N. Li et al. (Oct 2025)
Journal of Cellular and Molecular Medicine 29 19
BNIP3L/BNIP3‐Mediated Mitophagy Contributes to the Maintenance of Ovarian Cancer Stem Cells
Ovarian cancer remains the most lethal gynaecological malignancy,with tumour recurrence and chemoresistance posing significant therapeutic challenges. Emerging evidence suggests that cancer stem cells (CSCs),a rare subpopulation within tumours with self‐renewal and differentiation capacities,contribute to these hurdles. Therefore,elucidating the mechanisms that sustain CSCs is critical for improving treatment strategies. Mitophagy,a selective process for eliminating damaged mitochondria,plays a key role in maintaining cellular homeostasis,including CSC survival. Our study demonstrates that ovarian CSCs exhibit enhanced mitophagy,accompanied by elevated expression of the mitochondrial outer membrane receptors BNIP3 and BNIP3L. Knockdown of BNIP3 or BNIP3L significantly reduces mitophagy and impairs CSC self‐renewal,indicating that receptor‐mediated mitophagy is essential for CSC maintenance. Mechanistically,we identify that hyperactivated NF‐κB signalling drives the upregulation of BNIP3 and BNIP3L in ovarian CSCs. Inhibition of NF‐κB signalling,either via p65 knockdown or pharmacological inhibitors,effectively suppresses mitophagy. Furthermore,we demonstrate that elevated DNA‐PK expression contributes to the constitutive activation of NF‐κB signalling,thereby promoting mitophagy in ovarian CSCs. In summary,our findings establish that BNIP3/BNIP3L‐mediated mitophagy,driven by DNA‐PK‐dependent NF‐κB hyperactivation,is essential for CSC maintenance. Targeting the DNA‐PK/NF‐κB/BNIP3L‐BNIP3 axis to disrupt mitochondrial quality control in CSCs represents a promising therapeutic strategy to prevent ovarian cancer recurrence and metastasis.
View Publication
A. Becerra-Calixto et al. (Oct 2025)
Journal of Neuroinflammation 22
A neuroimmune cerebral assembloid model to study the pathophysiology of familial Alzheimer’s disease
Alzheimer’s disease (AD) is the leading cause of dementia globally. The accumulation of amyloid and tau proteins,neuronal cell death and neuroinflammation are seen with AD progression,resulting in memory and cognitive impairment. Microglia are crucial for AD progression as they engage with neural cells and protein aggregates to regulate amyloid pathology and neuroinflammation. Recent studies indicate that microglia contribute to the propagation of amyloid beta (Aβ) via their immunomodulatory functions including Aβ phagocytosis and inflammatory cytokine production. Three-dimensional cell culture techniques provide the opportunity to study pathophysiological changes in AD in human-derived samples that are difficult to recapitulate in animal models (e.g.,transgenic mice). However,these models often lack immune cells such as microglia,which play a critical role in AD pathophysiology. In this study,we developed a neuroimmune assembloid model by integrating cerebral organoids (COs) with induced microglia-like cells (iMGs) derived from human induced pluripotent stem cells from familial AD patient with PSEN2 mutation. After 120 days in culture,we found that iMGs were successfully integrated within the COs. Interestingly,our assembloids displayed histological,functional and transcriptional features of the pro-inflammatory environment seen in AD,including amyloid plaque-like and neurofibrillary tangle-like structures,reduced microglial phagocytic capability,and enhanced neuroinflammatory and apoptotic gene expression. In conclusion,our neuroimmune assembloid model effectively replicates the inflammatory phenotype and amyloid pathology seen in AD. The online version contains supplementary material available at 10.1186/s12974-025-03544-x.
View Publication
R. B. Kang et al. (Oct 2025)
Nature Communications 16
Human pancreatic α-cell heterogeneity and trajectory inference analyses reveal SMOC1 as a β-cell dedifferentiation gene
β-cell dysfunction and dedifferentiation towards an α-cell-like phenotype are hallmarks of type 2 diabetes. However,the cell subtypes involved in β-to-α-cell transition are unknown. Using single-cell and single-nucleus RNA-seq,RNA velocity,PAGA/cell trajectory inference,and gene commonality,we interrogated α-β-cell fate switching in human islets. We found five α-cell subclusters with distinct transcriptomes. PAGA analysis showed bifurcating cell trajectories in non-diabetic while unidirectional cell trajectories from β-to-α-cells in type 2 diabetes islets suggesting dedifferentiation towards α-cells. Ten genes comprised the common signature genes in trajectories towards α-cells. Among these,the α-cell gene SMOC1 was expressed in β-cells in type 2 diabetes. Enhanced SMOC1 expression in β-cells decreased insulin expression and secretion and increased β-cell dedifferentiation markers. Collectively,these studies reveal differences in α-β-cell trajectories in non-diabetes and type 2 diabetes human islets,identify signature genes for β-to-α-cell trajectories,and discover SMOC1 as an inducer of β-cell dysfunction and dedifferentiation. Subject terms: Cell signalling,Diabetes,Differentiation
View Publication
M. Amouzgar et al. (Oct 2025)
Nature Communications 16
A deep single cell mass cytometry approach to capture canonical and noncanonical cell cycle states
The cell cycle (CC) underpins diverse cell processes like cell differentiation,cell expansion,and tumorigenesis but current single-cell (sc) strategies study CC as: coarse phases,rely on transcriptomic signatures,use imaging modalities limited to adherent cells,or lack high-throughput multiplexing. To solve this,we develop an expanded,Mass Cytometry (MC) approach with 48 CC-related molecules that deeply phenotypes the diversity of scCC states. Using Cytometry by Time of Flight,we quantify scCC states across suspension and adherent cell lines,and stimulated primary human T cells. Our approach captures the diversity of scCC states,including atypical CC states beyond canonical definitions. Pharmacologically-induced CC arrest reveals that perturbations exacerbate noncanonical states and induce previously unobserved states. Notably,primary cells escaping CC inhibition demonstrated aberrant CC states compared to untreated cells. Our approach enables deeper phenotyping of CC biology that generalizes to diverse cell systems with simultaneous multiplexing and integration with MC platforms. Subject terms: Assay systems,Proteomics,Cell biology,Immunology,Systems biology
View Publication
K. Vekrellis et al. (Sep 2025)
NPJ Parkinson's Disease 11
Proteolytic activities of extracellular vesicles attenuate A-synuclein aggregation
Extracellular vesicles (EVs) are nano-sized lipid vesicles released into the extracellular space. We investigated the role of mouse brain-derived EVs in α-synuclein (α-syn) degradation and pathology transmission. Using sucrose gradient isolation and biochemical characterization,we found that EVs harbor active proteases that cleave both monomeric α-syn and pre-formed fibrils (PFFs). Protease activity and inhibitor profiling identified cathepsins B and S as key enzymes mediating this cleavage. EV-mediated proteolysis reduced the seeding capacity of α-syn PFFs in vitro and in vivo,whereas protease inhibition enhanced aggregation. Proteomic analysis revealed a restricted protease repertoire within EV cargo. Our findings suggest that EVs regulate extracellular α-syn levels via proteolysis,thereby modulating its prion-like spreading potential. We suggest that EVs represent a novel post-translational mechanism to regulate the levels of extracellular α-syn and may thus affect the spreading of α-syn pathology. Targeting this proteolytic capacity may offer new therapeutic interventions for mitigating synucleinopathies. Subject terms: Biochemistry,Cell biology,Neuroscience,Pathogenesis
View Publication
Z. Li et al. (Sep 2025)
Journal of Asthma and Allergy 18 4
Inhibition of LOXL2 Suppresses Nasal Mucosal Inflammation and Remodeling in Allergic Rhinitis
Tissue remodeling is a key feature of allergic rhinitis (AR),but its underlying molecular mechanisms remain unclear. Lysyl oxidase-like 2 (LOXL2),a regulator of tissue remodeling,has not been studied in AR. Proteomic analysis was performed on nasal mucosal tissues from 8 AR patients and 8 healthy controls (HCs) to identify differentially expressed proteins (DEPs). The top three upregulated DEPs and their association with tissue remodeling markers were validated by immunofluorescence,Western blot,and RT-qPCR in an independent cohort of 30 AR patients and 30 HCs. In vitro,human nasal epithelial cells (HNECs) were treated with IL-4,and the effects of candidate protein inhibitors on remodeling were assessed. An AR mouse model was used to evaluate the impact of these inhibitors on nasal inflammation and remodeling. Proteomic analysis revealed a disease-specific protein expression profile in the nasal mucosa of AR patients,with the top three upregulated proteins being LOXL2,TGF-β1,and TIRAP. Tissue validation showed that LOXL2 was significantly upregulated in the nasal mucosa of AR patients compared to HCs and was significantly correlated with EMT markers (TGF-β1,α-SMA,and E-cadherin). In vitro,IL-4 stimulation significantly upregulated LOXL2,TGF-β1,and α-SMA,while downregulating E-cadherin in a dose-dependent manner in human nasal epithelial cells. These effects were reversed by inhibition of LOXL2. Further investigations demonstrated that LOXL2 promotes tissue remodeling through activation of the TGF-β1/Smad signaling pathway. In the AR mouse model,LOXL2 inhibitors significantly reduced nasal mucosal inflammation and tissue remodeling. Our proteomic analysis suggests that LOXL2 may be involved in the pathological remodeling processes of AR,potentially through modulation of the TGF-β1/Smad signaling pathway. These findings provide preliminary evidence that LOXL2 could serve as a candidate biomarker and a possible therapeutic target in AR,warranting further investigation.
View Publication
S. Trushin et al. (Sep 2025)
eBioMedicine 120 4
Therapeutic assessment of a novel mitochondrial complex I inhibitor in in vitro and in vivo models of Alzheimer's disease
Despite recent approval of monoclonal antibodies that reduce amyloid (Aβ) accumulation,the development of disease-modifying strategies targeting the underlying mechanisms of Alzheimer's disease (AD) is urgently needed. We demonstrate that mitochondrial complex I (mtCI) represents a druggable target,where its weak inhibition activates neuroprotective signalling,benefiting AD mouse models with Aβ and p-Tau pathologies. Rational design and structure‒activity relationship studies yielded mtCI inhibitors profiled in a drug discovery funnel designed to address safety,selectivity,and efficacy. The lead compound C458 is highly protective against Aβ toxicity,has favourable pharmacokinetics,and minimal off-target effects. C458 exhibited excellent brain penetrance,activating neuroprotective pathways with a single dose. Preclinical studies in APP/PS1 mice were conducted using functional tests,metabolic assessment,in vivo 31 P-NMR spectroscopy,blood cytokine panels,ex vivo electrophysiology,and Western blotting. Chronic oral administration improved long-term potentiation,reduced oxidative stress and inflammation,and enhanced mitochondrial biogenesis,antioxidant signalling,and cellular energetics. Efficacy against Aβ and p-Tau was confirmed in human organoids. These studies provide further evidence that the restoration of mitochondrial function in response to mild energetic stress represents a promising disease-modifying strategy for AD. This research was supported by grants from NIH AG 5549-06,NS1 07265,AG 062135,UG3/UH3 NS 113776,and ADDF 291204 (all to ET); U19 AG069701 (to TK); the Alzheimer’s Association Research Fellowship grant 23AARF-1027342 (to TKON).
View Publication
A. Chakraborty et al. (Aug 2025)
International Journal of Molecular Sciences 26 17
Cholesterol is an essential plasma membrane component,and altered cholesterol metabolism has been linked to cholesterol accumulation in the airways of COPD and cystic fibrosis patients. However,its role in airway epithelial differentiation is not well understood. Tandem mass spectrometry-based proteomic analysis of differentiating primary human bronchial epithelial cells (phBECs) revealed an overall inhibition of the cholesterol biosynthesis pathway. We hypothesized that excess cholesterol impairs the differentiation of phBECs into a fully functional bronchial epithelium. PhBECs were differentiated in the presence of 80 µM cholesterol for 21 days,the main airway cell type populations monitored using qRT-PCR and immunofluorescent stainings,and epithelial barrier integrity was analyzed via transepithelial electrical resistance measurements. Chronic cholesterol exposure led to a significant increase in CC10 + secretory cells at the expense of ciliated cells. Pathway enrichment analysis suggested the tumor protein p53 as a master regulator of genes during normal differentiation of phBECs. Chronic cholesterol exposure drastically impaired the nuclear translocation of p53. Our findings suggest that this inhibition underlies the cholesterol-induced expansion of CC10 + secretory cell populations at the expense of ciliated cells. In conclusion,we identify cholesterol as an important regulator of normal bronchial epithelial cell differentiation through inhibition of p53 nuclear translocation.
View Publication
W. Yang et al. (Aug 2025)
Cancers 17 17
A Polyomavirus-Positive Merkel Cell Carcinoma Mouse Model Supports a Unified Origin for Somatic and Germ Cell Cancers
Cancer research has long focused on mutations in normal body cells,but this approach has not produced major breakthroughs for most cancers. Our study explores a different concept that some aggressive cancers may actually arise from early reproductive cells called primordial germ cells,which normally develop into eggs and sperm. We created a new experimental model showing how a virus can transform human primordial germ cell-like cells into virus-positive Merkel cell carcinoma,a rare but deadly skin cancer. This model shows that cancers can emerge through changes in developmental states rather than relying solely on genetic mutations. By linking cancer development to early germ cells,our findings suggest a unifying explanation for both germ cell cancers and body cancers. This new perspective may guide more effective approaches to study,diagnose,and treat cancer by focusing on early human development rather than only DNA mutations and later developmental stages.
View Publication