1
|
Zhu X, Mao Z, Zheng P, Wang L, Zhang F, Zi G, Liu H, Zhang H, Liu W, Zhou L. The role and research progress of epigenetic modifications in obstructive sleep apnoea-hypopnea syndrome and related complications. Respir Med 2025; 242:108099. [PMID: 40228610 DOI: 10.1016/j.rmed.2025.108099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 04/10/2025] [Accepted: 04/11/2025] [Indexed: 04/16/2025]
Abstract
Epigenetic modifications are heritable changes in gene expression that do not alter the DNA sequence. Histone modifications, non-coding RNA expression, and DNA methylation are examples of common epigenetic changes. Obstructive sleep apnoea-hypopnea syndrome (OSAHS) is the most common sleep-related breathing disorder, and its incidence is increasing annually, making it a hotspot of clinical research and significantly impacting health and well-being. The main cause of OSAHS is related to complications caused by repeated chronic intermittent hypoxia (CIH). Currently, polysomnography (PSG) and continuous positive airway pressure (CPAP) remain the gold standards for the diagnosis and treatment of OSAHS. However, their limitations-such as time consumption, high cost, and poor patient comfort-contribute to the paradox of high disease prevalence yet low rates of diagnosis and treatment, resulting in a substantial disease burden. In recent years, rapid advances in epigenetics have revealed that biomarkers such as microRNAs (miRNAs), circular RNAs (circRNAs), and other epigenetic modifications hold promise as non-invasive tools for the diagnosis and treatment of OSAHS and its related complications. Although numerous studies have explored epigenetic modifications in other diseases, this study focuses on how epigenetic modifications participate in the process of OSAHS and its related complications, with an aim of elucidating the pathogenesis of OSAHS from an epigenetic perspective and provide new directions for identifying molecular targets for the diagnosis and treatment of OSAHS and related complications.
Collapse
Affiliation(s)
- Xiaoyan Zhu
- Department of Respiratory and Critical Care Medicine, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhenyu Mao
- Department of Respiratory and Critical Care Medicine, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Pengdou Zheng
- Department of Respiratory and Critical Care Medicine, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lingling Wang
- Department of Respiratory and Critical Care Medicine, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fengqin Zhang
- Department of Respiratory and Critical Care Medicine, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Guisha Zi
- Department of Respiratory and Critical Care Medicine, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Huiguo Liu
- Department of Respiratory and Critical Care Medicine, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Huojun Zhang
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Wei Liu
- Department of Geriatrics, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, Hubei 430030, China.
| | - Ling Zhou
- Department of Respiratory and Critical Care Medicine, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
2
|
Wusiman R, Haimiti S, Abuduaini H, Yang M, Wang Y, Gu M, Sailike A, Gao L. Increased SUMO-activating enzyme subunit 1 promotes glycolysis and fibrotic phenotype of diabetic nephropathy. Biochem Pharmacol 2025; 237:116920. [PMID: 40194606 DOI: 10.1016/j.bcp.2025.116920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 02/21/2025] [Accepted: 04/02/2025] [Indexed: 04/09/2025]
Abstract
Renal fibrosis is a prominent feature of diabetic nephropathy (DN), and the connection between renal fibrosis and abnormal glycolysis is not fully understood. SUMO-activating enzyme subunit 1 (SAE1) plays a crucial role in the SUMO modification process and is related to abnormal glycolysis. Despite this, the specific role of SAE1 in DN and its mechanism are not well defined. To investigate this, a streptozotocin-induced diabetic CD1 mice model was used, with SAE1 suppression achieved through systemic administration of SAE1 siRNA. In parallel, human renal proximal tubular tubule HK2 cells transfected with siSAE1 were exposed to high glucose for in vitro studies. The study revealed that SAE1 levels were elevated in diabetic kidney, and the deletion of SAE1 mitigated renal fibrosis in DN mice. Such suppression in SAE1 was associated with the lower expression of hypoxia inducible factor-1α (HIF-1α) and 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3), these alterations subsequently improved abnormal glycolysis and mesenchymal transformations in vivo and in vitro. Further experiments discovered that SAE1 stabilized transcription factor HIF-1α expression through SUMOylation, promoting PFKFB3 transcription, which enhanced glycolysis characterized by increased PFK1 activity and lactate production. Additionally, pharmacological inhibition of PFKFB3 reduced renal fibrosis in DN mice, while overexpression of PFKFB3 partly restored the glycolysis and mesenchymal transformations inhibited by SAE1 knockdown in vitro. These data demonstrate that SAE1 promotes abnormal glycolysis by HIF-1α/PFKFB3 which is responsible for the fibrotic phenotype of diabetic kidney. Inhibition of SAE1 could be an alternative strategy in combating diabetes associated-kidney fibrosis via improving aberrant glycolysis.
Collapse
Affiliation(s)
- Reziwanguli Wusiman
- Department of Endocrinology and Metabolic Diseases, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, PR China
| | - Shayila Haimiti
- Department of Endocrinology and Metabolic Diseases, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, PR China
| | - Hanikezi Abuduaini
- Department of Endocrinology and Metabolic Diseases, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, PR China
| | - Miaoyan Yang
- Department of Endocrinology and Metabolic Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China
| | - Yitian Wang
- School of Basic Medicine, Ningxia Medical University, Yinchuan, PR China
| | - Meijun Gu
- Department of General Medicine, Bainiaohu Hospital, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, PR China
| | - Ali Sailike
- Department of General Medicine, Bainiaohu Hospital, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, PR China
| | - Lei Gao
- Department of Endocrinology and Metabolic Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China; Department of General Medicine, Bainiaohu Hospital, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, PR China.
| |
Collapse
|
3
|
Wang Y, Wu W, Xu Y, Wu C, Han Q, Lu T, Zhang H, Jiao L, Zhang Y, Liu B, Yu XY, Li Y. Ncl liquid-liquid phase separation and SUMOylation mediate the stabilization of HIF-1α expression and promote pyroptosis in ischemic hindlimb. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167706. [PMID: 39933290 DOI: 10.1016/j.bbadis.2025.167706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 01/02/2025] [Accepted: 02/03/2025] [Indexed: 02/13/2025]
Abstract
Liquid-liquid phase separation (LLPS) has emerged as a flexible intracellular compartment that modulates various pathological processes. Hypoxia-inducible factor-1α (HIF-1α) has been shown to play an essential role in inflammation after ischemic injury. However, the mechanisms underlying HIF-1α-induced inflammation in ischemic diseases have not been defined. This study found that HIF-1α mediated the progression of ischemia-induced muscle injury. After ischemic injury, SUMO1 is upregulated and rapidly activates NLRP3 inflammasome through the upregulation of HIF-1α, leading to enhanced inflammation and pyroptosis. Co-IP revealed an interaction between SUMO1 and HIF-1α and SUMOylation of HIF-1α at K477. Moreover, we demonstrated the important role of dynamic phase separation of Nucleolin (Ncl) in regulating HIF-1α mRNA stability through fluorescence recovery after photobleach (FRAP) analysis. The stability of HIF-1α is regulated by Ncl liquid-liquid phase separation and SUMOylation in ischemia-induced hindlimb injury. HIF-1α can promote the expression of NLRP3 and other inflammation-related molecules, leading to pyroptosis, suggesting that Ncl/LLPS/HIF-1α or SUMO1/HIF-1α pathway may be a new target for the treatment of inflammation in ischemic diseases. Although previous studies have found that HIF-1α is able to promote the expression of target genes after hypoxia, and these genes are used to maintain the stability of the intracellular environment to adapt to hypoxia. We found that HIF-1α is involved in the activation process of NLRP3 inflammasomes after hind limb ischemia, which enriches our understanding of the biological role of HIF-1α.
Collapse
Affiliation(s)
- Yanli Wang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Weiliang Wu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Yan Xu
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China
| | - Chengjie Wu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Qingfang Han
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Tonggan Lu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Huiling Zhang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Lijuan Jiao
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Yu Zhang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Bin Liu
- Department of Cardiology, the Second Hospital of Jilin University, Changchun, Jilin 130041, PR China
| | - Xi-Yong Yu
- NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Yangxin Li
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, PR China.
| |
Collapse
|
4
|
Qi H, Gao H, Li M, Sun T, Gu X, Wei L, Zhi M, Li Z, Fu D, Liu Y, Wei Z, Dou Y, Feng Q. Parvimonas micra promotes oral squamous cell carcinoma metastasis through TmpC-CKAP4 axis. Nat Commun 2025; 16:2305. [PMID: 40055343 PMCID: PMC11889085 DOI: 10.1038/s41467-025-57530-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 02/25/2025] [Indexed: 03/12/2025] Open
Abstract
Parvimonas micra (P. micra), an opportunistic oral pathogen associated with multiple cancers, has limited research on its role in oral squamous cell carcinoma (OSCC). This study shows that P. micra is enriched in OSCC tissues and positively correlated with tumor metastasis and stages. P. micra infection promotes OSCC metastasis by inducing hypoxia/HIF-1α, glycolysis, and autophagy. Mechanistically, P. micra surface protein TmpC binds to CKAP4, a receptor overexpressed in OSCC, facilitating bacterial attachment and invasion. This interaction activates HIF-1α and autophagy via CKAP4-RanBP2 and CKAP4-NBR1 pathways, driving metastasis. Targeting CKAP4 with masitinib or antibodies impairs P. micra attachment and abolishes P. micra-promoted OSCC metastasis in vitro and in vivo. Together, our findings identify P. micra as a pathogen that promotes OSCC metastasis and highlight that TmpC-CKAP4 interaction could be a potential therapeutic target for OSCC.
Collapse
Affiliation(s)
- Houbao Qi
- Department of Human Microbiome, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| | - Haiting Gao
- Department of Human Microbiome, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| | - Meihui Li
- Department of Human Microbiome, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| | - Tianyong Sun
- Department of Human Microbiome, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| | - Xiufeng Gu
- Department of Human Microbiome, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| | - Li Wei
- Department of Human Microbiome, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| | - Mengfan Zhi
- Department of Human Microbiome, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| | - Zixuan Li
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, Nanjing, 210093, China
| | - Dachuan Fu
- Department of Human Microbiome, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| | - Yiran Liu
- Department of Endocrinology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Ziyi Wei
- Department of Human Microbiome, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| | - Yu Dou
- Department of Human Microbiome, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| | - Qiang Feng
- Department of Human Microbiome, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China.
| |
Collapse
|
5
|
Yang C, Zhong H. Multiomics Analyses Demonstrate the Attenuation of Metabolic Cardiac Disorders Associated With Type 2 Diabetes by Stachydrine in Relation With the Transition of Gastrointestinal Microbiota. Chem Biol Drug Des 2025; 105:e70066. [PMID: 39994907 DOI: 10.1111/cbdd.70066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 01/20/2025] [Accepted: 01/31/2025] [Indexed: 02/26/2025]
Abstract
Stachydrine (STA) has therapeutic effects on heart disorders. The current study assessed its effects on Type 2 diabetes (T2D) induced cardiac disorders by focusing on the heart-gut axis. Mice were subjected to high-fat diet (HFD) and streptozocin (STZ) to induce cardiac disorders such as inflammation and structural deteriorations, which were handled with STA. Changes regarding the composition and metabolism of gastrointestinal (GI) microbiota were then determined using a multiomics strategy, including amplicon sequencing and metabolomics. The data showed that STA improved heart function, reduced intestinal permeability, and suppressed inflammation in mice in a dose-dependent manner. However, the compound had little influence on the overall alpha diversity of gut microbiota, while it did influence the beta diversity. The analyses based on the multiomics strategy demonstrated that certain GI microbial groups, including Paramuribaculum, Allobaculum, Bifidobacterium, and Adlercreutzia, responded to the STA administration, which contributed to the alternatives of metabolites in the gut. Correlation analyses showed that Duncaniella and Ruminococcus negatively impacted health, while Muribaculum, Paramuribaculum, and Prevotella positively influenced intestinal permeability and heart health. Collectively, STA attenuated T2D-induced cardiac disorders by improving heart structure and function and suppressing inflammation, during which the GI homeostasis of the T2D mice changed to an alternative state that was different from that of healthy mice.
Collapse
Affiliation(s)
- Chaoxing Yang
- Department of Cardiovascular Medicine, Ganzhou People's Hospital, Ganzhou, China
| | - Huaping Zhong
- Heart Center, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| |
Collapse
|
6
|
Liu A, Zhang Y, Lin Y, Li X, Wang S, Pu W, Liu X, Jiang Z, Xiao Z. A rat model of adenoid hypertrophy constructed by using ovalbumin and lipopolysaccharides to induce allergy, chronic inflammation, and chronic intermittent hypoxia. Animal Model Exp Med 2025; 8:353-362. [PMID: 38572767 PMCID: PMC11871125 DOI: 10.1002/ame2.12396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 01/18/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND Adenoid hypertrophy (AH) is a common pediatric disease that significantly impacts the growth and quality of life of children. However, there is no replicable and valid model for AH. METHODS An AH rat model was developed via comprehensive allergic sensitization, chronic inflammation induction, and chronic intermittent hypoxia (CIH). The modeling process involved three steps: female Sprague-Dawley rats (aged 4-5 weeks) were used for modeling. Allergen sensitization was induced via intraperitoneal administration and intranasal provocation using ovalbumin (OVA); chronic nasal inflammation was induced through intranasal lipopolysaccharide (LPS) administration for sustained nasal irritation; CIH akin to obstructive sleep apnea/hypopnea syndrome was induced using an animal hypoxia chamber. Postmodel establishment, behaviors, and histological changes in nasopharynx-associated lymphoid tissue (NALT) and nasal mucosa were assessed. Arterial blood gas analysis and quantification of serum and tissue levels of (interleukin) IL-4 and IL-13, OVA-specific immunoglobulin E (sIgE), eosinophil cationic protein (ECP), tumor necrosis factor (TNF-α), IL-17, and transforming growth factor (TGF)-β were conducted for assessment. The treatment group received a combination of mometasone furoate and montelukast sodium for a week and then was evaluated. RESULTS Rats exhibited notable nasal symptoms and hypoxia after modeling. Histopathological analysis revealed NALT follicle hypertrophy and nasal mucosa inflammatory cell infiltration. Elevated IL-4, IL-13, IL-17, OVA-sIgE, ECP, and TNF-α levels and reduced TGF-β levels were observed in the serum and tissue of model-group rats. After a week of treatment, the treatment group exhibited symptom and inflammatory factor improvement. CONCLUSION The model effectively simulates AH symptoms and pathological changes. But it should be further validated for genetic, immunological, and hormonal backgrounds in the currently used and other strains and species.
Collapse
Affiliation(s)
- Anqi Liu
- Department of PediatricsLonghua Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Yixing Zhang
- Department of PediatricsLishui Hospital of Traditional Chinese MedicineLishuiChina
| | - Yan Lin
- Department of PediatricsLonghua Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Xuejun Li
- Department of PediatricsLonghua Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Shuming Wang
- Department of PediatricsLonghua Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Wenyan Pu
- Department of PediatricsLonghua Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Xiuxiu Liu
- Department of PediatricsLonghua Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Zhiyan Jiang
- Department of PediatricsLonghua Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Zhen Xiao
- Department of PediatricsLonghua Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| |
Collapse
|
7
|
Ugur F, Topal K, Albayrak M, Taskin R, Topal M. Preliminary Investigation into the Association between Scoliosis and Hypoxia: A Retrospective Cohort Study on the Impact of Eliminating Hypoxic Factors on Scoliosis Outcomes. CHILDREN (BASEL, SWITZERLAND) 2024; 11:1134. [PMID: 39334666 PMCID: PMC11429953 DOI: 10.3390/children11091134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 09/06/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024]
Abstract
OBJECTIVE This study delves into the implications of adenoidectomy for scoliosis progression, investigating the intricate nexus of hypoxia, spinal curvature, and surgical intervention. With adenoidectomy being a common procedure for addressing pediatric sleep-disordered breathing, this research study explores its potential impact on spinal health. PATIENTS AND METHODS Employing a retrospective cohort design, this study gathered data from patients who underwent adenoidectomy, including those with scoliosis, between January 2017 and March 2023. Initial and follow-up evaluations involved clinical and radiological assessments, notably measuring the Cobb angle to quantify spinal curvature. RESULTS This study enrolled 218 patients under 10 years old. Among them, 18 exhibited Cobb angles of 10° or more, with a mean Cobb angle of 12.8°. In the follow-up evaluation, 83% of patients with initial Cobb angles of 10° or more were reached out to, along with 84.6% of those with Cobb angles below 10°. The postoperative follow-up revealed a notable decrease in Cobb angles for most patients, particularly those with an initial Cobb angle exceeding 10°. CONCLUSIONS This study underscores the potential connection between adenoidectomy, hypoxia, and scoliosis regression, highlighting the importance of early intervention for scoliosis management. Despite certain limitations, this investigation lays the foundation for future research involving larger patient cohorts and multifaceted analyses. The observed interactions between airway function, hypoxia, and spinal health open avenues for refining clinical strategies in scoliosis treatment.
Collapse
Affiliation(s)
- Fatih Ugur
- Department of Orthopaedics and Traumatology, Kastamonu University Medical Faculty, 37100 Kastamonu, Turkey; (R.T.); (M.T.)
| | - Kubra Topal
- Department of Otorhinolaryngology, Private Clinic, 37100 Kastamonu, Turkey;
| | - Mehmet Albayrak
- Department of Orthopaedics and Traumatology, Private Practice, 59000 Tekirdag, Turkey;
| | - Recep Taskin
- Department of Orthopaedics and Traumatology, Kastamonu University Medical Faculty, 37100 Kastamonu, Turkey; (R.T.); (M.T.)
| | - Murat Topal
- Department of Orthopaedics and Traumatology, Kastamonu University Medical Faculty, 37100 Kastamonu, Turkey; (R.T.); (M.T.)
| |
Collapse
|
8
|
Ding GY, Tan WM, Lin YP, Ling Y, Huang W, Zhang S, Shi JY, Luo RK, Ji Y, Wang XY, Zhou J, Fan J, Cai MY, Yan B, Gao Q. Mining the interpretable prognostic features from pathological image of intrahepatic cholangiocarcinoma using multi-modal deep learning. BMC Med 2024; 22:282. [PMID: 38972973 PMCID: PMC11229270 DOI: 10.1186/s12916-024-03482-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 06/13/2024] [Indexed: 07/09/2024] Open
Abstract
BACKGROUND The advances in deep learning-based pathological image analysis have invoked tremendous insights into cancer prognostication. Still, lack of interpretability remains a significant barrier to clinical application. METHODS We established an integrative prognostic neural network for intrahepatic cholangiocarcinoma (iCCA), towards a comprehensive evaluation of both architectural and fine-grained information from whole-slide images. Then, leveraging on multi-modal data, we conducted extensive interrogative approaches to the models, to extract and visualize the morphological features that most correlated with clinical outcome and underlying molecular alterations. RESULTS The models were developed and optimized on 373 iCCA patients from our center and demonstrated consistent accuracy and robustness on both internal (n = 213) and external (n = 168) cohorts. The occlusion sensitivity map revealed that the distribution of tertiary lymphoid structures, the geometric traits of the invasive margin, the relative composition of tumor parenchyma and stroma, the extent of necrosis, the presence of the disseminated foci, and the tumor-adjacent micro-vessels were the determining architectural features that impacted on prognosis. Quantifiable morphological vector extracted by CellProfiler demonstrated that tumor nuclei from high-risk patients exhibited significant larger size, more distorted shape, with less prominent nuclear envelope and textural contrast. The multi-omics data (n = 187) further revealed key molecular alterations left morphological imprints that could be attended by the network, including glycolysis, hypoxia, apical junction, mTORC1 signaling, and immune infiltration. CONCLUSIONS We proposed an interpretable deep-learning framework to gain insights into the biological behavior of iCCA. Most of the significant morphological prognosticators perceived by the network are comprehensible to human minds.
Collapse
Affiliation(s)
- Guang-Yu Ding
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, No.180, Feng Lin Road, Shanghai, 200032, China
| | - Wei-Min Tan
- School of Computer Science, Shanghai Key Laboratory of Intelligent Information Processing, Fudan University, No.2005, Song Hu Road, Shanghai, 200433, China
| | - You-Pei Lin
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, No.180, Feng Lin Road, Shanghai, 200032, China
| | - Yu Ling
- School of Computer Science, Shanghai Key Laboratory of Intelligent Information Processing, Fudan University, No.2005, Song Hu Road, Shanghai, 200433, China
| | - Wen Huang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Shu Zhang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, No.180, Feng Lin Road, Shanghai, 200032, China
| | - Jie-Yi Shi
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, No.180, Feng Lin Road, Shanghai, 200032, China
| | - Rong-Kui Luo
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yuan Ji
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiao-Ying Wang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, No.180, Feng Lin Road, Shanghai, 200032, China
| | - Jian Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, No.180, Feng Lin Road, Shanghai, 200032, China
- Institute of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Jia Fan
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, No.180, Feng Lin Road, Shanghai, 200032, China
- Institute of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Mu-Yan Cai
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, No.651 Dongfeng Road East, Guangzhou, 510060, China.
| | - Bo Yan
- School of Computer Science, Shanghai Key Laboratory of Intelligent Information Processing, Fudan University, No.2005, Song Hu Road, Shanghai, 200433, China.
| | - Qiang Gao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, No.180, Feng Lin Road, Shanghai, 200032, China.
- Institute of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
- State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200433, China.
| |
Collapse
|
9
|
Yi H, Zhu B, Zheng C, Ying Z, Cheng M. CXCL13/CXCR5 promote chronic postsurgical pain and astrocyte activation in rats by targeting NLRP3. Neuroreport 2024; 35:406-412. [PMID: 38526919 DOI: 10.1097/wnr.0000000000002023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
Chronic postsurgical pain (CPSP) with high incidence negatively impacts the quality of life. X-C motif chemokine 13 (CXCL13) has been associated with postsurgery inflammation and exacerbates neuropathic pain in patients with CPSP. This study was aimed to illustrate the relationship between CXCL13 and nod-like receptor protein-3 (NLRP3), which is also involved in CPSP. A CPSP model was constructed by skin/muscle incision and retraction (SMIR) in right medial thigh, and the rats were divided into three groups: Sham, SMIR, and SMIR + anti-CXCL13 (intrathecally injected with anti-CXCL13 antibody). Then, the paw withdrawal threshold (PWT) score of rats was recorded. Primary rat astrocytes were isolated and treated with recombinant protein CXCL13 with or without NLRP3 inhibitor INF39. The expressions of CXCL13, CXCR5, IL-1β, IL-18, GFAP, NLRP3, and Caspase-1 p20 were detected by real-time quantitative reverse transcription PCR, western blot, ELISA, immunocytochemistry, and immunofluorescence analyses. The anti-CXCL13 antibody alleviated SMIR-induced decreased PWT and increased expression of GFAP, CXCL13, CXCR5, NLRP3, and Caspase-1 p20 in spinal cord tissues. The production of IL-1β, IL-18, and expression of CXCL13, CXCR5, GFAP, NLRP3, and Caspase-1 p20 were increased in recombinant protein CXCL13-treated primary rat astrocytes in a dose-dependent manner. Treatment with NLRP3 inhibitor INF39 inhibited the function of recombinant protein CXCL13 in primary rat astrocytes. The CXCL13/CXCR5 signaling could promote neuropathic pain, astrocytes activation, and NLRP3 inflammasome activation in CPSP model rats by targeting NLRP3. NLRP3 may be a potential target for the management of CPSP.
Collapse
Affiliation(s)
- Hongda Yi
- Department of Anesthesiology, Hangzhou Women's Hospital, Hangzhou, China
| | | | | | | | | |
Collapse
|
10
|
Kang L, Wang X, Wang J, Guo J, Zhang W, Lei R. SENP1 knockdown-mediated CTCF SUMOylation enhanced its stability and alleviated lipopolysaccharide-evoked inflammatory injury in human lung fibroblasts via regulation of FOXA2 transcription. Biochim Biophys Acta Gen Subj 2024; 1868:130500. [PMID: 37914145 DOI: 10.1016/j.bbagen.2023.130500] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 10/24/2023] [Accepted: 10/27/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND Excessive inflammation is the main cause of treatment failure in neonatal pneumonia (NP). CCCTC-binding factor (CTCF) represents an important node in various inflammatory diseases. In the present study, we tried to clarify the function and underlying molecular mechanism of CTCF on an in vitro cellular model of NP, which was generated by simulating the human lung fibroblast cell line WI-38 with lipopolysaccharide (LPS). METHODS The SUMOylation level and protein interaction were verified by Co-immunoprecipitation assay. Cell viability was measured by Cell Counting Kit-8 assay. Inflammatory factors were examined by Enzyme-linked immunosorbent assay. Cell apoptosis was evaluated by TUNEL assay. The binding activity of CTCF to target promoter was tested by chromatin immunoprecipitation and luciferase reporter assay. RESULTS LPS treatment restrained cell viability, promoted the production of inflammatory factors, and enhanced cell apoptosis. CTCF overexpression played anti-inflammatory and anti-apoptotic roles. Furthermore, CTCF was modified by SUMOylation with small ubiquitin-like modifier protein 1 (SUMO1). Interfering with sumo-specific protease 1 (SENP1) facilitated CTCF SUMOylation and protein stability, thus suppressing LPS-evoked inflammatory and apoptotic injuries. Moreover, CTCF could bind to the forkhead box protein A2 (FOXA2) promoter region to promote FOXA2 expression. The anti-inflammatory and anti-apoptotic roles of CTCF are associated with FOXA2 activation. In addition, SENP1 knockdown increased FOXA2 expression by enhancing the abundance and binding ability of CTCF. CONCLUSIONS SUMOylation of CTCF by SENP1 knockdown enhanced its protein stability and binding ability and it further alleviated LPS-evoked inflammatory injury in human lung fibroblasts by positively regulating FOXA2 transcription.
Collapse
Affiliation(s)
- Le Kang
- Neonatal Intensive Care Unit, Zhumadian Central Hospital, 463100 Zhumadian, Henan Province, China.
| | - Xinhua Wang
- Neonatal Intensive Care Unit, Zhumadian Central Hospital, 463100 Zhumadian, Henan Province, China
| | - Jianfang Wang
- Department of Clinical Laboratory, Zhumadian Central Hospital, 463100 Zhumadian, Henan Province, China
| | - Jing Guo
- Neonatal Intensive Care Unit, Henan Children's Hospital, 450000 Zhengzhou, Henan Province, China
| | - Wang Zhang
- Neonatal Intensive Care Unit, Zhumadian Central Hospital, 463100 Zhumadian, Henan Province, China
| | - Ruirui Lei
- Department of Neonatology, Zhumadian Central Hospital, 463100 Zhumadian, Henan Province, China
| |
Collapse
|
11
|
Ugur F, Topal K, Albayrak M, Topal M. Is Obstructive Sleep Apnea-Associated Adenoid Hypertrophy Linked to Scoliotic Attitudes in Children? Cureus 2023; 15:e47307. [PMID: 37869050 PMCID: PMC10585604 DOI: 10.7759/cureus.47307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2023] [Indexed: 10/24/2023] Open
Abstract
Introduction Scoliosis, a multifaceted spinal deformity commonly affecting pediatric and adolescent populations, has spurred extensive scientific inquiry to understand its origins and impacts. Early-onset scoliosis (EOS), characterized by spinal curvature exceeding 10° before the age of 10, presents a unique challenge necessitating a comprehensive understanding of its etiological factors. Within this context, the potential role of hypoxia-induced by adenoid hypertrophy in contributing to the pathogenesis of EOS has emerged as an intriguing avenue of investigation. Materials and methods This retrospective study was conducted focusing on radiological and clinical data pertaining to children below 10 years of age who underwent isolated adenoidectomy for adenoid hypertrophy. Preoperative posteroanterior standing chest radiographs were utilized for scoliosis assessment, with Cobb angles serving as the primary measurement metric. To ensure accuracy and reliability, Cobb angle measurements were independently performed by two experienced observers. Statistical analyses encompassed the Mann-Whitney U test, Spearman correlation analysis, and intraclass correlation coefficient calculations to evaluate interobserver agreement. Results Among the cohort of 218 pediatric adenoidectomy patients, 177 individuals had radiographs suitable for EOS evaluation. The mean age of the participants was 5.72±2 years, with a nearly equal distribution of 52.5% male and 47.5% female patients. Strikingly, the study identified a 10.2% prevalence of coronal plane curvatures exceeding the critical threshold of 10°, indicative of EOS. The robust interobserver reliability was demonstrated by a commendable mean interclass correlation coefficient (ICC) value of 0.926, affirming consistent and accurate Cobb angle measurements between the observers. Conclusion In light of the heightened prevalence of EOS observed in children undergoing adenoidectomy, this study provides a compelling impetus for exploring the potential interrelationship between adenoid hypertrophy, hypoxia, and the emergence of early-onset scoliosis. The study underscores the importance of prospective research to elucidate the complex mechanisms connecting these factors, offering insights into potential risk factors and underlying pathogenic pathways associated with the development of early-onset scoliosis.
Collapse
Affiliation(s)
- Fatih Ugur
- Orthopedics and Traumatology, Faculty of Medicine, Kastamonu University, Kastamonu, TUR
| | - Kubra Topal
- Otolaryngology - Head and Neck Surgery, Private Practice, Kastamonu, TUR
| | - Mehmet Albayrak
- Orthopedics and Traumatology, Ozel Tekirdag Yasam Hospital, Tekirdag, TUR
| | - Murat Topal
- Orthopedics and Traumatology, Faculty of Medicine, Kastamonu University, Kastamonu, TUR
| |
Collapse
|
12
|
Yu J, Liu X, Ji H, Zhang Y, Zhan H, Zhang Z, Wen J, Wang Z. Distribution of serum uric acid concentration and its association with lipid profiles: a single-center retrospective study in children aged 3 to 12 years with adenoid and tonsillar hypertrophy. Lipids Health Dis 2023; 22:48. [PMID: 37024876 PMCID: PMC10077755 DOI: 10.1186/s12944-023-01806-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 03/14/2023] [Indexed: 04/08/2023] Open
Abstract
BACKGROUND Presently, there is no consensus regarding the optimal serum uric acid (SUA) concentration for pediatric patients. Adenoid and tonsillar hypertrophy is considered to be closely associated with pediatric metabolic syndrome and cardiovascular risk and is a common condition in children admitted to the hospital. Therefore, we aimed to evaluate the relationship between SUA and dyslipidemia and propose a reference range for SUA concentration that is associated with a healthy lipid profile in hospitalized children with adenoid and tonsillar hypertrophy. METHODS Preoperative data from 4922 children admitted for elective adenoidectomy and/or tonsillectomy surgery due to adenoid and tonsillar hypertrophy were collected. SUA concentrations were scaled to standard deviation (SD), and SUA deviations were expressed as SD from the mean SUA of children without dyslipidemia. RESULTS The mean SUA concentration of the participants was 4.27 ± 1.01 mg/dL, and the prevalence of hyperuricemia was 1.6% when it was defined using an SUA of ≥ 7.0 mg/dL. Participants with dyslipidemia (856, 17.4%) had a higher prevalence of hyperuricemia (3.4% vs. 1.2%, P < 0.001) and higher SUA concentrations (4.51 ± 1.15 vs. 4.22 ± 0.97 mg/dL, P < 0.001) than those with ortholiposis. The circulating lipid status of participants with SUAs < 1 SD below the mean value for the participants with ortholiposis (range 1.80-3.28 mg/dL) was more normal. Each 1-SD increase in SUA was associated with a 27% increase in the risk of dyslipidemia (OR = 1.270, 95% CI, 1.185-1.361). Adjustment for a number of potential confounders reduced the strength of the relationship, but this remained significant (OR = 1.125, 95% CI, 1.042-1.215). The higher risk of dyslipidemia was maintained for participants with SUAs > 1 SD above the mean value of the participants with ortholiposis. CONCLUSIONS SUA was independently associated with dyslipidemia in children with adenoid and tonsillar hypertrophy, and an SUA < 1 SD below the mean value for patients with ortholiposis was associated with a healthy lipid profile.
Collapse
Affiliation(s)
- Jiating Yu
- Henan Joint International Pediatric Urodynamic Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xin Liu
- Department of Clinical Laboratory, Key Clinical Laboratory of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Honglei Ji
- NHC Key Laboratory of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Yawei Zhang
- Department of Endocrinology and Metabolic Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hanqiang Zhan
- Department of Medical Record Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ziyin Zhang
- Department of Information, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jianguo Wen
- Henan Joint International Pediatric Urodynamic Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Zhimin Wang
- Department of Endocrinology and Metabolic Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
13
|
HUVECs affect HuT-78 cell apoptosis and cytokine production via the HIF-1α-PD-L1/PD-1 pathway under hypoxia. Int Immunopharmacol 2023; 118:110010. [PMID: 36924563 DOI: 10.1016/j.intimp.2023.110010] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 02/24/2023] [Accepted: 03/06/2023] [Indexed: 03/17/2023]
Abstract
We investigated whether human umbilical vein endothelial cells (HUVECs) under hypoxic conditions can suppress the production of cytokines in Hut-78 cells via the HIF-1α/PD-L1/PD-1 pathway, and the intervention effect of Nivolumab. HUVECs and HuT-78 cells were monocultured or cocultured in a tri-gas incubator with or without Nivolumab pretreatment. Real-time PCR, western blotting, and protein chips were used. Transcriptional regulation of PD-L1 and PD-1 by HIF-1α was analyzed by ChIP-qPCR and luciferase reporter gene assays. Apoptosis was assessed by flow cytometry. In HuT-78 cells, hypoxic monoculture significantly increased the expression of HIF-1α, PD-1, IL-2, IL-4, IL-6, IL-8, IL-10, TNF-α, IFN-α, and Bax, decreased the expression of Bcl-2, and resulted in increased apoptosis. In comparison to hypoxic monoculture, hypoxic coculture significantly reduced the expression of IL-2, IL-4, IL-6, IL-8, IL-10, TNF-α, and IFN-α, as well as Bcl-2, in HuT-78 cells. Meanwhile, Bax expression was significantly increased with elevated apoptosis in HuT-78 cells. However, pretreatment with Nivolumab significantly antagonized the reduction in cytokines and the elevation in apoptosis in HuT-78 cells. Chip-qPCR and luciferase reporter gene assays demonstrated that hypoxia significantly increased the binding of HIF-1α to the upstream regulatory regions of PD-1 at -63 and -66 bp and PD-L1 at -571 bp, promoting their transcription. Therefore, HUVECs under hypoxia can reduce cytokine production and inhibit their own apoptosis in co-culture with HuT-78 cells via the HIF-1α/PD-L1/PD-1 pathway. These findings provide new clues for exploring the combined use of immune checkpoint inhibitors and anti-angiogenic drugs in clinical settings.
Collapse
|
14
|
Cai H, Ma X, Lu D, Chen L, Bian X, Zhang N, Tang W, Liu X, Li Z. Mild Hypothermia Promotes Ischemic Tolerance and Survival of Neural Stem Cell Grafts by Enhancing Global SUMOylation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6503504. [PMID: 35669854 PMCID: PMC9166982 DOI: 10.1155/2022/6503504] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/07/2022] [Accepted: 04/18/2022] [Indexed: 11/17/2022]
Abstract
Cerebral infarct penumbra due to hypoxia and toxin accumulation is not conducive to the transplantation of neural stem cells (NSCs), although mild hypothermia can improve the local microenvironment of the ischemic penumbra and exert neuroprotective effects. However, insufficient understanding of the molecular mechanism by which mild hypothermia protects the brain limits widespread clinical application. This study evaluated the molecular mechanism of mild hypothermia-induced brain protection from the perspective of global protein small ubiquitin-like modifier (SUMO) modification, with the aim of improving NSC transplant survival rates in the penumbra to enhance neurological function. NSCs from neonatal rats were extracted to detect the effects of hypoxia and mild hypothermia on SUMOylation modification levels, cell stemness, and hypoxia-induced injury. Overexpression and knockdown of UBC9 in NSCs were used to evaluate their ability to maintain stemness and withstand hypoxic injury. Finally, a rat middle cerebral artery occlusion (MCAO) model was used to verify the effect of mild hypothermia treatment and UBC9 overexpression on neural function of NSCs following penumbra transplantation in rats. Results showed that hypoxia and mild hypothermia promoted both the SUMOylation modification and maintenance of NSC stemness. Overexpression of UBC9 enhanced the abilities of NSCs to maintain stemness and resist hypoxic injury, while UBC9 knockdown had the opposite effect. Following transplantation into the ischemic penumbra of MCAO model rats, mild hypothermia and Ubc9-overexpressing NSCs significantly reduced cerebral infarct areas and improved neurological function. In conclusion, this study demonstrated that global protein SUMOylation is an important molecular mechanism for NSCs to tolerate hypoxia, and mild hypothermia can further increase the degree of global SUMOylation to enhance the hypoxia tolerance of NSCs, which increases their survival during transplantation in situ and ability to perform nerve repair in the penumbra of cerebral infarction.
Collapse
Affiliation(s)
- Heng Cai
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China
- Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China
| | - Xiaofang Ma
- Central Laboratory, The Fifth Central Hospital of Tianjin, Tianjin 300450, China
- Tianjin Key Laboratory of Epigenetics for Organ Development of Preterm Infants, The Fifth Central Hospital of Tianjin, Tianjin 300450, China
| | - Dading Lu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China
- Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China
| | - Liangyu Chen
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China
- Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China
| | - Xiyun Bian
- Central Laboratory, The Fifth Central Hospital of Tianjin, Tianjin 300450, China
- Tianjin Key Laboratory of Epigenetics for Organ Development of Preterm Infants, The Fifth Central Hospital of Tianjin, Tianjin 300450, China
| | - Nan Zhang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China
- Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China
| | - Wei Tang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China
- Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China
| | - Xiaozhi Liu
- Central Laboratory, The Fifth Central Hospital of Tianjin, Tianjin 300450, China
- Tianjin Key Laboratory of Epigenetics for Organ Development of Preterm Infants, The Fifth Central Hospital of Tianjin, Tianjin 300450, China
| | - Zhiqing Li
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China
- Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China
| |
Collapse
|