1
|
Pan W, Shi Y, Sun F, Zou M, Piao H. Oleogel-mediated Topical Administration of Roflumilast and Paclitaxel as a Synergistic Strategy to Combat Imiquimod-induced Psoriasis. AAPS PharmSciTech 2025; 26:127. [PMID: 40342026 DOI: 10.1208/s12249-025-03118-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 04/17/2025] [Indexed: 05/11/2025] Open
Abstract
Psoriasis is a systemic immune disease with severe inflammation and skin thickening. Roflumilast (ROF) blocks cAMP hydrolysis, and paclitaxel (PTX) inhibits cell proliferation; both are effective in topical psoriasis treatment. However, the combination of ROF and PTX has not been reported. This study explored their synergistic mechanism and formulated a ROF-PTX oleogel with strong skin adhesion, low viscosity, enhanced skin penetration, and increased retention. The oleogel, prepared via direct gelation with jojoba oil as oil phase, PPG-15 as solvent, Transcutol as solubilizer, and hydrogenated castor oil as oleogelator. It showed 78.9% holding oil capacity and a viscosity of 0.4049 Pa·s, indicating excellent stability and adhesion. In the imiquimod-induced psoriasis model, the ROF:PTX (1:1) oleogel reduced Baker scores and splenic indices more effectively than ROF or PTX alone. Histological studies suggested that the combination was superior in reducing inflammation and skin thickening. The ROF:PTX (1:1) oleogel group exhibited lower Baker scores and epidermal thickness, demonstrating superior therapeutic efficacy. The H-SCORE revealed a 2.95-fold reduction in IL-17 levels compared to the model group, highlighting the potential of the ROF and PTX combination as an effective psoriasis treatment strategy.
Collapse
Affiliation(s)
- Wenxiu Pan
- Zhejiang Medicine Co.,Ltd., Zhejiang Province, Shaoxing, China
| | - Yunyi Shi
- Emory University, 201 Dowman Dr, Atlanta, GA, United States of America
| | - Fei Sun
- Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province, 110016, China
| | - Meijuan Zou
- Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province, 110016, China
| | - Hongyu Piao
- Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province, 110016, China.
| |
Collapse
|
2
|
Ordaz-Ramos A, Diaz-Blancas J, Martínez-Cruz A, Castro-Oropeza R, Zampedri C, Romero-Rodríguez DP, Rodriguez-Dorantes M, Melendez-Zajgla J, Maldonado V, Vazquez-Santillan K. RANKL regulates differentially breast cancer stem cell properties through its RANK and LGR4 receptors. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119888. [PMID: 39662745 DOI: 10.1016/j.bbamcr.2024.119888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 10/31/2024] [Accepted: 12/02/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND Breast cancer stem cells (BCSC) are a subpopulation responsible for cancer resistance and relapse. The receptor activator of nuclear factor kappa-Β ligand (RANKL) is a cytokine capable of activating RANK and LGR4 receptors. RANKL/RANK signaling maintains the self-renewal of BCSCs, however, the effect of RANKL via LGR4 remains unclear. Evidence from osteoclasts suggests that RANKL/LGR4 axis disrupts RANK signaling, leading to opposing cellular responses. Anti-RANKL inhibitors are potential agents for eradicating CSCs, but their effect on RANKL/LGR4 signal has not been demonstrated. OBJECTIVE This project aimed to elucidate the role of RANKL in regulating stemness depending on the expression of its receptors. METHODS We use in vitro and in vivo approaches to evaluate the effects of RANKL inhibition in stemness in low or high-LGR4 expressing cells. Furthermore, we analyze the effects of RANKL stimulation on the stemness of LGR4 or RANK overexpressing cells. Additionally, we evaluated the impact of RANKL/LGR4 signaling in the activity of Wnt/β-catenin and NF-κB signaling pathways. RESULTS Our findings indicated that elevated RANKL expression is related to a favorable prognosis in patients with high LGR4 levels. Furthermore, RANKL inhibition decreased BCSC properties in LGR4-low cell lines, while it promoted migration in LGR4-high cells. Additionally, the RANKL/RANK axis activated NF-κB signaling and enhanced BCSCs in RANK-overexpressing cells. In contrast, in LGR4-overexpressing cells, RANKL failed to activate NF-κB but instead inhibited the Wnt/β-catenin pathway, leading to a reduction in BCSCs. CONCLUSION Our findings suggest that RANKL exerts different responses according to the expression of its receptors.
Collapse
Affiliation(s)
- Alejandro Ordaz-Ramos
- Innovation and Precision Medicine Laboratory, Instituto Nacional de Medicina Genómica, Periférico Sur No.4809, Col Arenal Tepepan, Tlalpan, Mexico City C.P. 14610, Mexico; Posgrado en Ciencias Biológicas, Unidad de Posgrado, Edificio D, 1° Piso, Circuito de Posgrados, Ciudad Universitaria, Coyoacán, Mexico City C.P. 04510, Mexico
| | - Jorge Diaz-Blancas
- Epigenetics Laboratory, Instituto Nacional de Medicina Genómica, Periférico Sur No.4809, Col Arenal Tepepan, Tlalpan, Mexico City C.P. 14610, Mexico
| | - Aketzalli Martínez-Cruz
- Innovation and Precision Medicine Laboratory, Instituto Nacional de Medicina Genómica, Periférico Sur No.4809, Col Arenal Tepepan, Tlalpan, Mexico City C.P. 14610, Mexico
| | - Rosario Castro-Oropeza
- Molecular Oncology Laboratory, Unidad de Investigación Médica en Enfermedades Oncológicas, Hospital de Oncologia, Centro Medico Nacional Siglo XXI, IMSS, Avenida Cuahuhtemoc 330, Col Doctores, Cuauhtemoc, Mexico City C.P. 06720, Mexico
| | - Cecilia Zampedri
- Multidisciplinary Zebrafish Laboratory, Department of Bioengineer, Escuela de Ingenieria y Ciencias, Instituto Tecnologico y de Estudios Superiores Monterrey, Mexico City, Mexico
| | - Damaris P Romero-Rodríguez
- Flow Citometry Laboratory, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosió Villegas", Calzada de Tlalpan 4502, Col Belisario Dominguez Secc 16, Tlalpan, C.P. 14080, Mexico; Laboratorio Nacional Conahcyt de Investigación y Diagnóstico por Inmunocitofluorometría (LANCIDI), Mexico City, Mexico
| | - Mauricio Rodriguez-Dorantes
- Oncogenomics Laboratory, Instituto Nacional de Medicina Genómica, Periférico Sur No.4809, Col Arenal Tepepan, Tlalpan, Mexico City C.P. 14610, Mexico
| | - Jorge Melendez-Zajgla
- Functional Cancer Genomics Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14160, Mexico
| | - Vilma Maldonado
- Epigenetics Laboratory, Instituto Nacional de Medicina Genómica, Periférico Sur No.4809, Col Arenal Tepepan, Tlalpan, Mexico City C.P. 14610, Mexico
| | - Karla Vazquez-Santillan
- Innovation and Precision Medicine Laboratory, Instituto Nacional de Medicina Genómica, Periférico Sur No.4809, Col Arenal Tepepan, Tlalpan, Mexico City C.P. 14610, Mexico.
| |
Collapse
|
3
|
Pérez-Chacón G, Santamaría PG, Redondo-Pedraza J, González-Suárez E. RANK/RANKL Signaling Pathway in Breast Development and Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:309-345. [PMID: 39821032 DOI: 10.1007/978-3-031-70875-6_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
RANK pathway has attracted increasing interest as a promising target in breast cancer, given the availability of denosumab, an anti-RANKL drug. RANK signaling mediates progesterone-driven regulation of mammary gland development and favors breast cancer initiation by controlling mammary cell proliferation and stem cell fate. RANK activation promotes luminal mammary epithelial cell senescence, acting as an initial barrier to tumorigenesis but ultimately facilitating tumor progression and metastasis. Comprehensive analyses have demonstrated that RANK protein expression is an independent biomarker of poor prognosis in postmenopausal and estrogen receptor-negative breast cancer patients. RANK pathway also has multiple roles in immunity and inflammation, regulating innate and adaptive responses. In the tumor microenvironment, RANK and RANKL are expressed by different immune cell populations and contribute to the regulation of tumor immune surveillance, mainly driving immunosuppressive effects.Herein, we discuss the preventive and therapeutic potential of targeting RANK signaling in breast cancer given its tumor cell intrinsic and extrinsic effects. RANKL inhibition has been shown to induce mammary tumor cell differentiation and an antitumor immune response. Moreover, loss of RANK signaling increases sensitivity of breast cancer cells to chemotherapy, targeted therapies such as HER2 and CDK4/6 inhibitors, and immunotherapy. Finally, we describe clinical trials of denosumab for breast cancer prevention, such as those ongoing in women with high risk of developing breast cancer, large phase III clinical trials where the impact of adjuvant denosumab on disease-free survival has been assessed, and window trials to evaluate the immunomodulatory effects of denosumab in breast cancer and other solid tumors.
Collapse
Affiliation(s)
- Gema Pérez-Chacón
- Molecular Oncology, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | | | | | - Eva González-Suárez
- Molecular Oncology, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.
- Oncobell, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.
| |
Collapse
|
4
|
Gatting L, Ahmed S, Meccheri P, Newlands R, Kehagia AA, Waller J. Acceptability of artificial intelligence in breast screening: focus groups with the screening-eligible population in England. BMJ PUBLIC HEALTH 2024; 2:e000892. [PMID: 40018529 PMCID: PMC11816108 DOI: 10.1136/bmjph-2024-000892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 10/31/2024] [Indexed: 03/01/2025]
Abstract
Introduction Preliminary studies of artificial intelligence (AI) tools developed to support breast screening demonstrate the potential to reduce radiologist burden and improve cancer detection which could lead to improved breast cancer outcomes. This study explores the public acceptability of the use of AI in breast screening from the perspective of screening-eligible women in England. Methods 64 women in England, aged 50-70 years (eligible for breast screening) and 45-49 years (approaching eligibility), participated in 12 focus groups-8 online and 4 in person. Specific scenarios in which AI may be used in the mammogram reading process were presented. Data were analysed using a reflexive thematic analysis. Results Four themes described public perceptions of AI in breast screening found in this study: (1) Things going wrong and being missed summarises a predominant and pervasive concern about an AI tool being used in breast screening; (2) Speed of change and loss of control captures a positive association of AI with technological advances held by the women but also feelings of things being out of their control, and that they were being left behind and in the dark; (3) The importance of humans reports concern around the possibility that AI excludes humans and renders them redundant and (4) Desire for thorough research, staggered implementation and double-checking of scans included insistence that any AI be thoroughly trialled, tested and not solely relied on when initially implemented. Conclusions It will be essential that future decision-making and communication about AI implementation in breast screening (and, likely, in healthcare more widely) address concerns surrounding (1) the fallibility of AI, (2) lack of inclusion, control and transparency in relation to healthcare and technology decisions and (3) humans being left redundant and unneeded, while building on women's hopes for the technology.
Collapse
Affiliation(s)
- Lauren Gatting
- Cancer Prevention Group, School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
- King’s Technology Evaluation Centre, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
- Centre for Cancer Screening, Prevention and Early Diagnosis, Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - Syeda Ahmed
- School of Mental Health & Psychological Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Priscilla Meccheri
- School of Mental Health & Psychological Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Rumana Newlands
- Health Services Research Unit, University of Aberdeen, Aberdeen, UK
| | - Angie A Kehagia
- King’s Technology Evaluation Centre, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Jo Waller
- Cancer Prevention Group, School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
- Centre for Cancer Screening, Prevention and Early Diagnosis, Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| |
Collapse
|
5
|
Zhang C, Cheng H, Dou S, Wang Y, Ye X, Cui H, Chang X, Li Y. Near-infrared fluorescent molecular probes with cetuximab in the in vivo fluorescence imaging for epithelial ovarian cancer. J Ovarian Res 2024; 17:225. [PMID: 39543737 PMCID: PMC11566390 DOI: 10.1186/s13048-024-01547-5] [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: 07/22/2024] [Accepted: 10/27/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND Near-infrared fluorescence (NIRF) imaging is an excellent choice for image-guided surgery due to its simple operation and non-invasiveness. Developing tumor-specific fluorescent molecular probes is key to fluorescence imaging-guided surgery. EGFR (epidermal growth factor receptor) is closely related to the proliferation and growth of tumor cells and is highly expressed in epithelial ovarian cancer (EOC). The study aims to construct a NIR fluorescent molecular probe using cetuximab (an EGFR monoclonal antibody) and investigate its feasibility for targeting EOC in vivo through fluorescence imaging. METHODS We determined the expression of EGFR in EOC. NIR fluorescent molecular probe with cetuximab (cetuximab-Cy7) was chemically engineered and identified. The subcutaneous xenografted tumor model of EOC was induced using SKOV3-Luc cell line with positive expression of EGFR. Cetuximab-Cy7 was used for in vivo fluorescence imaging, and phosphate-buffered saline, free Cy7 dye and mouse isotype immunoglobulin G-Cy7 were used as controls. NIRF imaging system was performed to study the distribution and targeting of the probes. Tumors were imaged in situ and ex vivo, and fluorescent intensity was quantified. Resected specimens were analyzed to confirm diagnosis, and immunohistochemical (IHC) staining was used to identify EGFR expression. RESULTS EGFR expression was increased in EOC tissues than fallopian tube tissues. The high expression of EGFR was significantly correlated with well-differentiation, residual lesions ≤ 1 cm, no recurrence and increased survival. NIRF imaging showed that the cetuximab-Cy7 enabled detection of tumor lesions in EOC-bearing mice with the optimal dose of 30 µg. The suitable imaging time window may be 24-96 h post-injection. Ex vivo fluorescence imaging indicated that fluorescent signal was mainly detected in the tumor and the lung. IHC results confirmed that xenografts were EGFR positive. CONCLUSION Cetuximab-Cy7 can specifically target the tumors of EOC xenografted nude mice. This research lays the foundation for future studies on EOC surgery navigation.
Collapse
Affiliation(s)
- Chen Zhang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, No. 11 Xizhimen South Str., Xicheng District, Beijing, 100044, China
| | - Hongyan Cheng
- Department of Obstetrics and Gynecology, Peking University People's Hospital, No. 11 Xizhimen South Str., Xicheng District, Beijing, 100044, China
| | - Sha Dou
- Department of Obstetrics and Gynecology, Peking University People's Hospital, No. 11 Xizhimen South Str., Xicheng District, Beijing, 100044, China
| | - Yuanfen Wang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, No. 11 Xizhimen South Str., Xicheng District, Beijing, 100044, China
| | - Xue Ye
- Department of Obstetrics and Gynecology, Peking University People's Hospital, No. 11 Xizhimen South Str., Xicheng District, Beijing, 100044, China
| | - Heng Cui
- Department of Obstetrics and Gynecology, Peking University People's Hospital, No. 11 Xizhimen South Str., Xicheng District, Beijing, 100044, China.
| | - Xiaohong Chang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, No. 11 Xizhimen South Str., Xicheng District, Beijing, 100044, China.
| | - Yi Li
- Department of Obstetrics and Gynecology, Peking University People's Hospital, No. 11 Xizhimen South Str., Xicheng District, Beijing, 100044, China.
| |
Collapse
|
6
|
Silva S, Sousa JC, Nogueira C, Feijo R, Neto FM, Marinho LC, Sousa G, Denninghoff V, Tavora F. Relationship between the expressions of DLL3, ASC1, TTF-1 and Ki-67: First steps of precision medicine at SCLC. Oncotarget 2024; 15:750-763. [PMID: 39392394 PMCID: PMC11468345 DOI: 10.18632/oncotarget.28660] [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: 07/29/2024] [Accepted: 09/17/2024] [Indexed: 10/12/2024] Open
Abstract
This study presents an observational, cross-sectional analysis of 64 patients diagnosed with small cell lung cancer (SCLC) at a reference laboratory for thoracic pathology between 2022 and 2024. The primary objective was to evaluate the expression of Delta-like ligand 3 (DLL3) and other neuroendocrine markers such as Chromogranin, and Synaptophysin, utilizing both traditional immunohistochemistry and digital pathology tools. Patients were primarily older adults, with a median age of over 71, and most biopsies were obtained from lung parenchyma. Immunohistochemistry (IHC) was performed using specific monoclonal antibodies, with DLL3 showing variable expression across the samples. Notably, DLL3 was expressed in 72.3% of the cases, with varied intensities and a semi-quantitative H-score applied for more nuanced analysis. ASCL1 was expressed in 97% of cases, with the majority considered low-expressors. Only 11% had high expression. TTF-1, traditionally not a conventional marker for the diagnosis of SCLC, was positive in half of the cases, suggesting its potential as a biomarker. The study underscores the significant variability in the expression of neuroendocrine markers in SCLC, with implications for both diagnosis and potential therapeutic targeting. DLL3, particularly, shows promise as a therapeutic target due to its high expression rate in the cohort. The use of digital pathology software QuPath enhanced the accuracy and depth of analysis, allowing for detailed morphometric analysis and potentially informing more personalized treatment approaches. The findings emphasize the need for further research into the role of these markers in the management and treatment of SCLC, considering the poor prognosis and high mortality rate observed in the cohort.
Collapse
Affiliation(s)
- Samuel Silva
- Department of Pathology, Faculty of Medicine, Federal University of Ceará, Fortaleza (Ceará), Brazil
- ARGOS Laboratory, Fortaleza (Ceará), Brazil
| | | | - Cleto Nogueira
- Department of Pathology, Faculty of Medicine, Federal University of Ceará, Fortaleza (Ceará), Brazil
- ARGOS Laboratory, Fortaleza (Ceará), Brazil
| | - Raquel Feijo
- Department of Pathology, Faculty of Medicine, Federal University of Ceará, Fortaleza (Ceará), Brazil
- Messejana Heart and Lung Hospital, Fortaleza (Ceará), Brazil
| | | | - Laura Cardoso Marinho
- Department of Pathology, Faculty of Medicine, Federal University of Ceará, Fortaleza (Ceará), Brazil
- ARGOS Laboratory, Fortaleza (Ceará), Brazil
| | | | - Valeria Denninghoff
- Molecular Oncology Clinical Lab, University of Buenos Aires (UBA)—National Council for Scientific and Technical Research (CONICET), Buenos Aires, Argentina
- Liquid Biopsy and Cancer Interception Unit, GENYO, Centre for Genomics and Oncological Research (Pfizer/University of Granada/Andalusian Regional Government), Granada, Spain
| | - Fabio Tavora
- Department of Pathology, Faculty of Medicine, Federal University of Ceará, Fortaleza (Ceará), Brazil
- ARGOS Laboratory, Fortaleza (Ceará), Brazil
| |
Collapse
|
7
|
Wan YX, Qi XW, Lian YY, Liu ZY, Wang H, Qiu YQ, Zhang CG, Li WN, Jiang HL, Yang DH, Zhao W, Chen ZS, Huang JC. Electroacupuncture facilitates vascular normalization by inhibiting Glyoxalase1 in endothelial cells to attenuate glycolysis and angiogenesis in triple-negative breast cancer. Cancer Lett 2024; 598:217094. [PMID: 38945204 DOI: 10.1016/j.canlet.2024.217094] [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: 01/04/2024] [Revised: 06/13/2024] [Accepted: 06/27/2024] [Indexed: 07/02/2024]
Abstract
Recent therapeutic strategies for the treatment of triple-negative breast cancer (TNBC) have shifted the focus from vascular growth factors to endothelial cell metabolism. This study highlights the underexplored therapeutic potential of peri-tumoral electroacupuncture, a globally accepted non-pharmacological intervention for TNBC, and molecular mechanisms. Our study showed that peri-tumoral electroacupuncture effectively reduced the density of microvasculature and enhanced vascular functionality in 4T1 breast cancer xenografts, with optimal effects on day 3 post-acupuncture. The timely integration of peri-tumoral electroacupuncture amplified the anti-tumor efficacy of paclitaxel. Multi-omics analysis revealed Glyoxalase 1 (Glo1) and the associated methylglyoxal-glycolytic pathway as key mediators of electroacupuncture-induced vascular normalization. Peri-tumoral electroacupuncture notably reduced Glo1 expression in the endothelial cells of 4T1 xenografts. Using an in vivo matrigel plug angiogenesis assay, we demonstrated that either Glo1 knockdown or electroacupuncture inhibited angiogenesis. In contrast, Glo1 overexpression increased blood vessel formation. In vitro pharmacological inhibition and genetic knockdown of Glo1 in human umbilical vein endothelial cells inhibited proliferation and promoted apoptosis via downregulating the methylglyoxal-glycolytic pathway. The study using the Glo1-silenced zebrafish model further supported the role of Glo1 in vascular development. This study underscores the pivotal role of Glo1 in peri-tumoral electroacupuncture, spotlighting a promising avenue for enhancing vascular normalization and improving TNBC treatment outcomes.
Collapse
Affiliation(s)
- Yu-Xiang Wan
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xue-Wei Qi
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 100029, China
| | - Yan-Yan Lian
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Ze-Yu Liu
- Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Hui Wang
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yu-Qin Qiu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Chun-Guang Zhang
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Wen-Na Li
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Hong-Lin Jiang
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Dong-Hua Yang
- New York College of Traditional Chinese Medicine, Mineola, NY, 11501, USA
| | - Wei Zhao
- Guanganmen Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, 100029, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA.
| | - Jin-Chang Huang
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
8
|
Wunderle M, Heindl F, Behrens AS, Häberle L, Hack CC, Heusinger K, Huebner H, Gass P, Ruebner M, Schulz-Wendtland R, Erber R, Hartmann A, Beckmann MW, Dougall WC, Press MF, Fasching PA, Emons J. Correlation of RANK and RANKL with mammographic density in primary breast cancer patients. Arch Gynecol Obstet 2024; 310:1223-1233. [PMID: 38836929 PMCID: PMC11258178 DOI: 10.1007/s00404-024-07495-1] [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: 01/12/2024] [Accepted: 03/24/2024] [Indexed: 06/06/2024]
Abstract
PURPOSE The receptor activator of nuclear factor kappa B (RANK) and its ligand (RANKL) have been shown to promote proliferation of the breast and breast carcinogenesis. The objective of this analysis was to investigate whether tumor-specific RANK and RANKL expression in patients with primary breast cancer is associated with high percentage mammographic density (PMD), which is a known breast cancer risk factor. METHODS Immunohistochemical staining of RANK and RANKL was performed in tissue microarrays (TMAs) from primary breast cancer samples of the Bavarian Breast Cancer Cases and Controls (BBCC) study. For RANK and RANKL expression, histochemical scores (H scores) with a cut-off value of > 0 vs 0 were established. PMD was measured in the contralateral, non-diseased breast. Linear regression models with PMD as outcome were calculated using common predictors of PMD (age at breast cancer diagnosis, body mass index (BMI) and parity) and RANK and RANKL H scores. Additionally, Spearman rank correlations (ρ) between PMD and RANK and RANKL H score were performed. RESULTS In the final cohort of 412 patients, breast cancer-specific RANK and RANKL expression was not associated with PMD (P = 0.68). There was no correlation between PMD and RANK H score (Spearman's ρ = 0.01, P = 0.87) or RANKL H score (Spearman's ρ = 0.04, P = 0.41). RANK expression was highest in triple-negative tumors, followed by HER2-positive, luminal B-like and luminal A-like tumors, while no subtype-specific expression of RANKL was found. CONCLUSION Results do not provide evidence for an association of RANK and RANKL expression in primary breast cancer with PMD.
Collapse
Affiliation(s)
- Marius Wunderle
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Felix Heindl
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Annika S Behrens
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Lothar Häberle
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Biostatistics Unit, Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Carolin C Hack
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Katharina Heusinger
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Hanna Huebner
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Paul Gass
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Matthias Ruebner
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Rüdiger Schulz-Wendtland
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Institute of Diagnostic Radiology, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Ramona Erber
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Institute of Pathology, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Arndt Hartmann
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Institute of Pathology, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Matthias W Beckmann
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - William C Dougall
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4702, Australia
- Hematology and Oncology Research, Amgen, Inc, Seattle, WA, 98119, USA
| | - Michael F Press
- Department of Pathology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Peter A Fasching
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany.
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany.
| | - Julius Emons
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| |
Collapse
|
9
|
Zhou C, Cui H, Yang Y, Chen L, Feng M, Gao Y, Li D, Li L, Chen X, Li X, Cao Y. SEC61 translocon gamma subunit is correlated with glycolytic activity, epithelial mesenchymal transition and the immune suppressive phenotype of lung adenocarcinoma. Acta Biochim Biophys Sin (Shanghai) 2024; 56:1748-1760. [PMID: 38978503 PMCID: PMC11693863 DOI: 10.3724/abbs.2024109] [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: 01/26/2024] [Accepted: 04/08/2024] [Indexed: 07/10/2024] Open
Abstract
Lung adenocarcinoma (LUAD) remains a predominant cause of cancer-related mortality globally, underscoring the urgency for targeted therapeutic strategies. The specific role and impact of the SEC61 translocon gamma subunit (SEC61G) in LUAD progression and metastasis remain largely unexplored. In this study, we use a multifaceted approach, combining bioinformatics analysis with experimental validation, to elucidate the pivotal role of SEC61G and its associated molecular mechanisms in LUAD. Our integrated analyses reveal a significant positive correlation between SEC61G expression and the glycolytic activity of LUAD, as evidenced by increased fluorodeoxyglucose (FDG) uptake on positron emission tomography (PET)/CT scans. Further investigations show the potential influence of SEC61G on metabolic reprogramming, which contributes to the immunosuppressive tumor microenvironment (TME). Remarkably, we identify a negative association between SEC61G expression levels and the infiltration of critical immune cell populations within the TME, along with correlations with immune checkpoint gene expression and tumor heterogeneity scores in LUAD. Functional studies demonstrate that SEC61G knockdown markedly inhibits the migration of A549 and H2030 LUAD cells. This inhibitory effect is accompanied by a significant downregulation of key regulators of tumor progression, including hypoxia-inducible factor-1 alpha (HIF-1α), lactate dehydrogenase A, and genes involved in the epithelial-mesenchymal transition pathway. In conclusion, our comprehensive analyses position SEC61G as a potential prognostic biomarker intricately linked to glycolytic metabolism, the EMT pathway, and the establishment of an immune-suppressive phenotype in LUAD. These findings underscore the potential of SEC61G as a therapeutic target and predictive marker for immunotherapeutic responses in LUAD patients.
Collapse
Affiliation(s)
- Changshuai Zhou
- Department of NeurosurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Huanhuan Cui
- Department of NeurosurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Yuechao Yang
- Department of NeurosurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Lei Chen
- Department of NeurosurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Mingtao Feng
- Department of NeurosurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Yang Gao
- Department of NeurosurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Deheng Li
- Department of NeurosurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Liangdong Li
- Department of NeurosurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Xin Chen
- Department of NeurosurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Xiaoqiu Li
- Department of PathologyFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Yiqun Cao
- Department of NeurosurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| |
Collapse
|
10
|
Li X, Meng Y, Gu B. A novel immune‑related lncRNA as a prognostic biomarker in HER2 + breast cancer. Oncol Lett 2024; 27:269. [PMID: 38686356 PMCID: PMC11057035 DOI: 10.3892/ol.2024.14402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 03/15/2024] [Indexed: 05/02/2024] Open
Abstract
Human epidermal growth factor receptor 2 (HER2)+ breast cancer is characterized by high malignancy and poor prognosis. Long non-coding (lnc)RNAs are crucial in breast cancer progression and prognosis, especially in tumor-associated immune processes. The present study aimed to elucidate novel lncRNAs related to immune function that could serve as biomarkers for both diagnosis and prognosis of this cancer subtype. Using data from The Cancer Genome Atlas and The Immunology Database and Analysis Portal, correlation analysis was performed to identify differentially expressed lncRNAs and immune-related genes. Through receiver operating characteristic analysis, the diagnostic value of specific lncRNAs was identified and evaluated, with a focus on their capacity to distinguish between cancerous and non-cancerous states. The present research revealed 22 differentially expressed lncRNAs and 23 differentially expressed immune-related genes, with 19 immune-related lncRNAs. A total of 13 of these lncRNAs demonstrated diagnostic relevance. In particular, it was demonstrated that the expression of lncRNA CTC-537E7.2 was significantly correlated with patient survival, suggesting its potential as a prognostic marker. Additionally, the expression of lncRNA CTC-537E7.2 was significantly correlated with clinical parameters, such as hormone receptor status and patient demographics. Moreover, it exhibited associations with four distinct immune cell types and demonstrated involvement in the Janus kinase-signal transducer and activator of transcription pathway. Further assessment by in situ hybridization confirmed the increased expression of lncRNA CTC-537E7.2 in samples from HER2+ patients, reinforcing its significance. In summary, the present study uncovered a novel prognostic biomarker for HER2+ breast cancer, thereby laying the groundwork for investigating the underlying molecular mechanisms driving the development of this subtype of breast cancer.
Collapse
Affiliation(s)
- Xinwei Li
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, P.R. China
- Department of Clinical Laboratory, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, P.R. China
| | - Yue Meng
- Department of Clinical Laboratory, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, P.R. China
| | - Bing Gu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, P.R. China
- Department of Clinical Laboratory, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
11
|
Zheng S, Cheng X, Ke S, Zhang L, Wu H, He D, Cheng X. Bioinformatics analysis and validation of mesenchymal stem cells related gene MT1G in osteosarcoma. Aging (Albany NY) 2024; 16:8155-8170. [PMID: 38747739 PMCID: PMC11131992 DOI: 10.18632/aging.205809] [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: 10/05/2023] [Accepted: 03/18/2024] [Indexed: 05/21/2024]
Abstract
BACKGROUND Osteosarcoma (OS) is a primary malignant bone tumor arising from mesenchymal cells. The standard clinical treatment for OS involves extensive tumor resection combined with neoadjuvant chemotherapy or radiotherapy. OS's invasiveness, lung metastasis, and drug resistance contribute to a low cure rate and poor prognosis with this treatment. Metallothionein 1G (MT1G), observed in various cancers, may serve as a potential therapeutic target for OS. METHODS OS samples in GSE33382 and TARGET datasets were selected as the test cohorts. As the external validation cohort, 13 OS tissues and 13 adjacent cancerous tissues from The Second Affiliated Hospital of Nanchang University were collected. Patients with OS were divided into high and low MT1G mRNA-expression groups; differentially expressed genes (DEGs) were identified as MT1G-related genes. The biological function of MT1G was annotated using Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene Ontology (GO) and gene set enrichment analysis (GSEA). Gene expression correlation analysis and competing endogenous RNA (ceRNA) regulatory network construction were used to determine potential biological regulatory relationships of DEGs. Survival analysis assessed the prognostic value of MT1G. RESULTS MT1G expression increased in OS samples and presented higher in metastatic OS compared with non-metastatic OS. Functional analyses indicated that MT1G was mainly associated with spliceosome. A ceRNA network with DEGs was constructed. MT1G is an effective biomarker predicting survival and correlated with increased recurrence rates and poorer survival. CONCLUSIONS This research identified MT1G as a potential biomarker for OS prognosis, highlighting its potential as a therapy target.
Collapse
Affiliation(s)
- Sikuan Zheng
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
- The Second Clinical College, Medical College of Nanchang University, Nanchang, China
- Institute of Orthopedics of Jiangxi Province, Nanchang, China
| | - Xifu Cheng
- School of Ophthalmology and Optometry, Nanchang University, Nanchang, China
| | - Sulun Ke
- Nanchang University Queen Mary School, Jiangxi Medical College of Nanchang University, Nanchang University, Nanchang, China
| | - Linyi Zhang
- School of Ophthalmology and Optometry, Nanchang University, Nanchang, China
| | - Hui Wu
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
- The Second Clinical College, Medical College of Nanchang University, Nanchang, China
- Institute of Orthopedics of Jiangxi Province, Nanchang, China
| | - Dingwen He
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute of Orthopedics of Jiangxi Province, Nanchang, China
| | - Xigao Cheng
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute of Orthopedics of Jiangxi Province, Nanchang, China
| |
Collapse
|
12
|
Zhang Z, Ye S, Bernhardt SM, Nelson HD, Velie EM, Borges VF, Woodward ER, Evans DGR, Schedin PJ. Postpartum Breast Cancer and Survival in Women With Germline BRCA Pathogenic Variants. JAMA Netw Open 2024; 7:e247421. [PMID: 38639936 PMCID: PMC11031688 DOI: 10.1001/jamanetworkopen.2024.7421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/18/2024] [Indexed: 04/20/2024] Open
Abstract
Importance In young-onset breast cancer (YOBC), a diagnosis within 5 to 10 years of childbirth is associated with increased mortality. Women with germline BRCA1/2 pathogenic variants (PVs) are more likely to be diagnosed with BC at younger ages, but the impact of childbirth on mortality is unknown. Objective To determine whether time between most recent childbirth and BC diagnosis is associated with mortality among patients with YOBC and germline BRCA1/2 PVs. Design, Setting, and Participants This prospective cohort study included women with germline BRCA1/2 PVs diagnosed with stage I to III BC at age 45 years or younger between 1950 and 2021 in the United Kingdom, who were followed up until November 2021. Data were analyzed from December 3, 2021, to November 29, 2023. Exposure Time between most recent childbirth and subsequent BC diagnosis, with recent childbirth defined as 0 to less than 10 years, further delineated to 0 to less than 5 years and 5 to less than 10 years. Main Outcomes and Measures The primary outcome was all-cause mortality, censored at 20 years after YOBC diagnosis. Mortality of nulliparous women was compared with the recent post partum groups and the 10 or more years post partum group. Cox proportional hazards regression analyses were adjusted for age, tumor stage, and further stratified by tumor estrogen receptor (ER) and BRCA gene status. Results Among 903 women with BRCA PVs (mean [SD] age at diagnosis, 34.7 [6.1] years; mean [SD] follow-up, 10.8 [9.8] years), 419 received a BC diagnosis 0 to less than 10 years after childbirth, including 228 women diagnosed less than 5 years after childbirth and 191 women diagnosed 5 to less than 10 years after childbirth. Increased all-cause mortality was observed in women diagnosed within 5 to less than 10 years post partum (hazard ratio [HR], 1.56 [95% CI, 1.05-2.30]) compared with nulliparous women and women diagnosed 10 or more years after childbirth, suggesting a transient duration of postpartum risk. Risk of mortality was greater for women with ER-positive BC in the less than 5 years post partum group (HR, 2.35 [95% CI, 1.02-5.42]) and ER-negative BC in the 5 to less than 10 years post partum group (HR, 3.12 [95% CI, 1.22-7.97]) compared with the nulliparous group. Delineated by BRCA1 or BRCA2, mortality in the 5 to less than 10 years post partum group was significantly increased, but only for BRCA1 carriers (HR, 2.03 [95% CI, 1.15-3.58]). Conclusions and Relevance These findings suggest that YOBC with germline BRCA PVs was associated with increased risk for all-cause mortality if diagnosed within 10 years after last childbirth, with risk highest for ER-positive BC diagnosed less than 5 years post partum, and for ER-negative BC diagnosed 5 to less than 10 years post partum. BRCA1 carriers were at highest risk for poor prognosis when diagnosed at 5 to less than 10 years post partum. No such associations were observed for BRCA2 carriers. These results should inform genetic counseling, prevention, and treatment strategies for BRCA PV carriers.
Collapse
Affiliation(s)
- Zhenzhen Zhang
- Division of Oncological Sciences, Oregon Health & Science University, Portland
- Knight Cancer Institute, Oregon Health & Science University, Portland
| | - Shangyuan Ye
- Biostatistics Shared Resource, Knight Cancer Institute, Oregon Health & Science University, Portland
| | - Sarah M. Bernhardt
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland
| | - Heidi D. Nelson
- Kaiser Permanente Bernard D. Tyson School of Medicine, Pasadena, California
| | - Ellen M. Velie
- Zilber College of Public Health, University of Wisconsin-Milwaukee, Milwaukee
- Departments of Medicine and Pathology, Medical College of Wisconsin, Milwaukee
| | - Virginia F. Borges
- Young Women’s Breast Cancer Translational Program, Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora
| | - Emma R. Woodward
- Manchester Centre for Genomic Medicine, Manchester Academic Health Sciences Centre, Division of Evolution Infection and Genomic Science, St Mary’s Hospital, University of Manchester, Manchester, United Kingdom
- Prevent Breast Cancer Centre, University Hospital of South Manchester NHS Trust, Wythenshawe, Manchester, United Kingdom
- Manchester Centre for Genomic Medicine, St Mary’s Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom
- Manchester Breast Centre, University of Manchester, Manchester, United Kingdom
| | - D. Gareth R. Evans
- Manchester Centre for Genomic Medicine, Manchester Academic Health Sciences Centre, Division of Evolution Infection and Genomic Science, St Mary’s Hospital, University of Manchester, Manchester, United Kingdom
- Prevent Breast Cancer Centre, University Hospital of South Manchester NHS Trust, Wythenshawe, Manchester, United Kingdom
- Manchester Centre for Genomic Medicine, St Mary’s Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom
- Manchester Breast Centre, University of Manchester, Manchester, United Kingdom
| | - Pepper J. Schedin
- Knight Cancer Institute, Oregon Health & Science University, Portland
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland
| |
Collapse
|
13
|
Wen Z, Luo D, Wang S, Rong R, Evers BM, Jia L, Fang Y, Daoud EV, Yang S, Gu Z, Arner EN, Lewis CM, Solis Soto LM, Fujimoto J, Behrens C, Wistuba II, Yang DM, Brekken RA, O'Donnell KA, Xie Y, Xiao G. Deep Learning-Based H-Score Quantification of Immunohistochemistry-Stained Images. Mod Pathol 2024; 37:100398. [PMID: 38043788 PMCID: PMC11141889 DOI: 10.1016/j.modpat.2023.100398] [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: 07/17/2023] [Revised: 11/14/2023] [Accepted: 11/21/2023] [Indexed: 12/05/2023]
Abstract
Immunohistochemistry (IHC) is a well-established and commonly used staining method for clinical diagnosis and biomedical research. In most IHC images, the target protein is conjugated with a specific antibody and stained using diaminobenzidine (DAB), resulting in a brown coloration, whereas hematoxylin serves as a blue counterstain for cell nuclei. The protein expression level is quantified through the H-score, calculated from DAB staining intensity within the target cell region. Traditionally, this process requires evaluation by 2 expert pathologists, which is both time consuming and subjective. To enhance the efficiency and accuracy of this process, we have developed an automatic algorithm for quantifying the H-score of IHC images. To characterize protein expression in specific cell regions, a deep learning model for region recognition was trained based on hematoxylin staining only, achieving pixel accuracy for each class ranging from 0.92 to 0.99. Within the desired area, the algorithm categorizes DAB intensity of each pixel as negative, weak, moderate, or strong staining and calculates the final H-score based on the percentage of each intensity category. Overall, this algorithm takes an IHC image as input and directly outputs the H-score within a few seconds, significantly enhancing the speed of IHC image analysis. This automated tool provides H-score quantification with precision and consistency comparable to experienced pathologists but at a significantly reduced cost during IHC diagnostic workups. It holds significant potential to advance biomedical research reliant on IHC staining for protein expression quantification.
Collapse
Affiliation(s)
- Zhuoyu Wen
- Quantitative Biomedical Research Center, Peter O'Donnell Jr School of Public Health, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Danni Luo
- Quantitative Biomedical Research Center, Peter O'Donnell Jr School of Public Health, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Shidan Wang
- Quantitative Biomedical Research Center, Peter O'Donnell Jr School of Public Health, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ruichen Rong
- Quantitative Biomedical Research Center, Peter O'Donnell Jr School of Public Health, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Bret M Evers
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Liwei Jia
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Yisheng Fang
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Elena V Daoud
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Shengjie Yang
- Quantitative Biomedical Research Center, Peter O'Donnell Jr School of Public Health, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Zifan Gu
- Quantitative Biomedical Research Center, Peter O'Donnell Jr School of Public Health, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Emily N Arner
- Department of Surgery, The University of Texas Southwestern Medical Center, Dallas, Texas; Hamon Center for Therapeutic Oncology Research, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Cheryl M Lewis
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, Texas; Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Luisa M Solis Soto
- Division of Pathology and Laboratory Medicine, Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Junya Fujimoto
- Division of Pathology and Laboratory Medicine, Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Carmen Behrens
- Division of Cancer Medicine, Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ignacio I Wistuba
- Division of Pathology and Laboratory Medicine, Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Donghan M Yang
- Quantitative Biomedical Research Center, Peter O'Donnell Jr School of Public Health, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Rolf A Brekken
- Department of Surgery, The University of Texas Southwestern Medical Center, Dallas, Texas; Hamon Center for Therapeutic Oncology Research, The University of Texas Southwestern Medical Center, Dallas, Texas; Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Kathryn A O'Donnell
- Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas; Hamon Center for Regenerative Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas; Department of Molecular Biology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Yang Xie
- Quantitative Biomedical Research Center, Peter O'Donnell Jr School of Public Health, The University of Texas Southwestern Medical Center, Dallas, Texas; Hamon Center for Regenerative Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas; Department of Bioinformatics, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Guanghua Xiao
- Quantitative Biomedical Research Center, Peter O'Donnell Jr School of Public Health, The University of Texas Southwestern Medical Center, Dallas, Texas; Hamon Center for Regenerative Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas; Department of Bioinformatics, The University of Texas Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
14
|
Zhang X, Qin Y, Chen X, Xiong M, Shu S. Clinical Value of Human Endogenous Retrovirus-H Long Terminal Repeat Associating 2 (HHLA2) in Small Cell Lung Cancer. Technol Cancer Res Treat 2024; 23:15330338241240683. [PMID: 38613340 PMCID: PMC11015749 DOI: 10.1177/15330338241240683] [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: 09/28/2023] [Revised: 01/29/2024] [Accepted: 02/26/2024] [Indexed: 04/14/2024] Open
Abstract
Objective: Human endogenous retrovirus-H long terminal repeat associating 2 (HHLA2) is a new immune checkpoint in the B7 family, and the value of HHLA2 in small cell lung cancer (SCLC) is unknown. Methods: We retrospectively detected HHLA2 expression by immunohistochemistry in SCLC patients. Moreover, plasma biomarkers of SCLC were detected retrospectively. Results: Seventy-four percent of SCLC patients exhibited HHLA2 expression. HHLA2 staining was localised within the nucleus of SCLC cells, while no staining was detected in normal lung tissue specimens. The correlation between HHLA2 expression and clinical factors was also analysed. Limited stage (LS) SCLC was more common than extensive stage (ES) SCLC among patients with HHLA2 staining. SCLC patients without metastasis had higher HHLA2 expression than SCLC patients with metastasis. HHLA2 expression was more frequently detected in the group with a tumour size greater than 5 cm than in the group with a tumour size less than 5 cm. The proportion of patients with HHLA2-positive staining was greater in the stage III and IV SCLC groups than in the stage I and II SCLC groups. A high proportion of SCLC patients with HHLA2-positive staining had a survival time <2 years. Neuron-specific enolase (NSE), CEA and Ki-67 levels were measured. The NSE level in the HHLA2-positive group was significantly greater than that in the HHLA2-negative group. The CEA and Ki-67 levels did not significantly differ between the HHLA2-positive and HHLA2-negative patients, nor were age, sex, smoking status, nodal metastasis status, Karnofsky Performance Scale (KPS) score, or Ki-67 expression score. HHLA2-positive SCLC patients had higher tumour stages and shorter 2-year survival times than HHLA2-negative patients did. Conclusion: The new immune molecule HHLA2 may be an ideal clinical biomarker for predicting SCLC progression and could serve as a new immunotherapy target in SCLC.
Collapse
Affiliation(s)
- Xiuqin Zhang
- Respiratory and Critical Care Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yan Qin
- Department of Pathology Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Xu Chen
- Department of Basic Medicine, Jiangnan University, Wuxi, China
| | - Mengrui Xiong
- Department of Basic Medicine, Jiangnan University, Wuxi, China
| | - Song Shu
- Department of Pathology Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| |
Collapse
|
15
|
Behrens A, Wurmthaler L, Heindl F, Gass P, Häberle L, Volz B, Hack CC, Emons J, Erber R, Hartmann A, Beckmann MW, Ruebner M, Dougall WC, Press MF, Fasching PA, Huebner H. RANK and RANKL Expression in Tumors of Patients with Early Breast Cancer. Geburtshilfe Frauenheilkd 2024; 84:77-85. [PMID: 38178900 PMCID: PMC10764119 DOI: 10.1055/a-2192-2998] [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: 08/25/2023] [Accepted: 10/15/2023] [Indexed: 01/06/2024] Open
Abstract
Introduction The receptor activator of nuclear factor-κB (RANK) pathway was associated with the pathogenesis of breast cancer. Several studies attempted to link the RANK/RANKL pathway to prognosis; however, with inconsistent outcomes. We aimed to further contribute to the knowledge about RANK/RANKL as prognostic factors in breast cancer. Within this study, protein expression of RANK and its ligand, RANKL, in the tumor tissue was analyzed in association with disease-free survival (DFS) and overall survival (OS) in a study cohort of patients with early breast cancer. Patients and Methods 607 samples of female primary and early breast cancer patients from the Bavarian Breast Cancer Cases and Controls Study were analyzed to correlate the RANK and RANKL expression with DFS and OS. Therefore, expression was quantified using immunohistochemical staining of a tissue microarray. H-scores were determined with the cut-off value of 8.5 for RANK and 0 for RANKL expression, respectively. Results RANK and RANKL immunohistochemistry were assessed by H-score. Both biomarkers did not correlate (ρ = -0.04). According to molecular subtypes, triple-negative tumors and HER2-positive tumors showed a higher number of RANK-positive tumors (H-score ≥ 8.5), however, no subtype-specific expression of RANKL could be detected. Higher RANKL expression tended to correlate with a better prognosis. However, RANK and RANKL expression could not be identified as statistically significant prognostic factors within the study cohort. Conclusions Tumor-specific RANK and RANKL expressions are not applicable as prognostic factors for DFS and OS, but might be associated with subtype-specific breast cancer progression.
Collapse
Affiliation(s)
- Annika Behrens
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Bavarian Center for Cancer Research (BZKF), Erlangen, Germany
| | - Lena Wurmthaler
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Bavarian Center for Cancer Research (BZKF), Erlangen, Germany
| | - Felix Heindl
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Bavarian Center for Cancer Research (BZKF), Erlangen, Germany
| | - Paul Gass
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Bavarian Center for Cancer Research (BZKF), Erlangen, Germany
| | - Lothar Häberle
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Bavarian Center for Cancer Research (BZKF), Erlangen, Germany
- Biostatistics Unit, Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Bernhard Volz
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Ansbach University of Applied Sciences, Ansbach, Germany
| | - Carolin C. Hack
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Bavarian Center for Cancer Research (BZKF), Erlangen, Germany
| | - Julius Emons
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Bavarian Center for Cancer Research (BZKF), Erlangen, Germany
| | - Ramona Erber
- Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Bavarian Center for Cancer Research (BZKF), Erlangen, Germany
- Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Arndt Hartmann
- Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Bavarian Center for Cancer Research (BZKF), Erlangen, Germany
- Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Matthias W. Beckmann
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Bavarian Center for Cancer Research (BZKF), Erlangen, Germany
| | - Matthias Ruebner
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Bavarian Center for Cancer Research (BZKF), Erlangen, Germany
| | - William C. Dougall
- Hematology and Oncology Research, Amgen, Inc., Seattle, WA, USA
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Michael F. Press
- Department of Pathology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Peter A. Fasching
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Bavarian Center for Cancer Research (BZKF), Erlangen, Germany
| | - Hanna Huebner
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Bavarian Center for Cancer Research (BZKF), Erlangen, Germany
| |
Collapse
|
16
|
Yang Z, Wei X, Ji C, Ren X, Su W, Wang Y, Zhou J, Zhao Z, Zhou P, Zhao K, Yao B, Song N, Qin C. OGT/HIF-2α axis promotes the progression of clear cell renal cell carcinoma and regulates its sensitivity to ferroptosis. iScience 2023; 26:108148. [PMID: 37915611 PMCID: PMC10616330 DOI: 10.1016/j.isci.2023.108148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 07/24/2023] [Accepted: 10/02/2023] [Indexed: 11/03/2023] Open
Abstract
O-GlcNAc transferase (OGT) acts in the development of various cancers, but its role in clear cell renal cell carcinoma (ccRCC) remains unclear. In this study, we found that OGT was upregulated in ccRCC and this upregulation was associated with a worse survival. Moreover, OGT promoted the proliferation, clone formation, and invasion of VHL-mutated ccRCC cells. Mechanistically, OGT increased the protein level of hypoxia-inducible factor-2α (HIF-2α) (the main driver of the clear cell phenotype) by repressing ubiquitin‒proteasome system-mediated degradation. Interestingly, the OGT/HIF-2α axis conferred ccRCC a high sensitivity to ferroptosis. In conclusion, OGT promotes the progression of VHL-mutated ccRCC by inhibiting the degradation of HIF-2α, and agents that can modulate the OGT/HIF-2α axis may exert therapeutic effects on mutated VHL ccRCC.
Collapse
Affiliation(s)
- Zhou Yang
- The State Key Lab of Reproductive, Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiyi Wei
- The State Key Lab of Reproductive, Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Chengjian Ji
- The State Key Lab of Reproductive, Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xiaohan Ren
- The State Key Lab of Reproductive, Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Wei Su
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yichun Wang
- The State Key Lab of Reproductive, Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jingwan Zhou
- National Experimental Teaching Center of Basic Medical Science, Nanjing Medical University, Nanjing, China
| | - Zheng Zhao
- National Experimental Teaching Center of Basic Medical Science, Nanjing Medical University, Nanjing, China
| | - Pengcheng Zhou
- National Experimental Teaching Center of Basic Medical Science, Nanjing Medical University, Nanjing, China
| | - Kejie Zhao
- National Experimental Teaching Center of Basic Medical Science, Nanjing Medical University, Nanjing, China
| | - Bing Yao
- National Experimental Teaching Center of Basic Medical Science, Nanjing Medical University, Nanjing, China
- Department of Medical Genetics, Nanjing Medical University, Nanjing, China
| | - Ninghong Song
- The State Key Lab of Reproductive, Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Chao Qin
- The State Key Lab of Reproductive, Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| |
Collapse
|
17
|
Loibl S, Azim HA, Bachelot T, Berveiller P, Bosch A, Cardonick E, Denkert C, Halaska MJ, Hoeltzenbein M, Johansson ALV, Maggen C, Markert UR, Peccatori F, Poortmans P, Saloustros E, Saura C, Schmid P, Stamatakis E, van den Heuvel-Eibrink M, van Gerwen M, Vandecaveye V, Pentheroudakis G, Curigliano G, Amant F. ESMO Expert Consensus Statements on the management of breast cancer during pregnancy (PrBC). Ann Oncol 2023; 34:849-866. [PMID: 37572987 DOI: 10.1016/j.annonc.2023.08.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/14/2023] Open
Abstract
The management of breast cancer during pregnancy (PrBC) is a relatively rare indication and an area where no or little evidence is available since randomized controlled trials cannot be conducted. In general, advances related to breast cancer (BC) treatment outside pregnancy cannot always be translated to PrBC, because both the interests of the mother and of the unborn should be considered. Evidence remains limited and/or conflicting in some specific areas where the optimal approach remains controversial. In 2022, the European Society for Medical Oncology (ESMO) held a virtual consensus-building process on this topic to gain insights from a multidisciplinary group of experts and develop statements on controversial topics that cannot be adequately addressed in the current evidence-based ESMO Clinical Practice Guideline. The aim of this consensus-building process was to discuss controversial issues relating to the management of patients with PrBC. The virtual meeting included a multidisciplinary panel of 24 leading experts from 13 countries and was chaired by S. Loibl and F. Amant. All experts were allocated to one of four different working groups. Each working group covered a specific subject area with two chairs appointed: Planning, preparation and execution of the consensus process was conducted according to the ESMO standard operating procedures.
Collapse
Affiliation(s)
- S Loibl
- GBG c/o GBG Forschungs GmbH, Neu-Isenburg; Centre for Haematology and Oncology Bethanien, Frankfurt am Main, Frankfurt; Goethe University Frankfurt, Frankfurt am Main, Frankfurt, Germany.
| | - H A Azim
- Breast Cancer Center, School of Medicine, Tecnologico de Monterrey, San Pedro Garza Garcia, Nuevo Leon, Mexico
| | - T Bachelot
- Department of medical oncology, Centre Léon Bérard, Lyon, France
| | - P Berveiller
- Department of Gynecology and Obstetrics, Poissy-Saint Germain Hospital, Poissy; UMR 1198 - BREED, INRAE, Paris Saclay University, RHuMA, Montigny-Le-Bretonneux, France
| | - A Bosch
- Division of Oncology, Department of Clinical Sciences, Lund University, Lund; Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - E Cardonick
- Cooper Medical School at Rowan University, Camden, USA
| | - C Denkert
- Philipps-University Marburg and Marburg University Hospital (UKGM), Marburg, Germany
| | - M J Halaska
- Department of Obstetrics and Gynaecology, Third Faculty of Medicine, Charles University in Prague and Universital Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - M Hoeltzenbein
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Clinical Pharmacology and Toxicology, Embryotox Center of Clinical Teratology and Drug Safety in Pregnancy, Berlin, Germany
| | - A L V Johansson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden; Cancer Registry of Norway, Oslo, Norway
| | - C Maggen
- Department of Obstetrics and Prenatal Medicine, University Hospital Brussels, Brussels, Belgium
| | - U R Markert
- Placenta Lab, Department of Obstetrics, Jena University Hospital, Jena, Germany
| | - F Peccatori
- Gynecologic Oncology Department, European Institute of Oncology IRCCS, Milan, Italy
| | - P Poortmans
- Iridium Netwerk, Antwerp; University of Antwerp, Antwerp, Belgium
| | - E Saloustros
- Department of Oncology, University General Hospital of Larissa, Larissa, Greece
| | - C Saura
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - P Schmid
- Cancer Institute, Queen Mary University London, London, UK
| | - E Stamatakis
- Department of Anesthesiology, 'Alexandra' General Hospital, Athens, Greece
| | | | - M van Gerwen
- Gynecologic Oncology, Antoni van Leeuwenhoek-Netherlands Cancer Institute, Amsterdam; Department of Child and Adolescent Psychiatry and Psychosocial Care, Amsterdam UMC, University of Amsterdam; Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - V Vandecaveye
- Department of Radiology, University Hospitals Leuven, Leuven, Belgium
| | - G Pentheroudakis
- European Society for Medical Oncology (ESMO), Lugano, Switzerland
| | - G Curigliano
- Division of Early Drug Development, European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - F Amant
- Gynecologic Oncology, Antoni van Leeuwenhoek-Netherlands Cancer Institute, Amsterdam; Division Gynaecologic Oncology, UZ Leuven, Belgium
| |
Collapse
|
18
|
Fu C, Liu Z, An T, Li H, Hu X, Li X, Liu X, Wu D, Zhang R, Li K, Qiu Y, Wang H. Poliovirus receptor (PVR) mediates carboplatin-induced PD-L1 expression in non-small-cell lung cancer cells. Biochim Biophys Acta Gen Subj 2023; 1867:130439. [PMID: 37516256 DOI: 10.1016/j.bbagen.2023.130439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 07/04/2023] [Accepted: 07/26/2023] [Indexed: 07/31/2023]
Abstract
Programmed death-ligand-1 (PD-L1) is an immune suppressor that inhibits T cell based immunity. Anti-PD-L1/PD-1 immunotherapy benefits those patients receiving platinum-based combinational chemotherapy. However, the underlying mechanism is still largely unknown. In this study, we found that carboplatin could induce PD-L1 expression in NSCLC H292, A549 and H1299 cells in a dose-dependent manner. mRNA sequencing and the subsequent validation assays found that carboplatin significantly induced PVR expression, which is considered as an immuno-adhesion molecule. Mechanistically, PVR knockdown significantly abrogated carboplatin-induced PD-L1 expression. Functionally, knockdown of PVR significantly reversed the CD3+ T cells proliferation inhibition caused by carboplatin increased PD-L1. Moreover, the carboplatin-induced PVR and subsequent up-regulation of PD-L1 might be mediated via the EGFR, PI3K/AKT, and ERK signaling pathways. Immunohistochemical staining results showed that the PD-L1 expression was positively associated with PVR expression in clinical NSCLC samples. Our study reveals a novel regulatory mechanism of PD-L1 expression, provides evidence that carboplatin inhibits tumor immune response by up-regulating PD-L1 expression and explains the rationale for combining platinum-based chemotherapy with PD-L1/PD-1 inhibitors.
Collapse
Affiliation(s)
- Chen Fu
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, China
| | - Zongcai Liu
- The Laboratory of Endocrinology and Metabolism, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Taixue An
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Haixia Li
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xiumei Hu
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xin Li
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xinyao Liu
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Danjuan Wu
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ruyi Zhang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Kui Li
- Guangzhou Huayinkang Medical Laboratory Center Co., Ltd., Guangzhou 510515, China.
| | - Yurong Qiu
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Guangzhou Huayinkang Medical Laboratory Center Co., Ltd., Guangzhou 510515, China.
| | - Haifang Wang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
19
|
De Leon-Oliva D, Barrena-Blázquez S, Jiménez-Álvarez L, Fraile-Martinez O, García-Montero C, López-González L, Torres-Carranza D, García-Puente LM, Carranza ST, Álvarez-Mon MÁ, Álvarez-Mon M, Diaz R, Ortega MA. The RANK-RANKL-OPG System: A Multifaceted Regulator of Homeostasis, Immunity, and Cancer. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1752. [PMID: 37893470 PMCID: PMC10608105 DOI: 10.3390/medicina59101752] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/21/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023]
Abstract
The RANK-RANKL-OPG system is a complex signaling pathway that plays a critical role in bone metabolism, mammary epithelial cell development, immune function, and cancer. RANKL is a ligand that binds to RANK, a receptor expressed on osteoclasts, dendritic cells, T cells, and other cells. RANKL signaling promotes osteoclast differentiation and activation, which leads to bone resorption. OPG is a decoy receptor that binds to RANKL and inhibits its signaling. In cancer cells, RANKL expression is often increased, which can lead to increased bone resorption and the development of bone metastases. RANKL-neutralizing antibodies, such as denosumab, have been shown to be effective in the treatment of skeletal-related events, including osteoporosis or bone metastases, and cancer. This review will provide a comprehensive overview of the functions of the RANK-RANKL-OPG system in bone metabolism, mammary epithelial cells, immune function, and cancer, together with the potential therapeutic implications of the RANK-RANKL pathway for cancer management.
Collapse
Affiliation(s)
- Diego De Leon-Oliva
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (D.D.L.-O.); (S.B.-B.); (L.J.-Á.); (O.F.-M.); (C.G.-M.); (D.T.-C.); (L.M.G.-P.); (S.T.C.); (M.Á.Á.-M.); (M.Á.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Silvestra Barrena-Blázquez
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (D.D.L.-O.); (S.B.-B.); (L.J.-Á.); (O.F.-M.); (C.G.-M.); (D.T.-C.); (L.M.G.-P.); (S.T.C.); (M.Á.Á.-M.); (M.Á.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Department of Nursing and Physiotherapy, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
| | - Laura Jiménez-Álvarez
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (D.D.L.-O.); (S.B.-B.); (L.J.-Á.); (O.F.-M.); (C.G.-M.); (D.T.-C.); (L.M.G.-P.); (S.T.C.); (M.Á.Á.-M.); (M.Á.-M.)
- Surgery Service, University Hospital Principe de Asturias, 28801 Alcala de Henares, Spain
| | - Oscar Fraile-Martinez
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (D.D.L.-O.); (S.B.-B.); (L.J.-Á.); (O.F.-M.); (C.G.-M.); (D.T.-C.); (L.M.G.-P.); (S.T.C.); (M.Á.Á.-M.); (M.Á.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Cielo García-Montero
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (D.D.L.-O.); (S.B.-B.); (L.J.-Á.); (O.F.-M.); (C.G.-M.); (D.T.-C.); (L.M.G.-P.); (S.T.C.); (M.Á.Á.-M.); (M.Á.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Laura López-González
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain;
| | - Diego Torres-Carranza
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (D.D.L.-O.); (S.B.-B.); (L.J.-Á.); (O.F.-M.); (C.G.-M.); (D.T.-C.); (L.M.G.-P.); (S.T.C.); (M.Á.Á.-M.); (M.Á.-M.)
| | - Luis M. García-Puente
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (D.D.L.-O.); (S.B.-B.); (L.J.-Á.); (O.F.-M.); (C.G.-M.); (D.T.-C.); (L.M.G.-P.); (S.T.C.); (M.Á.Á.-M.); (M.Á.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Sara T. Carranza
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (D.D.L.-O.); (S.B.-B.); (L.J.-Á.); (O.F.-M.); (C.G.-M.); (D.T.-C.); (L.M.G.-P.); (S.T.C.); (M.Á.Á.-M.); (M.Á.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Miguel Ángel Álvarez-Mon
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (D.D.L.-O.); (S.B.-B.); (L.J.-Á.); (O.F.-M.); (C.G.-M.); (D.T.-C.); (L.M.G.-P.); (S.T.C.); (M.Á.Á.-M.); (M.Á.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Melchor Álvarez-Mon
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (D.D.L.-O.); (S.B.-B.); (L.J.-Á.); (O.F.-M.); (C.G.-M.); (D.T.-C.); (L.M.G.-P.); (S.T.C.); (M.Á.Á.-M.); (M.Á.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain;
- Immune System Diseases-Rheumatology Service, University Hospital Principe de Asturias, 28801 Alcala de Henares, Spain
| | - Raul Diaz
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Surgery Service, University Hospital Principe de Asturias, 28801 Alcala de Henares, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain;
| | - Miguel A. Ortega
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (D.D.L.-O.); (S.B.-B.); (L.J.-Á.); (O.F.-M.); (C.G.-M.); (D.T.-C.); (L.M.G.-P.); (S.T.C.); (M.Á.Á.-M.); (M.Á.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| |
Collapse
|
20
|
Jin P, Zhou Y, Liu Z, Chen X, Qi H. Downregulation of CLDN1 impairs trophoblast invasion and endovascular trophoblast differentiation in early-onset preeclampsia. Placenta 2023; 140:20-29. [PMID: 37523840 DOI: 10.1016/j.placenta.2023.07.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 07/07/2023] [Accepted: 07/13/2023] [Indexed: 08/02/2023]
Abstract
INTRODUCTION To investigate the role of claudin-1 (CLDN1) in trophoblast invasion and endovascular trophoblast (enEVT) differentiation in early-onset preeclampsia (EOPE). METHODS The expression and localization of CLDN1 in normal (n = 18) and EOPE (n = 20) placental tissues were detected by immunohistochemical (IHC) staining, quantitative real-time polymerase chain reaction (qRT‒PCR) and Western blotting. Next, invasion, migration and tube formation assays were performed to explore the involvement of CLDN1 in trophoblast invasion and enEVT differentiation in trophoblast cell lines (HTR8/SVneo). Then, invasion and enEVT markers were analyzed via Western blotting and qRT‒PCR, respectively. Finally, we established an EOPE mouse model to detect the Cldn1 protein level. RESULTS CLDN1 expression was significantly decreased in EOPE placental tissues. Knockdown of CLDN1 suppressed HTR8/SVneo cell invasion, migration and the ability to penetrate the endothelial tube. Conversely, overexpression of CLDN1 promoted trophoblast invasion and the ability to invade the endothelial tube. Inhibition of CLDN1 decreased the protein expression of VIM and SNAIL along with downregulating IL1B and PECAM1 mRNA levels, while overexpression of CLDN1 gave the opposite results. In the EOPE mouse model, we found a decrease in Cldn1 expression in EOPE mouse placentas. DISCUSSION These results suggest that the downregulation of CLDN1 in trophoblast cells is involved in the pathogenesis of early-onset preeclampsia by affecting trophoblast invasion and enEVT differentiation.
Collapse
Affiliation(s)
- Pingsong Jin
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Yijie Zhou
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Zheng Liu
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Xuehai Chen
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, 400016, China.
| | - Hongbo Qi
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University (CQMU-WCH), Chongqing, 401147, China; Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, 400016, China; Joint International Research Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, 400016, China.
| |
Collapse
|
21
|
Zhu Y, Wang Y, Li Y, Li Z, Kong W, Zhao X, Chen S, Yan L, Wang L, Tong Y, Shao H. Carnitine palmitoyltransferase 1A promotes mitochondrial fission by enhancing MFF succinylation in ovarian cancer. Commun Biol 2023; 6:618. [PMID: 37291333 PMCID: PMC10250469 DOI: 10.1038/s42003-023-04993-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 05/30/2023] [Indexed: 06/10/2023] Open
Abstract
Mitochondria are dynamic organelles that are important for cell growth and proliferation. Dysregulated mitochondrial dynamics are highly associated with the initiation and progression of various cancers, including ovarian cancer. However, the regulatory mechanism underlying mitochondrial dynamics is still not fully understood. Previously, our study showed that carnitine palmitoyltransferase 1A (CPT1A) is highly expressed in ovarian cancer cells and promotes the development of ovarian cancer. Here, we find that CPT1A regulates mitochondrial dynamics and promotes mitochondrial fission in ovarian cancer cells. Our study futher shows that CPT1A regulates mitochondrial fission and function through mitochondrial fission factor (MFF) to promote the growth and proliferation of ovarian cancer cells. Mechanistically, we show that CPT1A promotes succinylation of MFF at lysine 302 (K302), which protects against Parkin-mediated ubiquitin-proteasomal degradation of MFF. Finally, the study shows that MFF is highly expressed in ovarian cancer cells and that high MFF expression is associated with poor prognosis in patients with ovarian cancer. MFF inhibition significantly inhibits the progression of ovarian cancer in vivo. Overall, CPT1A regulates mitochondrial dynamics through MFF succinylation to promote the development of ovarian cancer. Moreover, our findings suggest that MFF is a potential therapeutic target for ovarian cancer.
Collapse
Affiliation(s)
- Yaqin Zhu
- National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, 710119, Xi'an, Shaanxi, China
| | - Yue Wang
- National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, 710119, Xi'an, Shaanxi, China
| | - Ying Li
- National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, 710119, Xi'an, Shaanxi, China
| | - Zhongqi Li
- National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, 710119, Xi'an, Shaanxi, China
| | - Wenhui Kong
- National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, 710119, Xi'an, Shaanxi, China
| | - Xiaoxuan Zhao
- National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, 710119, Xi'an, Shaanxi, China
| | - Shuting Chen
- National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, 710119, Xi'an, Shaanxi, China
| | - Liting Yan
- National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, 710119, Xi'an, Shaanxi, China
| | - Lenan Wang
- National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, 710119, Xi'an, Shaanxi, China
| | - Yunli Tong
- National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, 710119, Xi'an, Shaanxi, China
| | - Huanjie Shao
- National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, 710119, Xi'an, Shaanxi, China.
| |
Collapse
|
22
|
Wang S, Wang Y, Wang Y, Li Q, Zeng K, Li X, Feng X. Myc derived circRNA promotes triple-negative breast cancer progression via reprogramming fatty acid metabolism. Discov Oncol 2023; 14:67. [PMID: 37173608 PMCID: PMC10182216 DOI: 10.1007/s12672-023-00679-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023] Open
Abstract
Myc is a well-known proto-oncogene that is frequently amplified and activated in breast cancer, especially in triple-negative breast cancer (TNBC). However, the role of circular RNA (circRNA) generated by Myc remains unclear. Herein, we found that circMyc (hsa_circ_0085533) was remarkably upregulated in TNBC tissues and cell lines, which was attributed to gene amplification. Genetic knockdown of circMyc mediated by lentiviral vector significantly inhibited TNBC cell proliferation and invasion. Importantly, circMyc increased cellular triglycerides, cholesterols and lipid droplet contents. CircMyc was detected in both cytoplasm and nucleus, cytoplasmic circMyc could directly bind to HuR protein, facilitating the binding of HuR to SREBP1 mRNA, resulting in increasing SREBP1 mRNA stability. Nuclear circMyc bound to Myc protein, facilitating the occupation of Myc on SREBP1 promoter, leading to increasing SREBP1 transcription. As a result, the elevated SREBP1 increased the expression of its downstream lipogenic enzymes, enhancing lipogenesis and TNBC progression. Moreover, the orthotopic xenograft model showed that depletion of circMyc markedly inhibited lipogenesis and reduced tumor size. Clinically, high circMyc was closely related to larger tumor volume, later clinical stage and lymph node metastasis, functioning as an adverse prognostic factor. Collectively, our findings characterize a novel Myc-derived circRNA controlling TNBC tumorigenesis via regulation of metabolic reprogramming, implying a promising therapeutic target.
Collapse
Affiliation(s)
- Shengting Wang
- Department of Clinical Medicine, Xi'an Peihua University, 888 Changning Street, Xi'an, Shaanxi, 710125, China.
| | - Yufang Wang
- Department of Clinical Medicine, Xi'an Peihua University, 888 Changning Street, Xi'an, Shaanxi, 710125, China
| | - Yue Wang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Qian Li
- Department of Clinical Medicine, Xi'an Peihua University, 888 Changning Street, Xi'an, Shaanxi, 710125, China
| | - Kaixuan Zeng
- School of Medicine, Xi'an Jiaotong University, Xi'an, 710000, China
| | - Xiaoming Li
- Department of Clinical Medicine, Xi'an Peihua University, 888 Changning Street, Xi'an, Shaanxi, 710125, China
| | - Xinghua Feng
- Department of Clinical Medicine, Xi'an Peihua University, 888 Changning Street, Xi'an, Shaanxi, 710125, China
| |
Collapse
|
23
|
Ciscar M, Trinidad EM, Perez‐Chacon G, Alsaleem M, Jimenez M, Jimenez‐Santos MJ, Perez‐Montoyo H, Sanz‐Moreno A, Vethencourt A, Toss M, Petit A, Soler‐Monso MT, Lopez V, Gomez‐Miragaya J, Gomez‐Aleza C, Dobrolecki LE, Lewis MT, Bruna A, Mouron S, Quintela‐Fandino M, Al‐Shahrour F, Martinez‐Aranda A, Sierra A, Green AR, Rakha E, Gonzalez‐Suarez E. RANK is a poor prognosis marker and a therapeutic target in ER-negative postmenopausal breast cancer. EMBO Mol Med 2023; 15:e16715. [PMID: 36880458 PMCID: PMC10086586 DOI: 10.15252/emmm.202216715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 01/24/2023] [Accepted: 02/08/2023] [Indexed: 03/08/2023] Open
Abstract
Despite strong preclinical data, the therapeutic benefit of the RANKL inhibitor, denosumab, in breast cancer patients, beyond the bone, is unclear. Aiming to select patients who may benefit from denosumab, we hereby analyzed RANK and RANKL protein expression in more than 2,000 breast tumors (777 estrogen receptor-negative, ER- ) from four independent cohorts. RANK protein expression was more frequent in ER- tumors, where it associated with poor outcome and poor response to chemotherapy. In ER- breast cancer patient-derived orthoxenografts (PDXs), RANKL inhibition reduced tumor cell proliferation and stemness, regulated tumor immunity and metabolism, and improved response to chemotherapy. Intriguingly, tumor RANK protein expression associated with poor prognosis in postmenopausal breast cancer patients, activation of NFKB signaling, and modulation of immune and metabolic pathways, suggesting that RANK signaling increases after menopause. Our results demonstrate that RANK protein expression is an independent biomarker of poor prognosis in postmenopausal and ER- breast cancer patients and support the therapeutic benefit of RANK pathway inhibitors, such as denosumab, in breast cancer patients with RANK+ ER- tumors after menopause.
Collapse
Affiliation(s)
- Marina Ciscar
- Molecular Oncology, Spanish National Cancer Research Centre (CNIO)MadridSpain
- Oncobell, Bellvitge Biomedical Research Institute (IDIBELL)BarcelonaSpain
| | - Eva M Trinidad
- Oncobell, Bellvitge Biomedical Research Institute (IDIBELL)BarcelonaSpain
| | - Gema Perez‐Chacon
- Molecular Oncology, Spanish National Cancer Research Centre (CNIO)MadridSpain
| | - Mansour Alsaleem
- Nottingham Breast Cancer Research Centre, Academic Unit for Translational Medical Sciences, School of MedicineUniversity of Nottingham Biodiscovery Institute, University ParkNottinghamUK
- Present address:
Department of Applied Medical Science, Applied CollegeQassim UniversityUnayzahSaudi Arabia
| | - Maria Jimenez
- Molecular Oncology, Spanish National Cancer Research Centre (CNIO)MadridSpain
| | - Maria J Jimenez‐Santos
- Bioinformatics Unit, Structural Biology, Spanish National Cancer Research Centre (CNIO)MadridSpain
| | | | - Adrian Sanz‐Moreno
- Oncobell, Bellvitge Biomedical Research Institute (IDIBELL)BarcelonaSpain
| | - Andrea Vethencourt
- Oncobell, Bellvitge Biomedical Research Institute (IDIBELL)BarcelonaSpain
- Medical Oncology, Breast Unit, Catalan Institute of Oncology (ICO)University Hospital of BellvitgeBarcelonaSpain
| | - Michael Toss
- Nottingham Breast Cancer Research Centre, Academic Unit for Translational Medical Sciences, School of MedicineUniversity of Nottingham Biodiscovery Institute, University ParkNottinghamUK
| | - Anna Petit
- Pathology DepartmentUniversity Hospital of Bellvitge, IDIBELLBarcelonaSpain
| | | | - Victor Lopez
- Molecular Oncology, Spanish National Cancer Research Centre (CNIO)MadridSpain
| | | | - Clara Gomez‐Aleza
- Oncobell, Bellvitge Biomedical Research Institute (IDIBELL)BarcelonaSpain
| | - Lacey E Dobrolecki
- Molecular and Cellular Biology and RadiologyThe Lester and Sue Smith Breast Center, Baylor College of MedicineHoustonTexasUSA
| | - Michael T Lewis
- Molecular and Cellular Biology and RadiologyThe Lester and Sue Smith Breast Center, Baylor College of MedicineHoustonTexasUSA
| | - Alejandra Bruna
- Cancer Research UK Cambridge CentreCambridgeUK
- Present address:
Molecular Pathology DivisionCentre for Paediatric Oncology Experimental MedicineCentre for Cancer EvolutionThe Institute of Cancer ResearchLondonUK
| | - Silvana Mouron
- Breast Cancer Clinical Research Unit, Clinical Research ProgramSpanish National Cancer Research Centre (CNIO)MadridSpain
| | - Miguel Quintela‐Fandino
- Breast Cancer Clinical Research Unit, Clinical Research ProgramSpanish National Cancer Research Centre (CNIO)MadridSpain
| | - Fatima Al‐Shahrour
- Bioinformatics Unit, Structural Biology, Spanish National Cancer Research Centre (CNIO)MadridSpain
| | - Antonio Martinez‐Aranda
- Oncobell, Bellvitge Biomedical Research Institute (IDIBELL)BarcelonaSpain
- Medical Oncology, Breast Unit, Catalan Institute of Oncology (ICO)University Hospital of BellvitgeBarcelonaSpain
| | - Angels Sierra
- Oncobell, Bellvitge Biomedical Research Institute (IDIBELL)BarcelonaSpain
- Present address:
Laboratory of Experimental Oncological Neurosurgery, Neurosurgery ServiceHospital Clinic de Barcelona‐FCRBBarcelonaSpain
| | - Andrew R Green
- Nottingham Breast Cancer Research Centre, Academic Unit for Translational Medical Sciences, School of MedicineUniversity of Nottingham Biodiscovery Institute, University ParkNottinghamUK
| | - Emad Rakha
- Nottingham Breast Cancer Research Centre, Academic Unit for Translational Medical Sciences, School of MedicineUniversity of Nottingham Biodiscovery Institute, University ParkNottinghamUK
| | - Eva Gonzalez‐Suarez
- Molecular Oncology, Spanish National Cancer Research Centre (CNIO)MadridSpain
- Oncobell, Bellvitge Biomedical Research Institute (IDIBELL)BarcelonaSpain
| |
Collapse
|
24
|
Liu W, Pan W, Zou M, Jin S, Mi R, Cheng G, Piao H. Tacrolimus and paclitaxel co-loaded O/O ointment without surfactant: Synergistic combinations for the treatment of psoriasis. Eur J Pharm Biopharm 2023; 185:28-43. [PMID: 36828239 DOI: 10.1016/j.ejpb.2023.02.007] [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: 11/01/2022] [Revised: 02/07/2023] [Accepted: 02/17/2023] [Indexed: 02/24/2023]
Abstract
Psoriasis is an autoimmune disorder disease with abnormally activated T lymphocytes and thickening of the epidermis. The mechanism of the action of tacrolimus and paclitaxel are matched with the two only known pathogenesis of psoriasis. However, there has been no report on tacrolimus combined with paclitaxel in the treatment of psoriasis until now. The O/O ointment was prepared for the topical application to overcome poor solubility, poor skin penetration, and erratic absorption of the two drugs. A high-speed shearing method was adopted to prepare the ointment, in which propylene carbonate was used to solve tacrolimus and paclitaxel completely. The ointment showed excellent stability, slow release of the drugs, better retention in psoriatic skin, and good skin tolerance. The therapeutic efficacy of ointment was evaluated with imiquimod induced psoriatic model, and the level of expression of psoriatic biochemical markers was evaluated using the PASI score and immunohistochemistry. The cumulative PASI score was 10.8 for the imiquimod induced group, 7.8 for the tacrolimus ointment group, 8.3 for the paclitaxel ointment and 5.3 for the tacrolimus-paclitaxel (1:1) ointment group, respectively. Ointment group with tacrolimus and paclitaxel indicated a significant improvement in the phenotypic features of the psoriatic skin treated. Compared with the imiquimod group, tacrolimus-paclitaxel (1:1) ointment group was significantly reduced the level of IL-17. The results confirm that tacrolimus and paclitaxel co-loaded ointment can be an effective strategy for the treatment of psoriasis.
Collapse
Affiliation(s)
- Wenxue Liu
- Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, China; CSPC Zhongqi Pharmaceutical Technology (Shijiazhuang) Co., LTD, Hebei Province 050035, China
| | - Wenxiu Pan
- Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, China
| | - Meijuan Zou
- Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, China
| | - Sichen Jin
- Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, China
| | - Ru Mi
- Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, China
| | - Gang Cheng
- Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, China
| | - Hongyu Piao
- Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, China.
| |
Collapse
|
25
|
Meier ME, Hagelstein-Rotman M, Majoor BCJ, Geels RES, Appelman-Dijkstra NM, Bravenboer N. Expression of RANKL in breast cancer tissue in patients with fibrous dysplasia/McCune-Albright syndrome. Bone 2023; 169:116679. [PMID: 36652988 DOI: 10.1016/j.bone.2023.116679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/22/2022] [Accepted: 01/12/2023] [Indexed: 01/18/2023]
Abstract
BACKGROUND In fibrous dysplasia/McCune-Albright syndrome (FD/MAS), mosaic mutations in the GNAS gene lead to locally abnormal bone turnover. Additionally, patients with FD/MAS, particularly with thoracic lesions, have an increased risk for breast cancer. Development and progression of breast cancer has been associated with expression of Receptor Activator of NF-κB ligand (RANKL) in mammary tissue, and due to the GNAS mutation, RANKL is systemically increased in patients with FD/MAS. Yet it is unknown whether breast cancer in FD/MAS is also dependent on RANKL. We hypothesized that the GNAS mutation might induce RANKL overproduction and an oncogenic niche in mammary tissue, and examined RANKL expression in breast cancer tissue of patients with FD/MAS compared to controls. METHODS Nine patients with FD/MAS and breast cancer were included and clinical data were retrieved. Patients were matched to controls with breast cancer without FD/MAS based on age and tumor type. Three pregnant breast cancer patients were included as positive controls. Immunohistochemical detection of RANKL was performed on formalin-fixed paraffin-embedded breast cancer specimens. Staining intensity was classified as weak, moderate or intense. The area of positive RANKL staining divided by the total ductal-lobular area was assessed (positive area percentage, PAP). Number of patients with RANKL expression was compared between FD/MAS and control group by chi-square (χ2) test, the PAP by Mann-Whitney U test (MWU). RESULTS RANKL expression was observed in 3 patients with FD/MAS (38 %), mainly in healthy tissue, and none of the control patients (χ2p = 0.055). The FD/MAS group demonstrated considerably more intense staining than the control group, comparable to positive controls. The median PAP was 0.64 % (range 0.14-2.04 %) in the 3 FD/MAS patients with RANKL expression, 0.01 % (Q1-Q3: 0.0003-0.514 %) in the entire FD/MAS group, 0.006 % (Q1-Q3: 0.001-0.012 %) in the control group (MWU = 0.574), and 0.19 % (0.08-0.32 %) in the pregnant patients. All patients with FD/MAS and RANKL expression had thoracic bone lesions, but no correlation was observed between RANKL expression and presence of the GNAS mutation or FD disease burden. CONCLUSIONS The triad of a higher number of patients, higher positive area percentage and stronger intensity in the FD/MAS compared to the control group indicates that RANKL may be upregulated in mammary tissue in a subset of patients with FD/MAS, which may explain the increased risk for breast cancer, although the clinical significance remains unclear. Further research is needed to establish risk profiles for the development of RANKL-positive breast cancer and to improve early screening and treatment.
Collapse
Affiliation(s)
- M E Meier
- Center for Bone Quality, Department of Orthopaedic Surgery, Leiden University Medical Center, Leiden, the Netherlands.
| | - M Hagelstein-Rotman
- Center for Bone Quality, Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, the Netherlands
| | - B C J Majoor
- Center for Bone Quality, Department of Orthopaedic Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - R E S Geels
- Center for Bone Quality, Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, the Netherlands
| | - N M Appelman-Dijkstra
- Center for Bone Quality, Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, the Netherlands
| | - N Bravenboer
- Center for Bone Quality, Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, the Netherlands; Department of Clinical Chemistry, Amsterdam University Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
26
|
Bai B, Dong L, Feng M, Zhang Z, Lu Y, Xu Z, Liu Y. Prognostic and functional roles of EIF4G1 in lung squamous cell carcinoma. Hum Cell 2023; 36:1099-1107. [PMID: 36897548 PMCID: PMC10110680 DOI: 10.1007/s13577-023-00884-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 02/17/2023] [Indexed: 03/11/2023]
Abstract
Eukaryotic translation initiation factor 4 gamma 1 (EIF4G1) is highly expressed in many cancers and affects their occurrence and development. However, the effect of EIF4G1 on the prognosis, biological function and the relevant mechanism in lung squamous cell carcinoma (LSCC) is unclear. Through clinical cases, Cox's proportional hazard model and Kaplan-Meier plotter survival analysis, we find the expression levels of EIF4G1 are dependent on age and clinical stage, high expression of EIF4G1 could be used to predict the overall survival of LSCC patients. LSCC cell line NCI-H1703, NCI-H226 and SK-MES-1infected with EIF4G1 siRNA are used to detect the function of EIF4G1 with cell proliferation and tumorigenesis in vivo and vitro. The data show that EIF4G1 promotes tumor cell proliferation and the G1/S transition of cell cycle in LSCC, then the biological function of LSCC is effected by the AKT/mTOR pathway. Above all, these results have demonstrated that EIF4G1 promotes LSCC cell proliferation and may represent an indicator of prognosis in LSCC.
Collapse
Affiliation(s)
- Baoxin Bai
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200092, China
- Research Center for Translational Medicine, Shanghai East Hospital, GuiLin University School of Medicine, Guilin, 541004, China
- Hubei University of Medicine, No. 30, Renmin South Road, Maojian District, Shiyan, 442000, China
| | - Lin Dong
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200092, China
- Department of Cardiothoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Minghao Feng
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200092, China
- Department of Cardiothoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Zhiwen Zhang
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200092, China
| | - Ying Lu
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200092, China
| | - Zengguang Xu
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200092, China.
- Research Center for Translational Medicine, Shanghai East Hospital, GuiLin University School of Medicine, Guilin, 541004, China.
| | - Yali Liu
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
27
|
Tang ZN, Bi XF, Chen WL, Zhang CL. RANKL Promotes Chemotherapy Resistance in Breast Cancer Cells Through STAT3 Mediated Autophagy Induction. Clin Breast Cancer 2023; 23:388-396. [PMID: 36872108 DOI: 10.1016/j.clbc.2023.01.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/09/2023]
Abstract
BACKGROUND This study was to investigate the functional role and mechanism of receptor activator of nuclear factor-kappa B ligand (RANKL) associated autophagy and chemoresistance in breast cancer. MATERIALS AND METHODS Cell Counting Kit-8 (CCK-8) assay was used to detect the cell viability. Real-time polymerase chain reaction (PCR) was used for determining the relative mRNA levels of key genes and protein expression was assessed by Western blotting. Immunofluorescence was performed to evaluate the changes in the autophagy flux. Short hairpin (shRNA) was used to knockdown the expression of the target genes in breast cancer cells. Based on The Cancer Genome Atlas (TCGA) database, we explored the expression of receptor activator of nuclear factor-kappa B (RANK), autophagy and signal transducer and activator of transcription 3 (STAT3) signaling associated genes and analyzed their correlation with the prognosis of breast cancer patients. RESULTS The findings showed that receptor activator of nuclear factor-kappa B ligand (RANKL), the ligand of RANK, could effectively enhance the chemoresistance potential of breast cancer cells. Our results showed that RANKL induced autophagy and enhanced the expression of autophagy associated genes in breast cancer cells. The knockdown of RANK suppressed RANKL mediated autophagy induction in these cells. Furthermore, the inhibition of autophagy suppressed RANKL mediated chemoresistance in breast cancer cells. We found STAT3 signaling pathway was involved in RANKL-induced autophagy. Analysis of the expression of RANK, and autophagy and STAT3 signaling associated genes in breast cancer tissues showed that the expression of autophagy and STAT3 signaling associated genes was correlated with the prognosis of breast cancer patients. CONCLUSION The present study suggests that the RANKL/RANK axis may potentially mediate chemoresistance in breast cancer cells by inducing autophagy through the STAT3 signaling pathway.
Collapse
Affiliation(s)
- Zhen-Ning Tang
- Department of Surgical Oncology, General Hospital of Ningxia Medical University, 750004 Yinchuan, Ningxia, China
| | - Xiao-Fang Bi
- Department of Pathology, The First People's Hospital of Yinchuan, 750001 Yinchuan, Ningxia, China
| | - Wei-Liang Chen
- Department of Breast Surgery, Herbei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, 061001 Cangzhou, Hebei, China
| | - Chao-Lin Zhang
- Department of Surgical Oncology, General Hospital of Ningxia Medical University, 750004 Yinchuan, Ningxia, China.
| |
Collapse
|
28
|
Siddig A, Wan Abdul Rahman WF, Mohd Nafi SN, Sulong S, Yahya MM, Al-Astani Tengku Din TAD, Razali R, Musa KI. Comparing the Biology of Young versus Old Age Estrogen-Receptor-Positive Breast Cancer through Gene and Protein Expression Analyses. Biomedicines 2023; 11:200. [PMID: 36672708 PMCID: PMC9855392 DOI: 10.3390/biomedicines11010200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 12/26/2022] [Accepted: 01/09/2023] [Indexed: 01/15/2023] Open
Abstract
Background: Breast cancer developed at a young age (≤45 years) is hypothesized to have unique biology; however, findings in this field are controversial. Methods: We compared the whole transcriptomic profile of young vs. old-age breast cancer using DNA microarray. RNA was extracted from 13 fresh estrogen receptor (ER)-positive primary breast cancer tissues of untreated patients (7 = young age ≤45 years and 6 = old age ≥55 years). In silico validation for the differentially expressed genes (DEGs) by young-age patients was conducted using The Cancer Genome Atlas (TCGA) database. Next, we analyzed the protein expression encoded by two of the significantly down-regulated genes by young-age patients, Glycine N-acyltransferase-like 1 (GLYATL-1) and Ran-binding protein 3 like (RANBP3L), using immunohistochemical analysis in an independent cohort of 56 and 74 ER-positive pre-therapeutic primary breast cancer tissues, respectively. Results: 12 genes were significantly differentially expressed by young-age breast cancers (fold change >2 or <2- with FDR p-value < 0.05). TCGA data confirmed the differential expression of six genes. Protein expression analysis of GLYATL-1 and RANBP3L did not show heterogeneous expression between young and old-age breast cancer tissues. Loss of expression of GLYATL-1 was significantly (p-value 0.005) associated with positive lymph node status. Higher expression of RANBP3L was significantly associated with breast cancers with lower histopathological grades (p-value 0.038). Conclusions: At the transcriptomic level, breast cancer developed in young and old age patients seems homogenous. The variation in the transcriptomic profiles can be attributed to the other clinicopathological characteristics rather than the age of the patient.
Collapse
Affiliation(s)
- Alaa Siddig
- Department of Pathology, School of Medical Sciences, Universiti Sains Malaysia, Kelantan 16150, Malaysia
| | - Wan Faiziah Wan Abdul Rahman
- Department of Pathology, School of Medical Sciences, Universiti Sains Malaysia, Kelantan 16150, Malaysia
- Breast Cancer Awareness & Research Unit, Hospital Universiti Sains Malaysia, Kelantan 16150, Malaysia
| | - Siti Norasikin Mohd Nafi
- Department of Pathology, School of Medical Sciences, Universiti Sains Malaysia, Kelantan 16150, Malaysia
| | - Sarina Sulong
- Human Genome Centre, School of Medical Sciences, Universiti Sains Malaysia, Kelantan 16150, Malaysia
| | - Maya Mazuwin Yahya
- Breast Cancer Awareness & Research Unit, Hospital Universiti Sains Malaysia, Kelantan 16150, Malaysia
- Department of Surgery, School of Medical Sciences, Universiti Sains Malaysia, Kelantan 16150, Malaysia
| | - Tengku Ahmad Damitri Al-Astani Tengku Din
- Breast Cancer Awareness & Research Unit, Hospital Universiti Sains Malaysia, Kelantan 16150, Malaysia
- Department of Chemical Pathology, School of Medical Sciences, Universiti Sains Malaysia, Kelantan 16150, Malaysia
| | - Rozaimi Razali
- Department of Biomedical Sciences, College of Health Sciences, QU-Health, Qatar University, Doha P.O. Box 2703, Qatar
| | - Kamarul Imran Musa
- Department of Community Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kelantan 16150, Malaysia
| |
Collapse
|
29
|
Zhang G, Zhao X, Cai J, Li S, Li X, Li W, Shi P, Liu D, Zheng D, Zhang T, Feng R, Liu H. XCHT alleviates the pancreatic fibrosis via VDR/NLRP3 signaling pathway in a mouse model of CP. JOURNAL OF ETHNOPHARMACOLOGY 2023; 300:115689. [PMID: 36096349 DOI: 10.1016/j.jep.2022.115689] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Xiao Chai Hu Tang (XCHT) derived from the classic medical book Shang Han Lun (Treatise on Febrile Diseases) in the Eastern Han Dynasty, which has been widely used in China and other Asian countries for the treatment of inflammation and fibrosis of chronic pancreatitis (CP), but the therapeutic mechanism of XCHT in pancreatic fibrosis remains unclear. AIM OF THE STUDY This study aimed to evaluate the intervention effects and explore pharmacological mechanism of XCHT on inflammation and fibrosis in cerulein-induced CP model. MATERIALS AND METHODS Fifty male C57BL/6 mice were randomly divided into five main groups, 10 animals in each: Control, CP model (50 μg/kg cerulein), high dose XCHT-treated CP group (60 g/kg XCHT), medium dose XCHT-treated CP group (30 g/kg XCHT) and low dose XCHT-treated CP group (15 g/kg XCHT). Different doses of XCHT were given to mice by gavage twice a day for 2 weeks after the CP model induction. Pancreatic tissues were harvested and the pancreatic inflammation and fibrosis were evaluated by histological score, Sirius red staining, and alpha-smooth muscle actin (α-SMA) immunohistochemical staining. ELISA, IHC and RT-qPCR were performed to detect the expression of Vitamin D3 (VD3) and Vitamin D receptor (VDR) in serum and pancreatic tissues, respectively. The expressions of NLRP3 inflammasome related genes and molecules were assayed by WB, IHC and RT-qPCR. RESULTS The pathohistological results demonstrated that XCHT markedly inhibited the fibrosis and chronic inflammation of cerulein-induced CP, indicated by reduction of collagen I, collagen III, α-SMA, and NLRP3 expressions. XCHT significantly increased VD3 and VDR expression while reduced the pancreatic NLRP3 expression. Correspondingly, XCHT decreased the levels of NLRP3 downstream targets IL-1β, TNF-α and IL-6. CONCLUSIONS These results revealed that XCHT suppressed the pancreatic fibrosis and chronic inflammation in cerulein-induced CP model by enhancing the VD3/VDR expression and inhibiting the secretion of NLRP3-assoicated inflammatory factors.
Collapse
Affiliation(s)
- Guixian Zhang
- Department of Cancer Pharmacology, Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin Medicine and Health Research Center, Duolun Rd, Tianjin, 300020, China
| | - Xiumei Zhao
- Department of Cancer Pharmacology, Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin Medicine and Health Research Center, Duolun Rd, Tianjin, 300020, China
| | - Jun Cai
- Department of Cancer Pharmacology, Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin Medicine and Health Research Center, Duolun Rd, Tianjin, 300020, China
| | - Sainan Li
- Graduate School of Tianjin Medical University, Tianjin, 300070, China
| | - Xijing Li
- Department of Cancer Pharmacology, Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin Medicine and Health Research Center, Duolun Rd, Tianjin, 300020, China
| | - Wenchang Li
- Department of Cancer Pharmacology, Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin Medicine and Health Research Center, Duolun Rd, Tianjin, 300020, China
| | - Pengcheng Shi
- Department of Cancer Pharmacology, Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin Medicine and Health Research Center, Duolun Rd, Tianjin, 300020, China
| | - Dawei Liu
- Department of Cancer Pharmacology, Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin Medicine and Health Research Center, Duolun Rd, Tianjin, 300020, China
| | - Duo Zheng
- Department of Cancer Pharmacology, Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin Medicine and Health Research Center, Duolun Rd, Tianjin, 300020, China
| | - Ting Zhang
- Department of Cancer Pharmacology, Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin Medicine and Health Research Center, Duolun Rd, Tianjin, 300020, China
| | - Renrui Feng
- Department of Cancer Pharmacology, Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin Medicine and Health Research Center, Duolun Rd, Tianjin, 300020, China
| | - Hongbin Liu
- Department of Cancer Pharmacology, Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin Medicine and Health Research Center, Duolun Rd, Tianjin, 300020, China.
| |
Collapse
|
30
|
Yang Z, Huang R, Wang Y, Guan Q, Li D, Wu Y, Liao T, Wang Y, Xiang J. SIRT6 drives sensitivity to ferroptosis in anaplastic thyroid cancer through NCOA4-dependent autophagy. Am J Cancer Res 2023; 13:464-474. [PMID: 36895980 PMCID: PMC9989618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/26/2023] [Indexed: 03/11/2023] Open
Abstract
The sirtuin family has been reported to participate in the regulation of oxidative stress, cancer metabolism, aging, and so on. However, few studies have demonstrated its role in ferroptosis. Our previous studies confirmed that SIRT6 is upregulated in thyroid cancer and associated with cancer development by regulating glycolysis and autophagy. In this research, we aimed to elucidate the association between SIRT6 and ferroptosis. RSL3, erastin, ML210, and ML162 were applied to induce ferroptosis. Cell death and lipid peroxidation were measured by flow cytometry. We found that overexpression of SIRT6 significantly increased the sensitivity of cells to ferroptosis, whereas knockout of SIRT6 promoted resistance to ferroptosis. Furthermore, we demonstrated that SIRT6 induced NCOA4-dependent autophagic degradation of ferritin, thus driving sensitivity to ferroptosis. The clinically used ferroptosis inducer sulfasalazine showed promising therapeutic effects on SIRT6-upregulated thyroid cancer cells in vivo. In conclusion, our research demonstrated SIRT6-driven sensitivity to ferroptosis via NCOA4-dependent autophagy and proposed ferroptosis inducers as promising therapeutic agents for anaplastic thyroid cancer patients.
Collapse
Affiliation(s)
- Zhou Yang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University Shanghai 200032, China
| | - Renhong Huang
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine Shanghai 200025, China
| | - Yunjun Wang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University Shanghai 200032, China
| | - Qing Guan
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University Shanghai 200032, China
| | - Duanshu Li
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University Shanghai 200032, China
| | - Yi Wu
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University Shanghai 200032, China
| | - Tian Liao
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University Shanghai 200032, China
| | - Yu Wang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University Shanghai 200032, China
| | - Jun Xiang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University Shanghai 200032, China
| |
Collapse
|
31
|
Ma H, Qiu Q, Tan D, Chen Q, Liu Y, Chen B, Wang M. The Cancer-Associated Fibroblasts-Related Gene COMP Is a Novel Predictor for Prognosis and Immunotherapy Efficacy and Is Correlated with M2 Macrophage Infiltration in Colon Cancer. Biomolecules 2022; 13:biom13010062. [PMID: 36671447 PMCID: PMC9856124 DOI: 10.3390/biom13010062] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/23/2022] [Accepted: 12/23/2022] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Colon cancer is characterized by a sophisticated tumor microenvironment (TME). Cancer-associated fibroblasts (CAFs), which make up the majority of the stromal cells in TME, participate in tumor development and immune regulation. Further investigations of CAFs would facilitate an in-depth understanding of its role in colon cancer TME. METHODS In this study, we estimated CAF abundance based on The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases using the Microenvironment Cell Populations-counter (MCP-counter) algorithm. CAF-related genes were identified by differential gene expression analysis combined with weighted gene coexpression network analysis. For further selection, the least absolute shrinkage and selection operator (LASSO)-Cox regression was used, and the prognostic value of the selected gene was confirmed in numerous external cohorts. The function enrichment, immunological characteristics, tumor mutation signature, immunotherapy response, and drug sensitivity of the selected gene were subsequently explored. The bioinformatics analysis results were validated using immunohistochemistry on clinical samples from our institution. RESULTS According to our findings, cartilage oligomeric matrix protein (COMP) was uncovered as a candidate CAFs-driven biomarker in colon cancer and plays an important role in predicting prognosis in colon cancer. COMP upregulation was associated with enhanced stromal and immune activation, and immune cell infiltration, especially M2 macrophages. Genes that mutated differently between the high- and low-COMP expression subgroups may be correlated with TME change. Following verification, COMP reliably predicted the immunotherapy response and drug response. In addition, our experimental validation demonstrated that COMP overexpression is associated with colon cancer carcinogenesis and is strongly associated with CAFs and M2 macrophage infiltration. CONCLUSION Our study uncovered that COMP was a key CAFs-driven gene associated with M2 macrophage infiltration and acted as a convincing predictor for prognosis and immunotherapy response in colon cancer patients.
Collapse
Affiliation(s)
- He Ma
- Department of General Surgery, RuiJin Hospital Lu Wan Branch, Shanghai Jiaotong University School of Medicine, Shanghai 200020, China
| | - Qingqing Qiu
- Department of General Surgery, RuiJin Hospital Lu Wan Branch, Shanghai Jiaotong University School of Medicine, Shanghai 200020, China
| | - Dan Tan
- Department of General Surgery, RuiJin Hospital Lu Wan Branch, Shanghai Jiaotong University School of Medicine, Shanghai 200020, China
| | - Qiaofeng Chen
- Department of General Surgery, RuiJin Hospital Lu Wan Branch, Shanghai Jiaotong University School of Medicine, Shanghai 200020, China
| | - Yaping Liu
- Department of General Surgery, RuiJin Hospital Lu Wan Branch, Shanghai Jiaotong University School of Medicine, Shanghai 200020, China
| | - Bing Chen
- Central Laboratory, RuiJin Hospital Lu Wan Branch, Shanghai Jiaotong University School of Medicine, Shanghai 200020, China
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Correspondence: (B.C.); (M.W.)
| | - Mingliang Wang
- Department of General Surgery, RuiJin Hospital Lu Wan Branch, Shanghai Jiaotong University School of Medicine, Shanghai 200020, China
- Department of General Surgery, RuiJin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
- Correspondence: (B.C.); (M.W.)
| |
Collapse
|
32
|
Yan H, Cai X, Fu S, Zhang X, Zhang J. PRDX3 promotes resistance to cisplatin in gastric cancer cells. J Cancer Res Ther 2022; 18:1994-2000. [PMID: 36647961 DOI: 10.4103/jcrt.jcrt_970_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Objective This study aims to investigate peroxiredoxin 3 (PRDX3) expression in gastric cancer tissue and its effects on cisplatin resistance in gastric cancer cells and its possible mechanism. Methods PRDX3 expression in human gastric cancer tissue microarrays was detected via immunohistochemistry. The PRDX3 small interfering RNA (siPRDX3 group) and the negative control siNC (siNC group) were transfected into AGS and MKN-74 cell lines, respectively, whereas a blank control group was set up. Each group was treated with different cisplatin concentrations (0, 5, 10, 15, 20, 25, and 30 μg/ml), and the half-inhibitory concentration (IC50) of each group of the two cell lines was calculated using the CCK8 assay. The corresponding IC50 concentration of the siPRDX3 group in the two cell lines was used to treat cells of each group. Flow cytometry was used to detect cell apoptosis, and Western blotting was used to detect the expression levels of cleaved caspase-3 and Bax in each group. Results PRDX3 was overexpressed in gastric adenocarcinoma tissue compared with adjacent noncancer tissue (P = 0.0053). After cisplatin treatment, the IC50 in the siPRDX3 group of AGS cells (5.91 ± 0.18 μg/ml) and the siPRDX3 group of MKN-74 cells (3.48 ± 0.30 μg/ml) was significantly lower than in the corresponding siNC groups (10.01 ± 0.99 and 6.39 ± 0.70 μg/ml; P = 0.0022 and 0.0027, respectively). AGS cells (38.81% ± 1.69%) and MKN-74 cells (25.03% ± 2.80%) in the siPRDX3 group showed significantly higher apoptosis rates than in the corresponding siNC groups (23.17% ± 1.43% and 16.7% ± 1.39%; P = 0.0003 and 0.0099, respectively). The expression levels of cleaved caspase-3 and Bax were significantly higher in the siPRDX3 group of both cell lines than in the siNC group (P < 0.0001). Conclusion PRDX3 increases the gastric cancer cell resistance to cisplatin by reducing apoptosis and thus may serve as a target to overcome cisplatin resistance.
Collapse
Affiliation(s)
- Hao Yan
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital; Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Xinyu Cai
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital; Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Shanshan Fu
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong Province, China
| | - Xiubin Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong Province, China
| | - Jianna Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong Province, China
| |
Collapse
|
33
|
Ye C, Qin S, Qiu S, Zhao L, Miao J, Chen Y, Zhou T. A lncRNA-immune checkpoint-related gene signature predicts metastasis-free survival in prostate adenocarcinoma. Transl Androl Urol 2022; 11:1691-1705. [PMID: 36632155 PMCID: PMC9827409 DOI: 10.21037/tau-22-711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 11/24/2022] [Indexed: 12/24/2022] Open
Abstract
Background The 5-year overall survival rate in metastatic prostate adenocarcinoma (PRAD) is extremely low. Genomic studies of PRAD have improved our understanding of disease biology. However, the role of immune checkpoint genes (ICGs) in PRAD remains unclear. Methods Univariate and multivariate analyses were used to analyze genes associated with metastasis-free survival (MFS) in The Cancer Genome Atlas (TCGA)-PRAD dataset. The expressions of ADORA2A and TNFRSF18 were detected via immunohistochemical assay and real-time fluorescence quantitative PCR (RT-PCR) assay in our in-house cohort. The expression of long non-coding RNAs (lncRNAs) AL139287.1, SLC9A3-AS1, and SNHG12 were detected via RT-PCR assay in our in-house cohort. Stepwise regression, Cox regression, and nomogram analyses were used to evaluate the prognostic role of these genes in both the TCGA dataset and in-house cohort. The "pRRophetic" R package was used to evaluate drug sensitivity in the TCGA cohort according to the gene mRNA expression level. Results In our study, univariate and multivariate analyses revealed that the mRNA expressions of two ICGs, ADORA2A and TNFRSF18, were independent factors affecting MFS in PRAD patients. A prognostic 2-ICG model predicted the MFS of PRAD patients with medium-to-high accuracy in the TCGA dataset and in-house cohort. The expressions of AL139287.1, SLC9A3-AS1, and SNHG12 were correlated with ADORA2A and TNFRSF18. A prognostic lncRNA-ICG model predicted the MFS of PRAD patients with medium-to-high accuracy in the TCGA dataset and in-house cohort. In addition, correlation analyses between the sensitivity of doxorubicin, erlotinib, gemcitabine, or vinorelbine and AL139287.1, SLC9A3-AS1, SNHG12, ADORA2A, and TNFRSF18 were conducted. Conclusions Our results provide new targets for predicting tumor metastasis in PRAD and treating patients with metastatic PRAD.
Collapse
Affiliation(s)
- Chen Ye
- Department of Urology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Shengfei Qin
- Department of Urology, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shuang Qiu
- Department of Nuclear Medicine, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Lin Zhao
- Department of Urology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Jiaying Miao
- Department of Urology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yuangui Chen
- Department of Urology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Tie Zhou
- Department of Urology, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
34
|
Liu X, Meng X, Su X, Ren K, Ning C, Qi X, Zhang S. The mechanism of ginger and its processed products in the treatment of estradiol valerate coupled with oxytocin-induced dysmenorrhea in mice via regulating the TRP ion channel-mediated ERK 1/2/NF-κB signaling pathway. Food Funct 2022; 13:11236-11248. [PMID: 36222424 DOI: 10.1039/d2fo01845d] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
Ginger (Rhizoma zingiberis, RZ) has been used as a food, spice, supplement, flavoring agent, and as an edible herbal medicine. It is characterized by its pungency and aroma, and is rich in nutrients with remarkable pharmacological effects. It is used in traditional medicine clinics to treat diseases and symptoms, such as colds, headache, and primary dysmenorrhea (PD). In China, a variety of processed products of RZ are used as herbal medicines, such as baked ginger (BG) or ginger charcoal (GC) to treat different diseases and symptoms. However, the molecular mechanism of the therapeutic effect of RZ and its processed products (RZPPs, including BG or GC) against PD has not been well characterized. Moreover, whether the transient receptor potential (TRP) ion channels are involved in this process is not clear. In the present study, UHPLC-Q-TOF MS was adopted to analyse the differential quality markers (DQMs) between RZ and RZPPs. In addition, differential metabolomics (DMs) was acquired between RZ- and RZPPs-treated estradiol valerate coupled with an oxytocin-induced PD mouse uterus using untargeted metabolomics (UM). A correlation analysis between DQMs and DMs was also conducted. Benzenoids, lipids, and lipid-like molecules were the main DQMs between RZ and RZPPs. RZ and RZPPs were found to improve the pathological status of the uterus of a PD mouse, with significantly decreased serum levels of E2, PGF2α, TXB2 and remarkably increased levels of PROG and 6-keto-PGF1α. Moreover, RZ and RZPPs alleviated PD in mice via regulating the TRP ion channel-mediated ERK1/2/NF-κB signaling pathway. Our results indicate that the therapeutic effect of RZ and RZPPs against PD may be mediated by regulating the TRP ion channel-mediated ERK1/2/NF-κB signaling pathway, and provide a reference for the development of new dietary supplements or medicines.
Collapse
Affiliation(s)
- Xiaoqin Liu
- College of Chinese Materia Medica and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong 030619, Shanxi, China.
- Shanxi Key Laboratory of Traditional Herbal Medicines Processing, Jinzhong 030619, Shanxi, China
| | - Xianglong Meng
- College of Chinese Materia Medica and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong 030619, Shanxi, China.
- Shanxi Key Laboratory of Traditional Herbal Medicines Processing, Jinzhong 030619, Shanxi, China
| | - Xiaojuan Su
- College of Chinese Materia Medica and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong 030619, Shanxi, China.
- Shanxi Key Laboratory of Traditional Herbal Medicines Processing, Jinzhong 030619, Shanxi, China
| | - Kele Ren
- College of Chinese Materia Medica and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong 030619, Shanxi, China.
- Shanxi Key Laboratory of Traditional Herbal Medicines Processing, Jinzhong 030619, Shanxi, China
| | - Chenxu Ning
- College of Chinese Materia Medica and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong 030619, Shanxi, China.
- Shanxi Key Laboratory of Traditional Herbal Medicines Processing, Jinzhong 030619, Shanxi, China
| | - Xiaoming Qi
- College of Chinese Materia Medica and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong 030619, Shanxi, China.
- Shanxi Key Laboratory of Traditional Herbal Medicines Processing, Jinzhong 030619, Shanxi, China
| | - Shuosheng Zhang
- College of Chinese Materia Medica and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong 030619, Shanxi, China.
- Shanxi Key Laboratory of Traditional Herbal Medicines Processing, Jinzhong 030619, Shanxi, China
| |
Collapse
|
35
|
Huang X, Zhu J, Li Y, Yu Y, Tang J. La protein regulates protein expression by binding with the mRNAs of target genes and participates the pathological process of ovarian cancer. Front Oncol 2022; 12:763480. [PMID: 36110943 PMCID: PMC9468491 DOI: 10.3389/fonc.2022.763480] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 08/08/2022] [Indexed: 12/03/2022] Open
Abstract
Research on the mechanism and new targets of ovarian cancer is of great significance to reduce the high mortality and drug resistance of ovarian cancer. Human La protein has been found to be highly expressed in a variety of malignant tumors and plays a role in tumorigenesis and development through its RNA-binding function. However, its role and mechanism in ovarian cancer are not completely clear. The present study showed that La protein was highly expressed in serum and tissues of patients with ovarian cancer by ELISA and immunohistochemistry, and the high expression of La protein was associated with the increased degree of malignancy and poor prognosis by searching the KM plotter database. Interference of the La gene resulted in a significant decrease in the proliferation, migration, and invasion of ovarian cancer cells with growth block in the G1 phase and increasing apoptosis. By RNA binding protein immunoprecipitation, transcriptome sequencing, and proteomics, 14 downstream target genes were screened. The La protein might affect the protein expression of these 14 genes by binding with the mRNAs. Therefore, it played a role in the pathological process of ovarian cancer.
Collapse
|
36
|
SIRT4 functions as a tumor suppressor during prostate cancer by inducing apoptosis and inhibiting glutamine metabolism. Sci Rep 2022; 12:12208. [PMID: 35842463 PMCID: PMC9288510 DOI: 10.1038/s41598-022-16610-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 07/12/2022] [Indexed: 12/04/2022] Open
Abstract
Localized in the mitochondria, SIRT4 is a nicotinamide adenine dinucleotide (NAD +) -dependent adenosine diphosphate (ADP) -ribosyltransferase and is one of the least characterized members of the sirtuin family. Although it is well known that it shows deacetylase activity for energy metabolism, little is understood about its function in tumorigenesis. Recent research suggests that SIRT4 may work as both a tumor suppressor gene and an oncogene. However, the clinical significance of SIRT4 in prostate cancer remains unknown. In this study, we evaluated SIRT4 protein levels in cancerous prostate tissue and corresponding non-tumor prostate tissue via immunohistochemical staining on a tissue microarray including tissues from 89 prostate cancer patients. The association between SIRT4 expression and Gleason score was also determined. Further, shSIRT4 or stable prostate cancer cell lines (22RV1) overexpressing SIRT4 were constructed via lentiviral infection. Using Cell-Counting Kit-8 (CCK-8) assay, wound healing assay, migration, and invasion and apoptosis assays, the effects of SIRT4 on the migration, invasion ability, and proliferation of prostate cancer cells were investigated. We also determined the effect of SIRT4 on glutamine metabolism in 22RV1 cells. We found the protein levels of SIRT4 in prostate cancer tissues were significantly lower than those in their non-neoplastic tissue counterparts (P < 0.01); a lower SIRT4 level was also significantly associated with a higher Gleason score (P < 0.01). SIRT4 suppressed the migration, invasion capabilities, and proliferation of prostate cancer cells and induced cellular apoptosis. Furthermore, the invasion and migration of 22RV1 cells were mechanistically inhibited by SIRT4 via glutamine metabolism inhibition. In conclusion, the present study’s findings showed that SIRT4 protein levels are significantly associated with the Gleason score in patients with prostate cancer, and SIRT4 exerts a tumor-suppressive effect on prostate cancer cells by inhibiting glutamine metabolism. Thus, SIRT4 may serve as a potential novel therapeutic target for prostate cancer.
Collapse
|
37
|
Delineation of Pathogenomic Insights of Breast Cancer in Young Women. Cells 2022; 11:cells11121927. [PMID: 35741056 PMCID: PMC9221490 DOI: 10.3390/cells11121927] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/03/2022] [Accepted: 06/10/2022] [Indexed: 12/12/2022] Open
Abstract
The prognosis of breast cancer (BC) in young women (BCYW) aged ≤40 years tends to be poorer than that in older patients due to aggressive phenotypes, late diagnosis, distinct biologic, and poorly understood genomic features of BCYW. Considering the estimated predisposition of only approximately 15% of the BC population to BC-promoting genes, the underlying reasons for an increased occurrence of BCYW, at large, cannot be completely explained based on general risk factors for BC. This underscores the need for the development of next-generation of tissue- and body fluid-based prognostic and predictive biomarkers for BCYW. Here, we identified the genes associated with BCYW with a particular focus on the age, intrinsic BC subtypes, matched normal or normal breast tissues, and BC laterality. In young women with BC, we observed dysregulation of age-associated cancer-relevant gene sets in both cancer and normal breast tissues, sub-sets of which substantially affected the overall survival (OS) or relapse-free survival (RFS) of patients with BC and exhibited statically significant correlations with several gene modules associated with cellular processes such as the stroma, immune responses, mitotic progression, early response, and steroid responses. For example, high expression of COL1A2, COL5A2, COL5A1, NPY1R, and KIAA1644 mRNAs in the BC and normal breast tissues from young women correlated with a substantial reduction in the OS and RFS of BC patients with increased levels of these exemplified genes. Many of the genes upregulated in BCYW were overexpressed or underexpressed in normal breast tissues, which might provide clues regarding the potential involvement of such genes in the development of BC later in life. Many of BCYW-associated gene products were also found in the extracellular microvesicles/exosomes secreted from breast and other cancer cell-types as well as in body fluids such as urine, saliva, breast milk, and plasma, raising the possibility of using such approaches in the development of non-invasive, predictive and prognostic biomarkers. In conclusion, the findings of this study delineated the pathogenomics of BCYW, providing clues for future exploration of the potential predictive and prognostic importance of candidate BCYW molecules and research strategies as well as a rationale to undertake a prospective clinical study to examine some of testable hypotheses presented here. In addition, the results presented here provide a framework to bring out the importance of geographical disparities, to overcome the current bottlenecks in BCYW, and to make the next quantum leap for sporadic BCYW research and treatment.
Collapse
|
38
|
Tang R, Yi J, Lu S, Chen B, Liu B. Therapeutic Effect of Buyang Huanwu Decoction on the Gut Microbiota and Hippocampal Metabolism in a Rat Model of Cerebral Ischemia. Front Cell Infect Microbiol 2022; 12:873096. [PMID: 35774407 PMCID: PMC9237419 DOI: 10.3389/fcimb.2022.873096] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 05/11/2022] [Indexed: 12/04/2022] Open
Abstract
Buyang Huanwu decoction (BHD) is a well-known Chinese herbal prescription. It has been widely used in the clinical treatment of cerebral ischemia (CI) in China. However, the mechanism underlying the treatment of CI with BHD remains to be elucidated. In this study, we combined microbiomic and metabolomic strategies to explore the therapeutic effects of BHD on middle cerebral artery occlusion (MCAO) in rats. Our results showed that BHD could effectively improve neurological severity scores and alleviate neuronal damage in rats with MCAO. BHD could also reduce the level of peripheral proinflammatory cytokines and inhibit neuroinflammation. 16S rRNA sequencing showed that BHD could increase the relative abundances of the genera Lactobacillus, Faecalibacterium, Ruminococcaceae_UCG-002, etc., while decreasing the relative abundances of the genera Escherichia-Shigella, Klebsiella, Streptococcus, Coprococcus_2, Enterococcus, etc. Untargeted metabolomic analysis of hippocampal samples showed that 17 significantly differentially abundant metabolites and 9 enriched metabolic pathways were linked with BHD treatment. We also found that the regulatory effects of BHD on metabolites were correlated with the differentially abundant microbial taxa. The predicted function of the gut microbiota and the metabolic pathway enrichment results showed that purine metabolism, glutamatergic synapses, arginine and proline metabolism, and alanine, aspartic acid and glutamate metabolism were involved in the effects of BHD. These pathways may be related to pathological processes such as excitotoxicity, neuroinflammation, and energy metabolism disorder in CI. In summary, these findings suggest that regulation of hippocampal metabolism and of the composition and function of the gut microbiota may be important mechanisms underlying the effect of BHD in the treatment of CI.
Collapse
Affiliation(s)
- Rongmei Tang
- The First Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, China
| | - Jian Yi
- The First Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, China
| | - Shuangying Lu
- The First Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, China
| | - Bowei Chen
- The First Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, China
| | - Baiyan Liu
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
- *Correspondence: Baiyan Liu,
| |
Collapse
|
39
|
Kumar R, Abreu C, Toi M, Saini S, Casimiro S, Arora A, Paul AM, Velaga R, Rameshwar P, Lipton A, Gupta S, Costa L. Oncobiology and treatment of breast cancer in young women. Cancer Metastasis Rev 2022; 41:749-770. [PMID: 35488982 DOI: 10.1007/s10555-022-10034-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 04/14/2022] [Indexed: 12/20/2022]
Abstract
Female breast cancer emerged as the leading cancer type in terms of incidence globally in 2020. Although mortality due to breast cancer has improved during the past three decades in many countries, this trend has reversed in women less than 40 years since the past decade. From the biological standpoint, there is consensus among experts regarding the clinically relevant definition of breast cancer in young women (BCYW), with an age cut-off of 40 years. The idea that breast cancer is an aging disease has apparently broken in the case of BCYW due to the young onset and an overall poor outcome of BCYW patients. In general, younger patients exhibit a worse prognosis than older pre- and postmenopausal patients due to the aggressive nature of cancer subtypes, a high percentage of cases with advanced stages at diagnosis, and a high risk of relapse and death in younger patients. Because of clinically and biologically unique features of BCYW, it is suspected to represent a distinct biologic entity. It is unclear why BCYW is more aggressive and has an inferior prognosis with factors that contribute to increased incidence. However, unique developmental features, adiposity and immune components of the mammary gland, hormonal interplay and crosstalk with growth factors, and a host of intrinsic and extrinsic risk factors and cellular regulatory interactions are considered to be the major contributing factors. In the present article, we discuss the status of BCYW oncobiology, therapeutic interventions and considerations, current limitations in fully understanding the basis and underlying cause(s) of BCYW, understudied areas of BCYW research, and postulated advances in the coming years for the field.
Collapse
Affiliation(s)
- Rakesh Kumar
- Cancer Research Institute, Himalayan Institute of Medical Sciences, Swami Rama Himalayan University, Dehradun, India. .,Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Trivandrum, India. .,Department of Medicine, Division of Hematology and Oncology, Rutgers New Jersey Medical School, Newark, NJ, USA. .,Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA.
| | - Catarina Abreu
- Department of Medical Oncology, Hospital de Santa Maria- Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
| | - Masakazu Toi
- Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Sunil Saini
- Cancer Research Institute, Himalayan Institute of Medical Sciences, Swami Rama Himalayan University, Dehradun, India
| | - Sandra Casimiro
- Instituto de Medicina Molecular-João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Anshika Arora
- Cancer Research Institute, Himalayan Institute of Medical Sciences, Swami Rama Himalayan University, Dehradun, India
| | - Aswathy Mary Paul
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Trivandrum, India
| | - Ravi Velaga
- Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Pranela Rameshwar
- Department of Medicine, Division of Hematology and Oncology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Allan Lipton
- Hematology-Oncology, Department of Medicine, Penn State University School of Medicine, Hershey, PA, USA
| | - Sudeep Gupta
- Department of Medical Oncology, Tata Memorial Centre and Homi Bhabha National Institute, Mumbai, India
| | - Luis Costa
- Department of Medical Oncology, Hospital de Santa Maria- Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal.,Instituto de Medicina Molecular-João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
40
|
Song X, Wei C, Li X. The Signaling Pathways Associated With Breast Cancer Bone Metastasis. Front Oncol 2022; 12:855609. [PMID: 35372035 PMCID: PMC8965611 DOI: 10.3389/fonc.2022.855609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 02/16/2022] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Breast cancer (BC) is now the leading cause of cancer in women, and bone is the primary site of distant BC metastasis. BC bone metastasis seriously affects the quality of life of patients and increases the mortality rate. However, the mechanism of BC bone metastasis is not fully understood. MAIN BODY Paget's "seed and soil" hypothesis led experts to explore the relationship between surface markers and receptors in breast tumors and various growth factors in bone. The relevant breast tumor markers serve as "seeds", and the bone microenvironment that is suitable for the survival of the tumor serves as the "soil". These factors interact to make up an entire system and form feedback pathways that accelerate the production of various cytokines, attracting BC cells to migrate to bone tissue, which worsens the development of BC and seriously affects the prognosis of patients. This process is a vicious cycle. At present, there are seven major signaling pathways involved in BC bone metastasis: the OPG/RANK/RANKL signaling pathway, TGF-β signaling pathway, IGF system, PI3K-AKT-mTOR signaling pathway, Wnt signaling pathway and Hippo signaling pathway. In addition, FGF-FGFR signaling pathway, androgen-AR/LSD1-target gene pathway, Notch signaling pathway, JAK-STAT signaling pathway and CaN/NFATC1 signaling pathway also seem to be associated with BC bone metastasis. CONCLUSION This review focuses on the signaling pathways related to BC bone metastasis and explores the interactions among these pathways, which will lay a solid theoretical foundation for further understanding the mechanism of BC bone metastasis and developing effective targeted therapeutic drugs.
Collapse
Affiliation(s)
- Xuelian Song
- Department of Breast Surgery, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, China
| | - Changran Wei
- Department of The First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiangqi Li
- Department of Breast Surgery, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, China
| |
Collapse
|
41
|
Gong J, Ji Y, Liu X, Zheng Y, Zhen Y. Mithramycin suppresses tumor growth by regulating CD47 and PD-L1 expression. Biochem Pharmacol 2022; 197:114894. [PMID: 34968486 DOI: 10.1016/j.bcp.2021.114894] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 12/10/2021] [Accepted: 12/13/2021] [Indexed: 01/20/2023]
Abstract
Mithramycin A (MIT) has reacquired extensive research attention due to its anti-solid tumor activity and improved pharmacological production. Mechanismly, MIT was broadly used as a c-Myc inhibitor, and c-Myc regulated CD47 and PD-L1 expression which has been demonstrated. However, how MIT affects immune check-point molecules remains unknown. In this study, we found CD47 expression was higher in melanoma of pan-tissue array. MIT inhibited CD47 expression both in mRNA and protein level in melanoma cells (SK-MEL-28 and B16). MIT inhibited c-Myc, Sp-1 and CD47 expression in a concentration-dependent way. MIT inhibited the surface CD47 expression and promoted the phagocytosis of SK-MEL-28 cells by THP-1 cells. We found MIT inhibited tumor growth in melanoma allograft mice and CD47 expression in tumor mass. We also found MIT upregulated PD-L1 expression in cancer cells possibly via inhibiting PD-L1 ubiquitination, increasing ROS and IFN-γ. Combination of MIT and anti-PD-1 antibody showed enhanced antitumor activity compared to MIT and anti-PD-1 antibody alone in MC38 allograft mice. Using immune checkpoint array we found MIT inhibited expression of FasL and Galectin3. These results suggest that MIT inhibits CD47 expression, while improves PD-L1 expression. Furthermore, the combination of MIT and anti-PD-1 antibody exerts potent antitumor effect.
Collapse
Affiliation(s)
- Jianhua Gong
- Department of Oncology, Institute of Medicinal Biotechnology Chinese Academy of Medical Sciences & Peking Union Medical College, 1# Tiantan Xili, Beijing 100050, China; College of Life Sciences, North China University of Science and Technology, 21 Bohai Road, Caofeidian Xincheng, Tangshan 063210, Hebei, China.
| | - Yuying Ji
- Department of Oncology, Institute of Medicinal Biotechnology Chinese Academy of Medical Sciences & Peking Union Medical College, 1# Tiantan Xili, Beijing 100050, China; College of Life Sciences, North China University of Science and Technology, 21 Bohai Road, Caofeidian Xincheng, Tangshan 063210, Hebei, China
| | - Xiujun Liu
- Department of Oncology, Institute of Medicinal Biotechnology Chinese Academy of Medical Sciences & Peking Union Medical College, 1# Tiantan Xili, Beijing 100050, China; College of Life Sciences, North China University of Science and Technology, 21 Bohai Road, Caofeidian Xincheng, Tangshan 063210, Hebei, China
| | - Yanbo Zheng
- Department of Oncology, Institute of Medicinal Biotechnology Chinese Academy of Medical Sciences & Peking Union Medical College, 1# Tiantan Xili, Beijing 100050, China; College of Life Sciences, North China University of Science and Technology, 21 Bohai Road, Caofeidian Xincheng, Tangshan 063210, Hebei, China.
| | - Yongsu Zhen
- Department of Oncology, Institute of Medicinal Biotechnology Chinese Academy of Medical Sciences & Peking Union Medical College, 1# Tiantan Xili, Beijing 100050, China; College of Life Sciences, North China University of Science and Technology, 21 Bohai Road, Caofeidian Xincheng, Tangshan 063210, Hebei, China
| |
Collapse
|
42
|
Zhang D, Wu F, Song J, Meng M, Fan X, Lu C, Weng Q, Fang S, Zheng L, Tang B, Yang Y, Tu J, Xu M, Zhao Z, Ji J. A role for the NPM1/PTPN14/YAP axis in mediating hypoxia-induced chemoresistance to sorafenib in hepatocellular carcinoma. Cancer Cell Int 2022; 22:65. [PMID: 35135548 PMCID: PMC8822852 DOI: 10.1186/s12935-022-02479-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 01/20/2022] [Indexed: 12/21/2022] Open
Abstract
Background Tumor microenvironments are characterized by resistance to chemotherapeutic agents and radiotherapy. Hypoxia plays an important role in the development of tumor resistance, as well as the generation of metastatic potential. YAP also participates in the regulation of hypoxia-mediated chemoresistance, and is negatively regulated by protein tyrosine phosphatase non-receptor type 14 (PTPN14). Methods The PTPN14 expression in hepatocellular carcinoma (HCC) tissues were evaluated by qRT-PCR, western blot and tissue microarrays. The effect of PTPN14 on HCC progression was investigated in vitro and in vivo. Results Here, we report that PTPN14 expression was downregulated in HCC tissues and cell lines. Silencing PTPN14 significantly enhanced proliferation, migration, invasion of HepG2 cells in vitro and tumor growth and metastasis in vivo, whereas overexpression of PTPN14 significantly inhibited these abilities in SK-Hep1 cells. We also found that hypoxia-induced nuclear translocation and accumulation of PTPN14 led to resistance to sorafenib in HCC cells. Further mechanistic studies suggested that NPM1 regulates PTPN14 localization, and that NPM1 regulates YAP by retaining PTPN14 in the nucleus under hypoxic conditions. Conclusions These data suggest that a therapeutic strategy against chemoresistant HCC may involve disruption of NPM1-mediated regulation of YAP by retaining PTPN14 in the nucleus under hypoxic conditions. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-022-02479-0.
Collapse
Affiliation(s)
- Dengke Zhang
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Fazong Wu
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Jingjing Song
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Miaomiao Meng
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Xiaoxi Fan
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Chenying Lu
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Qiaoyou Weng
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Shiji Fang
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Liyun Zheng
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Bufu Tang
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Yang Yang
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Jianfei Tu
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Min Xu
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Zhongwei Zhao
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China.
| | - Jiansong Ji
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China.
| |
Collapse
|
43
|
Akkermansia muciniphila Ameliorates Acetaminophen-Induced Liver Injury by Regulating Gut Microbial Composition and Metabolism. Microbiol Spectr 2022; 10:e0159621. [PMID: 35107323 PMCID: PMC8809353 DOI: 10.1128/spectrum.01596-21] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The gut microbiota drives individual sensitivity to excess acetaminophen (APAP)-mediated hepatotoxicity. It has been reported that the bacterium Akkermansia muciniphila protects hosts against liver disease via the liver-gut axis, but its therapeutic potential for drug-induced liver injury remains unclear. In this study, we aimed to investigate the effect of A. muciniphila on APAP-induced liver injury and the underlying mechanism. Administration of A. muciniphila efficiently alleviated APAP-induced hepatotoxicity and reduced the levels of serum alanine aminotransferase (ALT) and aspartate transaminase (AST). A. muciniphila significantly attenuated APAP-induced oxidative stress and the inflammatory response, as evidenced by restoration of the reduced glutathione/oxidized glutathione (GSH/GSSG) balance, enhanced superoxide dismutase (SOD) activity, reduced proinflammatory cytokine production, and alleviation of macrophage and neutrophil infiltration. Moreover, A. muciniphila maintained gut barrier function, reshaped the perturbed microbial community and promoted short-chain fatty acid (SCFA) secretion. The beneficial effects of A. muciniphila were accompanied by alterations in hepatic gene expression at the transcriptional level and activation of the phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway. Our results suggested that A. muciniphila could be a potential pretreatment for APAP-induced liver injury. IMPORTANCE Our work revealed that A. muciniphila attenuated APAP-induced liver injury by alleviating oxidative stress and inflammation in the liver, and its hepatoprotective effect was accompanied by activation of the PI3K/Akt pathway and mediated by regulation of the composition and metabolic function of the intestinal microbiota. This finding suggested that the microbial community is a non-negligible impact on drug metabolism and probiotic administration could be a potential therapy for drug-induced liver injury.
Collapse
|
44
|
Guo D, Zhang X, Cui H, Yu D, Zhang H, Shi X, Pang C, Li J, Guo W, Zhang S. ACADL Functions as a Tumor Suppressor in Hepatocellular Carcinoma Metastasis by Inhibiting Matrix Metalloproteinase 14. Front Oncol 2022; 12:821484. [PMID: 35174091 PMCID: PMC8841782 DOI: 10.3389/fonc.2022.821484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/11/2022] [Indexed: 11/13/2022] Open
Abstract
High aggressiveness is the main reason for the poor prognosis of hepatocellular carcinoma (HCC) patients. However, its molecular mechanisms still remain largely unexplored. ACADL, a mitochondrial enzyme that facilitates the primary regulated step in mitochondrial fatty acid oxidation, plays a role in HCC growth inhibition. However, the function of ACADL in tumor metastasis is not well elucidated. We found that the reduced expression of ACADL is closely associated with the loss of tumor encapsulation, extrahepatic metastasis, and poor prognosis in HCC patients. Upregulation of ACADL significantly inhibited HCC migration and invasion ability. Whereas knockdown of ACADL markedly enhanced cell invasive capability. Expression of matrix metalloproteinase-14 (MMP14) was negatively associated with the content of ACADL in HCC specimens. MMP14-positive patients with a low expression of ACADL showed worse outcome. Treatment with MMP14 agonist reversed the inhibitory effect of ACADL on HCC metastasis. In addition, ACADL negatively regulated MMP14 expression by inhibiting the STAT3 signaling pathway, as the sustained activation of STAT3 effectively restored the level of MMP14 in ACADL-overexpressed cells. Collectively, these findings disclose that ACADL represses HCC metastasis via STAT3-MMP14 pathway. This study may propose a promising strategy for the precise treatment of metastatic HCC patients.
Collapse
Affiliation(s)
- Danfeng Guo
- Henan Key Laboratory for Digestive Organ Transplantation, Zhengzhou, China
- Henan Research Centre for Organ Transplantation, Zhengzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaodan Zhang
- Henan Key Laboratory for Digestive Organ Transplantation, Zhengzhou, China
- Henan Research Centre for Organ Transplantation, Zhengzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Honglei Cui
- Henan Key Laboratory for Digestive Organ Transplantation, Zhengzhou, China
- Henan Research Centre for Organ Transplantation, Zhengzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dongsheng Yu
- Henan Key Laboratory for Digestive Organ Transplantation, Zhengzhou, China
- Henan Research Centre for Organ Transplantation, Zhengzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huapeng Zhang
- Henan Key Laboratory for Digestive Organ Transplantation, Zhengzhou, China
- Henan Research Centre for Organ Transplantation, Zhengzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoyi Shi
- Henan Key Laboratory for Digestive Organ Transplantation, Zhengzhou, China
- Henan Research Centre for Organ Transplantation, Zhengzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chun Pang
- Henan Key Laboratory for Digestive Organ Transplantation, Zhengzhou, China
- Henan Research Centre for Organ Transplantation, Zhengzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jie Li
- Henan Key Laboratory for Digestive Organ Transplantation, Zhengzhou, China
- Henan Research Centre for Organ Transplantation, Zhengzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenzhi Guo
- Henan Key Laboratory for Digestive Organ Transplantation, Zhengzhou, China
- Henan Research Centre for Organ Transplantation, Zhengzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuijun Zhang
- Henan Key Laboratory for Digestive Organ Transplantation, Zhengzhou, China
- Henan Research Centre for Organ Transplantation, Zhengzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
45
|
Yu C, Qi H, Zhang Y, Zhao W, Wu G. Elevated Expression of Gamma-Glutamyl Hydrolase Is Associated With Poor Prognosis and Altered Immune Signature in Uterine Corpus Endometrial Carcinoma. Front Genet 2022; 12:764194. [PMID: 35082830 PMCID: PMC8785095 DOI: 10.3389/fgene.2021.764194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 12/21/2021] [Indexed: 01/21/2023] Open
Abstract
Uterine corpus endometrial carcinoma (UCEC) is a common malignant tumor of the female reproductive system with poor prognosis in advanced, recurrent, and metastatic cases. Identification of reliable molecular markers will help in the development of clinical strategies for early detection, diagnosis, and intervention. Gamma-glutamyl hydrolase (GGH) is a key enzyme in folate metabolism pathway. High expression of GGH is associated with severe clinicopathological features and poor prognosis of several cancers. High GGH expression is also related to cell resistance to antifolate drugs such as methotrexate. In this study we focused on the prognostic value of immunohistochemical GGH expression level in UCEC tissue and RNA-seq data from The Cancer Genome Atlas to establish associations with clinical features and outcomes. Further, we conducted comprehensive bioinformatics analyses to identify and functionally annotate differentially expressed genes (DEGs) associated with UCEC upregulation and assessed the effects of upregulation on immune infiltration. Both GGH mRNA and protein expression levels were elevated in tumor tissues, and higher expression was significantly associated with advanced clinicopathological features and poor prognosis by univariate analysis. Further multivariate analysis identified elevated GGH expression as an independent risk factor for poor outcome. Nomograms including GGH expression yielded a c-index for disease-specific survival prediction of 0.884 (95% confidence interval: 0.861–0.907). A total of 520 DEGs (111 upregulated and 409 downregulated) were identified between high and low GGH expression groups. Analysis using Gene ontology, Kyoto Encyclopedia of Genes and Genomes pathway, Gene set enrichment analysis, and protein‒protein interaction indicated significant associations of altered GGH expression with cell proliferation, immune response, and the occurrence and development of UCEC tumors. Finally, GGH expression level was associated with high Th2 cell and low natural killer CD56bright cell infiltration. Collectively, these findings indicate that GGH drives UCEC progression and could be a useful biomarker for survival prediction as well as a therapeutic target.
Collapse
Affiliation(s)
- Cong Yu
- School of Life Sciences, Qilu Normal University, Jinan, China
| | - Haining Qi
- Department of Obstetrics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yanhui Zhang
- Maternal and Child Health Care Hospital of Shandong Province, Shandong University, Jinan, China
| | - Wen Zhao
- College of Life Sciences, Shandong Normal University, Jinan, China
| | - Guoying Wu
- School of Life Sciences, Qilu Normal University, Jinan, China
| |
Collapse
|
46
|
Ding Y, Yan Y, Dong Y, Xu J, Su W, Shi W, Zou Q, Yang X. NLRP3 promotes immune escape by regulating immune checkpoints: A pan-cancer analysis. Int Immunopharmacol 2022; 104:108512. [PMID: 35026655 DOI: 10.1016/j.intimp.2021.108512] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 12/21/2021] [Accepted: 12/29/2021] [Indexed: 11/30/2022]
Abstract
NLRP3 plays a pathogenic role in tumorigenesis by regulating innate and acquired immunity, apoptosis, differentiation, and intestinal microbes in tumors. Our research aimed to investigate the role of NLRP3 in pan-cancers based on multi-omics data in the TCGA database. Most types of tumors showed increased expression of NLRP3. Among them, the overexpressed NLRP3 in liver hepatocellular carcinoma (LIHC) and ovarian cancer (OV) indicated worse overall survival (OS). Further analysis also confirmed overexpressed NLRP3 in colon cancer (COAD) indicated a high probability of microsatellite instability (MSI) and low tumor mutational burden (TMB), which indicated a better response to immune checkpoint inhibitors (ICIs). Interestingly, overexpression of NLRP3 was closely related to high infiltration of immune cells (T cells, B cells, etc.) and overexpressed immune checkpoints (PD-1, PD-L1, LAG3, etc.). These results demonstrated NLRP3 promoted immune escape in cancers. Finally, we investigated the expression of various immune checkpoints by treating NLRP3 inhibitor MCC950 during the co-culture of peripheral blood mononuclear cells (PBMC) and LIHC cell line Hep3B. MCC950 significantly repressed the expression of PD-L1 and LAG3, and promoted the apoptosis rate of Hep3B. In conclusion, our research demonstrated the role of NLRP3 in pan-cancer, especially in LIHC. Inhibition of NLRP3 promoted the killing effect of T cells to cancer cells by repressing the expression of immune checkpoints.
Collapse
Affiliation(s)
- Yue Ding
- Department of General Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Yilin Yan
- Hangzhou Children's Welfare Institute, China
| | - Yihui Dong
- Department of Paediatrics, Qingdao Eighth People's Hospital, Qingdao 266100, China
| | - Jingyuan Xu
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Wei Su
- Department of Oncology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Weijun Shi
- Department of General Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China.
| | - Qi Zou
- Department of General Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China.
| | - Xiaoping Yang
- Department of Hepatobiliary Pancreatic Surgery, the First Hospital of Ningbo City, Ningbo 315010, China.
| |
Collapse
|
47
|
Lou T, Liu C, Qu H, Zhang Z, Wang S, Zhuang H. FOXA1 can be modulated by HDAC3 in the progression of epithelial ovarian carcinoma. J Transl Med 2022; 20:19. [PMID: 34991620 PMCID: PMC8740004 DOI: 10.1186/s12967-021-03224-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 12/29/2021] [Indexed: 12/24/2022] Open
Abstract
FOXA1 is associated with malignant tumors, but the function of FOXA1 in EOC is unclear. HDAC3 can influence the proliferation, migration and invasion ability of EOC. In this study, we wanted to explore the function of FOXA1 in ovarian cancer and the relationship between HDAC3 and FOXA1.The expression of HDAC3 and FOXA1 was detected by immunohistochemical staining of primary lesions from 127 epithelial ovarian carcinoma patients. A proliferation assay, a Transwell assay, an apoptosis assay and animal experiments were used to assess the proliferation, invasion and apoptosis abilities of ovarian cancer cells before and after transfection with FOXA1. The relevance of the in vitro findings was confirmed in xenografts. The H-scores for FOXA1 and HDAC3 staining in FIGO stage III-IV were noticeably higher and predicted adverse clinical outcomes in patients with ovarian cancer. The expression level of HDAC3 was significantly correlated with the expression level of FOXA1. Invasion, proliferation and apoptosis capacity and tumor formation were decreased in the FOXA1-knockdown cells. Experiments in xenografts confirmed that HDAC3 mediated tumor formation. In conclusion, FOXA1 can be modulated by HDAC3 through the Wnt/β-catenin signaling pathway, and FOXA1 plays essential roles in the proliferation, apoptosis and invasion of EOC cell lines and xenograft experiments.
Collapse
Affiliation(s)
- Tong Lou
- Department of Obstetrics and Gynecology, Beijing Chaoyang Hospital, Capital Medical University, No.8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020, China
| | - Chongdong Liu
- Department of Obstetrics and Gynecology, Beijing Chaoyang Hospital, Capital Medical University, No.8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020, China
| | - Hong Qu
- Department of Obstetrics and Gynecology, Beijing Chaoyang Hospital, Capital Medical University, No.8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020, China
| | - Zhiqiang Zhang
- Department of Obstetrics and Gynecology, Beijing Chaoyang Hospital, Capital Medical University, No.8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020, China
| | - Shuzhen Wang
- Department of Obstetrics and Gynecology, Beijing Chaoyang Hospital, Capital Medical University, No.8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020, China
| | - Huiyu Zhuang
- Department of Obstetrics and Gynecology, Beijing Chaoyang Hospital, Capital Medical University, No.8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020, China.
| |
Collapse
|
48
|
Zhao P, Yun Q, Li A, Li R, Yan Y, Wang Y, Sun H, Damirin A. LPA3 is a precise therapeutic target and potential biomarker for ovarian cancer. Med Oncol 2022; 39:17. [PMID: 34982278 DOI: 10.1007/s12032-021-01616-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 11/19/2021] [Indexed: 12/15/2022]
Abstract
Current studies have demonstrated that significant increased LPA levels to be observed in ascites in patients with ovarian cancer. Although several studies have shown that Lysophosphatidic acid (LPA) related to the progression of ovarian cancer, which LPA receptors (LPARs) and G-coupled protein subtypes mediated in LPA actions have not been clearly elucidated. This study aimed to clarify the roles of LPA and it is subtype-specific LPARs mediating mechanisms in ovarian cancer integrated using bioinformatic analysis and biological experimental approaches. The big data analysis shown that LPA3 was the only differentially expressed LPA receptor among the six LPARs in ovarian cancer and further verified in immunohistochemistry of tissue microarrays. Also found that LPA3 was also highly expressed in ovarian cancer tissue and ovarian cancer cells. Importantly, LPA significantly promoted the proliferation and migration of LPA3-overexpressing ovarian cancer cells, while the LPA-induced actions blocked by Ki16425, a LPAR1/3 antagonist treated, and LPA3-shRNA transfected. In vivo study indicated that the LPA3-overexpressing cell-derived tumors metastasis, tumors volume, and tumors mass were apparently increased in xenografted nude mice. In addition, we also observed that LPA3 was differential high expression in ovarian cancer tissue of the patients. Our studies further confirmed the LPA3/Gi/MAPKs/NF-κB signals were involved in LPA-induced oncogenic actions in ovarian cancer cells. Our findings indicated that the LPA3 might be a novel precise therapeutic target and potential biomarker for ovarian cancer.
Collapse
Affiliation(s)
- Pengfei Zhao
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Qingru Yun
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Aodungerile Li
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Rong Li
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Yali Yan
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Yuewu Wang
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Hongju Sun
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China.
| | - Alatangaole Damirin
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China.
| |
Collapse
|
49
|
Breast carcinomas with osteoclast-like giant cells: a comprehensive clinico-pathological and molecular portrait and evidence of RANK-L expression. Mod Pathol 2022; 35:1624-1635. [PMID: 35697931 PMCID: PMC9596373 DOI: 10.1038/s41379-022-01112-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/08/2022] [Accepted: 05/12/2022] [Indexed: 11/28/2022]
Abstract
Breast carcinomas (BC) with osteoclast-like giant cells (OGC) are rare. Despite their distinct stromal features, their molecular characteristics remain unknown. Here, we report comprehensive clinico-pathological and molecular findings for 27 patients diagnosed with BC-OGC at Institut Curie between 2000 and 2021. Seventeen (63%) cases were invasive carcinomas of no special type (IC NST) with OGC (OGC-IC NST), four (15%) were mixed or multifocal cases with and without OGC (OGC-Mixed), and six (22%) were metaplastic carcinomas with OGC (OGC-MC). All OGC-IC NST and OGC-Mixed cases were ER+ HER2- tumors (most being luminal A based on transcriptomic subtyping, when available), while all OGC-MC were triple-negative. The median age at diagnosis was 46, 45 and 62 years for OGC-IC NST, OGC-Mixed and OGC-MC, respectively. Three patients developed distant metastases (one OGC-IC NST, two OGC-Mixed), one of whom died of metastatic disease (OGC-Mixed), and one other patient died of locally advanced disease (OGC-MC). Histopathological evaluation comparing 13 OGC-IC NST and 19 control IC NST without OGC confirmed that OGC-IC NST showed significantly higher density of vessels (by CD34 immunohistochemistry (IHC)), iron deposits (Perls stain), and CD68 and CD163-positive cell infiltrates. Genomic findings for nine OGC-IC NST and four OGC-MC were consistent with the underlying histologic subtype, including activating alterations of the PI3K/AKT/mTOR pathway in 7/13 cases. Using RNA-seq data, differential gene expression analysis between OGC-IC NST (n = 7) and control IC NST without OGC (n = 7) revealed significant overexpression of TNFSF11 (RANK-L), TNFRSF11A (RANK), CSF1 (M-CSF), CSF1R, and genes encoding osteoclastic enzymes (MMP9, ACP5, CTSK, CTSB) in OGC-IC NST, while OPG (osteoprotegerin) was underexpressed. We also confirmed for the first time RANK-L expression in BC with OGC by IHC (seen in 15 out of 16 cases, and only in 2 of 16 controls without OGC). These findings could offer a rationale for further investigating RANK-L as a therapeutic target in BC with OGC.
Collapse
|
50
|
Xu L, Xu Y, Zhu Z, Gu H, Chen C, Chen J. Tanshinone IIA attenuates renal injury during hypothermic preservation via the MEK/ERK1/2/GSK-3β pathway. BMC Complement Med Ther 2021; 21:257. [PMID: 34625061 PMCID: PMC8501657 DOI: 10.1186/s12906-021-03427-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 09/29/2021] [Indexed: 12/25/2022] Open
Abstract
Background Oxidative stress-induced injury during hypothermic preservation is a universal problem that delays graft function and decrease the success of organ transplantation. Tanshinone IIA (Tan IIA) was reported to exhibit a variety of biochemical activities, including protection against oxidative stress. Therefore, the specific molecular pathway by which Tan IIA protects renal tissues during preservation was investigated in this study. Methods In vivo study, Sprague-Dawley (SD) rats were divided into twelve groups and the kidneys were isolated and preserved in different solutions for 0, 24 or 48 h, respectively: control group (Celsior solution) and Tan II groups (Celsior solution containing 10, 50,100 μM). In vitro study, primary renal cell from SD rats was cultured which was treated H2O2 (800 μM) for 6 h to mimic oxidative stress injury. Four groups were finally divided: control group; H2O2 group; H2O2 + Tan IIA group; H2O2 + Tan IIA + G15 group. Results In present study, we demonstrate data indicating that a significant increase in the superoxide dismutase (SOD) activity and a decrease in the reactive oxygen species (ROS) content were observed in the kidneys and renal cells preserved with Tan IIA compared with those preserved with the Celsior solution alone after 24 h and 48 h of hypothermic preservation (P < 0.01). The expression of phosphorylated mitogen-activated protein kinase kinase (MEK), phosphorylated extracellular signal-regulated protein kinases 1/2 (ERK1/2), phosphorylated glycogen synthase kinase-3β (GSK-3β) and cleaved caspase-3 was lower in the kidneys and renal cells preserved with Tan IIA than in those preserved with the Celsior solution alone after 24 h and 48 h of hypothermic preservation (P < 0.01). The mitochondrial morphology was rescued and adenosine triphophate (ATP) production and mitochondrial membrane potential were increased in the Tan IIA groups. Finally, Tan IIA also decreased cell apoptosis. Conclusion It suggests that the supplementation of the standard Celsior solution with Tan IIA may significantly improve long-term kidney preservation. Tan IIA attenuated oxidative stress injury and decreased apoptosis levels via activation of the MEK/ERK1/2/GSK-3β signaling pathway during kidney hypothermic preservation. Supplementary Information The online version contains supplementary material available at 10.1186/s12906-021-03427-7.
Collapse
Affiliation(s)
- Linhao Xu
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.,Translational Medicine Research Center, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.,School of Basic Medical Sciences and Forensic Medicine, Hangzhou medical college, No. 481 Binwen Road, Binjiang District, Hangzhou, 310053, Zhejiang, China
| | - Yizhou Xu
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Zhoujing Zhu
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, 310051, China
| | - Huiquan Gu
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou medical college, No. 481 Binwen Road, Binjiang District, Hangzhou, 310053, Zhejiang, China
| | - Chaofeng Chen
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Jian Chen
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou medical college, No. 481 Binwen Road, Binjiang District, Hangzhou, 310053, Zhejiang, China.
| |
Collapse
|