1.Experimental study on promotion of skin radiation damage repair by icarin via HIF-2α/VEGF/Notch pathway to enhance the paracrine function of adipose-derived stem cells.
Yuer ZUO ; Shuangyi LI ; Siyu TAN ; Xiaohao HU ; Zhou LI ; Haoxi LI
Chinese Journal of Reparative and Reconstructive Surgery 2025;39(7):881-890
OBJECTIVE:
To investigate the effectiveness and preliminary mechanisms of icariin (ICA) in enhancing the reparative effects of adipose-derived stem cells (ADSCs) on skin radiation damagies in rats.
METHODS:
Twelve SPF-grade Sprague Dawley rats [body weight (220±10) g] were subjected to a single dose of 10 Gy X-ray irradiation on a 1.5 cm×1.5 cm area of their dorsal skin, with a dose rate of 200 cGy/min to make skin radiation damage model. After successful modelling, the rats were randomly divided into 4 groups ( n=3), and on day 2, the corresponding cells were injected subcutaneously into the irradiated wounds: group A received 0.1 mL of rat ADSCs (1×10 7cells/mL), group B received 0.1 mL of rat ADSCs (1×10 7cells/mL)+1 μmol/L ICA (0.1 mL), group C received 0.1 mL of rat ADSCs (1×10 7cells/mL) pretreated with a hypoxia-inducible factor 2α (HIF-2α) inhibitor+1 μmol/L ICA (0.1 mL), and group D received 0.1 mL of rat ADSCs (1×10 7cells/mL) pretreated with a Notch1 inhibitor+1 μmol/L ICA (0.1 mL). All treatments were administered as single doses. The skin injury in the irradiated areas of the rats was observed continuously from day 1 to day 7 after modelling. On day 28, the rats were sacrificed, and skin tissues from the irradiated areas were harvested for histological examination (HE staining and Masson staining) to assess the repair status and for quantitative collagen content detection. Immunohistochemical staining was performed to detect CD31 expression, while Western blot and real-time fluorescence quantitative PCR (qRT-PCR) were used to measure the protein and mRNA relative expression levels of vascular endothelial growth factor (VEGF), platelet-derived growth factor BB (PDGF-BB), fibroblast growth factor 2 (FGF-2), interleukin 10 (IL-10), transforming growth factor β (TGF-β), HIF-2α, and Notch1, 2, and 3.
RESULTS:
All groups exhibited skin ulcers and redness after irradiation. On day 3, exudation of tissue fluid was observed in all groups. On day 7, group B showed significantly smaller skin injury areas compared to the other 3 groups. On day 28, histological examination revealed that the epidermis was thickened and the dermal fibers were slightly disordered with occasional inflammatory cell aggregation in group A. In group B, the epidermis appeared more normal, the dermal fibers were more orderly, and there was an increase in new blood vessels without significant inflammatory cell aggregation. In contrast, groups C and D showed significantly increased epidermal thickness, disordered and disrupted dermal fibers. Group B had higher collagen fiber content than the other 3 groups, and group D had lower content than group A, with significant differences ( P<0.05). Immunohistochemical staining showed that group B had significantly higher CD31 expression than the other 3 groups, while groups C and D had lower expression than group A, with significant differences ( P<0.05). Western blot and qRT-PCR results indicated that group B had significantly higher relative expression levels of VEGF, PDGF-BB, FGF-2, IL-10, TGF-β, HIF-2α, and Notch1, 2, and 3 proteins and mRNAs compared to the other 3 groups ( P<0.05).
CONCLUSION
ICA may enhance the reparative effects of ADSCs on rat skin radiation damage by promoting angiogenesis and reducing inflammatory responses through the HIF-2α-VEGF-Notch signaling pathway.
Animals
;
Rats, Sprague-Dawley
;
Skin/pathology*
;
Rats
;
Vascular Endothelial Growth Factor A/genetics*
;
Basic Helix-Loop-Helix Transcription Factors/genetics*
;
Signal Transduction
;
Flavonoids/pharmacology*
;
Adipose Tissue/cytology*
;
Stem Cells/cytology*
;
Receptors, Notch/metabolism*
;
Radiation Injuries, Experimental/metabolism*
;
Wound Healing/drug effects*
;
Male
2.Effects of p38 phosphorylation on stemness maintenance and chemotherapy drug resistance of PANC-1 cells.
Xueying SHI ; Jinbo YU ; Shihai YANG ; Jin ZHAO
Chinese Journal of Cellular and Molecular Immunology 2025;41(2):116-124
Objective The aim of this study was to investigate the effect of p38 on stem cell maintenance of pancreatic cancer. Methods Human pancreatic cancer cells PANC-1 were treated with different concentrations of 5-fluorouracil(5-FU)(0.5×IC50, IC50, and 2×IC50) for 24 hours, and VX-702 (p38 phosphorylation inhibitor) was added, and the cells were inoculated in 6-well culture dishes with ultra-low adhesion to observe the changes of sphere tumors. The expression levels of cyclin-dependent kinase 2(CDK2), cyclin B1 and D1, Octamer-binding transcription factor 4(OCT4), SRY-box transcription factor 2(SOX2), Nanog and p38 were measured by Western blot. The mRNA expression levels of p38, OCT4, Nanog and SOX2 were tested by RT-PCR. Cell cycle, apoptosis, and the proportion of CD44+CD133+PANC-1 cells were evaluated by flow cytometry. Results The results showed that 5-FU inhibited the formation of tumor spheres in PANC-1 cells, increased CD44+CD133+cell fragments, down-regulated the expression of OCT4, Nanog and SOX2, and inhibited the stemness maintenance of PANC-1 tumor stem cells. Phosphorylation of PANC-1 cells was inhibited by a highly selective p38 MAPK inhibitor, VX-702(p38 mitogen-activated protein kinase inhibitor), which had the same effect as 5-FU treatment. When VX-702 combined with 5-FU was used to treat PANC-1 cells, the therapeutic effect was enhanced. Conclusion p38 inhibitors decreased PANC-1 cell activity and increased cell apoptosis. p38 inhibitors inhibit the stemness maintenance of pancreatic cancer stem cells.
Humans
;
Phosphorylation/drug effects*
;
Cell Line, Tumor
;
p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors*
;
Neoplastic Stem Cells/metabolism*
;
Drug Resistance, Neoplasm/drug effects*
;
Fluorouracil/pharmacology*
;
Pancreatic Neoplasms/pathology*
;
Apoptosis/drug effects*
;
SOXB1 Transcription Factors/genetics*
;
Octamer Transcription Factor-3/genetics*
3.Effects of human umbilical cord-derived mesenchymal stem cell therapy for cavernous nerve injury-induced erectile dysfunction in the rat model.
Wei WANG ; Ying LIU ; Zi-Hao ZHOU ; Kun PANG ; Jing-Kai WANG ; Peng-Fei HUAN ; Jing-Ru LU ; Tao ZHU ; Zuo-Bin ZHU ; Cong-Hui HAN
Asian Journal of Andrology 2025;27(4):508-515
Stem cell treatment may enhance erectile dysfunction (ED) in individuals with cavernous nerve injury (CNI). Nevertheless, no investigations have directly ascertained the implications of varying amounts of human umbilical cord-derived mesenchymal stem cells (HUC-MSCs) on ED. We compare the efficacy of three various doses of HUC-MSCs as a therapeutic strategy for ED. Sprague-Dawley rats (total = 175) were randomly allocated into five groups. A total of 35 rats underwent sham surgery and 140 rats endured bilateral CNI and were treated with vehicles or doses of HUC-MSCs (1 × 10 6 cells, 5 × 10 6 cells, and 1 × 10 7 cells in 0.1 ml, respectively). Penile tissues were harvested for histological analysis on 1 day, 3 days, 7 days, 14 days, 28 days, 60 days, and 90 days postsurgery. It was found that varying dosages of HUC-MSCs enhanced the erectile function of rats with bilateral CNI and ED. Moreover, there was no significant disparity in the effectiveness of various dosages of HUC-MSCs. However, the expression of endothelial markers (rat endothelial cell antigen-1 [RECA-1] and endothelial nitric oxide synthase [eNOS]), smooth muscle markers (alpha smooth muscle actin [α-SMA] and desmin), and neural markers (neurofilament [RECA-1] and neurogenic nitric oxide synthase [nNOS]) increased significantly with prolonged treatment time. Masson's staining demonstrated an increased in the smooth muscle cell (SMC)/collagen ratio. Significant changes were detected in the microstructures of various types of cells. In vivo imaging system (IVIS) analysis showed that at the 1 st day, the HUC-MSCs implanted moved to the site of damage. Additionally, the oxidative stress levels were dramatically reduced in the penises of rats administered with HUC-MSCs.
Male
;
Animals
;
Erectile Dysfunction/metabolism*
;
Rats, Sprague-Dawley
;
Mesenchymal Stem Cell Transplantation/methods*
;
Rats
;
Penis/pathology*
;
Humans
;
Disease Models, Animal
;
Umbilical Cord/cytology*
;
Peripheral Nerve Injuries/complications*
;
Mesenchymal Stem Cells
;
Nitric Oxide Synthase Type III/metabolism*
;
Actins/metabolism*
;
Nitric Oxide Synthase Type I/metabolism*
4.Exosomes derived from mesenchymal stem cells alleviate white matter damage in neonatal rats by targeting the NLRP3 inflammasome.
Chao WANG ; Yan-Ping ZHU ; BAYIERCAICIKE ; Yu-Qing FENG ; Yan-Mei WANG
Chinese Journal of Contemporary Pediatrics 2025;27(9):1119-1127
OBJECTIVES:
To investigate whether mesenchymal stem cell-derived exosomes (MSC-Exo) alleviate white matter damage (WMD) in neonatal rats by targeting the nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3).
METHODS:
Three-day-old Sprague-Dawley rats were randomly assigned to four groups: Sham, hypoxia-ischemia (HI), MSC-Exo, and MCC950 (NLRP3 inhibitor) (n=24 per group). The WMD model was established by unilateral common carotid artery ligation combined with hypoxia. Exosomes (1×108 particles/μL) were transplanted into the lateral ventricle using stereotaxic guidance. Fourteen days after modeling, hematoxylin-eosin staining was used to observe pathological changes in brain tissue, and transmission electron microscopy was used to assess myelinated axons. Western blotting was performed to detect the expression of myelin basic protein (MBP), NLRP3, caspase-1, and interleukin-1β (IL-1β). Immunohistochemistry was used to measure NLRP3, caspase-1, and IL-1β expression. Twenty-eight days post-modeling, behavioral changes were evaluated using the Morris water maze.
RESULTS:
In the HI group, marked inflammatory cell infiltration, extensive vacuolation, and decreased numbers of myelinated axons were observed compared to the Sham group. The MSC-Exo group showed reduced inflammatory infiltration, fewer vacuoles, and increased myelinated axons compared to the HI group, while the MCC950 group showed nearly normal cell morphology. Compared to the Sham group, the HI group exhibited decreased MBP expression, fewer platform crossings, shorter time in the target quadrant, increased expression of NLRP3, caspase-1, and IL-1β, and longer escape latency (all P<0.05). Compared to the HI group, the MSC-Exo and MCC950 groups showed increased MBP expression, more platform crossings, longer target quadrant stay, and reduced NLRP3, caspase-1, and IL-1β expression, as well as shorter escape latency (all P<0.05).
CONCLUSIONS
MSC-Exo may attenuate white matter damage in neonatal rats by targeting the NLRP3 inflammasome and promoting oligodendrocyte maturation.
Animals
;
NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors*
;
Rats, Sprague-Dawley
;
White Matter/pathology*
;
Inflammasomes/physiology*
;
Rats
;
Animals, Newborn
;
Mesenchymal Stem Cells
;
Interleukin-1beta/analysis*
;
Male
;
Caspase 1/analysis*
;
Hypoxia-Ischemia, Brain/therapy*
;
Myelin Basic Protein/analysis*
5.Human umbilical cord mesenchymal stem cells protect against neonatal white matter injury by activating the Nrf2/Keap1/HO-1 signaling pathway.
Chao WANG ; Meng-Xin WANG ; Yan-Ping ZHU
Chinese Journal of Contemporary Pediatrics 2025;27(11):1398-1407
OBJECTIVES:
To investigate whether human umbilical cord mesenchymal stem cells (HUC-MSCs) play protective effects against white matter injury (WMI) in neonatal rats via activation of the nuclear factor-erythroid 2-related factor 2 (Nrf2)/Kelch-like ECH-associated protein 1 (Keap1)/heme oxygenase-1 (HO-1) signaling pathway.
METHODS:
A neonatal WMI model was established in 3-day-old Sprague-Dawley rats by unilateral common carotid artery ligation combined with hypoxia. The study comprised two parts. (1) Rats were randomized into sham, hypoxia-ischemia (HI), and HUC-MSC groups (n=36 per group); brain tissues were collected at 7, 14, and 21 days after modeling. (2) Rats were randomized into sham, HI, HUC-MSC, and HUC-MSC+ML385 (Nrf2 inhibitor) groups (n=12 per group); tissues were collected 14 days after modeling. Hematoxylin-eosin staining assessed histopathology, and Luxol fast blue staining evaluated myelination. Immunohistochemistry examined the localization and expression of Nrf2, myelin basic protein (MBP), and proteolipid protein (PLP). Immunofluorescence assessed synaptophysin (SYP) and postsynaptic density-95 (PSD-95). Western blotting quantified Nrf2, Keap1, HO-1, SYP, PSD-95, MBP, and PLP. Spatial learning and memory were evaluated by the Morris water maze.
RESULTS:
At 7, 14, and 21 days after modeling, the sham group showed intact white matter, whereas the HI group exhibited white matter disruption, cellular vacuolation, and disorganized nerve fibers. These pathological changes were attenuated in the HUC-MSC group. Compared with the HI group, the HUC-MSC group showed increased Nrf2 immunopositivity and protein levels, increased HO-1 protein levels, and decreased Keap1 protein levels (P<0.05). Compared with the HI group, the HUC-MSC group had higher SYP and PSD-95 immunofluorescence intensities and protein levels, higher MBP and PLP positivity and protein levels, increased mean optical density of myelin, more platform crossings, and longer time in the target quadrant (all P<0.05). These improvements were reduced in the HUC-MSC+ML385 group compared with the HUC-MSC group (P<0.05).
CONCLUSIONS
HUC-MSCs may promote oligodendrocyte maturation and synaptogenesis after neonatal WMI by activating the Nrf2/Keap1/HO-1 pathway, thereby improving spatial cognitive function.
NF-E2-Related Factor 2/physiology*
;
Animals
;
Rats, Sprague-Dawley
;
Signal Transduction/physiology*
;
Humans
;
Rats
;
White Matter/pathology*
;
Kelch-Like ECH-Associated Protein 1/physiology*
;
Umbilical Cord/cytology*
;
Heme Oxygenase-1/physiology*
;
Animals, Newborn
;
Male
;
Mesenchymal Stem Cell Transplantation
;
Heme Oxygenase (Decyclizing)/physiology*
;
Mesenchymal Stem Cells/physiology*
;
Female
;
Hypoxia-Ischemia, Brain
6.Comparative Study of Diffuse Large B-Cell Lymphoma and Reactive Lymphoid Hyperplasia Lymph Node Derived Mesenchymal Stem Cells.
Yu-Shuo MA ; Zhi-He LIU ; Yang SUN ; Yu-Hang ZHANG ; Wen-Qiu WANG ; Li-Sheng WANG ; Xia ZHAO
Journal of Experimental Hematology 2025;33(5):1516-1523
OBJECTIVE:
To investigate the biological behavior, differentiation ability, and differential gene expression of lymph node mesenchymal stem cells (MSCs) in patients with diffuse large B-cell lymphoma (DLBCL) and reactive lymphoid hyperplasia (RLH), providing a theoretical basis for clinical chemotherapy resistance.
METHODS:
Lymph node MSCs from patients with DLBCL and RLH were separated, passaged and cultured. The cell morphology and growth status were observed. Flow cytometry was performed to detect the immune phenotype of MSCs. The in vitro directed differentiation ability of the two types of MSCs was observed. High-throughput sequencing was used to analyze the differential gene expression and enrichment of two groups of MSCs.
RESULTS:
The lymph node MSCs of patients with DLBCL and RLH had similar cell morphology and growth characteristics, and both groups of MSCs expressed CD90, CD105, and CD73 on the cell surface. Compared with lymph node MSCs derived from patients with RLH, lymph node MSCs derived from DLBCL patients showed stronger osteogenic and adipogenic differentiation abilities. High-throughput sequencing results displayed that lymph node MSCs derived from DLBCL patients significantly upregulated some genes such as TOP2A, LFNG, GRIA3, SEC14L2, SPON2, AURKA, LRRC15, FOXD1, HOXC9, CDC20 and remarkably downregulated some genes such as TBC1D8, LDLR, PCDHAC2, POLH, PKP2, ANKRD37, DMKN, HSD11B1, ARHGAP20, PTGS1,etc.
CONCLUSION
Lymph node MSCs in DLBCL patients exhibit unique biological behavior and gene expression profiles, which may be closely related to clinical chemotherapy resistance.
Humans
;
Mesenchymal Stem Cells/cytology*
;
Lymphoma, Large B-Cell, Diffuse/pathology*
;
Cell Differentiation
;
Lymph Nodes/pathology*
;
Pseudolymphoma/pathology*
7.Effect of astragaloside IV on osteogenic differentiation of BMSCs in osteoporotic rats via regulation of miR-21 and inhibition of the Notch signaling pathway.
Jingjing XIAO ; Xiaolan LIU ; Jianying HUANG ; Ben DOU
Journal of Central South University(Medical Sciences) 2025;50(7):1126-1136
OBJECTIVES:
The core pathology of osteoporosis lies in bone resorption exceeding bone formation; thus, promoting osteogenesis is a key therapeutic strategy. The osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) forms the biological basis of bone formation. Astragaloside IV (A-IV), a major active component of Astragalus membranaceus, is known to enhance osteogenesis, but its precise molecular mechanisms remain unclear. This study aims to investigate the effects of A-IV on the proliferation and osteogenic differentiation of BMSCs from osteoporotic rats and to elucidate its molecular mechanism through the regulation of microRNA-21 (miR-21) and Notch2 expression.
METHODS:
After 1 week of adaptive feeding, mature female SD rats were randomly divided into a sham-operated (Sham) group (n=4) and an ovariectomized (OVX) group (n=8) to establish an osteoporosis model. Twelve weeks after surgery, BMSCs were isolated from femoral bone marrow and cultured. Cells were divided into a S-BMSCs group (from Sham), an O-BMSCs group (from OVX), and an A-BMSCs group (from OVX-derived BMSCs treated with A-IV). S-BMSCs and O-BMSCs were induced for osteogenic differentiation using osteogenic induction medium, whereas A-BMSCs were treated with A-IV before induction. Flow cytometry was used to identify mesenchymal stem cell surface markers (CD29) and hematopoietic stem cell marker (CD34) to confirm BMSC characteristics. Cell proliferation was assessed using the methyl thiazolyl tetrazolium (MTT) assay. Alizarin red staining was performed to quantify calcium nodule formation, and alkaline phosphatase (ALP) activity assays were used to evaluate osteogenic differentiation. Real-time reverse transcription PCR (real-time RT-PCR) was used to detect changes in osteogenic-related genes, runt-related transcription factor 2 (Runx2) and osteopontin (OPN), as well as miR-21 expression. Western blotting was performed to assess Runx2, OPN, and Notch2 protein expression.
RESULTS:
Flow cytometry confirmed that O-BMSCs retained the phenotypic characteristics of mesenchymal stem cells. A-IV significantly enhanced the proliferation of BMSCs from osteoporotic rats (P<0.05), increased ALP activity, and upregulated the mRNA and protein expression of Runx2 and OPN (P<0.05). Bioinformatic and experimental analyses demonstrated that miR-21 directly targeted Notch2. A-IV treatment increased miR-21 expression while suppressing Notch2 protein expression and inhibiting activation of the Notch signaling pathway (P<0.05).
CONCLUSIONS
Astragaloside IV promotes the osteogenic differentiation of BMSCs derived from osteoporotic rats by upregulating miR-21 expression and inhibiting the key Notch signaling protein Notch2, thereby relieving the Notch2-mediated suppression of osteogenesis.
Animals
;
Triterpenes/pharmacology*
;
Saponins/pharmacology*
;
Osteogenesis/drug effects*
;
MicroRNAs/metabolism*
;
Rats, Sprague-Dawley
;
Female
;
Cell Differentiation/drug effects*
;
Mesenchymal Stem Cells/drug effects*
;
Signal Transduction/drug effects*
;
Osteoporosis/pathology*
;
Rats
;
Cells, Cultured
;
Receptor, Notch2/metabolism*
;
Receptors, Notch/metabolism*
;
Ovariectomy
;
Cell Proliferation/drug effects*
8.Dentate Gyrus Morphogenesis is Regulated by an Autism Risk Gene Trio Function in Granule Cells.
Mengwen SUN ; Weizhen XUE ; Hu MENG ; Xiaoxuan SUN ; Tianlan LU ; Weihua YUE ; Lifang WANG ; Dai ZHANG ; Jun LI
Neuroscience Bulletin 2025;41(1):1-15
Autism Spectrum Disorders (ASDs) are reported as a group of neurodevelopmental disorders. The structural changes of brain regions including the hippocampus were widely reported in autistic patients and mouse models with dysfunction of ASD risk genes, but the underlying mechanisms are not fully understood. Here, we report that deletion of Trio, a high-susceptibility gene of ASDs, causes a postnatal dentate gyrus (DG) hypoplasia with a zigzagged suprapyramidal blade, and the Trio-deficient mice display autism-like behaviors. The impaired morphogenesis of DG is mainly caused by disturbing the postnatal distribution of postmitotic granule cells (GCs), which further results in a migration deficit of neural progenitors. Furthermore, we reveal that Trio plays different roles in various excitatory neural cells by spatial transcriptomic sequencing, especially the role of regulating the migration of postmitotic GCs. In summary, our findings provide evidence of cellular mechanisms that Trio is involved in postnatal DG morphogenesis.
Animals
;
Dentate Gyrus/metabolism*
;
Mice
;
Morphogenesis/physiology*
;
Neurons/pathology*
;
Cell Movement
;
Mice, Inbred C57BL
;
Autism Spectrum Disorder/pathology*
;
Mice, Knockout
;
Neural Stem Cells
;
Male
;
Neurogenesis
;
Autistic Disorder/genetics*
9.Conditional Tnfaip6-Knockout in Inner Ear Hair Cells Does not Alter Auditory Function.
Yue QIU ; Song GAO ; Xiaoqiong DING ; Jie LU ; Xinya JI ; Wenli HAO ; Siqi CHENG ; Haolinag DU ; Yajun GU ; Chenjie YU ; Cheng CHENG ; Xia GAO
Neuroscience Bulletin 2025;41(3):421-433
Noise-induced hearing loss is a worldwide public health issue that is characterized by temporary or permanent changes in hearing sensitivity. This condition is closely linked to inflammatory responses, and interventions targeting the inflammatory gene tumor necrosis factor-alpha (TNFα) are known to mitigate cochlear noise damage. TNFα-induced proteins (TNFAIPs) are a family of translucent acidic proteins, and TNFAIP6 has a notable association with inflammatory responses. To date, there have been few reports on TNFAIP6 levels in the inner ear. To elucidate the precise mechanism, we generated transgenic mouse models with conditional knockout of Tnfaip6 (Tnfaip6 cKO). Evaluation of hair cell morphology and function revealed no significant differences in hair cell numbers or ribbon synapses between Tnfaip6 cKO and wild-type mice. Moreover, there were no notable variations in hair cell numbers or hearing function in noisy environments. Our results indicate that Tnfaip6 does not have a substantial impact on the auditory system.
Animals
;
Mice, Knockout
;
Hair Cells, Auditory, Inner/pathology*
;
Mice
;
Mice, Transgenic
;
Hearing Loss, Noise-Induced
;
Evoked Potentials, Auditory, Brain Stem/physiology*
10.Porphyromonas gingivalis potentiates stem-like properties of oral squamous cell carcinoma by modulating SCD1-dependent lipid synthesis via NOD1/KLF5 axis.
Wenli ZANG ; Fengxue GENG ; Junchao LIU ; Zengxu WANG ; Shuwei ZHANG ; Yuchao LI ; Ze LU ; Yaping PAN
International Journal of Oral Science 2025;17(1):15-15
Cancer stem cells (CSCs) are widely acknowledged as primary mediators to the initiation and progression of tumors. The association between microbial infection and cancer stemness has garnered considerable scholarly interest in recent years. Porphyromonas gingivalis (P. gingivalis) is increasingly considered to be closely related to the development of oral squamous cell carcinoma (OSCC). Nevertheless, the role of P. gingivalis in the stemness of OSCC cells remains uncertain. Herein, we showed that P. gingivalis was positively correlated with CSC markers expression in human OSCC specimens, promoted the stemness and tumorigenicity of OSCC cells, and enhanced tumor formation in nude mice. Mechanistically, P. gingivalis increased lipid synthesis in OSCC cells by upregulating the expression of stearoyl-CoA desaturase 1 (SCD1) expression, a key enzyme involved in lipid metabolism, which ultimately resulted in enhanced acquisition of stemness. Moreover, SCD1 suppression attenuated P. gingivalis-induced stemness of OSCC cells, including CSCs markers expression, sphere formation ability, chemoresistance, and tumor growth, in OSCC cells both in vitro and in vivo. Additionally, upregulation of SCD1 in P. gingivalis-infected OSCC cells was associated with the expression of KLF5, and that was modulated by P. gingivalis-activated NOD1 signaling. Taken together, these findings highlight the importance of SCD1-dependent lipid synthesis in P. gingivalis-induced stemness acquisition in OSCC cells, suggest that the NOD1/KLF5 axis may play a key role in regulating SCD1 expression and provide a molecular basis for targeting SCD1 as a new option for attenuating OSCC cells stemness.
Porphyromonas gingivalis/pathogenicity*
;
Stearoyl-CoA Desaturase/metabolism*
;
Humans
;
Carcinoma, Squamous Cell/pathology*
;
Mouth Neoplasms/metabolism*
;
Animals
;
Neoplastic Stem Cells/microbiology*
;
Mice, Nude
;
Mice
;
Nod1 Signaling Adaptor Protein/metabolism*
;
Kruppel-Like Transcription Factors/metabolism*
;
Cell Line, Tumor

Result Analysis
Print
Save
E-mail