1
|
Yang Y, Meng L, Hu X, Li X. Renal functional outcomes after nephrectomy in patients with localized renal cell carcinoma and diabetes mellitus: a systematic review and meta-analysis. Int Urol Nephrol 2024; 56:1859-1868. [PMID: 38300449 DOI: 10.1007/s11255-023-03885-7] [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: 07/08/2023] [Accepted: 10/31/2023] [Indexed: 02/02/2024]
Abstract
INTRODUCTION AND OBJECTIVE Diabetes mellitus (DM), one of the most common comorbidities in patients with renal cell carcinoma (RCC), was proven to be an important prognostic factor of overall survival for these patients. Regarding the influence on renal function after nephrectomy, evidence is still scant. This systematic review and meta-analysis was conducted to provide a more reliable analysis of the association between DM and long-term renal functional outcomes after nephrectomy. METHODS The PubMed, Web of Science, Embase and Cochrane Library (CENTRAL) databases were searched for eligible studies from inception to January 2023. Hazard ratios (HRs) with 95% confidence intervals (CIs) were extracted to evaluate the association between DM and renal functional outcomes using a random effects model. Stata 17.0 software was used for statistical analysis. RESULTS The meta-analysis included thirteen studies consisting of 8562 RCC patients who underwent nephrectomy. Preoperative comorbidity of DM was significantly associated with poor renal functional outcomes (HR = 1.91, 95% CI 1.48-2.48, p < 0.0001), regardless of ethnicity, follow-up time, body mass index (BMI) and age. However, in the radical nephrectomy subgroup, DM was not significantly associated with renal function decline (HR = 1.91, 95% CI 0.93-3.90, p = 0.0781). CONCLUSIONS The aggregate evidence indicated that preexisting DM may be associated with poor renal functional outcomes in patients with RCC after nephrectomy, especially in patients receiving partial nephrectomy. Urologists should focus more on the glycemic management of these patients after nephrectomy. More high-quality studies are needed to explore the influence of DM on renal function outcomes in postoperative patients.
Collapse
Affiliation(s)
- Yujia Yang
- Institute of Urology, Department of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
- West China School of Medicine, Sichuan University, Chengdu, 610041, China
| | - Linghao Meng
- Institute of Urology, Department of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
- West China School of Medicine, Sichuan University, Chengdu, 610041, China
| | - Xu Hu
- Institute of Urology, Department of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiang Li
- Institute of Urology, Department of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
2
|
Zhao Y, Lin S, Zeng W, Lin X, Qin X, Miu B, Gao S, Wu H, Liu J, Chen X. JS-K activates G2/M checkpoints through the DNA damage response and induces autophagy via CAMKKβ/AMPKα/mTOR pathway in bladder cancer cells. J Cancer 2024; 15:343-355. [PMID: 38169515 PMCID: PMC10758033 DOI: 10.7150/jca.86393] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/21/2023] [Indexed: 01/05/2024] Open
Abstract
The aim of this study was to investigate the effects of JS-K, a nitric oxide donor prodrug, on DNA damage and autophagy in bladder cancer (BCa) cells and to explore the potential related mechanisms. Through detecting proliferation viability, cell morphology observation and colony formation assay low concentrations of JS-K significantly inhibited BCa growth while having no effect on normal cells. JS-K induced an increase in the level of DNA damage protein γH2AX and a decrease in the level of DNA damage repair-related proteins PCNA and RAD51 in BCa cells, indicating that JS-K can induce DNA damage in BCa cells and inhibit DNA damage repair. JS-K induced G2/M phase block and calcium overload using flow cytometry analysis. Moreover, we also investigated the levels of cell G2/M cycle checkpoint-related protein and autophagy-associated protein by western blot. The results of our study demonstrated that JS-K induced BCa cells G2/M phase arrest due to upregulating proteins related to DNA damage-related G2/M checkpoint activation (p-ATM, p-ATR, p-Chk1, p-Chk2, and p-Cdc2) and down-regulation of Cyclin B1 protein. In addition, our study demonstrated that JS-K-induced autophagy in BCa cells was related to the CAMKKβ/AMPKα/mTOR pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jianjun Liu
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Xiaojun Chen
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| |
Collapse
|
3
|
Cheng Y, Qu Z, Jiang Q, Xu T, Zheng H, Ye P, He M, Tong Y, Ma Y, Bao A. Functional Materials for Subcellular Targeting Strategies in Cancer Therapy: Progress and Prospects. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023:e2305095. [PMID: 37665594 DOI: 10.1002/adma.202305095] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/26/2023] [Indexed: 09/05/2023]
Abstract
Neoadjuvant and adjuvant therapies have made significant progress in cancer treatment. However, tumor adjuvant therapy still faces challenges due to the intrinsic heterogeneity of cancer, genomic instability, and the formation of an immunosuppressive tumor microenvironment. Functional materials possess unique biological properties such as long circulation times, tumor-specific targeting, and immunomodulation. The combination of functional materials with natural substances and nanotechnology has led to the development of smart biomaterials with multiple functions, high biocompatibilities, and negligible immunogenicities, which can be used for precise cancer treatment. Recently, subcellular structure-targeting functional materials have received particular attention in various biomedical applications including the diagnosis, sensing, and imaging of tumors and drug delivery. Subcellular organelle-targeting materials can precisely accumulate therapeutic agents in organelles, considerably reduce the threshold dosages of therapeutic agents, and minimize drug-related side effects. This review provides a systematic and comprehensive overview of the research progress in subcellular organelle-targeted cancer therapy based on functional nanomaterials. Moreover, it explains the challenges and prospects of subcellular organelle-targeting functional materials in precision oncology. The review will serve as an excellent cutting-edge guide for researchers in the field of subcellular organelle-targeted cancer therapy.
Collapse
Affiliation(s)
- Yanxiang Cheng
- Department of Gynecology, Renmin Hospital, Wuhan University, No.238 Jiefang Road, Wuchang, Wuhan, 430060, P. R. China
| | - Zhen Qu
- Department of Blood Transfusion Research, Wuhan Blood Center (WHBC), HUST-WHBC United Hematology Optical Imaging Center, No.8 Baofeng 1st Road, Wuhan, Hubei, 430030, P. R. China
| | - Qian Jiang
- Department of Blood Transfusion Research, Wuhan Blood Center (WHBC), HUST-WHBC United Hematology Optical Imaging Center, No.8 Baofeng 1st Road, Wuhan, Hubei, 430030, P. R. China
| | - Tingting Xu
- Department of Clinical Laboratory, Wuhan Blood Center (WHBC), No.8 Baofeng 1st Road, Wuhan, Hubei, 430030, P. R. China
| | - Hongyun Zheng
- Department of Clinical Laboratory, Renmin Hospital, Wuhan University, No.238 Jiefang Road, Wuchang, Wuhan, 430060, P. R. China
| | - Peng Ye
- Department of Pharmacy, Renmin Hospital, Wuhan University, No.238 Jiefang Road, Wuchang, Wuhan, 430060, P. R. China
| | - Mingdi He
- Department of Blood Transfusion Research, Wuhan Blood Center (WHBC), HUST-WHBC United Hematology Optical Imaging Center, No.8 Baofeng 1st Road, Wuhan, Hubei, 430030, P. R. China
| | - Yongqing Tong
- Department of Clinical Laboratory, Renmin Hospital, Wuhan University, No.238 Jiefang Road, Wuchang, Wuhan, 430060, P. R. China
| | - Yan Ma
- Department of Blood Transfusion Research, Wuhan Blood Center (WHBC), HUST-WHBC United Hematology Optical Imaging Center, No.8 Baofeng 1st Road, Wuhan, Hubei, 430030, P. R. China
| | - Anyu Bao
- Department of Clinical Laboratory, Renmin Hospital, Wuhan University, No.238 Jiefang Road, Wuchang, Wuhan, 430060, P. R. China
| |
Collapse
|
4
|
Rosidi B, Priyatno D, Putra TP, Yusuf I. Metformin Induces a Caspase 3-Unrelated Apoptosis in Human Colorectal Cancer Cell Lines HCT116 and SW620. Cancer Manag Res 2023; 15:475-485. [PMID: 37312884 PMCID: PMC10259592 DOI: 10.2147/cmar.s385278] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 03/16/2023] [Indexed: 06/15/2023] Open
Abstract
Purpose To study the effects of metformin on the proliferation and growth of human colorectal cancer cell lines HCT116 and SW620. Materials and Methods The antiproliferative effect of metformin was assayed using an MTS reagent and its ability to inhibit colony formation was demonstrated using a clonogenic assay. Flow cytometry using YO-PRO-1/PI was performed to examine the effects of metformin on apoptosis and cell death of HCT116 and SW620. Caspase 3 activities were measured in caspase-3 activity tests using a caspase-3 activity kit. Furthermore, Western blots were performed with anti-PARP1, anti-caspase 3, and anti-cleaved caspase 3 to confirm whether caspase activation was present or not. Results Both MTS proliferation assays and clonogenic assays showed that metformin inhibited the proliferation and growth of HCT116 and SW620 cells in a concentration-dependent manner. Flow cytometric analysis identified early apoptosis and metformin-induced cell death in both cell lines. However, caspase 3 activity could not be detected. Cleavage of both PARP1 and pro-caspase 3 was not observed in the Western blot, confirming the absence of caspase 3 activations. Conclusion This present study suggests a caspase 3-unrelated apoptosis mechanism of metformin-induced cell death in human colorectal cancer cell lines HCT116 and SW620.
Collapse
Affiliation(s)
- Bustanur Rosidi
- Division of Proteomics, Mochtar Riady Institute for Nanotechnology and Medical Science Group, University of Pelita Harapan, Tangerang, Banten, Indonesia
| | - Diana Priyatno
- Division of Proteomics, Mochtar Riady Institute for Nanotechnology and Medical Science Group, University of Pelita Harapan, Tangerang, Banten, Indonesia
| | - Teguh Pribadi Putra
- Division of Proteomics, Mochtar Riady Institute for Nanotechnology and Medical Science Group, University of Pelita Harapan, Tangerang, Banten, Indonesia
| | - Irawan Yusuf
- Division of Proteomics, Mochtar Riady Institute for Nanotechnology and Medical Science Group, University of Pelita Harapan, Tangerang, Banten, Indonesia
- Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| |
Collapse
|
5
|
Zhao Y, Lin X, Zeng W, Qin X, Miao B, Gao S, Liu J, Li Z. Berberine inhibits the progression of renal cell carcinoma cells by regulating reactive oxygen species generation and inducing DNA damage. Mol Biol Rep 2023:10.1007/s11033-023-08381-w. [PMID: 37217616 DOI: 10.1007/s11033-023-08381-w] [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: 07/22/2022] [Accepted: 03/09/2023] [Indexed: 05/24/2023]
Abstract
BACKGROUND Berberine is a natural isoquinoline alkaloid that has been shown to have antitumor properties in a growing number of studies. However, its role in renal cell carcinoma remains unclear. This study investigates berberine's effect and mechanism in renal cell carcinoma. METHODS The methyl-tetrazolium, colony formation, and lactate dehydrogenase assay were used to detect proliferation and cytotoxicity, respectively. Flow cytometry, caspase-Glo 3/7 assay, and adenosine triphosphate assay were used to detect apoptosis and the adenosine triphosphate levels. Wound healing and transwell assay were used to examine the migration ability of renal cell carcinoma cells. Besides, the level of reactive oxygen species (ROS) was explored using a DCFH-DA-based kit. Additionally, western blot and Immunofluorescence assay was taken to determine the levels of relative proteins. RESULTS In vitro, our findings indicated that the proliferation and migration of renal cell carcinoma cells treated with berberine in various concentrations were inhibited, while the level of ROS and apoptosis rate were increased. Furthermore, The results of western blot showed that the expression of Bax, Bad, Bak, Cyto c, Clv-Caspase 3, Clv-Caspase 9, E-cadherin, TIMP-1and γH2AX were up-regulated, while Bcl-2, N-cadherin, Vimentin, Snail, Rad51 and PCNA were down-regulated after treating with berberine with various concentration. CONCLUSION The result of this study revealed that berberine inhibits renal cell carcinoma progression via regulating ROS generation and inducing DNA break.
Collapse
Affiliation(s)
- Yuwan Zhao
- Department of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, 524001, Zhanjiang, Guangdong, China
| | - Xinghua Lin
- Department of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, 524001, Zhanjiang, Guangdong, China
| | - Wenfeng Zeng
- Department of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, 524001, Zhanjiang, Guangdong, China
| | - Xingzhang Qin
- Department of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, 524001, Zhanjiang, Guangdong, China
| | - Bailiang Miao
- Department of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, 524001, Zhanjiang, Guangdong, China
| | - Sheng Gao
- Department of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, 524001, Zhanjiang, Guangdong, China
| | - Jianjun Liu
- Department of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, 524001, Zhanjiang, Guangdong, China.
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, 524001, Zhanjiang, Guangdong, China.
| | - Zhuo Li
- Department of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, 524001, Zhanjiang, Guangdong, China.
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, 524001, Zhanjiang, Guangdong, China.
| |
Collapse
|
6
|
Zhang Y, Zhou F, Guan J, Zhou L, Chen B. Action Mechanism of Metformin and Its Application in Hematological Malignancy Treatments: A Review. Biomolecules 2023; 13:250. [PMID: 36830619 PMCID: PMC9953052 DOI: 10.3390/biom13020250] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 02/03/2023] Open
Abstract
Hematologic malignancies (HMs) mainly include acute and chronic leukemia, lymphoma, myeloma and other heterogeneous tumors that seriously threaten human life and health. The common effective treatments are radiotherapy, chemotherapy and hematopoietic stem cell transplantation (HSCT), which have limited options and are prone to tumor recurrence and (or) drug resistance. Metformin is the first-line drug for the treatment of type 2 diabetes (T2DM). Recently, studies identified the potential anti-cancer ability of metformin in both T2DM patients and patients that are non-diabetic. The latest epidemiological and preclinical studies suggested a potential benefit of metformin in the prevention and treatment of patients with HM. The mechanism may involve the activation of the adenosine monophosphate-activated protein kinase (AMPK) signaling pathway by metformin as well as other AMPK-independent pathways to exert anti-cancer properties. In addition, combining current conventional anti-cancer drugs with metformin may improve the efficacy and reduce adverse drug reactions. Therefore, metformin can also be used as an adjuvant therapeutic agent for HM. This paper highlights the anti-hyperglycemic effects and potential anti-cancer effects of metformin, and also compiles the in vitro and clinical trials of metformin as an anti-cancer and chemosensitizing agent for the treatment of HM. The need for future research on the use of metformin in the treatment of HM is indicated.
Collapse
Affiliation(s)
| | | | | | | | - Baoan Chen
- Department of Hematology and Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| |
Collapse
|
7
|
Bonavida B. Historical Perspectives of the Role of NO/NO Donors in Anti-Tumor Activities: Acknowledging Dr. Keefer's Pioneering Research. Crit Rev Oncog 2023; 28:1-13. [PMID: 37824383 DOI: 10.1615/critrevoncog.2021035853] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
The role of nitric oxide (NO) in cancer has been a continuous challenge and particularly the contradictory findings in the literature reporting NO with either anti-cancer properties or pro-cancer properties. This dilemma was largely resolved by the level of NO/inducible nitric oxide synthase in the tumor environment as well as other cancer-associated gene activations in different cancers. The initial findings on the role of NO as an anti-cancer agent was initiated in the late 1990's in Dr. Larry Keefer's laboratory, who had been studying and synthesizing many compounds with releasing NO under different conditions. Using an experimental model with selected NO compounds they demonstrated for the first time that NO can inhibit tumor cell proliferation and sensitizes drug-resistant cancer cells to chemotherapy-induced cytotoxicity. This initial finding was the backbone and the foundation of subsequent reports by the Keefer's laboratory and followed by many others to date on NO-mediated anti-cancer activities and the clinical translation of NO donors in cancer therapy. Our laboratory initiated studies on NO-mediated anti-cancer therapy and chemo-immuno-sensitization following Keefer's findings and used one of his synthesized NO donors, namely, (Z)-1-[N-(2-aminoethyl)-N-(2-ammonioethyl)amino]diazen-1-ium-1,2-diolate (DETANONOate), throughout most of our studies. Many of Keefer's collaborators and other investigators have reported on the selected compound, O2-(2,4-dinitrophenyl) 1-[(4-ethoxycarbonyl)piperazin-1-yl] diazen-1-ium-1,2-diolate (JS-K), and its therapeutic role in many tumor model systems. Several lines of evidence that investigated the treatment with NO donors in various cancer models revealed that a large number of gene products are modulated by NO, thus emphasizing the pleiotropic effects of NO on cancers and the identification of many targets of therapeutic significance. The present review reports historically of several examples reported in the literature that emanated on NO-mediated anti-cancer activities by the Keefer's laboratory and his collaborators and other investigators including my laboratory at the University of California at Los Angeles.
Collapse
Affiliation(s)
- Benjamin Bonavida
- Department of Microbiology, Immunology, & Molecular Genetics, David Geffen School of Medicine at UCLA, Johnson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA 90025-1747, USA
| |
Collapse
|
8
|
Xu L, Wu X, Liu H, Dong G, Zhan J, Li G, Wang G, Liu T. Effects of combination docetaxel with NO treatment to enhance the anti-nasopharyngeal carcinoma efficiency in vitro and in vivo. Eur J Pharm Sci 2022; 178:106281. [PMID: 35995348 DOI: 10.1016/j.ejps.2022.106281] [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: 05/28/2022] [Revised: 08/05/2022] [Accepted: 08/18/2022] [Indexed: 02/05/2023]
Abstract
Nasopharyngeal carcinoma (NPC) is one of the major causes of death in Southern China. Due to the insidious location of NPC, the therapeutic effect of locoregionally advanced NPC is still unsatisfactory. In this work, to improve the treatment efficiency, combining DOC and JS-K to inhibit NPC cells (HNE-1) in vitro was investigated, as well as its possible mechanisms. Moreover, the in vivo effects of DOC and JS-K combination treatment were also evaluated in a xenograft model with HNE-1 cells. In vitro experiments including cell proliferation, migration ability, apoptosis, and expression levels of apoptosis-associated proteins revealed that the combination of DOC and JS-K was able to enhance antitumor effects. In vivo results further confirmed a significant treatment effect without obvious toxicity on mice. The present work provides a promising idea for the treatment of locally advanced NPC.
Collapse
Affiliation(s)
- Lingling Xu
- Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Xidong Wu
- Department of Drug Safety Evaluation, Jiangxi Testing Center of Medical Instruments, Nanchang, 330029, China
| | - Huiqin Liu
- Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China; Shantou University Medical College, Shantou, 515063, China
| | - Guangyuan Dong
- Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China; Shantou University Medical College, Shantou, 515063, China
| | - Jiandong Zhan
- Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Guanxue Li
- Pediatric Critical Care Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, 510280, China
| | - Guanhai Wang
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China.
| | - Tao Liu
- Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
9
|
Xing Y, Hu Y, Zou H, Xie H, Jiang T, Liu L. A novel O 2- (2,4-dinitrophenyl) diazeniumdiolate inhibits hepatocellular carcinoma migration, invasion, and EMT through the Wnt/β-catenin pathway. Toxicol In Vitro 2022; 84:105456. [PMID: 35985571 DOI: 10.1016/j.tiv.2022.105456] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 08/09/2022] [Accepted: 08/11/2022] [Indexed: 11/15/2022]
Abstract
Targeted Wnt/β-catenin pathway is considered to be a promising therapy for cancer metastasis. The novel O2 -(2,4-dinitrophenyl) diazeniumdiolate (JS-K) plays a potent inhibitory role in the proliferation of cancers. In this study, HepG2 and SMMC7721 were used to clarify the efficacy of JS-K inhibition of HCC metastasis. JS-K significantly inhibited cell motility through a wound-healing assay and restrained cell migration and invasion at noncytotoxic concentrations. However, the inhibitory effects of migration and invasion were abolished after the addition of NO scavenger, Carboxy-PTIO. In addition, JS-K inhibited the Wnt/β-catenin pathway by a decrease of p-GSK-3β at Ser9, cytosolic β-catenin, and nuclear β-catenin accumulation whereas an increase of p-β-catenin. Furthermore, the transcription regulators c-Myc, survivin, and Cyclin D1 were down-regulated after treating with JS-K. The inhibitory of the Wnt/β-catenin pathway was reversed after the addition of Carboxy-PTIO or LiCl. Meanwhile, JS-K also inhibited the epithelial-mesenchymal transition (EMT)-mediated cell migration and invasion. The characteristics of the inhibition were reflected by the upregulation of E-cadherin whereas the downregulation of Vimentin, Snail, and Slug. Taking together, these results demonstrated that JS-K inhibited HepG2 and SMMC7721 cells migration and invasion by reversing EMT via the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Yihao Xing
- Department of Pharmacy, School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang 471023, China
| | - Yile Hu
- Department of Pharmacy, School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang 471023, China
| | - Hanzhi Zou
- Department of Pharmacy, School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang 471023, China
| | - Huaxia Xie
- Department of Pharmacy, School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang 471023, China
| | - Tianci Jiang
- Department of Pharmacy, School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang 471023, China
| | - Ling Liu
- Department of Pharmacy, School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang 471023, China.
| |
Collapse
|
10
|
Markowicz-Piasecka M, Huttunen J, Zajda A, Sikora J, Huttunen KM. Sulfonamide metformin derivatives induce mitochondrial-associated apoptosis and cell cycle arrest in breast cancer cells. Chem Biol Interact 2021; 352:109795. [PMID: 34953865 DOI: 10.1016/j.cbi.2021.109795] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/10/2021] [Accepted: 12/20/2021] [Indexed: 11/03/2022]
Abstract
Metformin, an oral anti-diabetic drug, has attracted scientific attention due to its anti-cancer effects. This biguanide exerts preventive effects against cancer, and interferes with cancer-promoting signaling pathways at the cellular level. However, the direct cytotoxic or anti-proliferative effect of the drug is observed at very high concentrations, often exceeding 5-10 mM. This paper presents the synthesis of eight novel sulfonamide-based biguanides with improved cellular uptake in two breast cancer cell lines (MCF-7 and MDA-MB-231), and evaluates their effects on cancer cell growth. The synthesized sulfonamide-based analogues of metformin (1-5) were efficiently taken up in MCF-7 and MDA-MB-231 cells, and were characterized by stronger cytotoxic properties than those of metformin. Generally, compounds were more effective in MCF-7 than in MDA-MB-231. Compound 2, with an n-octyl chain, was the most active molecule with IC50 = 114.0 μmol/L in MCF-7 cells. The cytotoxicity of compound 2 partially results from its ability to induce early and late apoptosis. Increased intracellular reactive oxygen species (ROS) production and reduced mitochondrial membrane potential suggest that compound 2 promotes mitochondrial dysfunction and activates the mitochondrial-associated apoptosis-signaling pathway. In addition, compound 2 was also found to arrest cell cycle in the G0/G1 and G2/M phase and significantly inhibit cancer cell migration. In conclusion, this study supports the hypothesis that improved transporter-mediated cellular uptake of potential drug molecule is accompanied by its increased cytotoxicity. Therefore, compound 2 is a very good example of how chemical modification of a biguanide scaffold can affect its biological properties and improve anti-neoplastic potential.
Collapse
Affiliation(s)
- Magdalena Markowicz-Piasecka
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego1, 90-151, Lodz, Poland.
| | - Johanna Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1C, POB 1627, 70211, Kuopio, Finland
| | - Agnieszka Zajda
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego1, 90-151, Lodz, Poland
| | - Joanna Sikora
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego1, 90-151, Lodz, Poland.
| | - Kristiina M Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1C, POB 1627, 70211, Kuopio, Finland.
| |
Collapse
|
11
|
Fontana F, Limonta P. The multifaceted roles of mitochondria at the crossroads of cell life and death in cancer. Free Radic Biol Med 2021; 176:203-221. [PMID: 34597798 DOI: 10.1016/j.freeradbiomed.2021.09.024] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/22/2021] [Accepted: 09/27/2021] [Indexed: 12/15/2022]
Abstract
Mitochondria are the cytoplasmic organelles mostly known as the "electric engine" of the cells; however, they also play pivotal roles in different biological processes, such as cell growth/apoptosis, Ca2+ and redox homeostasis, and cell stemness. In cancer cells, mitochondria undergo peculiar functional and structural dynamics involved in the survival/death fate of the cell. Cancer cells use glycolysis to support macromolecular biosynthesis and energy production ("Warburg effect"); however, mitochondrial OXPHOS has been shown to be still active during carcinogenesis and even exacerbated in drug-resistant and stem cancer cells. This metabolic rewiring is associated with mutations in genes encoding mitochondrial metabolic enzymes ("oncometabolites"), alterations of ROS production and redox biology, and a fine-tuned balance between anti-/proapoptotic proteins. In cancer cells, mitochondria also experience dynamic alterations from the structural point of view undergoing coordinated cycles of biogenesis, fusion/fission and mitophagy, and physically communicating with the endoplasmic reticulum (ER), through the Ca2+ flux, at the MAM (mitochondria-associated membranes) levels. This review addresses the peculiar mitochondrial metabolic and structural dynamics occurring in cancer cells and their role in coordinating the balance between cell survival and death. The role of mitochondrial dynamics as effective biomarkers of tumor progression and promising targets for anticancer strategies is also discussed.
Collapse
Affiliation(s)
- Fabrizio Fontana
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, Milano, Italy.
| | - Patrizia Limonta
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, Milano, Italy.
| |
Collapse
|
12
|
Misirkic Marjanovic MS, Vucicevic LM, Despotovic AR, Stamenkovic MM, Janjetovic KD. Dual anticancer role of metformin: an old drug regulating AMPK dependent/independent pathways in metabolic, oncogenic/tumorsuppresing and immunity context. Am J Cancer Res 2021; 11:5625-5643. [PMID: 34873484 PMCID: PMC8640802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 10/29/2021] [Indexed: 06/13/2023] Open
Abstract
Metformin has been known to treat type 2 diabetes for decades and is widely prescribed antidiabetic drug. Recently, its anticancer potential has also been discovered. Moreover, metformin has low cost thus it has attained profound research interest. Comprehensing the complexity of the molecular regulatory networks in cancer provides a mode for advancement of research in cancer development and treatment. Metformin targets many pathways that play an important role in cancer cell survival outcome. Here, we described anticancer activity of metformin on the AMPK dependent/independent mechanisms regulating metabolism, oncogene/tumor suppressor signaling pathways together with the issue of clinical studies. We also provided brief overwiev about recently described metformin's role in cancer immunity. Insight in these complex molecular networks, will simplify application of metformin in clinical trials and contribute to improvement of anti-cancer therapy.
Collapse
Affiliation(s)
- Maja S Misirkic Marjanovic
- Department of Neurophysiology, Institute for Biological Research “Sinisa Stankovic”, National Institute of Republic of Serbia, University of BelgradeSerbia
| | - Ljubica M Vucicevic
- Department of Neurophysiology, Institute for Biological Research “Sinisa Stankovic”, National Institute of Republic of Serbia, University of BelgradeSerbia
| | - Ana R Despotovic
- Department of Neurophysiology, Institute for Biological Research “Sinisa Stankovic”, National Institute of Republic of Serbia, University of BelgradeSerbia
| | - Marina M Stamenkovic
- Department of Immunology, Institute of Microbiology and Immunology, Faculty of Medicine, University of BelgradeSerbia
| | - Kristina D Janjetovic
- Department of Neurophysiology, Institute for Biological Research “Sinisa Stankovic”, National Institute of Republic of Serbia, University of BelgradeSerbia
| |
Collapse
|
13
|
Hansel C, Barr S, Schemann AV, Lauber K, Hess J, Unger K, Zitzelsberger H, Jendrossek V, Klein D. Metformin Protects against Radiation-Induced Acute Effects by Limiting Senescence of Bronchial-Epithelial Cells. Int J Mol Sci 2021; 22:7064. [PMID: 34209135 PMCID: PMC8268757 DOI: 10.3390/ijms22137064] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 06/25/2021] [Accepted: 06/27/2021] [Indexed: 12/13/2022] Open
Abstract
Radiation-induced damage to normal lung parenchyma remains a dose-limiting factor in thorax-associated radiotherapy (RT). Severe early and late complications with lungs can increase the risk of morbidity in cancer patients after RT. Herein, senescence of lung epithelial cells following RT-induced cellular stress, or more precisely the respective altered secretory profile, the senescence-associated secretory phenotype (SASP), was suggested as a central process for the initiation and progression of pneumonitis and pulmonary fibrosis. We previously reported that abrogation of certain aspects of the secretome of senescent lung cells, in particular, signaling inhibition of the SASP-factor Ccl2/Mcp1 mediated radioprotection especially by limiting endothelial dysfunction. Here, we investigated the therapeutic potential of a combined metformin treatment to protect normal lung tissue from RT-induced senescence and associated lung injury using a preclinical mouse model of radiation-induced pneumopathy. Metformin treatment efficiently limited RT-induced senescence and SASP expression levels, thereby limiting vascular dysfunctions, namely increased vascular permeability associated with increased extravasation of circulating immune and tumor cells early after irradiation (acute effects). Complementary in vitro studies using normal lung epithelial cell lines confirmed the senescence-limiting effect of metformin following RT finally resulting in radioprotection, while fostering RT-induced cellular stress of cultured malignant epithelial cells accounting for radiosensitization. The radioprotective action of metformin for normal lung tissue without simultaneous protection or preferable radiosensitization of tumor tissue might increase tumor control probabilities and survival because higher radiation doses could be used.
Collapse
Affiliation(s)
- Christine Hansel
- Institute of Cell Biology (Cancer Research), University Hospital, Essen, University of Duisburg-Essen, 45122 Essen, Germany; (C.H.); (S.B.); (A.V.S.); (V.J.)
| | - Samantha Barr
- Institute of Cell Biology (Cancer Research), University Hospital, Essen, University of Duisburg-Essen, 45122 Essen, Germany; (C.H.); (S.B.); (A.V.S.); (V.J.)
| | - Alina V. Schemann
- Institute of Cell Biology (Cancer Research), University Hospital, Essen, University of Duisburg-Essen, 45122 Essen, Germany; (C.H.); (S.B.); (A.V.S.); (V.J.)
| | - Kirsten Lauber
- Department of Radiation Oncology, University Hospital, LMU München, 80539 Munich, Germany;
- German Cancer Consortium (DKTK), Partner Site Munich, 80539 Munich, Germany
- Clinical Cooperation Group ‘Personalized Radiotherapy in Head and Neck Cancer’ Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (J.H.); (K.U.); (H.Z.)
| | - Julia Hess
- Clinical Cooperation Group ‘Personalized Radiotherapy in Head and Neck Cancer’ Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (J.H.); (K.U.); (H.Z.)
- Research Unit Radiation Cytogenetics, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany
| | - Kristian Unger
- Clinical Cooperation Group ‘Personalized Radiotherapy in Head and Neck Cancer’ Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (J.H.); (K.U.); (H.Z.)
- Research Unit Radiation Cytogenetics, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany
| | - Horst Zitzelsberger
- Clinical Cooperation Group ‘Personalized Radiotherapy in Head and Neck Cancer’ Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (J.H.); (K.U.); (H.Z.)
- Research Unit Radiation Cytogenetics, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University Hospital, Essen, University of Duisburg-Essen, 45122 Essen, Germany; (C.H.); (S.B.); (A.V.S.); (V.J.)
| | - Diana Klein
- Institute of Cell Biology (Cancer Research), University Hospital, Essen, University of Duisburg-Essen, 45122 Essen, Germany; (C.H.); (S.B.); (A.V.S.); (V.J.)
| |
Collapse
|
14
|
Song A, Zhang C, Meng X. Mechanism and application of metformin in kidney diseases: An update. Biomed Pharmacother 2021; 138:111454. [PMID: 33714781 DOI: 10.1016/j.biopha.2021.111454] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/26/2021] [Accepted: 02/27/2021] [Indexed: 01/08/2023] Open
Abstract
Metformin is an oral antihyperglycemic drug widely used to treat type 2 diabetes mellitus (T2DM), acting via indirect activation of 5' Adenosine monophosphate-activated Protein Kinase (AMPK). Beyond the anti-diabetic effect, accumulative pieces of evidence have revealed that metformin also everts a beneficial effect in diverse kidney diseases. In various acute kidney diseases (AKI) animal models, metformin protects renal tubular cells from inflammation, apoptosis, reactive oxygen stress (ROS), endoplasmic reticulum (ER) stress, epithelial-mesenchymal transition (EMT) via AMPK activation. In diabetic kidney disease (DKD), metformin also alleviates podocyte loss, mesangial cells apoptosis, and tubular cells senescence through AMPK-mediated signaling pathways. Besides, metformin inhibits cystic fibrosis transmembrane conductance regulator (CFTR)-mediated fluids secretion and the mammalian target of rapamycin (mTOR)-involved cyst formation negatively regulated by AMPK in autosomal dominant polycystic kidney disease (APDKD). Furthermore, metformin also contributes to the alleviation of urolithiasis and renal cell carcinoma (RCC). As the common pathway for chronic kidney disease (CKD) progressing towards end-stage renal disease (ESRD), renal fibrosis is ameliorated by metformin, to a great extent dependent on AMPK activation. However, clinical data are not always consistent with preclinical data, some clinical investigations showed the unmeaningful even detrimental effect of metformin on T2DM patients with kidney diseases. Most importantly, metformin-associated lactic acidosis (MALA) is a vital issue restricting the application of metformin. Thus, we conclude the application of metformin in kidney diseases and uncover the underlying molecular mechanisms in this review.
Collapse
Affiliation(s)
- Anni Song
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xianfang Meng
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
15
|
Banerjee S, Katiyar P, Kumar V, Saini SS, Varshney R, Krishnan V, Sircar D, Roy P. Black pepper and piperine induce anticancer effects on leukemia cell line. Toxicol Res (Camb) 2021; 10:169-182. [PMID: 33884168 PMCID: PMC8045589 DOI: 10.1093/toxres/tfab001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 12/16/2020] [Accepted: 01/01/2021] [Indexed: 11/12/2022] Open
Abstract
The black pepper, most commonly used in Indian cuisines for ages, is considered as "king of spices." The present study evaluates the anticancer potential of black pepper and its main constituent, i.e. alkaloid piperine, against human leukemia cell line, K-562 cells. Gas chromatography-mass spectrometry (GC-MS) analysis confirmed the presence of piperine in black pepper extract. The methanolic extract of black pepper (BP-M) and pure piperine (PIP) showed a strong cytotoxic effect against this cell line. Both BP-M and PIP generated apoptotic bodies in K-562 cells and caused nuclear condensation as visualized by fluorescent microscopy, which was further confirmed by flow cytometry analysis. BP-M and PIP also generated reactive oxygen species in K-562 cells as established by flow cytometry. The translation of Bax, caspase-3 and caspase-9 genes was found to be upregulated with subsequent downregulation of Bcl-2 gene. The anti-proliferative effect of both BP-M and PIP was also observed by trypan blue staining and was further confirmed by the downregulated expression of proliferating cell nuclear antigen (PCNA). The molecular docking studies showed the binding of PIP with PCNA and Bcl-2 and supported the in vitro findings. The docking studies also proposed the binding of PIP to ADP binding pocket of Apaf-1 protein. Taken together, these findings signify the anticancer potential of both black pepper and PIP, thus proposing black pepper as a potent nutraceutical for preventing the progression of chronic myeloid leukemia.
Collapse
Affiliation(s)
- Somesh Banerjee
- Molecular Endocrinology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Parul Katiyar
- Molecular Endocrinology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Vijay Kumar
- Laboratory of Structural Microbiology, Regional Centre for Biotechnology, Faridabad 121001, Haryana, India
| | - Shashank Sagar Saini
- Plant Molecular Biology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Ritu Varshney
- Molecular Endocrinology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Vengadesan Krishnan
- Laboratory of Structural Microbiology, Regional Centre for Biotechnology, Faridabad 121001, Haryana, India
| | - Debabrata Sircar
- Plant Molecular Biology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Partha Roy
- Molecular Endocrinology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| |
Collapse
|
16
|
Mu N, Xu T, Gao M, Dong M, Tang Q, Hao L, Wang G, Li Z, Wang W, Yang Y, Hou J. Therapeutic effect of metformin in the treatment of endometrial cancer. Oncol Lett 2020; 20:156. [PMID: 32934724 DOI: 10.3892/ol.2020.12017] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 07/14/2020] [Indexed: 12/11/2022] Open
Abstract
The present review aims at reviewing the role of metformin in the treatment of endometrial cancer (EC). According to the literature, excessive estrogen levels and insulin resistance are established risk factors of EC. As a traditional insulin sensitizer and newly discovered anticancer agent, metformin directly and indirectly inhibits the development of EC. The direct mechanisms of metformin include inhibition of the LKB1-AMP-activated protein kinase-mTOR, PI3K-Akt and insulin-like growth factor 1-related signaling pathways, which reduces the proliferation and promotes the apoptosis of EC cells. In the indirect mechanism, metformin increases the insulin sensitivity of body tissues and decreases circulating insulin levels. Decreased levels of insulin increase the blood levels of sex hormone binding globulin, which leads to reductions in circulating estrogen and androgens. The aforementioned findings suggest that metformin serves an important role in the treatment of EC. Increased understanding of the mechanism of metformin in EC may provide novel insights into the treatment of this malignancy.
Collapse
Affiliation(s)
- Nan Mu
- Department of Gynecology and Obstetrics, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Tingting Xu
- Department of Gynecology and Obstetrics, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Mingxiao Gao
- Department of Cardiology, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Mei Dong
- Department of Cardiology, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Qing Tang
- Department of Gynecology and Obstetrics, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Li Hao
- Department of Gynecology and Obstetrics, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Guiqing Wang
- Department of Gynecology and Obstetrics, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Zenghui Li
- Department of Gynecology and Obstetrics, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Wenshuang Wang
- Department of Gynecology and Obstetrics, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Ying Yang
- Department of Gynecology and Obstetrics, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Jianqing Hou
- Department of Gynecology and Obstetrics, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong 264000, P.R. China
| |
Collapse
|