1
|
Li J, Qiu Z, Xie R, Ma L, Tang L, Yao M, Zhang Z, Chen L, Zhang L. CXCL2 Mediates Lactate Secretion and Promotes Aortic Dissection Formation through the p38/JNK/C-myc Pathways. Thromb Haemost 2025. [PMID: 40398663 DOI: 10.1055/a-2600-6318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
Aortic dissection (AD) is a critical cardiovascular condition with high mortality risk, and elevated lactate levels are linked to increased postoperative mortality in AD patients. CXCL2 is recognized for its significant involvement in cardiovascular pathologies. Nevertheless, the mechanisms by which CXCL2 regulates lactate levels in AD remain to be fully elucidated.We assessed CXCL2, CXCR2, p38/JNK, C-myc, MCT1, and MCT4 expression using Western blotting, real-time PCR, and immunostaining on AD patient samples, mouse models, and human aortic vascular smooth muscle cells (HAVSMCs). Lactate levels were measured with a lactate assay kit, and we investigated CXCL2's role in AD through vessel incubation.CXCL2 and CXCR2 expression was significantly upregulated in AD, accompanied by increased lactate secretion. Overexpression of CXCL2 not only stimulated lactate secretion but also expedited AD progression. In contrast, CXCR2 inhibition effectively countered the effects of CXCL2. Furthermore, CXCL2 elevated the expression of MCT1 and MCT4; inhibition of them alleviated CXCL2-induced AD and lactate accumulation. CXCL2 also activated the p38 and JNK signaling pathways and upregulated C-myc expression, leading to enhanced lactate secretion in HAVSMCs. Blocking these pathways reversed the effects induced by CXCL2.The CXCL2-CXCR2 axis regulated lactate secretion and drove the progression of AD through the activation of the p38/JNK/C-myc signaling pathways. These findings provide new insights into the molecular mechanisms underlying AD and identify potential therapeutic targets for clinical intervention.
Collapse
Affiliation(s)
- Jiakang Li
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Pathophysiology, The School of Basic Medical Sciences, Fuzhou, China
- Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China
| | - Zhihuang Qiu
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China
| | - Rumei Xie
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Pathophysiology, The School of Basic Medical Sciences, Fuzhou, China
- Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China
| | - Likang Ma
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China
| | - Lele Tang
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China
| | - Mengge Yao
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Pathophysiology, The School of Basic Medical Sciences, Fuzhou, China
- Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China
| | - Zhaofeng Zhang
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China
| | - Liangwan Chen
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China
| | - Li Zhang
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Pathophysiology, The School of Basic Medical Sciences, Fuzhou, China
- Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China
| |
Collapse
|
2
|
Tao DL, Chen JM, Wu JP, Zhao SS, Qi BF, Yang X, Fan YY, Song JK, Zhao GH. Neospora caninum hijacks host PFKFB3-driven glycolysis to facilitate intracellular propagation of parasites. Vet Res 2025; 56:94. [PMID: 40307939 PMCID: PMC12042381 DOI: 10.1186/s13567-025-01524-w] [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: 12/16/2024] [Accepted: 02/13/2025] [Indexed: 05/02/2025] Open
Abstract
Infection with Neospora caninum leads to reproductive failure in ruminants, such as cattle and goats; however, no effective vaccines or treatments are currently available to control this infection. Carefully regulating the glycolysis of host cells is essential for the intracellular survival of pathogens. Nonetheless, the impact of N. caninum infection on host cell glycolysis and the effects and mechanisms of host cell glycolysis on the intracellular survival of this parasite remains unclear. In this study, the analysis of metabolomics and transcriptomics revealed that N. caninum infection increases the expression of glycolysis-related enzymes and lactate production in caprine endometrial epithelial cells (EECs). The study's findings demonstrate that the inhibition of host cell glycolysis using 2-DG or sodium oxamate (an LDH-A inhibitor) inhibits host cell glycolysis and the intracellular propagation of N. caninum tachyzoites. Moreover, the addition of lactate further promotes the replication of N. caninum tachyzoites both in vivo and in vitro. Further investigation found that N. caninum infection induces host cell glycolysis via up-regulating 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) expression, while knockdown of PFKFB3 with small-interfering RNA or 3-PO significantly inhibits host cell glycolysis and the propagation of N. caninum tachyzoites both in vivo and in vitro. Additionally, a mechanistic study showed that N. caninum infection activates the JNK signalling pathway and inhibits the ubiquitination degradation of HIF-1α. Chromatin immunoprecipitation and dual-luciferase reporter assays revealed that N. caninum infection induces the expression of HIF-1α, which binds to the promoter region of pfkfb3. Our findings indicate that cellular glycolysis may serve as a potential therapeutic target for neosporosis, offering a novel insight for further investigating the intracellular survival mechanisms of N. caninum.
Collapse
Affiliation(s)
- De-Liang Tao
- Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Shaanxi, Yangling, China
| | - Jin-Ming Chen
- Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Shaanxi, Yangling, China
| | - Jiang-Ping Wu
- Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Shaanxi, Yangling, China
| | - Shan-Shan Zhao
- Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Shaanxi, Yangling, China
| | - Bu-Fan Qi
- Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Shaanxi, Yangling, China
| | - Xin Yang
- Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Shaanxi, Yangling, China
| | - Ying-Ying Fan
- Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Shaanxi, Yangling, China
| | - Jun-Ke Song
- Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Shaanxi, Yangling, China
| | - Guang-Hui Zhao
- Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Shaanxi, Yangling, China.
| |
Collapse
|
3
|
Xu Y, Wang J, Yuan R, Qin Z, Long K, Gao P. Targeting the immuno-inflammatory-microbial network: a key strategy for sepsis treatment. Front Immunol 2025; 16:1575516. [PMID: 40297590 PMCID: PMC12034552 DOI: 10.3389/fimmu.2025.1575516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 03/27/2025] [Indexed: 04/30/2025] Open
Abstract
Sepsis is a life-threatening condition caused by a dysregulated host response to infection, remaining a major global health challenge despite clinical advances. Therapeutic challenges arise from antibiotic misuse, incomplete understanding of its complex pathophysiology, and the unresolved interplay of immune dysregulation and microbiota disruption. Investigating microbial homeostasis in the shift from cytokine storm to immunosuppression may elucidate the interplay between microbial metabolites, immune dysfunction, and organ injury, providing a foundation for targeted therapies and drug development. Traditional Chinese Medicine (TCM) has demonstrated significant advantages in mitigating sepsis-associated cytokine storms and modulating gut microbiota homeostasis, offering a promising strategy for developing highly effective and less toxic targeted monomeric compounds. Elucidating the interactions within the immune-inflammation-microbiota network in sepsis paves the way for biomarker-driven personalized therapeutic approaches.
Collapse
Affiliation(s)
- Yue Xu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | | | | | | | | | - Peiyang Gao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
4
|
Jeon S, Baek H, Kim S, Kim Y, Kim J, Kim JW. Microalgae-Derived Microparticles Improve Immunomodulation via Combined Glycolysis and MAPK Activation. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:8619-8626. [PMID: 40145572 DOI: 10.1021/acs.langmuir.4c05088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
Natural polysaccharides possess potent immunostimulatory properties, but their poor solubility impedes efficiency of cellular delivery. This study focuses on extraction of microparticles (MPs) fromEuglena gracilis, a microalgae species characterized by abundant intracellular β-1,3-glucan and flexible cell membrane. We introduce anE. gracilis-derived MP (MPEG) system as a natural carrier for solubilized β-glucan. The MPEG system enhances β-glucan's solubility and loading efficiency through sequential sonication and cell extrusion. In vitro studies reveal that MPEG utilizes multiple endocytosis pathways, including phagocytosis, clathrin-mediated, and lipid raft-mediated routes, for effective β-glucan delivery into cells. Upon cellular internalization, MPEG components trigger dual activation of the MAPK signaling pathway and glycolysis in macrophages, leading to enhanced production of pro-inflammatory cytokines and lactic acid, ultimately strengthening innate immune responses. This MPEG system offers a promising approach to harnessing the immunostimulatory properties of natural polysaccharides while overcoming their solubility limitations, opening new avenues for targeted cellular delivery in immunomodulation therapies.
Collapse
Affiliation(s)
- Saetbyeol Jeon
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Hwira Baek
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Seulgi Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Youngseok Kim
- Technology Innovation Center, Shinsegae International Inc., Seoul 06015, Republic of Korea
| | - Junoh Kim
- Technology Innovation Center, Shinsegae International Inc., Seoul 06015, Republic of Korea
| | - Jin Woong Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
5
|
He J, Li X, Yu H, Xu C, Tian R, Zhou P, Yin Z. Inflammation-induced PFKFB3-mediated glycolysis promoting myometrium contraction through the PI3K-Akt-mTOR pathway in preterm birth mice. Am J Physiol Cell Physiol 2025; 328:C895-C907. [PMID: 39907705 DOI: 10.1152/ajpcell.00704.2024] [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: 09/23/2024] [Revised: 10/21/2024] [Accepted: 01/06/2025] [Indexed: 02/06/2025]
Abstract
Inflammation is a significant risk factor for preterm birth. Inflammation enhances glycolytic processes in various cell types and contributes to the development of myometrial contractions. However, the potential of inflammation to activate glycolysis in pregnant murine uterine smooth muscle cells (mUSMCs) and its role in promoting inflammatory preterm birth remain unexplored. In this study, lipopolysaccharide was employed to establish both cell and animal inflammation models. We found that inflammation of mUSMCs during late pregnancy could initiate glycolysis and promote cell contraction. Subsequently, the inhibition of glycolysis using the glycolysis inhibitor 2-deoxyglucose can reverse inflammation-induced cell contraction. The expression of 6-phosphofructokinase 2 kinase (PFKFB3) was significantly upregulated in mUSMCs following lipopolysaccharide stimulation. In addition, lactate accumulation and enhanced contraction were observed. Inhibition of PFKFB3 reversed the lactate accumulation and enhanced contraction induced by inflammation. We also found that inflammation activated the phosphatidylinositol 3-kinase (PI3K)-protein kinase B (Akt)-mammalian target of the rapamycin (mTOR) pathway, leading to the upregulation of PFKFB3 expression. The PI3K-Akt pathway inhibitor LY294002 and the mTOR pathway inhibitor rapamycin effectively inhibited the upregulation of PFKFB3 protein expression, lactate production, and the enhancement of cell contraction induced by lipopolysaccharide. This study indicates that inflammation regulates PFKFB3 through the PI3K-Akt-mTOR pathway, which enhances the glycolytic process in pregnant mUSMCs, ultimately leading to myometrial contraction.NEW & NOTEWORTHY Expression of PFKFB3, a key enzyme in glycolysis, was significantly upregulated both in the mUSMCs and myometrium of mice during late pregnancy after lipopolysaccharide stimulation. Activation of the PI3K-Akt-mTOR pathway enhanced PFKFB3 expression, which is involved in the initiation of glycolysis. Inflammation-activated PFKFB3 via the PI3K-Akt-mTOR pathway, which enhances the cellular glycolytic process and thus promotes myometrium contraction in pregnancy.
Collapse
Affiliation(s)
- Jing He
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
- NHC Key Laboratory of the Study of Abnormal Gametes and the Reproductive Tract, Anhui Medical University, Hefei, People's Republic of China
- Department of Obstetrics and Gynecology, Anqing Medical Center of Anhui Medical University, Anqing, People's Republic of China
| | - Xuan Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
- NHC Key Laboratory of the Study of Abnormal Gametes and the Reproductive Tract, Anhui Medical University, Hefei, People's Republic of China
- Anhui Province Key Laboratory of Reproductive Disorders and Obstetrics and Gynaecology Diseases, Hefei, People's Republic of China
| | - Huihui Yu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
- NHC Key Laboratory of the Study of Abnormal Gametes and the Reproductive Tract, Anhui Medical University, Hefei, People's Republic of China
- Anhui Province Key Laboratory of Reproductive Disorders and Obstetrics and Gynaecology Diseases, Hefei, People's Republic of China
| | - Chenyi Xu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
- Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, Hefei, People's Republic of China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, People's Republic of China
| | - Ruixian Tian
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
- Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, Hefei, People's Republic of China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, People's Republic of China
| | - Ping Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
- NHC Key Laboratory of the Study of Abnormal Gametes and the Reproductive Tract, Anhui Medical University, Hefei, People's Republic of China
- Anhui Province Key Laboratory of Reproductive Disorders and Obstetrics and Gynaecology Diseases, Hefei, People's Republic of China
| | - Zongzhi Yin
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
- NHC Key Laboratory of the Study of Abnormal Gametes and the Reproductive Tract, Anhui Medical University, Hefei, People's Republic of China
- Anhui Province Key Laboratory of Reproductive Disorders and Obstetrics and Gynaecology Diseases, Hefei, People's Republic of China
- Center for Big Data and Population Health of IHM, Hefei, People's Republic of China
| |
Collapse
|
6
|
Wang Y, Sheng Z, Li H, Tan X, Liu Y, Zhang W, Ma W, Ma L, Fan Y. The effects of Fraxini cortex and Andrographis herba on Escherichia coli-induced diarrhea in chicken. Poult Sci 2025; 104:104824. [PMID: 39874706 PMCID: PMC11810841 DOI: 10.1016/j.psj.2025.104824] [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: 11/04/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 01/30/2025] Open
Abstract
Escherichia coli (E. coli) is a type of pathogenic bacteria that often causes diarrhea in poultry. While antibiotics can control E. coli-induced diarrhea in chickens, it can lead the ongoing proliferation of antibiotic resistance. Traditional Chinese medicines (TCMs) that effectively protect against and treat chicken diarrhea caused by E. coli are an encouraging alternative. That enhance poultry immunity, curtail antibiotic resistance, and provide a secure, eco-friendly, and efficacious option for the livestock and poultry industry. In this study, the model of chicken diarrhea induced by E. coli was established, and different TCM formulas were used for treatment, and finally the formula with the best effect was screened out. The research also investigated the impact of these formulas on gut microbiota and serum metabolites in chickens, using 16S rRNA sequencing technology and metabolomics. Mass spectrometry technology and network pharmacology were used to analyze the optimal TCM formula corroborated by molecular docking and qPCR for further explore mechanism exploration. The findings indicated that Fraxini cortex and Andrographis herba dramatically lowered mortality rates and alleviated pathologic changes in cases of avian E. coli diarrhea (P < 0.05). Fraxini cortex and Andrographis herba significantly boosted the abundance of Bacteroidetes (P < 0.05) and mainly enhanced cysteine and methionine metabolic pathways. Moreover, 97 active ingredients in Fraxini cortex and Andrographis herba were identified, along with 1425 diarrhea-related targets, primarily enriched in the MAPK signaling pathway. Molecular docking and qPCR revealed that the crucial active ingredients in Fraxini cortex and Andrographis herba bonded effectively with disease targets and treated diarrhea by regulating the MAPK signaling pathway. This suggests that Fraxini cortex and Andrographis herba exerts an optimal effect on diarrhea by multi-target and multi-pathway regulation of metabolic pathways and gut microbiota.
Collapse
Affiliation(s)
- Yunying Wang
- College of Veterinary Medicine, Northwest A&F University, 712100, Yangling, PR China; Institute of Traditional Chinese Veterinary Medicine, Northwest A&F University, 712100, Yangling, PR China
| | - Zhenwei Sheng
- College of Veterinary Medicine, Northwest A&F University, 712100, Yangling, PR China; Institute of Traditional Chinese Veterinary Medicine, Northwest A&F University, 712100, Yangling, PR China
| | - Huicong Li
- College of Veterinary Medicine, Northwest A&F University, 712100, Yangling, PR China; Institute of Traditional Chinese Veterinary Medicine, Northwest A&F University, 712100, Yangling, PR China
| | - Xuewen Tan
- College of Veterinary Medicine, Northwest A&F University, 712100, Yangling, PR China; Institute of Traditional Chinese Veterinary Medicine, Northwest A&F University, 712100, Yangling, PR China
| | - Yingqiu Liu
- College of Veterinary Medicine, Northwest A&F University, 712100, Yangling, PR China; Institute of Traditional Chinese Veterinary Medicine, Northwest A&F University, 712100, Yangling, PR China
| | - Weimin Zhang
- College of Veterinary Medicine, Northwest A&F University, 712100, Yangling, PR China; Institute of Traditional Chinese Veterinary Medicine, Northwest A&F University, 712100, Yangling, PR China
| | - Wuren Ma
- College of Veterinary Medicine, Northwest A&F University, 712100, Yangling, PR China; Institute of Traditional Chinese Veterinary Medicine, Northwest A&F University, 712100, Yangling, PR China
| | - Lin Ma
- College of Veterinary Medicine, Northwest A&F University, 712100, Yangling, PR China.
| | - Yunpeng Fan
- College of Veterinary Medicine, Northwest A&F University, 712100, Yangling, PR China; Institute of Traditional Chinese Veterinary Medicine, Northwest A&F University, 712100, Yangling, PR China.
| |
Collapse
|
7
|
Cheng S, Li Y, Sun X, Liu Z, Guo L, Wu J, Yang X, Wei S, Wu G, Xu S, Yang F, Wu J. The impact of glucose metabolism on inflammatory processes in sepsis-induced acute lung injury. Front Immunol 2024; 15:1508985. [PMID: 39712019 PMCID: PMC11659153 DOI: 10.3389/fimmu.2024.1508985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 11/18/2024] [Indexed: 12/24/2024] Open
Abstract
Acute lung injury (ALI) is a prevalent and critical complication of sepsis, marked by high incidence and mortality rates, with its pathogenesis still not being fully elucidated. Recent research has revealed a significant correlation between the metabolic reprogramming of glucose and sepsis-associated ALI (S-ALI). Throughout the course of S-ALI, immune cells, including macrophages and dendritic cells, undergo metabolic shifts to accommodate the intricate demands of immune function that emerge as sepsis advances. Indeed, glucose metabolic reprogramming in S-ALI serves as a double-edged sword, fueling inflammatory immune responses in the initial stages and subsequently initiating anti-inflammatory responses as the disease evolves. In this review, we delineate the current research progress concerning the pathogenic mechanisms linked to glucose metabolic reprogramming in S-ALI, with a focus on the pertinent immune cells implicated. We encapsulate the impact of glucose metabolic reprogramming on the onset, progression, and prognosis of S-ALI. Ultimately, by examining key regulatory factors within metabolic intermediates and enzymes, We have identified potential therapeutic targets linked to metabolic reprogramming, striving to tackle the inherent challenges in diagnosing and treating Severe Acute Lung Injury (S-ALI) with greater efficacy.
Collapse
Affiliation(s)
- Shilei Cheng
- School of Anesthesiology, Shandong Second Medical University, Weifang, China
| | - Yufei Li
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Anesthesia and Respiratory Critical Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Anesthesiology, Jinan, China
- School of Pharmacy, Shandong University of Traditional Chinese Medicine (TCM), Jinan, China
| | - Xiaoliang Sun
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhirui Liu
- Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Liang Guo
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Anesthesia and Respiratory Critical Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Anesthesiology, Jinan, China
| | - Jueheng Wu
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Xiaohan Yang
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Sisi Wei
- Department of Anesthesiology, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, China
| | - Guanghan Wu
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Anesthesia and Respiratory Critical Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Anesthesiology, Jinan, China
| | - Shilong Xu
- School of Anesthesiology, Shandong Second Medical University, Weifang, China
| | - Fan Yang
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Anesthesia and Respiratory Critical Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Anesthesiology, Jinan, China
| | - Jianbo Wu
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Anesthesia and Respiratory Critical Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Anesthesiology, Jinan, China
| |
Collapse
|
8
|
Liang X, Hu X, Li J, Zhang B, Gu T, Wang H, Zhang M, Xia X, Guan S, Shangguan W, Miao S, Wang W, Zhang H, Zhao Z, Wang L. m6A methylation in myocardial tissue of septic mice analyzed using MeRIP/m6A-sequencing and RNA-sequencing. Funct Integr Genomics 2024; 24:173. [PMID: 39320434 DOI: 10.1007/s10142-024-01452-6] [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: 06/24/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 09/26/2024]
Abstract
Septic cardiomyopathy is a secondary myocardial injury caused by sepsis. N6-methyl-adenosine (m6A) modification is involved in the pathological progression of septic cardiomyopathy; however, the pathological mechanism remains unclear. In this study, we identified the overall m6A modification pattern in septic myocardial injury and determined its potential interactions with differentially expressed genes (DEGs). A sepsis mouse model exhibiting septic symptoms and myocardial tissue damage was induced by lipopolysaccharide (LPS). LPS-induced septic myocardial tissues and control myocardial tissues were subjected to methylated RNA immunoprecipitation sequencing and RNA sequencing to screen for differentially expressed m6A peaks and DEGs. We identified 859 significantly m6A-modified genes in septic myocardial tissues, including 432 upregulated and 427 downregulated genes. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses were performed to explore the biological importance of differentially expressed m6A methylated genes and DEGs. Differentially expressed m6A methylated genes were enriched in immune- and inflammation-related pathways. Conjoint analysis revealed co-expression of differentially expressed m6A genes and DEGs, including genes that were upregulated or downregulated and those showing opposite trends. High expression of m6A-related genes (WTAP and IGF2BP2), interleukin-17, and interleukin-17 pathway-related genes (MAPK11 and TRAF3IP2) was verified using reverse transcription-quantitative PCR. We confirmed the presence of m6A modification of the transcriptome and m6A-mediated gene expression in septic myocardial tissues.
Collapse
Affiliation(s)
- Xue Liang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Xiaotong Hu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Jiao Li
- Department of Cardiology, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, 300121, China
| | - Boyang Zhang
- Department of Emergency Medicine, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Tianshu Gu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Hualing Wang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Mingzhong Zhang
- Department of Traditional Chinese Medicine, People's Hospital of Linqing, Linqing Shandong, 252600, China
| | - Xiaodong Xia
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Siyu Guan
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Wenfeng Shangguan
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Shuai Miao
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Weiding Wang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Hao Zhang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Zhiqiang Zhao
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China.
| | - Lijun Wang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China.
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
9
|
Li M, Ren Q, Chen K, Yin R, Li W, Fang Z, Liu S, Lan L, Hong G. Regulation of macrophage polarization and glucose metabolism by the ERK/MAPK-HK1 signaling pathway in paraquat-induced acute lung injury. Chem Biol Interact 2024; 397:111062. [PMID: 38763349 DOI: 10.1016/j.cbi.2024.111062] [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: 03/09/2024] [Revised: 05/05/2024] [Accepted: 05/17/2024] [Indexed: 05/21/2024]
Abstract
Acute lung injury is the leading cause of paraquat (PQ) poisoning-related mortality. The mechanism by which macrophages are involved in PQ-induced acute lung injury remains unclear. In recent years, the role of metabolic reprogramming in macrophage functional transformation has received significant attention. The current study aimed to identify the role of altered macrophage glucose metabolism and molecular mechanisms in PQ poisoning-induced acute lung injury. We established a model of acute lung injury in PQ-intoxicated mice via the intraperitoneal injection of PQ. PQ exposure induces macrophage M1 polarization and promotes the release of inflammatory factors, which causes the development of acute lung injury in mice. In vitro analysis revealed that PQ altered glucose metabolism, which could be reversed by siRNA transfection to silence the expression of HK1, a key enzyme in glucose metabolism. RNA sequencing revealed that the ERK/MAPK pathway was the crucial molecular mechanism of PQ pathogenesis. Further, U0126, an ERK inhibitor, could inhibit PQ-induced HK1 activation and macrophage M1 polarization. These findings provide novel insights into the previously unrecognized mechanism of ERK/MAPK-HK1 activation in PQ poisoning.
Collapse
Affiliation(s)
- Mengxuan Li
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Qinghuan Ren
- Wenzhou Medical University, Wenzhou, 325000, China
| | - Kaiyuan Chen
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China; Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Ran Yin
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Wenwen Li
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Zuochun Fang
- Longgang Campus of the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Sunxiang Liu
- Longgang Campus of the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Linhua Lan
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Guangliang Hong
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China; Longgang Campus of the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
10
|
Hu X, Huang X, Yang Y, Sun Y, Zhao Y, Zhang Z, Qiu D, Wu Y, Wu G, Lei L. Dux activates metabolism-lactylation-MET network during early iPSC reprogramming with Brg1 as the histone lactylation reader. Nucleic Acids Res 2024; 52:5529-5548. [PMID: 38512058 PMCID: PMC11162783 DOI: 10.1093/nar/gkae183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 02/24/2024] [Accepted: 03/15/2024] [Indexed: 03/22/2024] Open
Abstract
The process of induced pluripotent stem cells (iPSCs) reprogramming involves several crucial events, including the mesenchymal-epithelial transition (MET), activation of pluripotent genes, metabolic reprogramming, and epigenetic rewiring. Although these events intricately interact and influence each other, the specific element that regulates the reprogramming network remains unclear. Dux, a factor known to promote totipotency during the transition from embryonic stem cells (ESC) to 2C-like ESC (2CLC), has not been extensively studied in the context of iPSC reprogramming. In this study, we demonstrate that the modification of H3K18la induced by Dux overexpression controls the metabolism-H3K18la-MET network, enhancing the efficiency of iPSC reprogramming through a metabolic switch and the recruitment of p300 via its C-terminal domain. Furthermore, our proteomic analysis of H3K18la immunoprecipitation experiment uncovers the specific recruitment of Brg1 during reprogramming, with both H3K18la and Brg1 being enriched on the promoters of genes associated with pluripotency and epithelial junction. In summary, our study has demonstrated the significant role of Dux-induced H3K18la in the early reprogramming process, highlighting its function as a potent trigger. Additionally, our research has revealed, for the first time, the binding of Brg1 to H3K18la, indicating its role as a reader of histone lactylation.
Collapse
Affiliation(s)
- Xinglin Hu
- Department of Histology and Embryology, Basic Medical Science College, Harbin Medical University, 194 Xuefu Rd, Nangang District, Harbin, Heilongjiang Province 150081, China
| | - Xingwei Huang
- Department of Histology and Embryology, Basic Medical Science College, Harbin Medical University, 194 Xuefu Rd, Nangang District, Harbin, Heilongjiang Province 150081, China
- Guangzhou Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou 510005 Guangdong Province, China
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510320, China
| | - Yue Yang
- Department of Histology and Embryology, Basic Medical Science College, Harbin Medical University, 194 Xuefu Rd, Nangang District, Harbin, Heilongjiang Province 150081, China
| | - Yuchen Sun
- Department of Histology and Embryology, Basic Medical Science College, Harbin Medical University, 194 Xuefu Rd, Nangang District, Harbin, Heilongjiang Province 150081, China
| | - Yanhua Zhao
- Department of Histology and Embryology, Basic Medical Science College, Harbin Medical University, 194 Xuefu Rd, Nangang District, Harbin, Heilongjiang Province 150081, China
| | - Zhijing Zhang
- Department of Histology and Embryology, Basic Medical Science College, Harbin Medical University, 194 Xuefu Rd, Nangang District, Harbin, Heilongjiang Province 150081, China
| | - Dan Qiu
- Department of Histology and Embryology, Basic Medical Science College, Harbin Medical University, 194 Xuefu Rd, Nangang District, Harbin, Heilongjiang Province 150081, China
| | - Yanshuang Wu
- Department of Histology and Embryology, Basic Medical Science College, Harbin Medical University, 194 Xuefu Rd, Nangang District, Harbin, Heilongjiang Province 150081, China
| | - Guangming Wu
- Guangzhou Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou 510005 Guangdong Province, China
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510320, China
| | - Lei Lei
- Department of Histology and Embryology, Basic Medical Science College, Harbin Medical University, 194 Xuefu Rd, Nangang District, Harbin, Heilongjiang Province 150081, China
| |
Collapse
|
11
|
Xu B, Liu Y, Li N, Geng Q. Lactate and lactylation in macrophage metabolic reprogramming: current progress and outstanding issues. Front Immunol 2024; 15:1395786. [PMID: 38835758 PMCID: PMC11148263 DOI: 10.3389/fimmu.2024.1395786] [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: 03/04/2024] [Accepted: 05/07/2024] [Indexed: 06/06/2024] Open
Abstract
It is commonly known that different macrophage phenotypes play specific roles in different pathophysiological processes. In recent years, many studies have linked the phenotypes of macrophages to their characteristics in different metabolic pathways, suggesting that macrophages can perform different functions through metabolic reprogramming. It is now gradually recognized that lactate, previously overlooked as a byproduct of glycolytic metabolism, acts as a signaling molecule in regulating multiple biological processes, including immunological responses and metabolism. Recently, lactate has been found to mediate epigenetic changes in macrophages through a newfound lactylation modification, thereby regulating their phenotypic transformation. This novel finding highlights the significant role of lactate metabolism in macrophage function. In this review, we summarize the features of relevant metabolic reprogramming in macrophages and the role of lactate metabolism therein. We also review the progress of research on the regulation of macrophage metabolic reprogramming by lactylation through epigenetic mechanisms.
Collapse
Affiliation(s)
- Bangjun Xu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yi Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
12
|
Yanar KE, Gür C, Değirmençay Ş, Aydın Ö, Aktaş MS, Baysal S. Insulin-like growth factor-1 expression levels in pro-inflammatory response in calves with neonatal systemic inflammatory response syndrome. Vet Immunol Immunopathol 2024; 268:110706. [PMID: 38159440 DOI: 10.1016/j.vetimm.2023.110706] [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: 11/17/2023] [Revised: 12/13/2023] [Accepted: 12/23/2023] [Indexed: 01/03/2024]
Abstract
The objective of this study was to investigate the mRNA expression of insulin-like growth factor-1 (IGF-1), pro-inflammatory cytokines (IL-1β, IL-6, IL-18, and TNF-α), serum immunoglobulin profiles (IgG and IgM), and lipid peroxidation status (MDA) in relation to pro-inflammatory cytokines. A case-controlled, prospective, and observational investigation was completed on 85 calves. Total RNA was isolated from whole blood samples of both the SIRS and healthy calves, followed by reverse transcription into cDNA. The resulting cDNAs were mixed with iTaq Universal SYBR Green Supermix and primers specific to the relevant genes using the Rotor-Gene Q instrument. After the reaction was completed, gene expressions were normalised against β-actin using the 2-ΔΔCT method. The mRNA levels of pro-inflammatory cytokines namely (IL-1β [SIRS: 2.15 ± 0.55, Control: 1.13 ± 0.62; P = 0.001], IL-6 [SIRS: 2.82 ± 0.52, Control: 0.91 ± 0.11; P < 0.001], IL-18 [SIRS: 1.92 ± 0.41, Control: 0.99 ± 0.13; P < 0.001], and TNF-α [SIRS: 2.59 ± 0.28, Control: 0.93 ± 0.09; P < 0.001]) and IGF-1 (SIRS: 3.55 ± 0.55, Control: 0.91 ± 0.15; P < 0.001) were up-regulated in calves with SIRS, while serum IgG (SIRS: 4.16 ± 0.26, Control: 1.73 ± 0.17; P < 0.001), IgM (SIRS: 1.55 ± 0.11, Control: 1.09 ± 0.13; P < 0.001), and MDA levels (SIRS: 41.12 ± 3.48, Control: 3.76 ± 0.81; P < 0.001) increased significantly in these calves. Furthermore, significant (P < 0.01) positive correlations were found in calves with SIRS in relation to the expression levels of IL-1β, IL-6, IL-18, TNF-α, IGF-1, serum immunoglobulins, and MDA levels. These results suggest that IGF-1 could be a valuable pro-inflammatory marker, considering its high positive correlation with the expression levels of pro-inflammatory cytokines (IL-1β, IL-6, IL-18, and TNF-α) and markers (MDA, IgG, and IgM) in calves with SIRS.
Collapse
Affiliation(s)
- Kerim Emre Yanar
- Department of Internal Medicine, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey.
| | - Cihan Gür
- Department of Biochemistry, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Şükrü Değirmençay
- Department of Internal Medicine, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Ömer Aydın
- Department of Internal Medicine, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Mustafa Sinan Aktaş
- Department of Internal Medicine, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Sümeyye Baysal
- Department of Internal Medicine, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| |
Collapse
|
13
|
Cheng Y, Wang P, Liu L. PFKFB3 Regulates the Growth and Migration of Ovarian Cancer Cells through Pyroptosis and Warburg Effect Progression. J Environ Pathol Toxicol Oncol 2024; 43:53-64. [PMID: 39016141 DOI: 10.1615/jenvironpatholtoxicoloncol.2024052948] [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: 07/18/2024] Open
Abstract
Ovarian cancer is one of the most common malignant tumors in female reproductive organs. Its incidence rate is second only to uterine body cancer and cervical cancer, posing a serious threat to women's health. Herein, we explored that PFKFB3 in cancer progression of ovarian cancer and its underlying mechanism. All the serum samples from ovarian cancer were collected by our hospital. PFKFB3 mRNA expressions in patients with ovarian cancer and ovarian cancer cell lines were up-regulated. PFKFB3 protein expressions in ovarian cancer cells were induced. ovarian cancer patients with high PFKFB3expression had lower survival rate. The PFKFB3gene promoted cell proliferation and EDU cells, and increased cell metastasis of ovarian cancer. Si-PFKFB3 reduced cell proliferation and EDU cells, and decreased cell metastasis of ovarian cancer. PFKFB3 gene up-regulation reduced caspase-3/9 activity levels of ovarian cancer. Si-PFKFB3 also promoted caspase-3/9 activity levels of ovarian cancer. PFKFB3 gene promoted Warburg effect progression of ovarian cancer. PFKFB3 gene reduced NLRP3-induced pyroptosis of ovarian cancer. PFKFB3 suppressed NLRP3 expression. NLRP3 was one target spot for PFKFB3 on pyroptosis of ovarian cancer. Taken together, we conclude that PFKFB3 suppressed NLRP3 axis to reduce pyroptosis and increase Warburg effect progression of ovarian cancer, and provide molecular insight into the mechanisms by which the PFKFB3 regulates pyroptosis of ovarian cancer.
Collapse
Affiliation(s)
- Ye Cheng
- Department of Obstetrics and Gynecology, Wuhan No. 1 Hospital, Wuhan 430030, China
| | - Ping Wang
- Department of Obstetrics and Gynecology, Wuhan No. 1 Hospital, Wuhan 430030, China
| | | |
Collapse
|
14
|
Xuan L, Xu Z, Luo J, Wang Y, Yan Y, Qu C, Xie Z, Skonieczna M, Zhou PK, Huang R. Lactate exacerbates lung damage induced by nanomicroplastic through the gut microbiota-HIF1a/PTBP1 pathway. Exp Mol Med 2023; 55:2596-2607. [PMID: 38036735 PMCID: PMC10766629 DOI: 10.1038/s12276-023-01129-3] [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: 07/17/2023] [Revised: 09/24/2023] [Accepted: 09/25/2023] [Indexed: 12/02/2023] Open
Abstract
Exposure to nanomicroplastics (nano-MPs) can induce lung damage. The gut microbiota is a critical modulator of the gut-lung axis. However, the mechanisms underlying these interactions have not been elucidated. This study explored the role of lactate, a key metabolite of the microbiota, in the development of lung damage induced by nano-MPs (LDMP). After 28 days of exposure to nano-MPs (50-100 nm), mice mainly exhibited damage to the lungs and intestinal mucosa and dysbiosis of the gut microbiota. Lactate accumulation was observed in the lungs, intestines and serum and was strongly associated with the imbalance in lactic acid bacteria in the gut. Furthermore, no lactate accumulation was observed in germ-free mice, while the depletion of the gut microbiota using a cocktail of antibiotics produced similar results, suggesting that lactate accumulation in the lungs may have been due to changes in the gut microbiota components. Mechanistically, elevated lactate triggers activation of the HIF1a/PTBP1 pathway, exacerbating nano-MP-induced lung damage through modulation of the epithelial-mesenchymal transition (EMT). Conversely, mice with conditional knockout of Ptbp1 in the lungs (Ptbp1flfl) and PTBP1-knockout (PTBP1-KO) human bronchial epithelial (HBE) cells showed reversal of the effects of lactate through modulation of the HIF1a/PTBP1 signaling pathway. These findings indicate that lactate is a potential target for preventing and treating LDMP.
Collapse
Affiliation(s)
- Lihui Xuan
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province, 410078, China
| | - Zheng Xu
- Translational Medicine Research Center, Naval Medical University, 800, Xiangyin Road, 200433, Shanghai, People's Republic of China
| | - Jinhua Luo
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province, 410078, China
| | - Yin Wang
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province, 410078, China
| | - Yuhui Yan
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province, 410078, China
| | - Can Qu
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province, 410078, China
| | - Zuozhong Xie
- Department of Otolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan Province, China
| | - Magdalena Skonieczna
- Department of Systems Biology and Engineering, Silesian University of Technology, Institute of Automatic Control, Akademicka 16, Gliwice, 44-100, Poland
- Biotechnology Centre, Silesian University of Technology, Krzywoustego 8, Gliwice, 44-100, Poland
| | - Ping-Kun Zhou
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| | - Ruixue Huang
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province, 410078, China.
| |
Collapse
|
15
|
Zhang Y, Li N, Yang L, Jia W, Li Z, Shao Q, Zhan X. Quantitative phosphoproteomics reveals molecular pathway network alterations in human early-stage primary hepatic carcinomas: potential for 3P medical approach. EPMA J 2023; 14:477-502. [PMID: 37605650 PMCID: PMC10439880 DOI: 10.1007/s13167-023-00335-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/25/2023] [Indexed: 08/23/2023]
Abstract
Objective Hepatic carcinoma is one of the most common types of malignant tumors in the digestive system, and its biological characteristics determine its high rate of metastasis and recurrence after radical resection, leading to a poor prognosis for patients. Increasing evidence demonstrates that phosphoproteins and phosphorylation-mediated molecular pathways influence the occurrence and development of hepatic carcinoma. It is urgent need to develop early-stage biomarkers for improving diagnosis, therapy, medical service, and prognostic assessment. We hypothesize that phosphoproteome and phosphorylation-mediated signaling pathway networks significantly differ in human early-stage primary hepatic carcinomas relative to control liver tissues, which will identify the key differentially phosphorylated proteins and phosphorylation-mediated signaling pathway network alterations in human early-stage primary hepatic carcinoma to innovate predictive diagnosis, prognostic assessment, and personalized medical services and progress beyond the state of the art in the framework of predictive, preventive, and personalized medicine (PPPM). Methods Tandem mass tag (TMT)-based quantitative proteomics coupled with TiO2 enrichment of phosphopeptides was used to identify phosphorylation profiling, and bioinformatics was used to analyze the pathways and biological functions of phosphorylation profiling between early-stage hepatic carcinoma tissues and tumor-adjacent normal control tissues. Furthermore, the integrative analysis with transcriptomic data from TCGA database obtained differently expressed genes (DEGs) corresponding to differentially phosphorylated proteins (DPPs) and overall survival (OS)-related DPPs. Results A total of 1326 phosphopeptides derived from 858 DPPs in human early-stage primary hepatic carcinoma were identified. KEGG pathway network analysis of 858 DPPs revealed 33 statistically significant signaling pathways, including spliceosome, glycolysis/gluconeogenesis, B-cell receptor signaling pathway, HIF-1 signaling pathway, and fatty acid degradation. Gene Ontology (GO) analysis of 858 DPPs revealed that protein phosphorylation was involved in 57 biological processes, 40 cellular components, and 37 molecular functions. Protein-protein interaction (PPI) network constructed multiple high-combined scores and co-expressed DPPs. Integrative analysis of transcriptomic data and DPP data identified 105 overlapped molecules (DPPs; DEGs) between hepatic carcinoma tissues and control tissues and 125 OS-related DPPs. Overlapping Venn plots showed 14 common molecules among datasets of DPPs, DEGs, and OS-related DDPs, including FTCD, NDRG2, CCT2, PECR, SLC23A2, PNPLA7, ANLN, HNRNPM, HJURP, MCM2, STMN1, TCOF1, TOP2A, and SSRP1. The drug sensitivities of OS-related DPPs were identified, including LMOD1, CAV2, UBE2E2, RAPH1, ANXA5, HDLBP, CUEDC1, APBB1IP, VCL, SRSF10, SLC23A2, EPB41L2, ESR1, PLEKHA4, SAFB2, SMARCAD1, VCAN, PSD4, RDH16, NOP56, MEF2C, BAIAP2L2, NAGS, SRSF2, FHOD3, and STMN1. Conclusions Identification and annotation of phosphoproteomes and phosphorylation-mediated signaling pathways in human early-stage primary hepatic carcinoma tissues provided new directions for tumor prevention and treatment, which (i) helps to enrich phosphorylation functional research and develop new biomarkers; (ii) enriches phosphorylation-mediated signaling pathways to gain a deeper understanding of the underlying mechanisms of early-stage primary hepatic carcinoma; and (iii) develops anti-tumor drugs that facilitate targeted phosphorylated sites. We recommend quantitative phosphoproteomics in early-stage primary hepatic carcinoma, which offers great promise for in-depth insight into the molecular mechanism of early-stage primary hepatic carcinoma, the discovery of effective therapeutic targets/drugs, and the construction of reliable phosphorylation-related biomarkers for patient stratification, predictive diagnosis, prognostic assessment, and personalized medical services in the framework of PPPM. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-023-00335-3.
Collapse
Affiliation(s)
- Yuping Zhang
- Department of General Surgery, The Third Xiangya Hospital, Central South University, 138 Tongzi Po Road, Changsha, Hunan 410013 People’s Republic of China
| | - Na Li
- Medical Science and Technology Innovation Center, Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Lamei Yang
- Medical Science and Technology Innovation Center, Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Wenshuang Jia
- Medical Science and Technology Innovation Center, Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Zhijun Li
- Medical Science and Technology Innovation Center, Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Qianwen Shao
- Medical Science and Technology Innovation Center, Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Xianquan Zhan
- Medical Science and Technology Innovation Center, Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| |
Collapse
|