1
|
Wang L, Wang X, Wu J, Chen J, He Z, Wang J, Zhang X. Magnesium Ions Induce Endothelial Cell Differentiation into Tip Cell and Enhance Vascularized Bone Regeneration. Adv Healthc Mater 2025:e2500274. [PMID: 40346783 DOI: 10.1002/adhm.202500274] [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: 01/16/2025] [Revised: 04/07/2025] [Indexed: 05/12/2025]
Abstract
Vascularization has been considered an essential strategy for bone regeneration and can be promoted by magnesium ions (Mg2+). During angiogenesis, the differentiation of endothelial cells (ECs) into tip cell is a critical step since it controls the growth direction and pattern of new vascular sprouts. While several studies have noted the pro-angiogenic effects of Mg2+, however, their specific influence on tip cell formation is unclear. Therefore, this research seeks to examine the impact of Mg2+ on tip cells and elucidate the potential mechanisms involved. The results reveal that Mg2+ shows good compatibility and stimulates ECs to migrate and invade in vitro. Moreover, Mg2+ enhances EC spheroids sprouting and elevates the expression of genes linked to tip cells. The underlying mechanisms are that Mg2+ facilitates tip cell differentiation via the VEGFA-VEGFR2/Notch1 signaling pathway crosstalk and promotes migration and filopodia formation of tip cells and proliferation of stalk cells by inducing YAP nuclear translocation, culminating in the maturation of vascular networks. Furthermore, EC spheroids stimulated by Mg2+ load in hydrogel enhance vascularized bone regeneration in vivo. These findings enrich the understanding of how Mg2+ influence blood vessel formation and provide practical strategies for the development and design of magnesium-based biomaterials.
Collapse
Affiliation(s)
- Liang Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xu Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jicenyuan Wu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Junyu Chen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Zihan He
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Prosthodontics and Implantology, The Affiliated Stomatological Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Jian Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xin Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| |
Collapse
|
2
|
Antonis S, Efthymia P, Maria K, Eleftherios P, Sotiris K, Neoklis G, Fuminori T, George A, Tasuku H, Apostolos K. The effect of combined oral contraceptive pills on angiogenesis in endometriotic lesions. Hormones (Athens) 2025:10.1007/s42000-025-00636-4. [PMID: 39982662 DOI: 10.1007/s42000-025-00636-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 02/06/2025] [Indexed: 02/22/2025]
Abstract
PURPOSE Neoangiogenesis is necessary for adhesion and invasion of endometriotic lesions. We hypothesize that by blocking angiogenetic pathways we can suppress endometriosis. Oral contraceptive pills (OCs) are routinely used in endometriosis to suppress symptoms of the disease. In the current study, we attempt to evaluate the effects of OCs on various angiogenetic factors in women with endometriosis. METHODS Sixty women with endometriosis were randomly divided into two groups. Group A consisted of 30 women who received OCs in a cyclical manner for 3 months before surgery and group B of 30 women who did not. Biopsy specimens of ovarian endometrioma were collected. We used qRT-PCR to study the mRNA expression levels of VEGF, TF, PAR-2, SP1, and FGF1. RESULTS The levels of mRNA of all angiogenic factors were found to be elevated in women who received OCs compared with women who did not. This difference was statistically significant for VEGF, TF, FGF1, SP1 (p < 0.001), and PAR-2 (p = 0.046). CONCLUSION OC administration does not inhibit neoangiogenesis in endometriotic lesions; on the contrary, angiogenetic pathways might be upregulated.
Collapse
Affiliation(s)
- Siampalis Antonis
- Dept. of Obstetrics & Gynecology, Patras University School of Medicine, Patras, Greece
| | | | - Keramida Maria
- Dept. of Obstetrics & Gynecology, Patras University School of Medicine, Patras, Greece
| | - Panteris Eleftherios
- Laboratory of Forensic Medicine and Toxicology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| | | | - Georgopoulos Neoklis
- Dept. of Obstetrics & Gynecology, Patras University School of Medicine, Patras, Greece
| | - Taniguchi Fuminori
- Dept. of Obstetrics & Gynecology, Tottori University Faculty of Medicine, Yonago, Japan
| | - Adonakis George
- Dept. of Obstetrics & Gynecology, Patras University School of Medicine, Patras, Greece
| | - Harada Tasuku
- Dept. of Obstetrics & Gynecology, Tottori University Faculty of Medicine, Yonago, Japan
| | - Kaponis Apostolos
- Dept. of Obstetrics & Gynecology, Patras University School of Medicine, Patras, Greece.
| |
Collapse
|
3
|
Wen Y, Chen J, Long T, Chen F, Wang Z, Chen S, Zhang G, Li M, Zhang S, Kang H, Feng W, Wang G. miR-6760-5p suppresses neoangiogenesis by targeting Yes-associated protein 1 in patients with moyamoya disease undergoing indirect revascularization. Gene 2025; 937:149152. [PMID: 39662645 DOI: 10.1016/j.gene.2024.149152] [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: 07/10/2024] [Revised: 11/22/2024] [Accepted: 12/05/2024] [Indexed: 12/13/2024]
Abstract
OBJECTIVE The aim of this research was to investigate the specific regulatory role of miR-6760-5p in angiogenesis in moyamoya disease. METHODS HUVECs were transfected with miR-6760-5p inhibitor and mimics fragments, then subjected to assays for cell proliferation, migration, and tube formation. Subsequently, downstream target genes of miR-6760-5p were predicted and the protein expression levels of these genes were evaluated. The presence of miR-6760-5p and YAP1 was verified by a dual luciferase reporter gene test, followed by an assessment of the effects of YAP1 and miR-6760-5p on the HUVECs. RESULTS Comparatively to the control group, increased expression of miR-6760-5p decreased cell growth, movement, and tube formation. YAP1 gene was discovered as a target controlled by miR-6760-5p, with subsequent investigation confirming YAP1 as a gene regulated by miR-6760-5p. Additionally, miR-6760-5p was found to counteract the angiogenesis-promoting effect of YAP1. CONCLUSION The results of this research suggest a possible link between the miR-6760-5p gene found in the cerebrospinal fluid of individuals with moyamoya disease and the process of vascularization in this particular condition. The findings indicate that miR-6760-5p may be a new molecular indicator and potential target for the diagnosis of moyamoya disease.
Collapse
Affiliation(s)
- Yunyu Wen
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China; The Institute of Brain Disease is part of Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China
| | - Junda Chen
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China
| | - Tinghan Long
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China
| | - Fangzhou Chen
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China
| | - Zhibin Wang
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China
| | - Siyuan Chen
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China
| | - Guozhong Zhang
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China
| | - Mingzhou Li
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China
| | - Shichao Zhang
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China
| | - Huibin Kang
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China
| | - Wenfeng Feng
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China; The Institute of Brain Disease is part of Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China.
| | - Gang Wang
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China; The Institute of Brain Disease is part of Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China.
| |
Collapse
|
4
|
Filindris T, Papakonstantinou E, Keramida M, Panteris E, Kalogeropoulos S, Georgopoulos N, Taniguchi F, Adonakis G, Harada T, Kaponis A. The effect of GnRH-a on the angiogenesis of endometriosis. Hormones (Athens) 2024; 23:509-515. [PMID: 38639888 PMCID: PMC11436414 DOI: 10.1007/s42000-024-00559-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 04/03/2024] [Indexed: 04/20/2024]
Abstract
PURPOSE Neoangiogenesis is necessary for adhesion and invasiveness of endometriotic lesions in women affected by endometriosis. Vascular endothelial growth factor (VEGF) is one of the main components of angiogenesis and is part of the major pathway tissue factor (TF)-protease activated receptor-2 (PAR-2)-VEGF that leads to neoangiogenesis. Specificity protein 1 (SP1) is a transcriptional factor that has recently been studied for its crucial role in angiogenesis via a specific pathway. We hypothesize that by blocking angiogenetic pathways we can suppress endometriotic lesions. Gonadotrophin-releasing hormone-agonists (GnRH-a) are routinely used, especially preoperatively, in endometriosis. It would be of great interest to clarify which angiogenetic pathways are affected and, thereby, pave the way for further research into antiangiogenetic effects on endometriosis. METHODS We used quantitative real-time polymerase chain reaction (qRT-PCR) to study mRNA expression levels of TF, PAR-2, VEGF, and SP1 in endometriotic tissues of women who underwent surgery for endometriosis and received GnRH-a (leuprolide acetate) preoperatively. RESULTS VEGF, TF, and PAR-2 expression is significantly lower in patients who received treatment (p < 0,001) compared to those who did not, whereas SP1 expression is not altered (p = 0.779). CONCLUSIONS GnRH-a administration does affect some pathways of angiogenesis in endometriotic lesions, but not all of them. Therefore, supplementary treatments that affect the SP1 pathway of angiogenesis should be developed to enhance the antiangiogenetic effect of GnRH-a in patients with endometriosis. TRIAL REGISTRATION Clinicaltrial.gov ID: NCT06106932.
Collapse
Affiliation(s)
- Theodoros Filindris
- Dept. of Obstetrics & Gynecology, Patras University School of Medicine, Patras, Greece
| | | | - Maria Keramida
- Dept. of Obstetrics & Gynecology, Patras University School of Medicine, Patras, Greece
| | - Eleftherios Panteris
- Laboratory of Forensic Medicine and Toxicology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| | | | - Neoklis Georgopoulos
- Dept. of Obstetrics & Gynecology, Patras University School of Medicine, Patras, Greece
| | - Fuminori Taniguchi
- Dept. of Obstetrics & Gynecology, Tottori University Faculty of Medicine, Yonago, Japan
| | - George Adonakis
- Dept. of Obstetrics & Gynecology, Patras University School of Medicine, Patras, Greece
| | - Tasuku Harada
- Dept. of Obstetrics & Gynecology, Tottori University Faculty of Medicine, Yonago, Japan
| | - Apostolos Kaponis
- Dept. of Obstetrics & Gynecology, Patras University School of Medicine, Patras, Greece.
| |
Collapse
|
5
|
Zhang Q, Xia Y, Wang L, Wang Y, Bao Y, Zhao GS. Targeted anti-angiogenesis therapy for advanced osteosarcoma. Front Oncol 2024; 14:1413213. [PMID: 39252946 PMCID: PMC11381227 DOI: 10.3389/fonc.2024.1413213] [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] [Received: 04/06/2024] [Accepted: 08/08/2024] [Indexed: 09/11/2024] Open
Abstract
To date, despite extensive research, the prognosis of advanced osteosarcoma has not improved significantly. Thus, patients experience a reduced survival rate, suggesting that a reevaluation of current treatment strategies is required. Recently, in addition to routine surgery, chemotherapy and radiotherapy, researchers have explored more effective and safer treatments, including targeted therapy, immunotherapy, anti-angiogenesis therapy, metabolic targets therapy, and nanomedicine therapy. The tumorigenesis and development of osteosarcoma is closely related to angiogenesis. Thus, anti-angiogenesis therapy is crucial to treat osteosarcoma; however, recent clinical trials found that it has insufficient efficacy. To solve this problem, the causes of treatment failure and improve treatment strategies should be investigated. This review focuses on summarizing the pathophysiological mechanisms of angiogenesis in osteosarcoma and recent advances in anti-angiogenesis treatment of osteosarcoma. We also discuss some clinical studies, with the aim of providing new ideas to improve treatment strategies for osteosarcoma and the prognosis of patients.
Collapse
Affiliation(s)
- Qiao Zhang
- Department of Pain and Rehabilitation, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yuxuan Xia
- Department of Clinical Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - LiYuan Wang
- Department of Spine Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yang Wang
- Department of Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yixi Bao
- Department of Clinical Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guo-Sheng Zhao
- Department of Spine Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
6
|
Shi Z, Zhang M, Fan H, Chen Y, Dong S, Zhou F, Wang B, Liu J, Jin J, Luo Y, Chen Q, Wang W, Zhang C, Chen Y. The marine Penicillium sp. GGF16-1-2 metabolite dicitrinone G inhibits pancreatic angiogenesis by regulating the activation of NLRP3 inflammasome. J Nat Med 2024; 78:78-90. [PMID: 37897512 DOI: 10.1007/s11418-023-01749-z] [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: 05/04/2023] [Accepted: 08/24/2023] [Indexed: 10/30/2023]
Abstract
Citrinin derivatives have been found to have various pharmacological activities, such as anti-inflammatory, anti-tumor, and antioxidant effects. Dicitrinone G (DG) was a new citrinin dimer isolated from marine-derived fungus Penicillium sp. GGF 16-1-2 which has potential activity. Here, we aim to investigate whether DG has anti-pancreatic cancer activity. In xenograft tumor model, 2 × 106 BXPC-3 cells were injected into the hind flank of NU/NU nude mice by subcutaneously for 2 weeks followed by treating with DG (0.25, 0.5, 1 mg/kg) and 5-FU (30 mg/kg) for 4 weeks. Tumor volume and weight were measured, and the expression of CD31, IL-18, NLRP3, and Caspase-1 in tumor tissue were detected. In vitro, HUVECs were treated with conditioned medium (CM) derived from BXPC-3 cells, the effects of DG on angiogenesis were detected by tube formation and western blot analysis. In vivo studies showed that the tumor growth and angiogenesis were greatly suppressed. The tumor weight inhibition rates of DG and 5-FU groups were about 42.36%, 38.94%, 43.80%, and 31.88%. Furthermore, the expression of CD31 and Caspase-1 were decreased. In vitro, CM derived from BXPC-3 cells which treated with DG could inhibit the tube formation and expression of pro-angiogenic NICD in HUVECs. Our study suggests that DG could suppress angiogenesis via the NLRP3/IL-18 pathway and may have the potential to inhibit tumor development.
Collapse
Affiliation(s)
- Zhimian Shi
- School of Pharmaceutical Sciences, Guangzhou Higher Education Mega Center, Guangzhou University of Chinese Medicine, 232, Waihuan East Road, Panyu, Guangzhou, 510000, China
- Guangdong Key Laboratory of TCM Pathogenesis and Prescriptions Realted to Heart and Spleen, Guangzhou Higher Education Mega Center, 232, Waihuan East Road, Panyu, Guangzhou, 510000, China
| | - Minyi Zhang
- School of Pharmaceutical Sciences, Guangzhou Higher Education Mega Center, Guangzhou University of Chinese Medicine, 232, Waihuan East Road, Panyu, Guangzhou, 510000, China
- Guangdong Key Laboratory of TCM Pathogenesis and Prescriptions Realted to Heart and Spleen, Guangzhou Higher Education Mega Center, 232, Waihuan East Road, Panyu, Guangzhou, 510000, China
| | - Hao Fan
- School of Pharmaceutical Sciences, Guangzhou Higher Education Mega Center, Guangzhou University of Chinese Medicine, 232, Waihuan East Road, Panyu, Guangzhou, 510000, China
| | - Yijun Chen
- School of Pharmaceutical Sciences, Guangzhou Higher Education Mega Center, Guangzhou University of Chinese Medicine, 232, Waihuan East Road, Panyu, Guangzhou, 510000, China
- Guangdong Key Laboratory of TCM Pathogenesis and Prescriptions Realted to Heart and Spleen, Guangzhou Higher Education Mega Center, 232, Waihuan East Road, Panyu, Guangzhou, 510000, China
| | - Su Dong
- Department of Pharmacy, People's Hospital of Dongxihu District, Wuhan, 430040, Hubei, China
| | - Fengguo Zhou
- School of Pharmaceutical Sciences, Guangzhou Higher Education Mega Center, Guangzhou University of Chinese Medicine, 232, Waihuan East Road, Panyu, Guangzhou, 510000, China
| | - Bin Wang
- Department of Cardiovascular Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Jingya Liu
- School of Pharmaceutical Sciences, Guangzhou Higher Education Mega Center, Guangzhou University of Chinese Medicine, 232, Waihuan East Road, Panyu, Guangzhou, 510000, China
| | - Jiaqi Jin
- School of Pharmaceutical Sciences, Guangzhou Higher Education Mega Center, Guangzhou University of Chinese Medicine, 232, Waihuan East Road, Panyu, Guangzhou, 510000, China
| | - Yong Luo
- School of Pharmaceutical Sciences, Guangzhou Higher Education Mega Center, Guangzhou University of Chinese Medicine, 232, Waihuan East Road, Panyu, Guangzhou, 510000, China
- Guangdong Key Laboratory of TCM Pathogenesis and Prescriptions Realted to Heart and Spleen, Guangzhou Higher Education Mega Center, 232, Waihuan East Road, Panyu, Guangzhou, 510000, China
| | - Qiuhe Chen
- School of Pharmaceutical Sciences, Guangzhou Higher Education Mega Center, Guangzhou University of Chinese Medicine, 232, Waihuan East Road, Panyu, Guangzhou, 510000, China
- Guangdong Key Laboratory of TCM Pathogenesis and Prescriptions Realted to Heart and Spleen, Guangzhou Higher Education Mega Center, 232, Waihuan East Road, Panyu, Guangzhou, 510000, China
| | - Wei Wang
- School of Pharmaceutical Sciences, Guangzhou Higher Education Mega Center, Guangzhou University of Chinese Medicine, 232, Waihuan East Road, Panyu, Guangzhou, 510000, China.
- Guangdong Key Laboratory of TCM Pathogenesis and Prescriptions Realted to Heart and Spleen, Guangzhou Higher Education Mega Center, 232, Waihuan East Road, Panyu, Guangzhou, 510000, China.
| | - Cuixian Zhang
- School of Pharmaceutical Sciences, Guangzhou Higher Education Mega Center, Guangzhou University of Chinese Medicine, 232, Waihuan East Road, Panyu, Guangzhou, 510000, China.
| | - Yang Chen
- School of Pharmaceutical Sciences, Guangzhou Higher Education Mega Center, Guangzhou University of Chinese Medicine, 232, Waihuan East Road, Panyu, Guangzhou, 510000, China.
- Guangdong Key Laboratory of TCM Pathogenesis and Prescriptions Realted to Heart and Spleen, Guangzhou Higher Education Mega Center, 232, Waihuan East Road, Panyu, Guangzhou, 510000, China.
| |
Collapse
|
7
|
Yin W, Li X, Liu P, Li Y, Liu J, Yu S, Tai S. Digestive system deep infiltrating endometriosis: What do we know. J Cell Mol Med 2023; 27:3649-3661. [PMID: 37632165 PMCID: PMC10718155 DOI: 10.1111/jcmm.17921] [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: 06/19/2023] [Revised: 08/06/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023] Open
Abstract
Digestive system infiltrating endometriosis (DSIE) is an uncommon form of endometriosis in the digestive system. DSIE often occurs in the intestines (especially the sigmoid rectum), liver, gallbladder and pancreas. Clinically, DSIE presents with the same symptoms as endometriosis, including cyclic pain, bleeding and infertility, in addition to specific biliary/intestinal obstruction and gastrointestinal bleeding. Compared to general endometriosis, DSIE has unique biological behaviour and pathophysiological mechanisms. Most DSIEs are deep invasive endometrioses, characterized by metastasis to the lymph nodes and lymphatic vessels, angiogenesis, peripheral nerve recruitment, fibrosis and invasion of surrounding tissues. DSIE-related peripheral angiogenesis is divided into three patterns: angiogenesis, vasculogenesis and inosculation. These patterns are regulated by interactions between multiple hypoxia-hormone cytokines. The nerve growth factors regulate the extensive neurofibril recruitment in DSIE lesions, which accounts for severe symptoms of deep pain. They are also associated with fibrosis and the aggressiveness of DSIE. Cyclic changes in DSIE lesions, recurrent inflammation and oxidative stress promote repeated tissue injury and repair (ReTIAR) mechanisms in the lesions, accelerating fibril formation and cancer-related mutations. Similar to malignant tumours, DSIE can also exhibit aggressiveness derived from collective cell migration mediated by E-cadherin and N-cadherin. This often makes DSIE misdiagnosed as a malignant tumour of the digestive system in clinical practice. In addition to surgery, novel treatments are urgently required to effectively eradicate this lesion.
Collapse
Affiliation(s)
- Wenze Yin
- Department of Hepatic SurgerySecond Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - Xiaoqing Li
- Department of PathologySecond Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - Peng Liu
- Laboratory of Medical GeneticsHarbin Medical UniversityHarbinChina
| | - Yingjie Li
- Department of PathologySix Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - Jin Liu
- Department of PathologySecond Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - Shan Yu
- Department of PathologySecond Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - Sheng Tai
- Department of Hepatic SurgerySecond Affiliated Hospital of Harbin Medical UniversityHarbinChina
| |
Collapse
|
8
|
Dai XS, Wei QH, Guo X, Ding Y, Yang XQ, Zhang YX, Xu XY, Li C, Chen Y. Ferulic acid, ligustrazine, and tetrahydropalmatine display the anti-proliferative effect in endometriosis through regulating Notch pathway. Life Sci 2023; 328:121921. [PMID: 37429417 DOI: 10.1016/j.lfs.2023.121921] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/30/2023] [Accepted: 07/06/2023] [Indexed: 07/12/2023]
Abstract
AIMS With an ambiguous anti-proliferative mechanism, the combination of ferulic acid, ligustrazine, and tetrahydropalmatine (FLT) shows good anti-endometriosis (EMS) activity. In EMS, the expression of Notch pathway and its role in proliferation are not yet unclear. In this study, we sought to uncover the role of Notch pathway's effect and FLT's anti-proliferative mechanism on EMS proliferation. MAIN METHODS In autograft and allograft EMS models, the proliferating markers (Ki67, PCNA), Notch pathway, and the effect of FLT on them were detected. Then, the anti-proliferative influence of FLT was measured in vitro. The proliferating ability of endometrial cells was investigated with a Notch pathway activator (Jagged 1 or VPA) or inhibitor (DAPT) alone, or in combination with FLT separately. KEY FINDINGS FLT presented the inhibitory effect on ectopic lesions in 2 EMS models. The proliferating markers and Notch pathway were promoted in ectopic endometrium, but FLT showed the counteraction. Meantime, FLT restrained the endometrial cell growth and clone formation along with a reduction in Ki67 and PCNA. Jagged 1 and VPA stimulated the proliferation. On the contrary, DAPT displayed the anti-proliferating effect. Furthermore, FLT exhibited an antagonistic effect on Jagged 1 and VPA by downregulating Notch pathway and restraining proliferation. FLT also displayed a synergistic effect on DAPT. SIGNIFICANCE This study indicated that the overexpressing Notch pathway induced EMS proliferation. FLT attenuated the proliferation by inhibiting Notch pathway.
Collapse
Affiliation(s)
- Xue-Shan Dai
- College of Pharmaceutical Sciences & Chinese Medicine, Southwest University, Chongqing, China; Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, Chongqing, China; Engineering Research Center of Coptis Development and Utilization, Ministry of Education, College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China; National Demonstration Center for Experimental Pharmacy Education, Southwest University, Chongqing, China
| | - Qing-Hua Wei
- College of Pharmaceutical Sciences & Chinese Medicine, Southwest University, Chongqing, China; Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, Chongqing, China; Engineering Research Center of Coptis Development and Utilization, Ministry of Education, College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China; National Demonstration Center for Experimental Pharmacy Education, Southwest University, Chongqing, China
| | - Xin Guo
- College of Pharmaceutical Sciences & Chinese Medicine, Southwest University, Chongqing, China; Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, Chongqing, China; Engineering Research Center of Coptis Development and Utilization, Ministry of Education, College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China; National Demonstration Center for Experimental Pharmacy Education, Southwest University, Chongqing, China
| | - Yi Ding
- College of Pharmaceutical Sciences & Chinese Medicine, Southwest University, Chongqing, China; Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, Chongqing, China; Engineering Research Center of Coptis Development and Utilization, Ministry of Education, College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China; National Demonstration Center for Experimental Pharmacy Education, Southwest University, Chongqing, China
| | - Xiao-Qian Yang
- College of Pharmaceutical Sciences & Chinese Medicine, Southwest University, Chongqing, China; Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, Chongqing, China; Engineering Research Center of Coptis Development and Utilization, Ministry of Education, College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China; National Demonstration Center for Experimental Pharmacy Education, Southwest University, Chongqing, China
| | - Yu-Xin Zhang
- College of Pharmaceutical Sciences & Chinese Medicine, Southwest University, Chongqing, China; Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, Chongqing, China; Engineering Research Center of Coptis Development and Utilization, Ministry of Education, College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China; National Demonstration Center for Experimental Pharmacy Education, Southwest University, Chongqing, China
| | - Xiao-Yu Xu
- College of Pharmaceutical Sciences & Chinese Medicine, Southwest University, Chongqing, China; Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, Chongqing, China; Engineering Research Center of Coptis Development and Utilization, Ministry of Education, College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China; National Demonstration Center for Experimental Pharmacy Education, Southwest University, Chongqing, China
| | - Cong Li
- Department of Obstetrics & Gynecology, First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yi Chen
- College of Pharmaceutical Sciences & Chinese Medicine, Southwest University, Chongqing, China; Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, Chongqing, China; Engineering Research Center of Coptis Development and Utilization, Ministry of Education, College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China; National Demonstration Center for Experimental Pharmacy Education, Southwest University, Chongqing, China.
| |
Collapse
|
9
|
Zhang M, Shi Z, Peng X, Cai D, Peng R, Lin Y, Dai L, Li J, Chen Y, Xiao J, Dong S, Wang W, Chen Y, He H. NLRP3 inflammasome-mediated Pyroptosis induce Notch signal activation in endometriosis angiogenesis. Mol Cell Endocrinol 2023:111952. [PMID: 37268099 DOI: 10.1016/j.mce.2023.111952] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 05/06/2023] [Accepted: 05/09/2023] [Indexed: 06/04/2023]
Abstract
Endometriosis is characterized by the presence of endometrial tissue outside the uterus that not only causes severe pelvic pain and infertility but also increased risk for ovarian carcinogenesis in women of reproductive age. Here, we found that angiogenesis was increased and accompanied with up-regulation of Notch1 in human endometriotic tissue sample, which is associated with pyroptosis induced by activation of endothelial NLRP3 inflammasome. Further, in endometriosis model induced in wild type and NLRP3-deficient (NLRP3-KO) mice, we found that deficiency of NLRP3 suppressing the development of endometriosis. In vitro, inhibiting the activation of NLRP3 inflammasome prevents LPS/ATP-induced tube formation in endothelial cells. Meanwhile, knockdown NLRP3 expression by gRNA disrupt the interaction between NICD and HIF-1α under the inflammatory microenvironment. This study demonstrates that activation of NLRP3 inflammasome-mediated pyroptosis affects angiogenesis in endometriosis via Notch1-dependent manner.
Collapse
Affiliation(s)
- Minyi Zhang
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Zhimian Shi
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Xianglan Peng
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, Guangdong, China
| | - Dongpeng Cai
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Rui Peng
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, Guangdong, China
| | - Yike Lin
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Linfeng Dai
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Jieyi Li
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Yulin Chen
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Jing Xiao
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
| | - Su Dong
- Department of Pharmacy, People's Hospital of Dongxihu District, Wuhan, 430040, Hubei, China
| | - Wei Wang
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China.
| | - Yang Chen
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China.
| | - Hong He
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, Guangdong, China.
| |
Collapse
|
10
|
Beter M, Abdollahzadeh A, Pulkkinen HH, Huang H, Orsenigo F, Magnusson PU, Ylä-Herttuala S, Tohka J, Laakkonen JP. SproutAngio: an open-source bioimage informatics tool for quantitative analysis of sprouting angiogenesis and lumen space. Sci Rep 2023; 13:7279. [PMID: 37142637 PMCID: PMC10160097 DOI: 10.1038/s41598-023-33090-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 04/06/2023] [Indexed: 05/06/2023] Open
Abstract
Three-dimensional image analyses are required to improve the understanding of the regulation of blood vessel formation and heterogeneity. Currently, quantitation of 3D endothelial structures or vessel branches is often based on 2D projections of the images losing their volumetric information. Here, we developed SproutAngio, a Python-based open-source tool, for fully automated 3D segmentation and analysis of endothelial lumen space and sprout morphology. To test the SproutAngio, we produced a publicly available in vitro fibrin bead assay dataset with a gradually increasing VEGF-A concentration ( https://doi.org/10.5281/zenodo.7240927 ). We demonstrate that our automated segmentation and sprout morphology analysis, including sprout number, length, and nuclei number, outperform the widely used ImageJ plugin. We also show that SproutAngio allows a more detailed and automated analysis of the mouse retinal vasculature in comparison to the commonly used radial expansion measurement. In addition, we provide two novel methods for automated analysis of endothelial lumen space: (1) width measurement from tip, stalk and root segments of the sprouts and (2) paired nuclei distance analysis. We show that these automated methods provided important additional information on the endothelial cell organization in the sprouts. The pipelines and source code of SproutAngio are publicly available ( https://doi.org/10.5281/zenodo.7381732 ).
Collapse
Affiliation(s)
- M Beter
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, P.O.Box 1627, 70211, Kuopio, Finland
| | - A Abdollahzadeh
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, P.O.Box 1627, 70211, Kuopio, Finland
| | - H H Pulkkinen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, P.O.Box 1627, 70211, Kuopio, Finland
| | - H Huang
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - F Orsenigo
- Vascular Biology Unit, IFOM ETS - The AIRC Institute of Molecular Oncology, Milan, Italy
| | - P U Magnusson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - S Ylä-Herttuala
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, P.O.Box 1627, 70211, Kuopio, Finland
- Heart Center, Kuopio University Hospital, Kuopio, Finland
- Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland
| | - J Tohka
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, P.O.Box 1627, 70211, Kuopio, Finland
| | - J P Laakkonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, P.O.Box 1627, 70211, Kuopio, Finland.
| |
Collapse
|
11
|
Lin C, Zeng S, Li M. miR-424-5p combined with miR-17-5p has high diagnostic efficacy for endometriosis. Arch Gynecol Obstet 2023; 307:169-177. [PMID: 35366691 DOI: 10.1007/s00404-022-06492-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/25/2022] [Indexed: 02/02/2023]
Abstract
PURPOSE Endometriosis (EMT) is a chronic benign disease with high prevalence. This study investigated the diagnostic value of serum miR-17-5p, miR-424-5p, and their combined expressions for EMT. METHODS Total 80 EMT patients of reproductive age who underwent laparoscopy or laparotomy and were confirmed by pathological examination were included as the study subjects, and another 80 healthy women of reproductive age receiving gynecological examination and ultrasonography with no pelvic abnormalities were selected as the control group. The whole blood samples of enrolled subjects were collected and clinical characteristics were recorded. The miR-17-5p, miR-424-5p, VEGFA, IL-4, and IL-6 levels in the serum were measured. ROC curve was used to evaluate the diagnostic efficacy of miR-17-5p and miR-424-5p expressions for EMT. Pearson correlation was performed to analyze the correlation of miR-17-5p and miR-424-5p with clinical indexes in EMT patients. RESULTS miR-17-5p and miR-424-5p were downregulated in EMT patients. For diagnosing EMT, the AUC of miR-17-5p was 0.865 and cutoff value was 0.890 (91.3% sensitivity and 85% specificity), the AUC of miR-424-5p was 0.737, and cutoff value was 0.915 (98.8% sensitivity and 61.2% specificity), and the AUC of miR-424-5p combined with miR-17-5p was 0.938 and cutoff value was 2.205 (93.8% sensitivity and 88.7% specificity), with the diagnostic efficacy higher than miR-424-5p or miR-17-5p alone. miR-17-5p and miR-424-5p expressions were negatively correlated with dysmenorrhea, infertility, pelvic pain, and rASRM stage, but not with age, BMI, menstrual disorder, and nulliparity. VEGFA, IL-4, IL-6, and CA-125 were increased in EMT patients and were inversely associated with miR-17-5p and miR-424-5p. CONCLUSION miR-424-5p combined with miR-17-5p has high diagnostic efficacy for EMT.
Collapse
Affiliation(s)
- Chunli Lin
- Department of Gynecology, Hunan Province Maternal and Child Health Care Hospital, 53 Xiangchun Road, Kaifu District, Changsha, 410008, Hunan, China.
| | - Saili Zeng
- Department of Respiratory Medicine, The Second Hospital of University of South China, 30 Jiefang Road, Shigu District, Hengyang, 421000, Hunan, China.
| | - Miaojie Li
- Department of Gynecology, People's Hospital of Yuxi City, Yuxi, 653100, Yunnan, China
| |
Collapse
|
12
|
Yan WK, Liu YN, Song SS, Kang JW, Zhang Y, Lu L, Wei SW, Xu QX, Zhang WQ, Liu XZ, Wu Y, Su RW. Zearalenone affects the growth of endometriosis via estrogen signaling and inflammatory pathways. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 241:113826. [PMID: 36068753 DOI: 10.1016/j.ecoenv.2022.113826] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 06/22/2022] [Accepted: 06/27/2022] [Indexed: 06/15/2023]
Abstract
Endometriosis is a chronic, inflammatory, estrogen-dependent gynecological disease characterized by the growth of endometrial stromal cells and glands outside the uterine cavity in response to hormones, which commonly occurs in reproductive-age women. Zearalenone (ZEA) is a toxic metabolite produced by Fusarium, which acts as estrogen activity because of the similarity of its structure to estrogen. In this study, we used an endometriosis mouse model: 15 days after ovariectomy, endometrial fragments were sutured on the pelvic wall, and exogenous estrogen was supplied using an estrogen-releasing silicone tube embedded subcutaneously. Mice were treated with different doses of ZEA by gavage for 21 days. The results show that ZEA significantly inhibited the growth of ectopic endometrium in a dose-dependent manner. The proliferation of cells decreased while apoptosis increased in the ectopic tissues of ZEA-treated mice compared to the vehicle group. The expression of estrogen receptor-α and its downstream targets MUC1 and p-AKT decreased, indicating an impaired estrogen signaling activity by ZEA treatment. In addition, the decreased expression of pro-inflammatory cytokine Tnf-α, Il-1β, and Il-6, the lower number of macrophages and neutrophils cells, and the inhibited NF-κB signaling pathway suggest the inflammatory response in the ectopic endometrium was also suppressed by ZEA treatment. However, when the exogenous estrogen supply is removed, ZEA, in turn, plays an estrogen-like role that promotes cell proliferation in the ectopic endometrium. In summary, our data suggest ZEA acts as an antagonist in endometriotic tissue when estrogen is sufficient but turns to estrogenic activity in the absence of estrogen in the development of endometriosis. ZEA also inhibits ectopic tissue growth by inhibiting inflammatory response in the endometriosis model.
Collapse
Affiliation(s)
- Wan-Kun Yan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Ying-Nan Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Shan-Shan Song
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Jin-Wen Kang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Yu Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Lei Lu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Shu-Wen Wei
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Qi-Xin Xu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Wang-Qing Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Xiao-Zheng Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Yao Wu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Ren-Wei Su
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China.
| |
Collapse
|
13
|
Notch Signaling Induced by Endoplasmic Reticulum Stress Regulates Cumulus-Oocyte Complex Expansion in Polycystic Ovary Syndrome. Biomolecules 2022; 12:biom12081037. [PMID: 36008931 PMCID: PMC9405998 DOI: 10.3390/biom12081037] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/16/2022] [Accepted: 07/25/2022] [Indexed: 12/10/2022] Open
Abstract
Endoplasmic reticulum (ER) stress activated in granulosa cells contributes to the pathophysiology of polycystic ovary syndrome (PCOS). In addition, recent studies have demonstrated that Notch signaling plays multiple roles in the ovary via cell-to-cell interactions. We hypothesized that ER stress activated in granulosa cells of antral follicles in PCOS induces Notch signaling in these cells, and that activated Notch signaling induces aberrant cumulus-oocyte complex (COC) expansion. Expression of Notch2 and Notch-target transcription factors was increased in granulosa cells of PCOS patients and model mice. ER stress increased expression of Notch2 and Notch-target transcription factors in cultured human granulosa-lutein cells (GLCs). Inhibition of Notch signaling abrogated ER stress-induced expression of genes associated with COC expansion in cultured human GLCs, as well as ER stress-enhanced expansion of cumulus cells in cultured murine COCs. Furthermore, inhibition of Notch signaling reduced the areas of COCs in PCOS model mice with activated ER stress in the ovary, indicating that Notch signaling regulates COC expansion in vivo. Our findings suggest that Notch2 signaling is activated in granulosa cells in PCOS and regulates COC expansion. It remains to be elucidated whether aberrant COC expansion induced by the ER stress-Notch pathway is associated with ovulatory dysfunction in PCOS patients.
Collapse
|
14
|
Huang J, Mao Y, Li Q, Hong H, Tang N, Kang X, Huang Y, Liu J, Gong Q, Yao Y, Li L. Kallistatin prevents ovarian hyperstimulation syndrome by regulating vascular leakage. J Cell Mol Med 2022; 26:4613-4623. [PMID: 35866203 PMCID: PMC9357611 DOI: 10.1111/jcmm.17491] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 06/12/2022] [Accepted: 06/28/2022] [Indexed: 11/30/2022] Open
Abstract
Angiogenesis and increased permeability are essential pathological basis for the development of ovarian hyperstimulation syndrome (OHSS). Kallistatin (KS) is an endogenous anti-inflammatory and anti-angiogenic factor that participates in a variety of diseases, but its role in OHSS remains unknown. In this study, treating a human ovarian granulosa-like tumour cell line KGN and human primary granulosa cells (PGCs) with human chorionic gonadotropin (hCG) reduced the expression of KS, but increased the expression of VEGF. Furthermore, we found that KS could attenuate the protein level of VEGF in both KGN cells and human PGCs. More interestingly, we observed that exogenous supplementation of KS significantly inhibited a series of signs of OHSS in mice, including weight gain, ovarian enlargement, increased vascular permeability and up-regulation of VEGF expression. In addition, KS was proved to be safe on mice ovulation, progression of normal pregnancy and fetus development. Collectively, these findings demonstrated that KS treatment prevented OHSS, at least partially, through down-regulating VEGF expression. For the first time, these results highlight the potential preventive value of KS in OHSS.
Collapse
Affiliation(s)
- Jianfang Huang
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Department of Obstetrics and Gynecology, Center for Reproductive Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Reproductive Medicine, Ganzhou People's Hospital, Ganzhou, Jiangxi, China
| | - Yuling Mao
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Department of Obstetrics and Gynecology, Center for Reproductive Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Quanxin Li
- Department of Biochemistry and Molecular Biology, GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Honghai Hong
- Department of Clinical Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ni Tang
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Department of Obstetrics and Gynecology, Center for Reproductive Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiangjin Kang
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Department of Obstetrics and Gynecology, Center for Reproductive Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuling Huang
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Department of Obstetrics and Gynecology, Center for Reproductive Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jianqiao Liu
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Department of Obstetrics and Gynecology, Center for Reproductive Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qing Gong
- Department of Biochemistry and Molecular Biology, GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yachao Yao
- Department of Laboratory Medicine, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Lei Li
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Department of Obstetrics and Gynecology, Center for Reproductive Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Key Laboratory for Reproductive Medicine of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
15
|
Yu J, Lee S, Song J, Lee SR, Kim S, Choi H, Kang H, Hwang Y, Hong YK, Jeon NL. Perfusable micro-vascularized 3D tissue array for high-throughput vascular phenotypic screening. NANO CONVERGENCE 2022; 9:16. [PMID: 35394224 PMCID: PMC8994007 DOI: 10.1186/s40580-022-00306-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 03/15/2022] [Indexed: 05/14/2023]
Abstract
Microfluidic organ-on-a-chip technologies have enabled construction of biomimetic physiologically and pathologically relevant models. This paper describes an injection molded microfluidic platform that utilizes a novel sequential edge-guided patterning method based on spontaneous capillary flow to realize three-dimensional co-culture models and form an array of micro-vascularized tissues (28 per 1 × 2-inch slide format). The MicroVascular Injection-Molded Plastic Array 3D Culture (MV-IMPACT) platform is fabricated by injection molding, resulting in devices that are reliable and easy to use. By patterning hydrogels containing human umbilical endothelial cells and fibroblasts in close proximity and allowing them to form vasculogenic networks, an array of perfusable vascularized micro-tissues can be formed in a highly efficient manner. The high-throughput generation of angiogenic sprouts was quantified and their uniformity was characterized. Due to its compact design (half the size of a 96-well microtiter plate), it requires small amount of reagents and cells per device. In addition, the device design is compatible with a high content imaging machine such as Yokogawa CQ-1. Furthermore, we demonstrated the potential of our platform for high-throughput phenotypic screening by testing the effect of DAPT, a chemical known to affect angiogenesis. The MV-IMPACT represent a significant improvement over our previous PDMS-based devices in terms of molding 3D co-culture conditions at much higher throughput with added reliability and robustness in obtaining vascular micro-tissues and will provide a platform for developing applications in drug screening and development.
Collapse
Affiliation(s)
- James Yu
- Interdisciplinary Program in Bioengineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Somin Lee
- Interdisciplinary Program in Bioengineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Jiyoung Song
- Department of Mechanical Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Seung-Ryeol Lee
- Department of Mechanical Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Suryong Kim
- Department of Mechanical Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Hyeri Choi
- Interdisciplinary Program in Bioengineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Habin Kang
- Interdisciplinary Program in Bioengineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Yunchan Hwang
- Department of Electrical Engineering and Computer Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Young-Kwon Hong
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Noo Li Jeon
- Interdisciplinary Program in Bioengineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea.
- Department of Mechanical Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea.
- Institute of Advanced Machines and Design, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea.
| |
Collapse
|
16
|
Impact of Musashi-1 and Musashi-2 Double Knockdown on Notch Signaling and the Pathogenesis of Endometriosis. Int J Mol Sci 2022; 23:ijms23052851. [PMID: 35269992 PMCID: PMC8911246 DOI: 10.3390/ijms23052851] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/25/2022] [Accepted: 03/02/2022] [Indexed: 12/12/2022] Open
Abstract
The stem cell marker and RNA-binding protein Musashi-1 is overexpressed in endometriosis. Musashi-1-siRNA knockdown in Ishikawa cells altered the expression of stem cell related genes, such as OCT-4. To investigate the role of both human Musashi homologues (MSI-1 and MSI-2) in the pathogenesis of endometriosis, immortalized endometriotic 12-Z cells and primary endometriotic stroma cells were treated with Musashi-1- and Musashi-2-siRNA. Subsequently, the impact on cell proliferation, cell apoptosis, cell necrosis, spheroid formation, stem cell phenotype and the Notch signaling pathway was studied in vitro. Using the ENDOMET Turku Endometriosis database, the gene expression of stem cell markers and Notch signaling pathway constituents were analyzed according to localization of the endometriosis lesions. The database analysis demonstrated that expression of Musashi and Notch pathway-related genes are dysregulated in patients with endometriosis. Musashi-1/2-double-knockdown increased apoptosis and necrosis and reduced stem cell gene expression, cell proliferation, and the formation of spheroids. Musashi silencing increased the expression of the anti-proliferation mediator p21. Our findings suggest the therapeutic potential of targeting the Musashi–Notch axis. We conclude that the Musashi genes have an impact on Notch signaling and the pathogenesis of endometriosis through the downregulation of proliferation, stemness characteristics and the upregulation of apoptosis, necrosis and of the cell cycle regulator p21.
Collapse
|
17
|
Moldovan GE, Miele L, Fazleabas AT. Notch signaling in reproduction. Trends Endocrinol Metab 2021; 32:1044-1057. [PMID: 34479767 PMCID: PMC8585702 DOI: 10.1016/j.tem.2021.08.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/27/2021] [Accepted: 08/06/2021] [Indexed: 12/22/2022]
Abstract
The Notch signaling pathway is conserved among mammalian species and controls proliferation, differentiation, and cell death in many organs throughout the body including the reproductive tract. Notch signaling plays critical roles in the development and function of both the male and female reproductive systems. Specifically, within the female reproductive tract, Notch signaling is hormone regulated and mediates key reproductive events important for ovarian and uterine function. In this review, we highlight the tissues that express Notch receptors, ligands, and downstream effectors and distinguish how these molecules regulate reproductive function in male and female mice, non-human primates, and humans. Finally, we describe some of the aberrations in Notch signaling in female reproductive pathologies and identify opportunities for future investigation.
Collapse
Affiliation(s)
- Genna E Moldovan
- Department of Obstetrics, Gynecology, and Reproductive Biology, Michigan State University, Grand Rapids, MI 49503, USA
| | - Lucio Miele
- Department of Genetics, Louisiana State University Health Sciences Center and Stanley S. Scott Cancer Center, New Orleans, LA 70112, USA
| | - Asgerally T Fazleabas
- Department of Obstetrics, Gynecology, and Reproductive Biology, Michigan State University, Grand Rapids, MI 49503, USA.
| |
Collapse
|
18
|
Grambow E, Sorg H, Sorg CGG, Strüder D. Experimental Models to Study Skin Wound Healing with a Focus on Angiogenesis. Med Sci (Basel) 2021; 9:medsci9030055. [PMID: 34449673 PMCID: PMC8395822 DOI: 10.3390/medsci9030055] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/16/2021] [Accepted: 08/18/2021] [Indexed: 12/11/2022] Open
Abstract
A large number of models are now available for the investigation of skin wound healing. These can be used to study the processes that take place in a phase-specific manner under both physiological and pathological conditions. Most models focus on wound closure, which is a crucial parameter for wound healing. However, vascular supply plays an equally important role and corresponding models for selective or parallel investigation of microcirculation regeneration and angiogenesis are also described. In this review article, we therefore focus on the different levels of investigation of skin wound healing (in vivo to in virtuo) and the investigation of angiogenesis and its parameters.
Collapse
Affiliation(s)
- Eberhard Grambow
- Department of General, Visceral, Thoracic, Vascular and Transplantation Surgery, Rostock University Medical Center, 18057 Rostock, Germany
- Correspondence:
| | - Heiko Sorg
- Department of Health, University of Witten/Herdecke, Alfred-Herrhausen-Str. 50, 58455 Witten, Germany;
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, Klinikum Westfalen, Am Knappschaftskrankenhaus 1, 44309 Dortmund, Germany
| | - Christian G. G. Sorg
- Chair of Management and Innovation in Health Care, Department of Management and Entrepreneurship, Faculty of Management, Economics and Society, Witten/Herdecke University, Alfred-Herrhausen-Straße 50, 58455 Witten, Germany;
| | - Daniel Strüder
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery “Otto Körner”, Rostock University Medical Center, 18057 Rostock, Germany;
| |
Collapse
|
19
|
Nenicu A, Yordanova K, Gu Y, Menger MD, Laschke MW. Differences in growth and vascularization of ectopic menstrual and non-menstrual endometrial tissue in mouse models of endometriosis. Hum Reprod 2021; 36:2202-2214. [PMID: 34109385 DOI: 10.1093/humrep/deab139] [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: 01/11/2021] [Revised: 03/29/2021] [Indexed: 11/12/2022] Open
Abstract
STUDY QUESTION Is there a difference in the growth and vascularization between murine endometriotic lesions originating from menstrual or non-menstrual endometrial fragments? SUMMARY ANSWER Endometriotic lesions developing from menstrual and non-menstrual tissue fragments share many similarities, but also exhibit distinct differences in growth and vascularization, particularly under exogenous estrogen stimulation. WHAT IS KNOWN ALREADY Mouse models are increasingly used in endometriosis research. For this purpose, menstrual or non-menstrual endometrial fragments serve for the induction of endometriotic lesions. So far, these two fragment types have never been directly compared under identical experimental conditions. STUDY DESIGN, SIZE, DURATION This was a prospective experimental study in a murine peritoneal and dorsal skinfold chamber model of endometriosis. Endometrial tissue fragments from menstruated (n = 15) and non-menstruated (n = 21) C57BL/6 mice were simultaneously transplanted into the peritoneal cavity or dorsal skinfold chamber of non-ovariectomized (non-ovx, n = 17), ovariectomized (ovx, n = 17) and ovariectomized, estrogen-substituted (ovx+E2, n = 17) recipient animals and analyzed throughout an observation period of 28 and 14 days, respectively. PARTICIPANTS/MATERIALS, SETTING, METHODS The engraftment, growth and vascularization of the newly developing endometriotic lesions were analyzed by means of high-resolution ultrasound imaging, intravital fluorescence microscopy, histology and immunohistochemistry. MAIN RESULTS AND THE ROLE OF CHANCE Menstrual and non-menstrual tissue fragments developed into peritoneal endometriotic lesions without differences in growth, microvessel density and cell proliferation in non-ovx mice. Lesion formation out of both fragment types was markedly suppressed in ovx mice. In case of non-menstrual tissue fragments, this effect could be reversed by estrogen supplementation. In contrast, endometriotic lesions originating from menstrual tissue fragments exhibited a significantly smaller volume in ovx+E2 mice, which may be due to a reduced hormone sensitivity. Moreover, menstrual tissue fragments showed a delayed vascularization and a reduced blood perfusion after transplantation into dorsal skinfold chambers when compared to non-menstrual tissue fragments, indicating different vascularization modes of the two fragment types. To limit the role of chance, the experiments were conducted under standardized laboratory conditions. Statistical significance was accepted for a value of P < 0.05. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION Endometriotic lesions were induced by syngeneic tissue transplantation into recipient mice without the use of pathological endometriotic tissue of human nature. Therefore, the results obtained in this study may not fully relate to human patients with endometriosis. WIDER IMPLICATIONS OF THE FINDINGS The present study significantly contributes to the characterization of common murine endometriosis models. These models represent important tools for studies focusing on the basic mechanisms of endometriosis and the development of novel therapeutic strategies for the treatment of this frequent gynecological disease. The presented findings indicate that the combination of different experimental models and approaches may be the most appropriate strategy to study the pathophysiology and drug sensitivity of a complex disease such as endometriosis under preclinical conditions. STUDY FUNDING/COMPETING INTEREST(S) There was no specific funding of this study. The authors have no conflicts of interest to declare.
Collapse
Affiliation(s)
- A Nenicu
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - K Yordanova
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Y Gu
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - M D Menger
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - M W Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| |
Collapse
|
20
|
Angiogenic Properties of NK Cells in Cancer and Other Angiogenesis-Dependent Diseases. Cells 2021; 10:cells10071621. [PMID: 34209508 PMCID: PMC8303392 DOI: 10.3390/cells10071621] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 06/25/2021] [Accepted: 06/26/2021] [Indexed: 12/23/2022] Open
Abstract
The pathogenesis of many serious diseases, including cancer, is closely related to disturbances in the angiogenesis process. Angiogenesis is essential for the progression of tumor growth and metastasis. The tumor microenvironment (TME) has immunosuppressive properties, which contribute to tumor expansion and angiogenesis. Similarly, the uterine microenvironment (UME) exerts a tolerogenic (immunosuppressive) and proangiogenic effect on its cells, promoting implantation and development of the embryo and placenta. In the TME and UME natural killer (NK) cells, which otherwise are capable of killing target cells autonomously, enter a state of reduced cytotoxicity or anergy. Both TME and UME are rich with factors (e.g., TGF-β, glycodelin, hypoxia), which support a conversion of NK cells to the low/non-cytotoxic, proangiogenic CD56brightCD16low phenotype. It is plausible that the phenomenon of acquiring proangiogenic and low cytotoxic features by NK cells is not only limited to cancer but is a common feature of different angiogenesis-dependent diseases (ADDs). In this review, we will discuss the role of NK cells in angiogenesis disturbances associated with cancer and other selected ADDs. Expanding the knowledge of the mechanisms responsible for angiogenesis and its disorders contributes to a better understanding of ADDs and may have therapeutic implications.
Collapse
|
21
|
Suszczyk D, Skiba W, Jakubowicz-Gil J, Kotarski J, Wertel I. The Role of Myeloid-Derived Suppressor Cells (MDSCs) in the Development and/or Progression of Endometriosis-State of the Art. Cells 2021; 10:cells10030677. [PMID: 33803806 PMCID: PMC8003224 DOI: 10.3390/cells10030677] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/15/2021] [Accepted: 03/15/2021] [Indexed: 02/06/2023] Open
Abstract
Endometriosis (EMS) is a common gynecological disease characterized by the presence of endometrial tissue outside the uterus. Approximately 10% of women around the world suffer from this disease. Recent studies suggest that endometriosis has potential to transform into endometriosis-associated ovarian cancer (EAOC). Endometriosis is connected with chronic inflammation and changes in the phenotype, activity, and function of immune cells. The underlying mechanisms include quantitative and functional disturbances of neutrophils, monocytes/macrophages (MO/MA), natural killer cells (NK), and T cells. A few reports have shown that immunosuppressive cells such as regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) may promote the progression of endometriosis. MDSCs are a heterogeneous population of immature myeloid cells (dendritic cells, granulocytes, and MO/MA precursors), which play an important role in the development of immunological diseases such as chronic inflammation and cancer. The presence of MDSCs in pathological conditions correlates with immunosuppression, angiogenesis, or release of growth factors and cytokines, which promote progression of these diseases. In this paper, we review the impact of MDSCs on different populations of immune cells, focusing on their immunosuppressive role in the immune system, which may be related with the pathogenesis and/or progression of endometriosis and its transformation into ovarian cancer.
Collapse
Affiliation(s)
- Dorota Suszczyk
- Independent Laboratory of Cancer Diagnostics and Immunology, Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, Staszica 16, 20-081 Lublin, Poland; (D.S.); (W.S.)
| | - Wiktoria Skiba
- Independent Laboratory of Cancer Diagnostics and Immunology, Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, Staszica 16, 20-081 Lublin, Poland; (D.S.); (W.S.)
| | - Joanna Jakubowicz-Gil
- Department of Functional Anatomy and Cytobiology, Maria Curie-Sklodowska University, Akademicka 19, 20-033 Lublin, Poland;
| | - Jan Kotarski
- Department of Gynaecologic Oncology and Gynaecology, Medical University of Lublin, Staszica 16, 20-081 Lublin, Poland;
| | - Iwona Wertel
- Independent Laboratory of Cancer Diagnostics and Immunology, Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, Staszica 16, 20-081 Lublin, Poland; (D.S.); (W.S.)
- Correspondence:
| |
Collapse
|
22
|
Yao Q, Xing Y, Wang Z, Liang J, Lin Q, Huang M, Chen Y, Lin B, Xu X, Chen W. MiR-16-5p suppresses myofibroblast activation in systemic sclerosis by inhibiting NOTCH signaling. Aging (Albany NY) 2020; 13:2640-2654. [PMID: 33411678 PMCID: PMC7880343 DOI: 10.18632/aging.202308] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 11/11/2020] [Indexed: 04/07/2023]
Abstract
Systemic sclerosis (SSc) is a prototypic fibrotic disease characterized by localized or diffuse skin thickening and fibrosis. Tissue fibrosis is driven by myofibroblasts, and factors affecting myofibroblast activation may also be involved in the development of SSc. In this study, we examined molecular mechanisms underlying SSc by focusing on myofibroblast activation processes. Bioinformatics analysis conducted to identify differentially expressed miRNAs (DEMs) and genes (DEGs) revealed that microRNA-16-5p (miR-16-5p) was downregulated and NOTCH2 was upregulated in SSc patients. In vitro experiments confirmed that miR-16-5p was able to bind directly to NOTCH2 and inhibit myofibroblast activation. Moreover, miR-16-5p-dependent inhibition of NOTCH2 decreased collagen and α-SMA expression. MiR-16-5p downregulation and NOTCH2 upregulation was also confirmed in vivo in SSc patients, and NOTCH2 activation promoted fibrosis progression in vitro. These results indicate that miR-16-5p suppresses myofibroblast activation by suppressing NOTCH signaling.
Collapse
Affiliation(s)
- Qicen Yao
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Hainan Medical University, Haikou 570311, Hainan Province, China
| | - Yixi Xing
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Hainan Medical University, Haikou 570311, Hainan Province, China
| | - Zaiyan Wang
- Department of Respiratory Medicine, The affiliated Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Jin Liang
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Hainan Medical University, Haikou 570311, Hainan Province, China
| | - Qianqi Lin
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Hainan Medical University, Haikou 570311, Hainan Province, China
| | - Meiqiong Huang
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Hainan Medical University, Haikou 570311, Hainan Province, China
| | - Yiling Chen
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Hainan Medical University, Haikou 570311, Hainan Province, China
| | - Bo Lin
- Department of Pharmacy, The Second Affiliated Hospital of Hainan Medical University, Haikou 570311, Hainan Province, China
| | - Xiayu Xu
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Hainan Medical University, Haikou 570311, Hainan Province, China
| | - Weifei Chen
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Hainan Medical University, Haikou 570311, Hainan Province, China
| |
Collapse
|
23
|
Yetkin-Arik B, Kastelein AW, Klaassen I, Jansen CHJR, Latul YP, Vittori M, Biri A, Kahraman K, Griffioen AW, Amant F, Lok CAR, Schlingemann RO, van Noorden CJF. Angiogenesis in gynecological cancers and the options for anti-angiogenesis therapy. Biochim Biophys Acta Rev Cancer 2020; 1875:188446. [PMID: 33058997 DOI: 10.1016/j.bbcan.2020.188446] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 10/02/2020] [Accepted: 10/04/2020] [Indexed: 02/06/2023]
Abstract
Angiogenesis is required in cancer, including gynecological cancers, for the growth of primary tumors and secondary metastases. Development of anti-angiogenesis therapy in gynecological cancers and improvement of its efficacy have been a major focus of fundamental and clinical research. However, survival benefits of current anti-angiogenic agents, such as bevacizumab, in patients with gynecological cancer, are modest. Therefore, a better understanding of angiogenesis and the tumor microenvironment in gynecological cancers is urgently needed to develop more effective anti-angiogenic therapies, either or not in combination with other therapeutic approaches. We describe the molecular aspects of (tumor) blood vessel formation and the tumor microenvironment and provide an extensive clinical overview of current anti-angiogenic therapies for gynecological cancers. We discuss the different phenotypes of angiogenic endothelial cells as potential therapeutic targets, strategies aimed at intervention in their metabolism, and approaches targeting their (inflammatory) tumor microenvironment.
Collapse
Affiliation(s)
- Bahar Yetkin-Arik
- Ocular Angiogenesis Group, Department of Ophthalmology, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands; Department of Medical Biology, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - Arnoud W Kastelein
- Department of Obstetrics and Gynaecology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands.
| | - Ingeborg Klaassen
- Ocular Angiogenesis Group, Department of Ophthalmology, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands; Department of Medical Biology, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - Charlotte H J R Jansen
- Department of Obstetrics and Gynaecology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - Yani P Latul
- Department of Obstetrics and Gynaecology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - Miloš Vittori
- Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Aydan Biri
- Department of Obstetrics and Gynecology, Koru Ankara Hospital, Ankara, Turkey
| | - Korhan Kahraman
- Department of Obstetrics and Gynecology, Bahcesehir University School of Medicine, Istanbul, Turkey
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Frederic Amant
- Department of Oncology, KU Leuven, Leuven, Belgium; Center for Gynaecological Oncology, Antoni van Leeuwenhoek, Amsterdam, the Netherlands; Center for Gynaecological Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands; Center for Gynaecological Oncology, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Christianne A R Lok
- Center for Gynaecological Oncology, Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - Reinier O Schlingemann
- Ocular Angiogenesis Group, Department of Ophthalmology, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands; Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Cornelis J F van Noorden
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands; Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| |
Collapse
|
24
|
Li C, Tan Y, Wu J, Ma Q, Bai S, Xia Z, Wan X, Liang J. Resveratrol Improves Bnip3-Related Mitophagy and Attenuates High-Fat-Induced Endothelial Dysfunction. Front Cell Dev Biol 2020; 8:796. [PMID: 32923443 PMCID: PMC7457020 DOI: 10.3389/fcell.2020.00796] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 07/28/2020] [Indexed: 12/16/2022] Open
Abstract
Statin treatment reduces cardiovascular risk. However, individuals with well-controlled low-density lipoprotein (LDL) levels may remain at increased risk owing to persistent high triglycerides and low high-density lipoprotein cholesterol. Because resveratrol promotes glucose metabolism and mitigates cardiovascular disorders, we explored its mechanism of protective action on high-fat-induced endothelial dysfunction. Human umbilical venous endothelial cells were treated with oxidized LDL (ox-LDL) in vitro. Endothelial function, cell survival, proliferation, migration, and oxidative stress were analyzed through western blots, quantitative polymerase chain reaction, ELISA, and immunofluorescence. ox-LDL induced endothelial cell apoptosis, proliferation arrest, and mobilization inhibition, all of which resveratrol reduced. ox-LDL suppressed the activities of mitochondrial respiration complex I and III and reduced levels of intracellular antioxidative enzymes, resulting in reactive oxygen species overproduction and mitochondrial dysfunction. Resveratrol treatment upregulated Bnip3-related mitophagy and prevented ox-LDL-mediated mitochondrial respiration complexes inactivation, sustaining mitochondrial membrane potential and favoring endothelial cell survival. We found that resveratrol enhanced Bnip3 transcription through hypoxia-inducible factor 1 (HIF1) and 5' AMP-activated protein kinase (AMPK). Inhibition of AMPK and HIF1 abolished resveratrol-mediated protection of mitochondrial redox balance and endothelial viability. Together, these data demonstrate resveratrol reduces hyperlipemia-related endothelial damage by preserving mitochondrial homeostasis.
Collapse
Affiliation(s)
- Chen Li
- Department of Cardiology, Foshan Hospital Affiliated with Southern Medical University (The Second People's Hospital of Foshan), Foshan, China
| | - Ying Tan
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiandi Wu
- Department of Cardiology, Foshan Hospital Affiliated with Southern Medical University (The Second People's Hospital of Foshan), Foshan, China
| | - Qinghui Ma
- Department of Oncology Hematology, Foshan Hospital Affiliated with Southern Medical University (The Second People's Hospital of Foshan), Foshan, China
| | - Shuchang Bai
- Department of Cardiology, Foshan Hospital Affiliated with Southern Medical University (The Second People's Hospital of Foshan), Foshan, China
| | - Zhangqing Xia
- Department of Cardiology, Foshan Hospital Affiliated with Southern Medical University (The Second People's Hospital of Foshan), Foshan, China
| | - Xiaoliang Wan
- Department of Cardiology, Foshan Hospital Affiliated with Southern Medical University (The Second People's Hospital of Foshan), Foshan, China
| | - Jianqiu Liang
- Department of Cardiology, Foshan Hospital Affiliated with Southern Medical University (The Second People's Hospital of Foshan), Foshan, China
| |
Collapse
|
25
|
Harmsen MJ, Wong CFC, Mijatovic V, Griffioen AW, Groenman F, Hehenkamp WJK, Huirne JAF. Role of angiogenesis in adenomyosis-associated abnormal uterine bleeding and subfertility: a systematic review. Hum Reprod Update 2020; 25:647-671. [PMID: 31504506 PMCID: PMC6737562 DOI: 10.1093/humupd/dmz024] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 06/11/2019] [Accepted: 06/19/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Adenomyosis commonly occurs with abnormal uterine bleeding (AUB) and is associated with subfertility and a higher miscarriage rate. Recent evidence showed abnormal vascularization in the endometrium in patients with adenomyosis, suggesting a role of angiogenesis in the pathophysiology of AUB and subfertility in adenomyosis and providing a possible treatment target. OBJECTIVE AND RATIONALE We hypothesized that the level of abnormal vascularization and expression of angiogenic markers is increased in the ectopic and eutopic endometrium of adenomyosis patients in comparison with the endometrium of control patients. This was investigated through a search of the literature. SEARCH METHODS A systematic search was performed in PubMed and Embase until February 2019. Combinations of terms for angiogenesis and adenomyosis were applied as well as AUB, subfertility or anti-angiogenic therapy. The main search was limited to clinical studies carried out on premenopausal women. Original research articles focusing on markers of angiogenesis in the endometrium of patients with adenomyosis were included. Studies in which no comparison was made to control patients or which were not published in a peer-reviewed journal were excluded. A second search was performed to explore the therapeutic potential of targeting angiogenesis in adenomyosis. This search also included preclinical studies. OUTCOMES A total of 20 articles out of 1669 hits met our selection criteria. The mean vascular density (MVD) was studied by quantification of CD31, CD34, von Willebrand Factor (vWF) or factor-VIII-antibody-stained microvessels in seven studies. All these studies reported a significantly increased MVD in ectopic endometrium, and out of the six articles that took it into account, four studies reported a significantly increased MVD in eutopic endometrium compared with control endometrium. Five articles showed a significantly higher vascular endothelial growth factor expression in ectopic endometrium and three articles in eutopic endometrium compared with control endometrium. The vascular and pro-angiogenic markers α-smooth muscle actin, endoglin, S100A13, vimentin, matrix metalloproteinases (MMPs), nuclear factor (NF)-kB, tissue factor (TF), DJ-1, phosphorylated mammalian target of rapamycin, activin A, folli- and myostatin, CD41, SLIT, roundabout 1 (ROBO1), cyclooxygenase-2, lysophosphatidic acid (LPA) 1,4-5, phospho signal transducer and activator of transcription 3 (pSTAT3), interleukin (IL)-6, IL-22 and transforming growth factor-β1 were increased in ectopic endometrium, and the markers S100A13, MMP-2 and -9, TF, follistatin, myostatin, ROBO1, LPA1 and 4-5, pSTAT3, IL-6 and IL-22 were increased in eutopic endometrium, compared with control endometrium. The anti-angiogenic markers E-cadherin, eukaryotic translation initiation factor 3 subunit and gene associated with retinoic-interferon-induced mortality 19 were decreased in ectopic endometrium and IL-10 in eutopic endometrium, compared with control endometrium. The staining level of vWF and two pro-angiogenic markers (NF-κB nuclear p65 and TF) correlated with AUB in patients with adenomyosis. We found no studies that investigated the possible relationship between markers of angiogenesis and subfertility in adenomyosis patients. Nine articles reported on direct or indirect targeting of angiogenesis in adenomyosis-either by testing hormonal therapy or herbal compounds in clinical studies or by testing angiogenesis inhibitors in preclinical studies. However, there are no clinical studies on the effectiveness of such therapy for adenomyosis-related AUB or subfertility. WIDER IMPLICATIONS The results are in agreement with our hypothesis that increased angiogenesis is present in the endometrium of patients with adenomyosis compared with the endometrium of control patients. It is likely that increased angiogenesis leads to fragile and more permeable vessels resulting in adenomyosis-related AUB and possibly subfertility. While this association has not sufficiently been studied yet, our results encourage future studies to investigate the exact role of angiogenesis in the etiology of adenomyosis and related AUB or subfertility in women with adenomyosis in order to design curative or preventive therapeutic strategies.
Collapse
Affiliation(s)
- Marissa J Harmsen
- Department of Obstetrics and Gynecology, Amsterdam UMC, location VUMC, Amsterdam, The Netherlands.,Angiogenesis Laboratory, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Caroline F C Wong
- Department of Obstetrics and Gynecology, Amsterdam UMC, location VUMC, Amsterdam, The Netherlands.,Angiogenesis Laboratory, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Velja Mijatovic
- Department of Obstetrics and Gynecology, Amsterdam UMC, location VUMC, Amsterdam, The Netherlands
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Freek Groenman
- Department of Obstetrics and Gynecology, Amsterdam UMC, location VUMC, Amsterdam, The Netherlands
| | - Wouter J K Hehenkamp
- Department of Obstetrics and Gynecology, Amsterdam UMC, location VUMC, Amsterdam, The Netherlands
| | - Judith A F Huirne
- Department of Obstetrics and Gynecology, Amsterdam UMC, location VUMC, Amsterdam, The Netherlands
| |
Collapse
|
26
|
Song Y, Su RW, Joshi NR, Kim TH, Lessey BA, Jeong JW, Fazleabas AT. Interleukin-6 (IL-6) Activates the NOTCH1 Signaling Pathway Through E-Proteins in Endometriotic Lesions. J Clin Endocrinol Metab 2020; 105:dgaa096. [PMID: 32119078 PMCID: PMC7096313 DOI: 10.1210/clinem/dgaa096] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 02/26/2020] [Indexed: 01/28/2023]
Abstract
CONTEXT NOTCH signaling is activated in endometriotic lesions, but the exact mechanisms remains unclear. IL-6, which is increased in the peritoneal fluid of women with endometriosis, induces NOTCH1 through E-proteins including E2A and HEB in cancer. OBJECTIVE To study the role of E-proteins in inducing NOTCH1 expression under the regulation of IL-6 in endometriosis. SETTING AND DESIGN The expression of E-proteins and NOTCH1 was first investigated in endometrium of women with endometriosis and the baboon model of endometriosis. Regulation of E-proteins and NOTCH1 expression was examined after IL-6 stimulation and siRNA mediated inhibition of E2A or/and HEB in human endometriotic epithelial cells (12Z) in vitro, and subsequently following IL-6 treatment in the mouse model of endometriosis in vivo. RESULTS E2A, HEB, and NOTCH1 were significantly upregulated in glandular epithelium (GE) of ectopic endometrium compared to eutopic endometrium in both women and the baboon model. IL-6 treatment upregulated the expression of NOTCH1 together with E2A and HEB in 12Z cells. Small interfering RNA inhibition of E2A and HEB or HEB alone decreased NOTCH1 expression. Binding efficiency of both E2A and HEB was significantly higher at the binding sites on the human NOTCH1 promoter after IL-6 treatment. Finally, IL-6 treatment resulted in a significantly increased number of endometriotic lesions along with increased expression of E2A, HEB, and NOTCH1 in GE of the lesions compared with the vehicle group in an endometriosis mouse model. CONCLUSIONS IL-6 induced NOTCH1 expression is mediated by E-proteins in the ectopic GE cells, which may promote endometriotic lesion development.
Collapse
Affiliation(s)
- Yong Song
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, Michigan, USA
| | - Ren-Wei Su
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Niraj R Joshi
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, Michigan, USA
| | - Tae Hoon Kim
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, Michigan, USA
| | - Bruce A Lessey
- Center for Fertility, Endocrinology and Menopause, Wake Forest University, Winston-Salem, North Carolina, USA
| | - Jae-Wook Jeong
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, Michigan, USA
| | - Asgerally T Fazleabas
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, Michigan, USA
| |
Collapse
|
27
|
Rudzitis-Auth J, Fuß SA, Becker V, Menger MD, Laschke MW. Inhibition of erythropoietin-producing hepatoma receptor B4 (EphB4) signalling suppresses the vascularisation and growth of endometriotic lesions. Br J Pharmacol 2020; 177:3225-3239. [PMID: 32144768 DOI: 10.1111/bph.15044] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 02/27/2020] [Accepted: 02/27/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND PURPOSE The development of endometriotic lesions is crucially dependent on the formation of new blood vessels. In the present study, we analysed whether this process is regulated by erythropoietin-producing hepatoma receptor B4 (EphB4) signalling. EXPERIMENTAL APPROACH We first assessed the anti-angiogenic action of the EphB4 inhibitor NVP-BHG712 in different in vitro angiogenesis assays. Then, endometriotic lesions were surgically induced in the dorsal skinfold chamber and peritoneal cavity of NVP-BHG712- or vehicle-treated BALB/c mice. This allowed to study the effect of EphB4 inhibition on their vascularisation and growth by means of intravital fluorescence microscopy, high-resolution ultrasound imaging, histology and immunohistochemistry. KEY RESULTS Non-cytotoxic doses of NVP-BHG712 suppressed the migration, tube formation and sprouting activity of both human dermal microvascular endothelial cells (HDMEC) and mouse aortic rings. Accordingly, we also detected a lower blood vessel density in NVP-BHG712-treated endometriotic lesions. This was associated with a reduced lesion growth due to a significantly lower number of proliferating stromal cells when compared to vehicle-treated controls. CONCLUSIONS AND IMPLICATIONS Inhibition of EphB4 signalling suppresses the vascularisation and growth of endometriotic lesions. Hence, EphB4 represents a promising pharmacological target for the treatment of endometriosis.
Collapse
Affiliation(s)
| | - Sophia A Fuß
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Vivien Becker
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Michael D Menger
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Matthias W Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| |
Collapse
|
28
|
Wang J, Toan S, Zhou H. Mitochondrial quality control in cardiac microvascular ischemia-reperfusion injury: New insights into the mechanisms and therapeutic potentials. Pharmacol Res 2020; 156:104771. [PMID: 32234339 DOI: 10.1016/j.phrs.2020.104771] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 03/13/2020] [Accepted: 03/19/2020] [Indexed: 12/17/2022]
Abstract
Thrombolytic therapy and revascularization strategies create a complete recanalization of the occluded epicardial coronary artery in patients with myocardial infarction (MI). However, about 35 % of patients still experience an impaired myocardial reperfusion, which is termed a no-reflow phenomenon mainly caused by cardiac microvascular ischemia-reperfusion (I/R) injury. Mitochondria are essential for microvascular endothelial cells' survival, both because of their roles as metabolic energy producers and as regulators of programmed cell death. Mitochondrial structure and function are regulated by a mitochondrial quality control (MQC) system, a series of processes including mitochondrial biogenesis, mitochondrial dynamics/mitophagy, mitochondrial proteostasis, and mitochondria-mediated cell death. Our review discusses the MQC mechanisms and how they are linked to cardiac microvascular I/R injury. Additionally, we will summarize the molecular basis that results in defective MQC mechanisms and present potential therapeutic interventions for improving MQC in cardiac microvascular I/R injury.
Collapse
Affiliation(s)
- Jin Wang
- Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, China
| | - Sam Toan
- Department of Chemical Engineering, University of Minnesota-Duluth, Duluth, MN 55812, USA
| | - Hao Zhou
- Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, China.
| |
Collapse
|
29
|
Lu K, Zhao J, Liu W. Macrophage stimulating 1-induced inflammation response promotes aortic aneurysm formation through triggering endothelial cells death and activating the NF-κB signaling pathway. J Recept Signal Transduct Res 2020; 40:374-382. [PMID: 32156191 DOI: 10.1080/10799893.2020.1738484] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Aortic aneurysm formation is associated with endothelial cells dysfunction through an undefined mechanism. Macrophage stimulating 1 (Mst1) and NF-κB signaling pathway have been found to be related to inflammation response in endothelial cell damage. The goal of our study is to explore the role of Mst1 in regulating endothelial cell viability with a focus on NF-κB signaling pathway and inflammation response. Endothelial cell viability and death were determined via immunofluorescence and ELISA. Agonist of NF-κB signaling pathway and siRNA against Mst1 were used. The results in our study demonstrated that Mst1 transcription and expression were significantly elevated after exposure to oxidative stress in endothelial cells. Once loss of Mst1 through transfection of siRNA (si-Mst1), endothelial cell viability and survival rate were rapidly increased in response to oxidative stress. In addition, we also found that Mst1 controlled inflammation response and mitochondrial function in endothelial cells. Re-activation of NF-κB signaling pathway was followed by an activation of inflammation response and mitochondrial dysfunction, as evidenced by increased expression of inflammation factors and decreased ATP synthesis. Altogether, our results identify Mst1 as the primary factors responsible for endothelial cells dysfunction in aneurysms formation through inducing inflammation response, endothelial apoptosis, and NF-κB signaling pathway activation.
Collapse
Affiliation(s)
- Kai Lu
- Daqing Oilfield General Hospital, Daqing, P. R. China
| | - Jianfei Zhao
- Daqing Oilfield General Hospital, Daqing, P. R. China
| | - Weili Liu
- Daqing Oilfield General Hospital, Daqing, P. R. China
| |
Collapse
|
30
|
Jiang H, Bi K, Wang K, Lu Z, Xu Y, Guo P, Li C, Wei Z, Chen Y, Cao Y. Reduction of myeloid derived suppressor cells by inhibiting Notch pathway prevents the progression of endometriosis in mice model. Int Immunopharmacol 2020; 82:106352. [PMID: 32143006 DOI: 10.1016/j.intimp.2020.106352] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/15/2020] [Accepted: 02/24/2020] [Indexed: 12/23/2022]
Abstract
Growing evidence suggested that immune dysregulation is one of the crucial drivers to the development of endometriosis (EMS). Myeloid derived suppressor cells (MDSCs) represent a heterogeneous subset of immature myeloid cells, and have been reported to promote the onset and progression of EMS. Notch signaling pathway played a major role in immunological reactions. Studies have found Notch signaling pathway could regulate MDSCs. However, how the biological effects of Notch signaling pathway on MDSCs may work in EMS is still unknown. In our study, we first built an endometriosis induced mice model. Then we treated mice with DAPT, a Notch signaling pathway inhibitor, or saline. We found that the DAPT could prevent the progression of EMS. The ADAM17, Notch1, Jagged1 and Hes1 were overexpressed in EMS mice, however, when mice were treated with DAPT, the overexpression was reduced. Meanwhile, we found a lower level of MDSCs in the DAPT treated EMS mice as compared to EMS mice without DAPT, accompanied by an increase of T helper (TH) 17 cells and a decrease of regulatory T cells (Tregs). We also investigated the reactive oxygen species (ROS) in peritoneal and endometriotic cells. Our results showed that ROS level decreased in both peritoneal and endometriotic cells in the study group treated with DAPT. Overall, our study indicates for the first time that blockage of Notch signaling could lessen MDSCs and ROS, and therefore preventing the development of endometriosis.
Collapse
Affiliation(s)
- Huanhuan Jiang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Kaihuan Bi
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Kangxia Wang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Zhimin Lu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Yuping Xu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No 81 Meishan Road, Hefei 230032, Anhui, China; Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Peipei Guo
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China; Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Caihua Li
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Zhaolian Wei
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No 81 Meishan Road, Hefei 230032, Anhui, China; Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Ya Chen
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Yunxia Cao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China.
| |
Collapse
|
31
|
Tian Y, Lv W, Lu C, Zhao X, Zhang C, Song H. LATS2 promotes cardiomyocyte H9C2 cells apoptosis via the Prx3-Mfn2-mitophagy pathways. J Recept Signal Transduct Res 2019; 39:470-478. [PMID: 31829064 DOI: 10.1080/10799893.2019.1701031] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Context: The pathogenesis of cardiomyocyte death is closely associated with mitochondrial homeostasis via poorly understood mechanisms.Objective: The aim of our study is to explore the contribution of large tumor suppressor kinase 2 (LATS2) to the apoptosis of cardiomyocyte H9C2 cells.Materials and Methods: Adenovirus-mediated LATS2 overexpression was carried out in H9C2 cells. The cell viability and apoptosis rate were measured via an MTT assay, TUNEL staining, western blotting, an ELISA, and an LDH release assay. Mitophagy was quantified using immunofluorescence and western blotting.Results: The overexpression of LATS2 in H9C2 cells drastically promoted cell death. Molecular investigations showed that LATS2 overexpression was associated with mitochondrial injury, as evidenced by increased mitochondrial ROS production, reduced antioxidant factor levels, increased cyt-c liberation into the nucleus and activated mitochondrial caspase-9-dependent apoptotic pathway activity. Furthermore, our results demonstrated that LATS2-mediated mitochondrial malfunction by repressing mitophagy and that the reactivation of mitophagy could sustain mitochondrial integrity and homeostasis in response to LATS2 overexpression. Furthermore, we found that LATS2 inhibited mitophagy by inactivating the Prx3-Mfn2 axis. The reactivation of Prx3-Mfn2 pathways abrogated the LATS2-mediated inhibition of mitochondrial apoptosis in H9C2 cells.Conclusions: The overexpression of LATS2 induces mitochondrial stress by repressing protective mitophagy in a manner dependent on Prx3-Mfn2 pathways, thus reducing the survival of H9C2 cells.
Collapse
Affiliation(s)
| | - Wei Lv
- Tianjin First Central Hospital, Tianjin, China
| | - Chengzhi Lu
- Tianjin First Central Hospital, Tianjin, China
| | | | - Chunguang Zhang
- North District Maternal and Child Health Family Planning Service Center, Qingdao, China
| | - Haoming Song
- Department of Cardiology, Shanghai Tongji Hospital, Shanghai, China
| |
Collapse
|
32
|
Laganà AS, Garzon S, Götte M, Viganò P, Franchi M, Ghezzi F, Martin DC. The Pathogenesis of Endometriosis: Molecular and Cell Biology Insights. Int J Mol Sci 2019; 20:E5615. [PMID: 31717614 PMCID: PMC6888544 DOI: 10.3390/ijms20225615] [Citation(s) in RCA: 271] [Impact Index Per Article: 45.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 11/01/2019] [Accepted: 11/07/2019] [Indexed: 12/15/2022] Open
Abstract
The etiopathogenesis of endometriosis is a multifactorial process resulting in a heterogeneous disease. Considering that endometriosis etiology and pathogenesis are still far from being fully elucidated, the current review aims to offer a comprehensive summary of the available evidence. We performed a narrative review synthesizing the findings of the English literature retrieved from computerized databases from inception to June 2019, using the Medical Subject Headings (MeSH) unique ID term "Endometriosis" (ID:D004715) with "Etiology" (ID:Q000209), "Immunology" (ID:Q000276), "Genetics" (ID:D005823) and "Epigenesis, Genetic" (ID:D044127). Endometriosis may origin from Müllerian or non-Müllerian stem cells including those from the endometrial basal layer, Müllerian remnants, bone marrow, or the peritoneum. The innate ability of endometrial stem cells to regenerate cyclically seems to play a key role, as well as the dysregulated hormonal pathways. The presence of such cells in the peritoneal cavity and what leads to the development of endometriosis is a complex process with a large number of interconnected factors, potentially both inherited and acquired. Genetic predisposition is complex and related to the combined action of several genes with limited influence. The epigenetic mechanisms control many of the processes involved in the immunologic, immunohistochemical, histological, and biological aberrations that characterize the eutopic and ectopic endometrium in affected patients. However, what triggers such alterations is not clear and may be both genetically and epigenetically inherited, or it may be acquired by the particular combination of several elements such as the persistent peritoneal menstrual reflux as well as exogenous factors. The heterogeneity of endometriosis and the different contexts in which it develops suggest that a single etiopathogenetic model is not sufficient to explain its complex pathobiology.
Collapse
Affiliation(s)
- Antonio Simone Laganà
- Department of Obstetrics and Gynecology, “Filippo Del Ponte” Hospital, University of Insubria, Piazza Biroldi 1, 21100 Varese, Italy; (S.G.); (F.G.)
| | - Simone Garzon
- Department of Obstetrics and Gynecology, “Filippo Del Ponte” Hospital, University of Insubria, Piazza Biroldi 1, 21100 Varese, Italy; (S.G.); (F.G.)
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, D-48149 Münster, Germany;
| | - Paola Viganò
- Reproductive Sciences Laboratory, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Via Olgettina 60, 20136 Milan, Italy;
| | - Massimo Franchi
- Department of Obstetrics and Gynecology, AOUI Verona, University of Verona, Piazzale Aristide Stefani 1, 37126 Verona, Italy;
| | - Fabio Ghezzi
- Department of Obstetrics and Gynecology, “Filippo Del Ponte” Hospital, University of Insubria, Piazza Biroldi 1, 21100 Varese, Italy; (S.G.); (F.G.)
| | - Dan C. Martin
- School of Medicine, University of Tennessee Health Science Center, 910 Madison Ave, Memphis, TN 38163, USA;
- Virginia Commonwealth University, 907 Floyd Ave, Richmond, VA 23284, USA
| |
Collapse
|
33
|
Wang Y, Zhang X, Wang P, Shen Y, Yuan K, Li M, Liang W, Que H. Sirt3 overexpression alleviates hyperglycemia-induced vascular inflammation through regulating redox balance, cell survival, and AMPK-mediated mitochondrial homeostasis. J Recept Signal Transduct Res 2019; 39:341-349. [PMID: 31680596 DOI: 10.1080/10799893.2019.1684521] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Context: Sirtuin-3 (Sirt3), a NAD-dependent deacetylase, has been reported to be involved in many biological processes.Objective: The present study aimed to investigate the effect and mechanism of Sirt3 on diabetic mice and human umbilical vein endothelial cells (HUVECs) under high glucose (HG) condition.Materials and methods: HUVECs were cultured under HG and inflammation pathway was determined via qPCR, western blots, and immunofluorescence.Results: Sirt3 expression was reduced in the progression of diabetic nephropathy. Overexpression of Sirt3 sustains renal function and retard the development of diabetic nephropathy. Mechanistically, Sirt3 overexpression attenuated hyperglycemia-mediated endothelial cells apoptosis in kidney. Besides, Sirt3 overexpression repressed oxidative injury and blocked caspase-9-related apoptosis pathway. Moreover, we found that Sirt3 overexpression was associated with AMPK activation and the latter elevates PGC1α-related mitochondrial protective system, especially mitochondrial autophagy. Loss of opa1 and/or inhibition of AMPK could depress mitochondrial autophagy and exacerbates mitochondrial function, finally contributing to the death of human renal mesangial cells.Conclusions: Our results demonstrated the beneficial effects of Sirt3 in the progression of diabetic nephropathy. Increased Sirt3-activated AMPK pathway, augments PGC1α-related mitochondrial protective system, sustained redox balance and closed caspase-9-involved apoptosis pathway in the setting of diabetic nephropathy.
Collapse
Affiliation(s)
- Yunfei Wang
- Department of Traditional Chinese Medicine Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xue Zhang
- Department of Vascular Surgery, South Campus, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Peng Wang
- Department of Vascular Surgery, South Campus, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yiting Shen
- Department of Traditional Chinese Medicine Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Kai Yuan
- Department of Vascular Surgery, South Campus, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Maoran Li
- Department of Vascular Surgery, South Campus, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Liang
- Department of Vascular Surgery, South Campus, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Huafa Que
- Department of Traditional Chinese Medicine Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
34
|
Ramirez Williams L, Brüggemann K, Hubert M, Achmad N, Kiesel L, Schäfer SD, Greve B, Götte M. γ-Secretase inhibition affects viability, apoptosis, and the stem cell phenotype of endometriotic cells. Acta Obstet Gynecol Scand 2019; 98:1565-1574. [PMID: 31424097 DOI: 10.1111/aogs.13707] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 08/13/2019] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Stem cells mediate cyclic regeneration of the endometrium. The upregulated expression of receptors and modulators of the notch signaling pathway in endometriosis suggests an involvement in the pathogenetic process. Here, we investigated the effects of notch pathway inhibition by a γ-secretase inhibitor (GSI) on stemness-associated properties of the epithelial endometriotic cell line 12Z and of primary endometriotic stroma cells. MATERIAL AND METHODS 12Z cells and primary endometriotic stroma cells of 7 patients were treated with or without GSI, and analyzed for changes in gene expression by TaqMan low-density arrays, quantitative PCR, and flow cytometry. The functional impact of GSI treatment was studied by MTT assay, cell cycle analysis, colony formation assay, annexin V apoptosis assay, and aldehyde dehydrogenase activity assays. RESULTS In 12Z cells, GSI treatment reduced aldehyde dehydrogenase activity and colony formation, and induced a shift to the G2/M phase of the cell cycle. Cell viability was decreased and apoptosis was increased in both cell models. GSI further induced transcriptional downregulation of the stemness-associated factors leukemia inhibitory factor receptor (LIFR), sex-determining region Y (SRY)- box 2, interferon-induced transmembrane protein 1, and hes-related family bHLH transcription factor with YRPW motif 1, in 12Z cells and in primary cell cultures. Downregulation of LIFR expression by GSI was confirmed at the protein level by flow cytometry. CONCLUSIONS Our in vitro data suggest that application of GSI may be a worthwhile approach in the treatment of endometriosis that warrants further investigation.
Collapse
Affiliation(s)
- Laura Ramirez Williams
- Department of Gynecology and Obstetrics, Münster University Hospital, Albert-Schweitzer-Campus 1, Münster, Germany
| | - Kathrin Brüggemann
- Department of Gynecology and Obstetrics, Münster University Hospital, Albert-Schweitzer-Campus 1, Münster, Germany
| | - Marina Hubert
- Department of Gynecology and Obstetrics, Münster University Hospital, Albert-Schweitzer-Campus 1, Münster, Germany
| | - Nurjannah Achmad
- Department of Gynecology and Obstetrics, Münster University Hospital, Albert-Schweitzer-Campus 1, Münster, Germany
| | - Ludwig Kiesel
- Department of Gynecology and Obstetrics, Münster University Hospital, Albert-Schweitzer-Campus 1, Münster, Germany
| | - Sebastian D Schäfer
- Department of Gynecology and Obstetrics, Münster University Hospital, Albert-Schweitzer-Campus 1, Münster, Germany
| | - Burkhard Greve
- Department of Radiotherapy-Radiooncology, Münster University Hospital, Albert-Schweitzer-Campus 1, Münster, Germany
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Albert-Schweitzer-Campus 1, Münster, Germany
| |
Collapse
|
35
|
Zhong J, Tan Y, Lu J, Liu J, Xiao X, Zhu P, Chen S, Zheng S, Chen Y, Hu Y, Guo Z. Therapeutic contribution of melatonin to the treatment of septic cardiomyopathy: A novel mechanism linking Ripk3-modified mitochondrial performance and endoplasmic reticulum function. Redox Biol 2019; 26:101287. [PMID: 31386965 PMCID: PMC6692063 DOI: 10.1016/j.redox.2019.101287] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 07/09/2019] [Accepted: 07/26/2019] [Indexed: 12/30/2022] Open
Abstract
The basic pathophysiological mechanisms underlying septic cardiomyopathy have not yet been completely clarified. Disease-specific treatments are lacking, and care is still based on supportive modalities. The aim of our study was to assess the protective effects of melatonin on septic cardiomyopathy, with a focus on the interactions between receptor-interacting protein kinase 3 (Ripk3), the mitochondria, endoplasmic reticulum (ER) and cytoskeletal degradation in cardiomyocytes. Ripk3 expression was increased in heart samples challenged with LPS, followed by myocardial inflammation, cardiac dysfunction, myocardial breakdown and cardiomyocyte death. The melatonin treatment attenuated septic myocardial injury in a comparable manner to the genetic depletion of Ripk3. Molecular investigations revealed that Ripk3 intimately regulated mitochondrial function, ER stress, cytoskeletal homeostasis and cardioprotective signaling pathways. Melatonin-mediated inhibition of Ripk3 improved mitochondrial bioenergetics, reduced mitochondria-initiated oxidative damage, sustained mitochondrial dynamics, ameliorated ER stress, normalized calcium recycling, and activated cardioprotective signaling pathways (including AKT, ERK and AMPK) in cardiomyocytes. Interestingly, Ripk3 overexpression mediated resistance to melatonin therapy following the infection of LPS-treated hearts with an adenovirus expressing Ripk3. Altogether, our findings identify Ripk3 upregulation as a novel risk factor for the development of sepsis-related myocardial injury, and melatonin restores the physiological functions of the mitochondria, ER, contractile cytoskeleton and cardioprotective signaling pathways. Additionally, our data also reveal a new, potentially therapeutic mechanism by which melatonin protects the heart from sepsis-mediated dysfunction, possibly by targeting Ripk3.
Collapse
Affiliation(s)
- Jiankai Zhong
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, 528308, Guangdong, China
| | - Ying Tan
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Jianhua Lu
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, 528308, Guangdong, China
| | - Jichen Liu
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xiaochan Xiao
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Pinji Zhu
- Medical School of Chinese PLA, PLA General Hospital, Beijing, 100853, China
| | - Sainan Chen
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Sulin Zheng
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, 528308, Guangdong, China
| | - Yuying Chen
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, 528308, Guangdong, China
| | - Yunzhao Hu
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, 528308, Guangdong, China
| | - Zhigang Guo
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
36
|
Liu Y, Fu Y, Hu X, Chen S, Miao J, Wang Y, Zhou Y, Zhang Y. Caveolin-1 knockdown increases the therapeutic sensitivity of lung cancer to cisplatin-induced apoptosis by repressing Parkin-related mitophagy and activating the ROCK1 pathway. J Cell Physiol 2019; 235:1197-1208. [PMID: 31270811 DOI: 10.1002/jcp.29033] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 06/12/2019] [Indexed: 12/12/2022]
Abstract
Chemotherapy is the first-line treatment option for patients with lung cancer. However, therapeutic resistance occurs through an incompletely understood mechanism. Our research wants to investigate the influence of Caveolin-1 (Cav-1) on the therapeutic sensitivity of lung cancer in vitro. Results in this study demonstrated that Cav-1 levels were markedly inhibited in A549 lung cancer cells after exposure to cisplatin. Knockdown of caveolin further enhanced cisplatin-triggered cancer death in A549 cells. The functional investigation demonstrated that Cav-1 inhibition amplified the mitochondrial stress signaling induced by cisplatin, as evidenced by the mitochondrial reactive oxygen species burst, cellular metabolic disruption, mitochondrial membrane potential reduction, and mitochondrial caspase-9-related apoptosis activation. At the molecular level, cav-1 augmented cisplatin-mediated mitochondrial damage by inhibiting Parkin-related mitochondrial autophagy. Mitophagy activation effectively attenuated the promotive impact of Cav-1 knockdown on mitochondrial damage and cell death. Furthermore, our data indicated that Cav-1 affected Parkin-related mitophagy by activating the Rho-associated coiled-coil kinase 1 (ROCK1) pathway; inhibition of the ROCK1 axis prevented cav-1 knockdown-mediated cell death and mitochondrial damage. Taken together, our results provide ample data illuminate the necessary action exerted by Cav-1 on affecting cisplatin-related therapeutic resistance. Silencing of Cav-1 inhibited Parkin-related mitophagy, thus amplifying cisplatin-mediated mitochondrial apoptotic signaling. This finding identifies the Cav-1/ROCK1/Parkin/mitophagy axis as a potential target to overcome cisplatin-related resistance in lung cancer cells.
Collapse
Affiliation(s)
- Yi Liu
- Department of Thoracic Surgery, Beijing Chaoyang Hospital, Beijing, Chaoyang, China
| | - Yili Fu
- Department of Thoracic Surgery, Beijing Chaoyang Hospital, Beijing, Chaoyang, China
| | - Xianoxing Hu
- Department of Thoracic Surgery, Beijing Chaoyang Hospital, Beijing, Chaoyang, China
| | - Shuo Chen
- Department of Thoracic Surgery, Beijing Chaoyang Hospital, Beijing, Chaoyang, China
| | - Jinbai Miao
- Department of Thoracic Surgery, Beijing Chaoyang Hospital, Beijing, Chaoyang, China
| | - Yang Wang
- Department of Thoracic Surgery, Beijing Chaoyang Hospital, Beijing, Chaoyang, China
| | - Ying Zhou
- Department of Pulmonary and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, Yangpu, China
| | - Yuan Zhang
- Department of Pulmonary and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, Yangpu, China
| |
Collapse
|
37
|
Song H, Wang M, Xin T. Mst1 contributes to nasal epithelium inflammation via augmenting oxidative stress and mitochondrial dysfunction in a manner dependent on Nrf2 inhibition. J Cell Physiol 2019; 234:23774-23784. [PMID: 31165471 DOI: 10.1002/jcp.28945] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 05/22/2019] [Accepted: 05/23/2019] [Indexed: 12/12/2022]
Abstract
Nasal epithelium inflammation plays an important role in transmitting and amplifying damage signals for the lower airway. However, the molecular basis of nasal epithelium inflammation damage has not been fully addressed. Mst1 is reported to modulate inflammation via multiple effects. Thus, the aim of our study is to understand the pathological mechanism underlying Mst1-related nasal epithelium inflammation in vitro. Our result indicated that Mst1 expression was rapidly increased in response to tumor necrosis factor-α (TNF-α) treatment in vitro and this effect was a dose-dependent manner. Interestingly, knockdown of Mst1 via transfecting small interfering RNA markedly reversed cell viability in the presence of TNF-α. Further, we found that Mst1 deficiency reduced cellular oxidative stress and attenuated mitochondrial dysfunction, as evidenced by reversed mitochondrial complex-I activity, decreased mitochondrial permeability transition pore opening rate, and stabilized mitochondrial membrane potential. Besides, we found that Nrf2 expression was increased after deletion of Mst1 whereas silencing of Nrf2 abolished the protective effects of Mst1 deletion on nasal epithelium survival and mitochondrial homeostasis. Moreover, Nrf2 overexpression also protected nasal epithelium against TNF-α-induced inflammation damage. Altogether, our data confirm that the Mst1 activation and Nrf2 downregulation seem to be the potential mechanisms responsible for the inflammation-mediated injury in nasal epithelium via mediating mitochondrial damage and cell oxidative stress.
Collapse
Affiliation(s)
- Henge Song
- Department of Respiratory Medicine, Tianjin Dongli Hospital, Tianjin, China
| | - Mengmeng Wang
- Department of Rheumatism and Immunology, Tianjin First Central Hospital, Tianjin, China
| | - Ting Xin
- Department of Cardiology, Tianjin First Central Hospital, Tianjin, China
| |
Collapse
|
38
|
Zhang J, Sun L, Li W, Wang Y, Li X, Liu Y. Overexpression of macrophage stimulating 1 enhances the anti-tumor effects of IL-24 in esophageal cancer via inhibiting ERK-Mfn2 signaling-dependent mitophagy. Biomed Pharmacother 2019; 114:108844. [PMID: 30981108 DOI: 10.1016/j.biopha.2019.108844] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 03/26/2019] [Accepted: 04/01/2019] [Indexed: 12/31/2022] Open
Abstract
Although cytokine-based therapy is a promising tool to control the progression of esophageal cancer, low therapeutic responses largely compromise treatment efficacy through unidentified mechanisms. The goal of our study was to explore the roles of macrophage stimulating 1 (Mst1) and mitophagy in enhancing IL-24-based cytokine therapy in esophageal cancer. Our data demonstrated that IL-24 application promoted cancer death by inducing mitochondrial stress, as manifested by mitochondrial ROS overproduction, mitochondrial potential dissipation, cellular ATP deprivation and mitochondrial death activation. Overexpression of Mst1 enhanced IL-24-mediated mitochondrial damage and further augmented IL-24-induced death in esophageal cancer. Molecular investigations illustrated that the IL-24-activated mitochondrial response is accompanied by activation of mitophagy, a protective mechanism to attenuate mitochondrial damage. However, Mst1 overexpression inhibited mitophagy activity, which was achieved by inactivating the ERK-Mfn2 signaling pathway. The re-activation of mitophagy abolished the cancer-killing effects of Mst1 overexpression on esophageal cancer. Altogether, our data demonstrate that IL-24-related therapeutic resistance is associated with mitophagy activation. Mst1 overexpression inhibits mitophagy activity via suppressing the ERK-Mfn2 pathway, ultimately augmenting IL-24-inducd esophageal cancer death via enhanced mitochondrial stress.
Collapse
Affiliation(s)
- Jianpeng Zhang
- Department of Thoracic Surgery, Beijing Luhe Hospital, Capital Medical University, 82 Xinhua South Road, Tongzhou District, Beijing, 101149, PR China.
| | - Lin Sun
- Department of Thoracic Surgery, Beijing Luhe Hospital, Capital Medical University, 82 Xinhua South Road, Tongzhou District, Beijing, 101149, PR China.
| | - Weiqiang Li
- Department of Thoracic Surgery, Beijing Luhe Hospital, Capital Medical University, 82 Xinhua South Road, Tongzhou District, Beijing, 101149, PR China.
| | - Yanyu Wang
- Department of Thoracic Surgery, Beijing Luhe Hospital, Capital Medical University, 82 Xinhua South Road, Tongzhou District, Beijing, 101149, PR China.
| | - Xinzhen Li
- Department of Thoracic Surgery, Beijing Luhe Hospital, Capital Medical University, 82 Xinhua South Road, Tongzhou District, Beijing, 101149, PR China.
| | - Yang Liu
- Department of Thoracic Surgery, Beijing Luhe Hospital, Capital Medical University, 82 Xinhua South Road, Tongzhou District, Beijing, 101149, PR China.
| |
Collapse
|
39
|
Zhang Y, Wang M, Xu X, Liu Y, Xiao C. Matrine promotes apoptosis in SW480 colorectal cancer cells via elevating MIEF1-related mitochondrial division in a manner dependent on LATS2-Hippo pathway. J Cell Physiol 2019; 234:22731-22741. [PMID: 31119752 DOI: 10.1002/jcp.28838] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 04/26/2019] [Accepted: 04/30/2019] [Indexed: 12/16/2022]
Abstract
Matrine, an alkaloid compound isolated from Sophora flavescens Ait, has been shown to exert cancer-killing actions in a variety of tumors; however, its anticancer mechanism in colorectal cancer (CRC) is not clear. The goal of our study was to characterize the anticancer effects and molecular mechanisms of matrine in SW480 CRC cells in vitro. Matrine treatment reduced mitochondrial metabolic function and ATP levels, repressed mitochondrial membrane potential, evoked mitochondrial reactive oxygen species accumulation, and promoted cyt-c-related mitochondrial apoptosis activation. In addition, we found that matrine treatment activated mitochondrial fission through upregulating mitochondrial elongation factor 1 (MIEF1); silencing of MIEF1 prevented matrine-mediated mitochondrial damage and reversed the decrease in SW480 cell viability. Moreover, matrine treatment affected MIEF1 expression via the large tumor suppressor-2 (LATS2)-Hippo axis, and LATS2 deficiency suppressed the anticancer actions exerted by matrine on SW480 cancer cells. In summary, we show for the first time that matrine inhibits SW480 cell survival by activating MIEF1-related mitochondrial division via the LATS2-Hippo pathway. These findings explain the anticancer mechanisms of matrine in CRC and also identify the LATS2-MIEF1 signaling pathway as an effective target for the treatment of CRC.
Collapse
Affiliation(s)
- Yawei Zhang
- Department of General Surgery, Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China
| | - Meiping Wang
- Department of General Surgery, Fuzhou General Hospital (Dongfang Hospital), Fuzhou, Fujian, China
| | - Xianfeng Xu
- Department of Critical Care Medicine, Changle People's Hospital, Fuzhou, Fujian, China
| | - Yonghong Liu
- Department of General Surgery, First People's Hospital of Yuhang District, Hangzhou, China
| | - Chunhong Xiao
- Department of General Surgery, Fuzhou General Hospital (Dongfang Hospital), Fuzhou, Fujian, China
| |
Collapse
|
40
|
Zhang X, Li F, Cui Y, Liu S, Sun H. Mst1 overexpression combined with Yap knockdown augments thyroid carcinoma apoptosis via promoting MIEF1-related mitochondrial fission and activating the JNK pathway. Cancer Cell Int 2019; 19:143. [PMID: 31139020 PMCID: PMC6530088 DOI: 10.1186/s12935-019-0860-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 05/13/2019] [Indexed: 02/06/2023] Open
Abstract
Background Cancer cell viability is strongly modulated by the Hippo pathway, which includes mammalian STE20-like protein kinase 1 (Mst1) and yes-associated protein (Yap). Although the roles of Mst1 and Yap in thyroid carcinoma cell death have been fully addressed, no study has determined whether differential modification of Mst1 and Yap could further suppress thyroid carcinoma progression. The aim of our study was to explore the antiapoptotic effects exerted by combined Mst1 overexpression and Yap knockdown in thyroid carcinoma MDA-T32 cells in vitro. Methods Mst1 adenovirus and Yap shRNA were transfected into MDA-T32 cells to overexpress Mst1 and inhibit Yap, respectively. Cell viability and death were determined via an MTT assay, a TUNEL assay and western blotting. Mitochondrial function, mitochondrial fission and pathway studies were performed via western blotting and immunofluorescence. Results The results of our study showed that combined Mst1 overexpression and Yap knockdown further augmented MDA-T32 cell death by mediating mitochondrial damage. In addition, cancer cell migration and proliferation were suppressed by combined Mst1 overexpression and Yap knockdown. At the molecular level, mitochondrial membrane potential, ATP production, respiratory function, and caspase-9-related apoptosis were activated by combined Mst1 overexpression and Yap knockdown. Further, we found that fatal mitochondrial fission was augmented by combined Mst1 overexpression and Yap knockdown in a manner dependent on the JNK-MIEF1 pathway. Inhibition of JNK-MIEF1 pathway activity abolished the proapoptotic effects exerted by Mst1/Yap on MDA-T32 cells. Conclusions Taken together, our data suggest that Mst1 activation and Yap inhibition coordinate to augment thyroid cancer cell death by controlling the JNK-MIEF1-mitochondria pathway, suggesting that differential regulation of the core Hippo pathway components is potentially a novel therapeutic tool for the treatment of thyroid cancer. Electronic supplementary material The online version of this article (10.1186/s12935-019-0860-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaoli Zhang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, #45, Chang Chun Street, Beijing, 100053 China
| | - Fei Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, #45, Chang Chun Street, Beijing, 100053 China
| | - Yeqing Cui
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, #45, Chang Chun Street, Beijing, 100053 China
| | - Shuang Liu
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, #45, Chang Chun Street, Beijing, 100053 China
| | - Haichen Sun
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, #45, Chang Chun Street, Beijing, 100053 China
| |
Collapse
|
41
|
Xing J, Xu H, Liu C, Wei Z, Wang Z, Zhao L, Ren L. Melatonin ameliorates endoplasmic reticulum stress in N2a neuroblastoma cell hypoxia-reoxygenation injury by activating the AMPK-Pak2 pathway. Cell Stress Chaperones 2019; 24:621-633. [PMID: 30976981 PMCID: PMC6527732 DOI: 10.1007/s12192-019-00994-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 03/27/2019] [Accepted: 04/03/2019] [Indexed: 12/21/2022] Open
Abstract
Endoplasmic reticulum (ER) stress has been identified as a primary factor involved in brain ischemia-reperfusion injury progression. p21-activated kinase 2 (Pak2) is a novel ER function regulator. The aim of our study is to explore the influence of Pak2 on ER stress and determine whether melatonin attenuates ER stress-mediated cell death by modulating Pak2 expression in vitro using N2a cells. The results of our study demonstrated that hypoxia-reoxygenation (HR) injury repressed the levels of Pak2, an effect that was accompanied by activation of ER stress. In addition, decreased Pak2 was associated with oxidative stress, calcium overload, and caspase-12-mediated apoptosis activation in HR-treated N2a cells. Interestingly, melatonin treatment reversed the decreased Pak2 expression under HR stress. Knockdown of Pak2 abolished the protective effects of melatonin on ER stress, oxidative stress, and caspase-12-related N2a cells death. Additionally, we found that Pak2 was regulated by melatonin via the AMPK pathway; inhibition of AMPK prevented melatonin-mediated Pak2 upregulation, a result that was accompanied by an increase in N2a cell death. Altogether, these results identify the AMPK-Pak2 axis as a new signaling pathway responsible for ER stress and N2a cell viability under HR injury. Modulation of the AMPK-Pak2 cascade via supplementation of melatonin might be considered an effective approach to attenuate reperfusion-mediated N2a cell damage via repression of ER stress.
Collapse
Affiliation(s)
- Jin Xing
- Department of Neurosurgery, Shanghai Pudong Hospital, Shanghai Fu Dan University School of Medicine, Shanghai, 201399, China
| | - Hao Xu
- Department of Neurosurgery, Shanghai Pudong Hospital, Shanghai Fu Dan University School of Medicine, Shanghai, 201399, China
| | - Chaobo Liu
- Department of Neurosurgery, Shanghai Pudong Hospital, Shanghai Fu Dan University School of Medicine, Shanghai, 201399, China
| | - Zilong Wei
- Department of Neurosurgery, Shanghai Pudong Hospital, Shanghai Fu Dan University School of Medicine, Shanghai, 201399, China
| | - Zhihan Wang
- Department of Neurosurgery, Shanghai Pudong Hospital, Shanghai Fu Dan University School of Medicine, Shanghai, 201399, China
| | - Liang Zhao
- Department of Neurosurgery, Shanghai Pudong Hospital, Shanghai Fu Dan University School of Medicine, Shanghai, 201399, China
| | - Li Ren
- Department of Neurosurgery, Shanghai Pudong Hospital, Shanghai Fu Dan University School of Medicine, Shanghai, 201399, China.
| |
Collapse
|
42
|
Zhang M, Wang S, Tang L, Wang X, Zhang T, Xia X, Fang X. Downregulated circular RNA hsa_circ_0067301 regulates epithelial-mesenchymal transition in endometriosis via the miR-141/Notch signaling pathway. Biochem Biophys Res Commun 2019; 514:71-77. [PMID: 31023528 DOI: 10.1016/j.bbrc.2019.04.109] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 04/14/2019] [Indexed: 11/29/2022]
Abstract
Endometriosis is a common gynecologic disorder with enigmatic etiopathogenesis and is characterized by tumor-like biological behaviors. Epithelial-mesenchymal transition (EMT) has been recognized as a core mechanism of endometriosis. Recently, circular RNAs (circRNAs) have attracted considerable attention because they play an important role in the progression of cancer. However, little is known about the function of circRNAs in endometriosis. This study is intended to investigate the involvement of circRNAs and microRNAs in the process of EMT in ovarian endometriosis in vitro. We found that relative RNA levels of hsa_circ_0067301 and miR-141-5p were significantly reduced in ectopic endometrium when compared to control endometrium. Hsa_circ_0067301 knockdown could promote the proliferation and migration in Ishikawa and End1/E6E7 cells, concomitant with increased the relative protein expression against Notch-1, Hes-1, N-cadherin, and vimentin but reduced expression of E-cadherin. After co-transfection with the miR-141-5p inhibitor, the miR-141-5p that competes for binding to hsa_circ_0067301 was reduced, reversed EMT and partially restored the expression of Notch-1 and Hes-1. Results demonstrate the hsa_circ_0067301/miR-141-5p/Notch-1 axis plays an important regulatory role in the process of EMT in endometriosis. The study highlighted the importance of circRNAs in ovarian endometriosis and provided unique insights into the molecular basis concerning the pathogenesis of endometriosis.
Collapse
Affiliation(s)
- Mengmeng Zhang
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, PR China
| | - Sixue Wang
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, PR China
| | - Lu Tang
- Department of Gynecology, Baoan Maternal and Child Health Hospital, Jinan University, Shenzhen, Guangdong 518000, PR China
| | - Xi Wang
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, PR China
| | - Tingting Zhang
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, PR China
| | - Xiaomeng Xia
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, PR China
| | - Xiaoling Fang
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, PR China.
| |
Collapse
|
43
|
Yao W, Zhu S, Li P, Zhang S. Large tumor suppressor kinase 2 overexpression attenuates 5-FU-resistance in colorectal cancer via activating the JNK-MIEF1-mitochondrial division pathway. Cancer Cell Int 2019; 19:97. [PMID: 31011291 PMCID: PMC6460675 DOI: 10.1186/s12935-019-0812-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 04/01/2019] [Indexed: 02/06/2023] Open
Abstract
Background 5-Fluorouracil (5-FU) is a standard treatment for colorectal cancer, but most patients develop 5-FU resistance. Here, we conducted experiments to identify an effective approach to augment 5-FU-based treatment in colorectal cancer in vitro. Methods SW480 cells were in the present study and treated with 5-FU. Besides, LATS2 adenovirus vectors were infected into SW480 cells. Western blotting, immunofluorescence and ELISA were used to evaluate cell death and mitochondrial function. Pathway blocker was used to verify the role of MAPK-JNK pathway in SW480 cell death. Results An obvious drop in large tumor suppressor kinase 2 (LATS2) expression was observed in SW480 cells after treatment with 5-FU. In addition, upregulation of LATS2 expression through infection with LATS2 adenovirus further increased the reduction of SW480 cell viability induced by 5-FU. Functional exploration showed that 5-FU treatment suppressed mitochondrial membrane potential, enhanced cyt-c release into the nucleus, induced an oxidative injury environment by promoting ROS production, and eventually upregulated Bax-related mitochondrial apoptosis. Besides, LATS2 overexpression in combination with 5-FU treatment further perturbed mitochondrial homeostasis, and this effect was achieved by elevating mitochondrial division. Mechanistically, LATS2 overexpression and 5-FU co-treatment amplified mitochondrial division by upregulating MIEF1 expression in a manner dependent on MAPK-JNK axis. Knockdown of MIEF1 using an siRNA-mediated loss of function assay and/or inhibition of the MAPK-JNK pathway using the specific inhibitor SP600125 abolished LATS2/5-FU-mediated deleterious effects on mitochondrial performance and SW480 cell viability. Conclusions In light of the above findings, LATS2 downregulation could be a potential mechanism of low response to 5-FU treatment. Overexpression of LATS2 to further disrupt mitochondrial function via the JNK-MIEF1 signalling pathway might be a method to optimize 5-FU-based chemotherapy.
Collapse
Affiliation(s)
- Weilong Yao
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, No. 95, Yong'an Road, Xicheng District, Beijing, 100050 People's Republic of China
| | - Shengtao Zhu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, No. 95, Yong'an Road, Xicheng District, Beijing, 100050 People's Republic of China
| | - Peng Li
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, No. 95, Yong'an Road, Xicheng District, Beijing, 100050 People's Republic of China
| | - Shutian Zhang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, No. 95, Yong'an Road, Xicheng District, Beijing, 100050 People's Republic of China
| |
Collapse
|
44
|
Li J, Li N, Yan S, Lu Y, Miao X, Gu Z, Shao Y. Liraglutide protects renal mesangial cells against hyperglycemia‑mediated mitochondrial apoptosis by activating the ERK‑Yap signaling pathway and upregulating Sirt3 expression. Mol Med Rep 2019; 19:2849-2860. [PMID: 30816450 DOI: 10.3892/mmr.2019.9946] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 01/11/2019] [Indexed: 11/05/2022] Open
Abstract
Diabetic nephropathy results from hyperglycemia‑mediated renal glomerular cell death via mitochondrial apoptosis. There is an emerging requirement for novel approaches with mitochondrial protective effects that alleviate the hyperglycemia‑induced loss of functional cells during diabetic renal damage. Liraglutide, a type of glucagon‑like peptide‑1 agonist, has been suggested to inhibit the progression of obesity and hyperglycemia. However, the contributions and mechanism of action of liraglutide on hyperglycemia‑mediated cell mitochondrial apoptosis in diabetic kidneys have not been illustrated. The present study demonstrated that liraglutide may protect human renal mesangial cells (HRMCs) against hyperglycemia‑induced cell death by inhibiting mitochondrial apoptosis. Liraglutide administration also maintained HRMC viability and promoted HRMC proliferation within a high glucose stress environment. Functional studies demonstrated that hyperglycemia triggered mitochondrial dysfunction, including mitochondrial potential reduction, mitochondrial permeability transition pore opening, reactive oxygen species overproduction and the activation of the mitochondrial apoptotic pathway. However, liraglutide treatment preserved mitochondrial function and prevented activation of mitochondrial apoptosis by upregulating sirtuin 3 (Sirt3) expression. Deletion of Sirt3 abrogated the protective effects of liraglutide on mitochondrial homeostasis following high glucose challenge. In addition, molecular analysis confirmed that liraglutide upregulated Sirt3 via activating the extracellular signal‑regulated kinase‑Yes‑associated protein (ERK‑Yap) signaling pathway. Inhibition of the ERK‑Yap axis negated the action of liraglutide on Sirt3 activation, leading to mitochondrial injury and HRMC apoptosis. Taken together, the present study illustrated that liraglutide protected renal mesangial cells from hyperglycemia‑mediated mitochondrial apoptosis by upregulating Sirt3 expression and activation of the ERK‑Yap signaling pathway.
Collapse
Affiliation(s)
- Jian Li
- Department of Geriatric Endocrinology, Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Nan Li
- Department of Geriatric Endocrinology, Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Shuangtong Yan
- Department of Geriatric Endocrinology, Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Yanhui Lu
- Department of Geriatric Endocrinology, Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Xinyu Miao
- Department of Geriatric Endocrinology, Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Zhaoyan Gu
- Department of Geriatric Endocrinology, Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Yinghong Shao
- Department of Outpatients, Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| |
Collapse
|
45
|
Zhou J, Shi M, Li M, Cheng L, Yang J, Huang X. Sirtuin 3 inhibition induces mitochondrial stress in tongue cancer by targeting mitochondrial fission and the JNK-Fis1 biological axis. Cell Stress Chaperones 2019; 24:369-383. [PMID: 30656603 PMCID: PMC6439076 DOI: 10.1007/s12192-019-00970-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 01/05/2019] [Accepted: 01/09/2019] [Indexed: 12/30/2022] Open
Abstract
Sirtuin 3 (Sirt3)-modified mitochondrial fission participates in the progression of several types of cancers. However, its role in tongue cancer requires investigation. The aim of our study is to determine whether Sirt3 knockdown regulates the viability of tongue cancer cells via modulating mitochondrial fission. Two types of tongue cancer cells were used in the present study, and siRNA was transfected into the cells to suppress Sirt3 expression. Mitochondrial function and cell apoptosis were determined via immunofluorescence, Western blotting, ELISA, and qPCR assays. A pathway blocker was applied to verify the role of the JNK-Fis1 signaling pathway in regulation of mitochondrial fission. The present study showed that loss of Sirt3 promoted tongue cancer cell death in a manner dependent on mitochondrial apoptosis. Mitochondrial oxidative stress, energy metabolism disorder, mitochondrial cyt-c liberation, and mitochondrial apoptosis activation were observed after Sirt3 silencing. Furthermore, we demonstrated that Sirt3 knockdown activated mitochondrial stress via triggering Fis1-related mitochondrial fission and that inhibition of Fis1-related mitochondrial fission abrogated the pro-apoptotic effect of Sirt3 knockdown on tongue cancer cells. To this end, we found that Sirt3 modulated Fis1 expression via the c-Jun N-terminal kinases (JNK) signaling pathway and that blockade of the JNK pathway attenuated mitochondrial stress and repressed apoptosis in Sirt3 knockdown cells. Altogether, our results identified a tumor-suppressive role for Sirt3 deficiency in tongue cancer via activation of the JNK-Fis1 axis and subsequent initiation of fatal mitochondrial fission. Given these findings, strategies to repress Sirt3 activity and enhance the JNK-Fis1-mitochondrial fission cascade have clinical benefits for patients with tongue cancer.
Collapse
Affiliation(s)
- Jichi Zhou
- Department of Oral and Maxillofacial Surgery, Beijing Stomatological Hospital, Capital Medical University, Tiantanxili 4, Dongcheng District, Beijing, 100050, China
| | - Menghan Shi
- Department of Oral and Maxillofacial Surgery, Beijing Stomatological Hospital, Capital Medical University, Tiantanxili 4, Dongcheng District, Beijing, 100050, China
| | - Man Li
- Department of Oral and Maxillofacial Surgery, Beijing Stomatological Hospital, Capital Medical University, Tiantanxili 4, Dongcheng District, Beijing, 100050, China
| | - Long Cheng
- Department of Oral and Maxillofacial Surgery, Beijing Stomatological Hospital, Capital Medical University, Tiantanxili 4, Dongcheng District, Beijing, 100050, China
| | - Jinsuo Yang
- Department of Oral and Maxillofacial Surgery, Beijing Stomatological Hospital, Capital Medical University, Tiantanxili 4, Dongcheng District, Beijing, 100050, China
| | - Xin Huang
- Department of Oral and Maxillofacial Surgery, Beijing Stomatological Hospital, Capital Medical University, Tiantanxili 4, Dongcheng District, Beijing, 100050, China.
| |
Collapse
|
46
|
Wei B, Wang M, Hao W, He X. Mst1 facilitates hyperglycemia-induced retinal pigmented epithelial cell apoptosis by evoking mitochondrial stress and activating the Smad2 signaling pathway. Cell Stress Chaperones 2019; 24:259-272. [PMID: 30632063 PMCID: PMC6363619 DOI: 10.1007/s12192-018-00963-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 12/07/2018] [Accepted: 12/12/2018] [Indexed: 01/12/2023] Open
Abstract
Hyperglycemia induces retinal pigmented epithelial cell apoptosis and mitochondrial stress via poorly understood mechanisms. The goal of our current study is to explore whether mammalian sterile 20-like kinase 1 (Mst1) is involved in the pathogenesis of hyperglycemia-mediated retinal pigmented epithelial cell apoptosis by triggering mitochondrial abnormalities and activating the Smad2 signaling pathway. Retinal pigmented epithelial ARPE-19 cells were presented with a high-glucose challenge in vitro. Cell viability and apoptosis were measured via western blotting, ELISAs, and immunofluorescence assays. Mitochondrial function was detected via JC-1 staining, mitochondrial ROS flow cytometry, western blotting, and ELISAs. Loss- and gain-of-function assays were performed via cell transfection and transduction with Mst1 siRNA and Smad2 adenovirus, respectively. The results indicated that hyperglycemia treatment upregulated the levels of Mst1, an effect that was accompanied by an increase in ARPE-19 cell apoptosis. Loss of Mst1 attenuated hyperglycemia-induced cell apoptosis, and this effect seemed to be associated with mitochondrial protection. In response to hyperglycemia stimulus, mitochondrial stress was noted in ARPE-19 cells, including mitochondrial ROS overproduction, mitochondrial respiratory metabolism dysfunction, mitochondrial fission/fusion imbalance, and mitochondrial apoptosis activation. Further, we provided evidence to support the crucial role played by Smad2 in promoting Mst1-mediated cell apoptosis and mitochondrial stress. Overexpression of Smad2 abrogated the beneficial effects of Mst1 deletion on ARPE-19 cell viability and mitochondrial protection. Altogether, our results identified Mst1 as a novel mediator controlling the fate of retinal pigmented epithelial cells and mitochondrial homeostasis via the Smad2 signaling pathway. Based on this finding, strategies to repress Mst1 upregulation and block Smad2 activation are vital to alleviate hyperglycemia-mediated retinal pigmented epithelial cell damage.
Collapse
Affiliation(s)
- Bing Wei
- Department of Medicine, He University, No.66, Sishui Street, Hunnan District, Shenyang City, Liaoning Province, China
| | - Min Wang
- Department of Medicine, He University, No.66, Sishui Street, Hunnan District, Shenyang City, Liaoning Province, China
| | - Wei Hao
- Department of Medicine, He University, No.66, Sishui Street, Hunnan District, Shenyang City, Liaoning Province, China
| | - Xiangdong He
- Department of Medicine, He University, No.66, Sishui Street, Hunnan District, Shenyang City, Liaoning Province, China.
| |
Collapse
|