISPR/Cas12, and CRISPR/Cas13. Initially, we provided a concise overview of the nucleic acid detection mechanism utilizing the CRISPR/Cas system. Subsequently, we dissect the molecular mechanisms of CRISPR tools, compare their clinical efficacy against traditional methods, and explore frontier innovations such as amplification-free detection and AI integration.
Conclusion: Ultimately, we argue that CRISPR diagnostics must evolve beyond technical optimization to embrace ecological adaptability, ensuring that precision medicine serves as a bridge-rather than a barrier-to global health equity.
Background: The recurrence and metastasis of oesophageal squamous cell cancer (ESCC) following radiation therapy are major treatment challenges. Cancer-associated fibroblasts (CAFs) are key in the ESCC microenvironment, yet their role in post-radiation recurrence remains unclear.
Materials and Methods: KYSE150 ESCC cells were co-implanted with non-irradiated (0 Gy) or irradiated (8 Gy) CAFs in nude mice. CAF-derived extracellular vesicles (EVs) were isolated via differential centrifugation and analysed by electron microscopy and immunoblotting. Transwell assays evaluated EVs' effects on ESCC cell migration and invasion in vitro. RNA sequencing identified differentially expressed microRNAs, and functional experiments verified the role of miR-193a-3p. Plasma samples from 32 ESCC patients and tissue samples from 76 ESCC patients were analysed for miR-193a-3p expression.
Results: Irradiated CAFs promoted the lung metastasis of ESCC cells in vivo, and their EVs enhanced ESCC cell invasion, migration and metastasis. Elevated miR-193a-3p levels in EVs from irradiated CAFs increased miR-193a-3p expression in ESCC cells. This effect was effectively attenuated by RNase and Triton X-100 (degrading microRNAs encapsulated in EVs), or GW4869 (inhibiting EVs biogenesis and secretion)—indicating that miR-193a-3p functions in an EV-dependent manner. Knockdown of miR-193a-3p diminished the invasion, migration and epithelial–mesenchymal transition (EMT)-promoting activities of CAF-derived EVs. Luciferase assays confirmed PTEN as a target of miR-193a-3p; miR-193a-3p overexpression decreased PTEN and increased p-Akt expression. In vivo, coinjection of miR-193a-3p-knockdown CAFs with KYSE150 ESCC cells resulted in smaller tumours, fewer lung metastases, increased PTEN and E-cadherin, and decreased p-Akt and Snail expression. Clinically, radiation increased plasma exosomal miR-193a-3p levels, and high miR-193a-3p expression was correlated with shorter survival, identifying miR-193a-3p as an independent predictor of poor prognosis in ESCC patients.
Conclusion: EVs from irradiated CAFs promote ESCC metastasis via the miR-193a-3p-mediated PTEN/Akt signalling pathway. Targeting this EVs-mediated interaction represents a promising strategy for improving ESCC radiotherapy outcomes.
Background: Triple-negative breast cancer (TNBC), which lacks hormone receptors and HER2 expression, presents substantial therapeutic challenges in breast cancer treatment. The efficacy of immunotherapy frequently suffers from the immunosuppressive nature of tumour microenvironment (TME). Hence, discovering effective targets to hinder TNBC progression and bolster immunotherapy's effectiveness is paramount. Previous research from our team indicated notable upregulation of c-Myc in TNBC, and suppressing c-Myc enhances the efficacy of PD-L1 blockade in murine models; nevertheless, the precise mechanisms underlying this phenomenon remain elusive.
Methods: We analysed c-Myc expression and implemented a systematic drug library screening strategy alongside c-Myc knockdown to pinpoint potential synergistic agents in TNBC cells. To decipher the regulatory mechanisms of this synergy on cellular malignancy, we conducted cell cycle analysis, cell interaction assays, and RNA-sequencing. Additionally, we established orthotopic and lung metastasis murine models assess how combination therapy influences PD-L1 blockade efficacy.
Results: Elevated c-Myc was observed in TNBC and the Ras inhibitor Salirasib was identified as a potent synergistic agent from cell cycle drug library in c-Myc-overexpressing TNBC. The application of Salirasib combined with c-Myc knockdown markedly suppressed tumour cell aggressiveness and induced apoptosis in vitro. Mechanistically, RNA sequencing revealed that the combination therapy blocked MCM2-mediated DNA replication in TNBC cells, causing G1/S phase arrest and enhancing tumour suppression. In vivo, the combination significantly improved PD-L1 blockade efficacy, leading to reduction of tumour volume, inhibition of lung metastases, and remodelling of the immune microenvironment in murine TNBC models.
Conclusions: In summary, our investigation identifies a molecular vulnerability in c-Myc-driven TNBC, where Ras inhibition reinforces c-Myc-targeted therapy and potentiates immune checkpoint blockade, presenting a promising strategy to improve immunotherapy efficacy in TNBC.
Background: Targeted drug delivery systems have garnered increasing research interest in cancer threapy. Bacteria have emerged as a promising vehicle due to their innate ability to the tumour microenvironment (TME) and their intrinsic immune-stimulating properties. This review explores the application of bacteria in oncology, emphasizing the tumour-targeting behaviour of specific strains, their immunomodulatory functions, and their potential as delivery platforms for the controlled release of therapeutic agents.
Main text: This review synthesizes recent advances in bacteria-mediated cancer therapy, focusing on the mechanisms underlying bacterial targeting of hypoxic and immunosuppressive regions within the tumor microenvironment (TME). We discuss how genetic modification has been employed to design recombinant bacterial strains with enhanced tumor specificity and amplified therapeutic effects. Furthermore, the integration of bacteria with nanotechnology has facilitated the development of hybrid systems capable of targeted drug delivery and triggered-release mechanisms. The combination of bacterial therapy with other treatment modalities—such as photodynamic (PDT) and sonodynamic therapies (SDT)—is also examined, emphasizing their synergistic potential in overcoming tumor heterogeneity and enhancing anti-tumor immunity. Finally, we survey the current clinical progress of bacteria-based therapeutics and offer perspectives on the future role of artificial intelligence (AI) in improving the design and application of these living medicines.
Conclusions: Bacteria-based delivery systems represent a multifunctional and innovative strategy in the evolution of targeted cancer therapies. Through genetic modification and nanobiotechnology approaches, bacteria can be customized to mediate multi-effect synergistic treatments for cancer, enhancing the precision, safety, and efficacy of cancer therapies. With the ongoing integration of advanced technologies, including AI, there is great potential to overcome existing limitations and accelerate the clinical translation of bacterial anticancer therapies. This interdisciplinary effort is poised to open new avenues for next-generation cancer treatments and lay the foundation for future directions in cancer research and therapeutic practice.
Background: Intracerebral haemorrhage (ICH) progresses rapidly with complex pathology and limited treatment options, making it a severe subtype of stroke. The extravasation of blood into the brain parenchyma triggers a cascade of inflammatory responses, contributing to secondary injury. Single-nucleus RNA sequencing (snRNA-seq) data have enabled more profound insights into the cellular heterogeneity and dynamic interactions within the haemorrhagic brain. Immune cells play a crucial role in shaping neuroinflammation. However, the lack of comprehensive longitudinal studies limits our understanding of the temporal evolution of these inflammatory processes, posing a challenge to the development of targeted therapeutic strategies.
Methods: We used snRNA-seq in collagenase-induced ICH mouse models at Days 1, 3, 7, 14 and 28 post-injury, alongside naive controls, to profile the dynamics of gene expression over time.
Results: We obtained 281 577 high-quality transcriptional profiles representing 21 distinct cell types. Co-expression network analysis revealed a prominent ‘inflammation module’ that remained active throughout ICH. Integrative single-cell transcriptomic and immunofluorescence staining suggested that the various Mif-expressing cells may contribute to local inflammation, potentially engaging macrophages via receptor–ligand pairs such as Cd44 and Cd74. Over time, microglia appeared to serve as key recipients of pro-inflammatory signals increasingly. During the resolution phase, oligodendrocytes exhibited transcriptional signatures consistent with enhanced maturation and remyelination, which T cell-mediated interactions may have facilitated.
Conclusions: These findings offer a systems-level perspective on cell-type–specific responses and immune-mediated interactions during ICH progression and resolution.
Raman spectroscopy is a versatile analytical technique for highly specific molecular characterisation of cells, biofluids and tissues. Confocal Raman microspectroscopy combines optical microscopy with Raman spectroscopy to spatially resolve biochemical changes in tissue samples. This work focuses on research articles that utilise confocal Raman microspectroscopy in human or murine tissue sections for identifying disease-linked spectra-pathological features. For scientists and clinicians who seek ideas in incorporating confocal Raman microspectroscopy into their experimental workflows, this piece provides a curated selection of studies (spanning cancer and cardiovascular diseases) that highlight key spectroscopic and biomedical insights. The lack of standardisation and the fragmentation of research protocols are major challenges that limit study reproducibility and prevent systematic cross-validation. Moving forward, confocal Raman microspectroscopy, coupled with robust computational approaches, will continue to detect disease-specific spatiotemporal biomolecular signatures, and integration with complementary imaging or omics methods will keep enhancing its ability to analyse complex biological systems and uncover disease progression mechanisms.
Background: KRASG12C is the most common KRAS mutation in lung adenocarcinoma (LUAD), yet clinical responses to KRASG12C-selective inhibitors (G12Ci) and immunotherapy remain variable.
Methods: Transcriptomic analysis of KRASG12C-mutant LUAD was performed using machine learning algorithms to classify molecular subtypes. Subtype-specific features, including genomic alterations, tumour microenvironment and therapeutic vulnerabilities, were systematically evaluated.
Results: We identified three distinct molecular subtypes (KC1, KC2 and KC3) of KRASG12C-mutant LUAD through transcriptomic analysis using machine learning algorithms. KC1 subtype is characterised by a neuroendocrine phenotype associated with SMARCA4 loss-of-function and frequent STK11 co-mutations, with a relatively good prognosis. It exhibits poor immune infiltration and demonstrates resistance to G12Ci and immunotherapy but shows sensitivity to MEK1/2 inhibitors; KC2 subtype exhibits a highly malignant phenotype with high proliferation, increased glucose metabolism, and the poorest prognosis. It is enriched with T-cell infiltration and responds best to G12Ci monotherapy and immunotherapy. KC3 subtype is distinguished by well differentiation and the best survival, with an immune-enriched microenvironment featuring abundant immune-suppressive cancer-associated fibroblasts. It demonstrates limited sensitivity to G12Ci and a moderate response to immunotherapy. Notably, KC1‒3 subtype-specific molecular signatures predict drug sensitivity more accurately than classical KRASG12C signalling models.
Conclusions: These findings illuminate the intricate interplay between tumour subtypes, microenvironmental factors and therapeutic responses, offering a robust framework for improved patient stratification and the development of personalised therapeutic strategies KRASG12C-mutant LUAD.
Background: Obesity has emerged as a global health challenge. Although GLP-1 receptor agonists are showing considerable promise in weight loss, their clinical utility is partly limited by gastrointestinal adverse reactions and non-fat weight loss side effects. UCP1-mediated adipose thermogenesis is a critical process for body temperature maintenance and weight management. However, the lack of effective and specific adipose thermogenesis therapies has restricted its clinical application. We aimed to explore the potential of inducing adipose-specific UCP1 overexpression via modified plasmids as an innovative therapeutic approach for obesity.
Methods: We replaced the cytomegalovirus (CMV) promoter in the plasmids with two types of adipose-specific promoters: mouse adiponectin (mADP) promoter and human adiponectin (hADP) promoter, to selectively overexpress UCP1 in adipocytes. The expression level of UCP1, weight loss, metabolic homeostasis and adipose thermogenesis effects were evaluated by immunohistochemistry, western blot, weight measurements, thermography, and comprehensive lab animal monitoring system.
Results: The experiments demonstrated that the mADP promoter-modified plasmids failed to drive UCP1 overexpression. In contrast, the hADP promoter-modified Ucp1 overexpression (hADP-Ucp1 OE) plasmids achieved robust adipose-specific UCP1 protein expression both in vitro and in vivo. In vitro experiments revealed that delivery of the hADP promoter-modified UCP1 overexpression (hADP-UCP1 OE) plasmids reduced lipid droplet size and enhanced energy consumption in human adipocytes. In obese mice, administration of the hADP-Ucp1 OE plasmids resulted in significant weight loss and improved metabolic homeostasis.
Conclusions: These findings highlight the therapeutic potential of hADP-UCP1 OE plasmids in obesity management.
Breast cancer (BC) is the most common malignancy in women, yet the dynamics of the intratumoural microbiome during tumour initiation, progression, and treatment remain poorly understood. Prior studies are predominantly cross-sectional and limited by indirect microbial inference from RNA-seq data. This study presents a comprehensive analysis of intratumoural microbiota across breast tissue samples by high-depth 16S rRNA sequencing (11 W tags), featuring two longitudinally paired cohorts for dynamic microbial profiling during tumour progression and treatment. Samples included 165 benign nodules (82 non-transforming, 83 that later progressed to cancer with matched malignant tissues); 180 primary BC tissues and 165 benign controls; and 101 neoadjuvant therapy (NAT) specimens (15 pCR, 86 non-pCR, with paired pre/post-treatment samples). We identified a cluster of taxa (Aeromicrobium, Halomonas, Dietzia, Nesterenkonia, Delftia, Nitriliruptor) depleted in nodules undergoing malignant transformation, declining with disease progression and partially restored after NAT, with transient enrichment early in transformation. Opposing trends were observed for Paenibacillus and Methyloversatilis. These changes corresponded to shifts in amino acid, lipid, and glycan metabolism. FISH and TEM analyses identified Paenibacillus pasadenensis and Halomonas hamiltonii within tumour cells, with opposing effects on tumour proliferation and activation. In addition, we developed two predictive models with high clinical relevance: one stratifying malignancy risk in nodules, and another predicting NAT response, both of which achieved strong performance in external validation. This longitudinal characterisations of intratumoural microbiota during breast tumourigenesis and treatment offer novel insights for precision oncology and microbiome-based interventions in breast cancer.
Background: Mounting evidence shows that myeloid-derived suppressor cells (MDSCs) reprogramming can significantly enhance the outcomes of immunotherapy. However, the therapeutic potential of targeting MDSCs alone is limited by persistent immunosuppressive cytokines and cellular crosstalk. In our previous study, we found that novel cryo-thermal therapy (CTT) can drive MDSCs maturation and induce CD4+ T helper type (Th)1-dominant differentiation, improving long-term survival in spontaneous high metastatic mouse models. Considering the established roles of Interleukin (IL)-6 and IL-17A in non-small cell lung cancer (NSCLC) progression and immune evasion, we developed a combination strategy integrating cytokine neutralization with CTT (combination therapy) in LLC1 tumor-bearing mice. Although the combination therapy successfully promoted MDSCs maturation and Th1 differentiation, the underlying mechanistic basis remained unclear.
Methods: The combination therapy was implemented in LLC1 tumor-bearing mice. We then observed its impacts on MDSCs maturation and Th1 differentiation and explored the related mechanisms by examining various aspects including the expression of CD40, the reactive oxygen species (ROS)-nuclear factor-kappa B (NF-κB) pathway, and the induction of tumor necrosis factor-α (TNF-α).
Results: It was observed that the combination therapy increased the expression of CD40 on MDSCs through the ROS-NF-κB pathway-dependent TNF-α induction. This TNF-α-mediated CD40 upregulation facilitated Th1 polarization via CD40L engagement on CD4+ T cells. Our results provided the first mechanistic evidence that autocrine TNF-α production by reprogrammed MDSCs governs CD40 expression following combination therapy.
Conclusion: Our research elucidated the methods and mechanisms of MDSCs reprogramming and offered a promising therapeutic strategy for patients with NSCLC and other types of cancer.
Objectives: Although extensive research on septic cardiomyopathy has been conducted, effective therapies are still limited. Ubiquitin-specific peptidase 20 (USP20), a deubiquitinating enzyme, is critical in regulating protein ubiquitination and various cellular processes. whether USP20 is involved in the pathogenesis of septic cardiomyopathy remains unclear. This study investigated the impact of USP20 on septic cardiomyopathy.
Methods: The cardiomyocyte-specific USP20 knockout mice (USP20CKO) and NLRP3 knockout mice (NLRP3-/-) were used in the present study. A sepsis mouse model was established using lipopolysaccharide (LPS) administration and the cecal ligation and puncture (CLP) procedure. Recombinant adeno-associated virus serotype 9 (AAV9) was used to achieve overexpression of USP20. Myocardial function, histopathological changes, and pyroptosis levels in heart tissues were evaluated. Liquid chromatography tandem mass spectrometry (LC-MS/MS) analysis and co-immunoprecipitation (co-IP) were performed to identify the molecular mechanism of USP20 in septic cardiomyopathy.
Results: Our results showed that USP20 was downregulated in the myocardium of septic mice. Cardiomyocyte-specific USP20 deficiency worsened myocardial injury and cardiac dysfunction induced by LPS and CLP. LC-MS/MS analysis and co-IP revealed NLRP3 as a substrate protein of USP20. Mechanistically, USP20 removed K63-linked ubiquitin from K243 via its active site C154, inhibiting NLRP3's interaction with ASC and suppressing its activation and subsequent pyroptosis. Moreover, overexpressing USP20 in cardiomyocytes reduced LPS-induced myocardial injury. Additionally, the protective effect of USP20 against LPS-induced damage was nullified in the absence of NLRP3 in mice.
Conclusions: These findings suggest that cardiomyocyte-derived USP20 is crucial in septic cardiomyopathy progression and may serve as a novel therapeutic target for managing septic cardiomyopathy.
Background: Intrahepatic cholangiocarcinoma (ICC) exhibits poor prognosis and limited therapeutic options. Ferroptosis represents a promising therapeutic strategy, yet resistance mechanisms remain poorly understood. This study investigated the role of mucin 1 (MUC1) in regulating ferroptosis sensitivity in ICC.
Methods: Bioinformatic analyses of GEO and TCGA datasets identified ferroptosis-related factors in ICC. MUC1 expression was validated in ICC cell lines and clinical specimens. Ferroptosis sensitivity was assessed through RSL3-induced cell death assays, lipid peroxidation measurements, and iron detection. Mechanistic studies employed immunoprecipitation-mass spectrometry, co-immunoprecipitation, kinase assays, and deubiquitination assays. In vivo efficacy was evaluated using subcutaneous tumor models.
Results: MUC1 was identified as a critical ferroptosis suppressor in ICC. MUC1 overexpression conferred RSL3 resistance by inhibiting lipid peroxidation and reducing ferrous iron accumulation, independent of the GPX4-glutathione pathway. Mechanistically, MUC1 recruited Src kinase, which phosphorylated deubiquitinating enzyme ubiquitin-specific protease 10 (USP10) at tyrosines 359 and 364, enhancing ferroptosis suppressor protein 1 (FSP1) deubiquitination at lysine 246 and stabilizing FSP1 protein. Concurrently, Src phosphorylated N-myristoyltransferase 1 (NMT1) at tyrosine 41, augmenting FSP1 membrane localization through myristoylation. This dual mechanism potentiated the FSP1- coenzyme Q10 (CoQ10) antioxidant system. MUC1 knockdown significantly enhanced ferroptotic sensitivity in vitro and suppressed tumor growth in vivo.
Conclusions: MUC1 orchestrates ferroptosis resistance in ICC through the Src-USP10/NMT1-FSP1 axis. Targeting this signaling cascade represents a potential therapeutic strategy for overcoming ferroptosis resistance in ICC.
Background: Epigenetic regulation plays a crucial role in skeletal degenerative diseases, including osteoporosis. As an epigenetic reader, bromodomain protein 4 (Brd4) is known as a key driver of gene activation; however, its role in maintaining skeletal homeostasis remains largely unknown.
Methods: We examined Brd4 expression in bone specimens from osteoporotic patients and mouse models, and generated two types of Brd4 conditional knockout mice using Lyz2-Cre and Ctsk-Cre systems. Bone mass, osteoclast differentiation, and metabolic activity were assessed under physiological and pathological conditions, including ovariectomy and lipopolysaccharide (LPS) challenge. Mechanistic analyses were performed using transcriptomic screening, gene overexpression, and pharmacological interventions.
Results: Brd4 expression was markedly elevated in bones from osteoporotic patients and mice compared with normal controls. Deletion of Brd4 increased basal bone mass and prevented bone loss induced by ovariectomy or LPS, primarily by suppressing osteoclastogenesis through inhibition of glycolysis. Unbiased screening identified solute carrier family 9 member B2 (Slc9b2) as a downstream effector of Brd4. Overexpression of Slc9b2 partially rescued the impaired osteoclastogenesis caused by Brd4 depletion. Moreover, phosphatidylserine-containing nanoliposomes loaded with Brd4-targeting PROTACs (e.g., dBET6) effectively suppressed osteoclastogenesis and alleviated pathological bone loss.
Conclusions: Brd4 serves as a crucial regulator of osteoclast metabolism and differentiation. Targeting Brd4 represents a promising therapeutic strategy for the prevention and treatment of osteoporosis and pathological bone loss.
Background: A subset of people living with HIV (PLWH) exhibit poor immune recovery despite effective antiretroviral therapy (ART), remaining at risk of disease progression. The immunometabolic mechanisms underlying this immunological non-response remain unclear.
Methods: We integrated transcriptomic and immunophenotypic approaches to characterise immune differences between immunological responders (IRs) and non-responders (INRs). Public datasets were analysed to identify differentially expressed genes (DEGs), followed by enrichment analysis, predictive modelling, immune infiltration assessment, and regulatory network construction. In parallel, flow cytometry was performed to assess T and B cell subsets in an independent cohort including IRs, INRs, treatment-naïve patients (TNPs), and healthy controls (HCs).
Results: DEGs between IRs and INRs were enriched in mitochondrial and ribosomal pathways. INRs showed reduced Th1, Th17, and Tfh cells, alongside increased markers of immune activation and exhaustion. Predictive modelling identified five hub genes (ATP5O, PIGY, UQCRQ, COX7C, and BLVRB) associated with immune recovery, and clustering based on their expression defined two transcriptionally distinct subtypes. Flow cytometry further confirmed that INRs exhibited diminished CD4+ T cell counts, increased PD-1+ and HLA-DR+ expression, and reduced resting memory B cells, reflecting persistent immune dysfunction.
Conclusions: This study underscores the pivotal role of immunometabolic dysregulation in shaping heterogeneous immune responses to ART. By integrating computational and experimental data, we identified key biomarkers and regulatory pathways associated with immune recovery. Our findings highlight the central influence of metabolic processes on immune restoration outcomes and propose personalised metabolic interventions as a promising strategy to enhance therapeutic efficacy in HIV-infected individuals.
Sarcomas are a heterogeneous group of mesenchymal malignancies with poor prognosis and limited response to standard therapies, including immune checkpoint inhibitors (ICIs). Tumour-intrinsic factors—such as telomere maintenance mechanisms (TMMs) and metabolic reprogramming—play central roles in driving immune evasion and therapeutic resistance. Telomerase activation and alternative lengthening of telomeres sustain replicative immortality while influencing the tumour immune microenvironment. In parallel, metabolic adaptations, including glutamine dependency and arginine auxotrophy, further suppress antitumour immunity. Together, TMMs and metabolism form an integrated axis that shapes immune modulation and treatment outcomes. Recent advances—ranging from telomerase-based vaccines and TMM-targeted immunotherapies to metabolic modulators combined with ICIs—demonstrate the translational promise of targeting this axis. This review synthesises current knowledge on telomere‒metabolism crosstalk in sarcomas, highlights its impact on immunotherapy response, and outlines future directions for biomarker-driven, combinatorial strategies to overcome resistance and improve patient outcomes.
Background: Immunosuppressive tumour-associated macrophages (TAMs) represent a promising target for cancer immunotherapy; however, existing TAM-directed therapies have shown limited clinical efficacy. C-type lectin domain family 4 member E (CLEC4E), a pro-inflammatory molecule expressed on macrophages, was recently found to be highly enriched in TAMs. This study aims to elucidate the role of CLEC4E in TAMs and identify potential therapeutic agents targeting CLEC4E, and to clarify the mechanism of Bromodomain and extraterminal domain (BET) inhibitor NHWD-870 in downregulating CLEC4E.
Methods: We first assessed the correlation between CLEC4E expression and survival in melanoma patients. Clec4eflox/flox Lyz2-cre (knockout) and Clec4eflox/flox (control) mice were generated and implanted with melanoma or ovarian cancer models. Single-cell RNA sequencing was performed to characterise macrophage phenotypic changes following CLEC4E knockout, with validation via RT-PCR, flow cytometry and proteomic sequencing. A drug screen identified BET inhibitors targeting CLEC4E, and their mechanisms were further investigated using RNA silencing, Chromatin Immunoprecipitation (ChIP)-seq and luciferase reporter assays.
Results: In melanoma patients, high CLEC4E+ TAM infiltration was associated with poor prognosis. CLEC4E knockout significantly suppressed tumour growth compared to control mice. TAMs from knockout mice exhibited downregulated proliferation markers and upregulated genes related to antigen presentation and pro-inflammatory responses. Mechanistically, CLEC4E deletion inhibited TAM proliferation via the Erk signalling pathway, enhanced TAM‒T cell interactions, and increased granzyme B expression in T cells. The BET inhibitor NHWD-870 was shown to disrupt BRD4‒CEBPβ interaction, leading to downregulation of CLEC4E expression.
Conclusions: CLEC4E+ TAMs promote an immunosuppressive microenvironment by enhancing their own proliferation and impairing anti-tumour functions, thereby limiting T-cell cytotoxicity. Targeting the BRD4/CEBPβ/CLEC4E axis with BET inhibitors represents a promising therapeutic strategy for reprogramming TAMs and enhancing anti-tumour immunity.
Background: Numerous studies have demonstrated the promising efficacy of granulocyte colony-stimulating factor (G-CSF) in the treatment of couples with unexplained recurrent pregnancy loss (URPL) during early pregnancy. While neutrophils are recognised as the main effectors mediating immunoregulation, their G-CSF-mobilised phenotype and mechanisms regulating maternal–fetal immunity remain unclear.
Methods: Single-cell RNA sequencing (scRNA-seq) and single-cell T-cell receptor sequencing (scTCR-seq) were conducted to uncover the immune reconstitution dynamics of peripheral blood under G-CSF stimulation. Integrative analysis of transcriptomic-proteomic profiles with functional validation revealed a unique immunomodulatory neutrophil population. Further, we used spatial transcriptomics, flow cytometry and immunohistochemistry to explore the spatial distribution characteristics of this population at the maternal–fetal interface, and validated its therapeutic efficacy in animal models.
Results: G-CSF-mobilised peripheral blood (G-PB) displayed immune hyporesponsiveness. Unique neutrophils expressing high levels of CD177 and the S100A gene family expanded substantially in response to G-CSF. These neutrophils exhibited a comparatively immature morphology and impaired T-cell responses via contact-dependent arginase 1 release, as well as upregulation of T-cell immune checkpoints. A reduction of CD177+S100Ahi neutrophils was observed in both peripheral blood and decidua of URPL patients relative to healthy pregnant women. Functional validation in abortion-prone murine models confirmed that exogenous supplementation of G-CSF or adoptive transfer of CD177+S100Ahi neutrophils could successfully improve the pregnancy outcomes.
Conclusion: G-CSF played a crucial regulatory role in improving pregnancy outcomes by selectively expanding CD177+S100Ahi neutrophils with polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) properties, providing a solid theoretical foundation for the treatment of patients with URPL using G-CSF.