1
|
Kang YM, Kim HM, Lee J, Baek JS, Lee M, An HJ. Indole-3-carbinol alleviates allergic skin inflammation via periostin/thymic stromal lymphopoietin suppression in atopic dermatitis. Chin Med 2024; 19:177. [PMID: 39722037 DOI: 10.1186/s13020-024-01042-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 11/27/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Atopic dermatitis (AD) is a chronic multifactorial inflammatory skin disorder with a complex etiology. Despite its increasing prevalence, treatment of AD is still limited. Indole-3-carbinol (I3C) is found in cruciferous vegetables and is formed when these vegetables are cut, chewed, or cooked; it exerts diverse pharmacological activities. METHODS HaCaT keratinocytes stimulated with tumor necrosis factor-α and interferon-γ mixture and NC/Nga mice stimulated with 2,4-dinitrochlorobenzen (DNCB) were used for AD models, in vitro and in vivo, respectively. RESULTS The results showed that I3C reduced the expression of pro-inflammatory cytokines, thymic stromal lymphopoietin (TSLP), and periostin in in vitro model. Oral administration of I3C alleviated AD-like skin inflammatory symptoms, including serum IgE levels, epidermal thickening, inflammatory cell infiltration, transepidermal water loss, and scratching behavior. Moreover, I3C decreased the expression of TSLP and periostin and recovered the expression of skin barrier proteins by regulating Aryl Hydrocarbon Receptor and inhibiting the mitogen-activated protein kinase and nuclear factor-κB pathways in the skin of DNCB-induced AD mice. CONCLUSIONS I3C is suggested as a potential therapeutic alternative for the treatment of AD by repressing allergic inflammatory pathways.
Collapse
Affiliation(s)
- Yun-Mi Kang
- Department of Herbology, College of Korean Medicine, Sangji University, Wonju, Gangwon-Do, 26339, Republic of Korea
- Korean Medicine (KM)-Application Center, Korea Institute of Oriental Medicine (KIOM), 70 Cheomdan-Ro, Dong-Gu, Daegu, 41062, Republic of Korea
| | - Hye-Min Kim
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Herbology, College of Korean Medicine, Sangji University, Wonju, Gangwon-Do, 26339, Republic of Korea
| | - Junho Lee
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Jong-Suep Baek
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Minho Lee
- Department of Life Science, Dongguk University-Seoul, Ilsandong-Gu, Goyang-Si, 10326, Gyeonggi-do, Republic of Korea.
| | - Hyo-Jin An
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea.
- Department of Integrated Drug Development and Natural Products, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
2
|
Pallozzi M, De Gaetano V, Di Tommaso N, Cerrito L, Santopaolo F, Stella L, Gasbarrini A, Ponziani FR. Role of Gut Microbial Metabolites in the Pathogenesis of Primary Liver Cancers. Nutrients 2024; 16:2372. [PMID: 39064815 PMCID: PMC11280141 DOI: 10.3390/nu16142372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Hepatobiliary malignancies, which include hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), are the sixth most common cancers and the third leading cause of cancer-related death worldwide. Hepatic carcinogenesis is highly stimulated by chronic inflammation, defined as fibrosis deposition, and an aberrant imbalance between liver necrosis and nodular regeneration. In this context, the gut-liver axis and gut microbiota have demonstrated a critical role in the pathogenesis of HCC, as dysbiosis and altered intestinal permeability promote bacterial translocation, leading to chronic liver inflammation and tumorigenesis through several pathways. A few data exist on the role of the gut microbiota or bacteria resident in the biliary tract in the pathogenesis of CCA, and some microbial metabolites, such as choline and bile acids, seem to show an association. In this review, we analyze the impact of the gut microbiota and its metabolites on HCC and CCA development and the role of gut dysbiosis as a biomarker of hepatobiliary cancer risk and of response during anti-tumor therapy. We also discuss the future application of gut microbiota in hepatobiliary cancer management.
Collapse
Affiliation(s)
- Maria Pallozzi
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, 00168 Rome, Italy; (M.P.); (V.D.G.); (N.D.T.); (L.C.); (F.S.); (L.S.); (A.G.)
| | - Valeria De Gaetano
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, 00168 Rome, Italy; (M.P.); (V.D.G.); (N.D.T.); (L.C.); (F.S.); (L.S.); (A.G.)
| | - Natalia Di Tommaso
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, 00168 Rome, Italy; (M.P.); (V.D.G.); (N.D.T.); (L.C.); (F.S.); (L.S.); (A.G.)
| | - Lucia Cerrito
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, 00168 Rome, Italy; (M.P.); (V.D.G.); (N.D.T.); (L.C.); (F.S.); (L.S.); (A.G.)
| | - Francesco Santopaolo
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, 00168 Rome, Italy; (M.P.); (V.D.G.); (N.D.T.); (L.C.); (F.S.); (L.S.); (A.G.)
| | - Leonardo Stella
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, 00168 Rome, Italy; (M.P.); (V.D.G.); (N.D.T.); (L.C.); (F.S.); (L.S.); (A.G.)
| | - Antonio Gasbarrini
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, 00168 Rome, Italy; (M.P.); (V.D.G.); (N.D.T.); (L.C.); (F.S.); (L.S.); (A.G.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Francesca Romana Ponziani
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, 00168 Rome, Italy; (M.P.); (V.D.G.); (N.D.T.); (L.C.); (F.S.); (L.S.); (A.G.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
3
|
Wang Y, Wang B, Cao W, Xu X. PTX3 activates POSTN and promotes the progression of glioblastoma via the MAPK/ERK signalling axis. Biochem Biophys Res Commun 2024; 703:149665. [PMID: 38359612 DOI: 10.1016/j.bbrc.2024.149665] [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: 12/24/2023] [Revised: 02/02/2024] [Accepted: 02/08/2024] [Indexed: 02/17/2024]
Abstract
BACKGROUND Intrinsic brain tumours such as glioblastoma (GBM) are believed to develop from neuroglial stem or progenitor cells. GBM accounts for approximately half of gliomas. GBM has a poor prognosis and a low 5-year survival rate. Pentraxin 3 (PTX3) is overexpressed in GBM, but the potential mechanism is unclear. METHODS Glioblastoma data from the TCGA and CGGA databases were used to analyse PTX3 expression. Subsequently, in vivo and in vitro experiments were conducted to verify the effect of PTX3 silencing in glioma cells on EMT like process and GSC maintenance. The JASPAR database was used to predict the downstream genes of PTX3. POSTN is a novel target gene of PTX3 in gliomas, and this finding was validated using a luciferase reporter gene assay. Western blotting and KEGG enrichment analysis were used to predict the downstream pathway of POSTN, and it was found that the MAPK/ERK pathway might be related to the function of POSTN. RESULTS GBM tissues have higher levels of PTX3 expression than normal brain tissues (NBTs). In functional tests, PTX3 promoted the EMT like process of GBM cells while maintaining the stem cell characteristics of GBM stem cells and enhancing their self-renewal. Moreover, we performed a dual luciferase reporter experiment to confirm that PTX3 binds to the POSTN promoter region. In addition, the expression of key proteins in the MAPK/ERK signalling pathway was increased after PTX3 overexpression. CONCLUSION POSTN is a direct target of PTX3 that promotes GBM growth via the MAPK/ERK signalling pathway.
Collapse
Affiliation(s)
- Yuhang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu, China
| | - Binbin Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu, China
| | - Wenping Cao
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu, China.
| | - Xiupeng Xu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu, China.
| |
Collapse
|
4
|
Tian X, He Y, Qi L, Liu D, Zhou D, Liu Y, Gong W, Han Z, Xia Y, Li H, Wang J, Zhu K, Chen L, Guo H, Zhao Q. Autophagy Inhibition Contributes to Apoptosis of PLK4 Downregulation-induced Dormant Cells in Colorectal Cancer. Int J Biol Sci 2023; 19:2817-2834. [PMID: 37324947 PMCID: PMC10266079 DOI: 10.7150/ijbs.79949] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 05/23/2023] [Indexed: 06/17/2023] Open
Abstract
Dormant cancer cells account for cancer recurrence, distant metastasis and drug resistance which lead to poor prognosis in colorectal cancer (CRC). However, little is known about the molecular mechanisms regulating tumor cell dormancy and how to eliminate dormant cancer cells. Recent studies indicate autophagy affects dormant tumor cell survival. Here, we found that polo-like kinases 4 (PLK4), a central regulator of the cell cycle and proliferation, plays a crucial role in regulating CRC cells dormancy both in vitro and in vivo. Downregulation of PLK4 induced dormancy and inhibited migration and invasion in different CRC cell lines. Clinically, PLK4 expression was correlated with the dormancy markers (Ki67, p-ERK, p-p38) and late recurrence in CRC tissues. Mechanistically, downregulation of PLK4 induced autophagy contributed to restoring phenotypically aggressive tumor cells to a dormant state through the MAPK signaling pathway, and inhibition of autophagy would trigger apoptosis of dormant cells. Our findings reveal that downregulation of PLK4-induced autophagy contributes to tumor dormancy and autophagy inhibition leads to apoptosis of CRC dormant cells. Our study is the first to report that downregulation PLK4 induced autophagy is an early event in CRC dormancy and highlights autophagy inhibitor as a potential therapeutic target for dormant cell elimination.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Lu Chen
- ✉ Corresponding author: Qiang Zhao, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China, Tel.: +86 22 23537796, fax: +86 22 23537796, E-mail: ; Hua Guo, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China, Tel.: +86 22 23537796, fax: +86 22 23537796, E-mail: ; Lu Chen, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China, Tel.: +86 22 23537796, fax: +86 22 23537796, E-mail:
| | - Hua Guo
- ✉ Corresponding author: Qiang Zhao, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China, Tel.: +86 22 23537796, fax: +86 22 23537796, E-mail: ; Hua Guo, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China, Tel.: +86 22 23537796, fax: +86 22 23537796, E-mail: ; Lu Chen, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China, Tel.: +86 22 23537796, fax: +86 22 23537796, E-mail:
| | - Qiang Zhao
- ✉ Corresponding author: Qiang Zhao, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China, Tel.: +86 22 23537796, fax: +86 22 23537796, E-mail: ; Hua Guo, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China, Tel.: +86 22 23537796, fax: +86 22 23537796, E-mail: ; Lu Chen, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China, Tel.: +86 22 23537796, fax: +86 22 23537796, E-mail:
| |
Collapse
|
5
|
Chen L, He Y, Han Z, Gong W, Tian X, Guo L, Guo H, Song T, Chen L. The impact of decreased expression of SVEP1 on abnormal neovascularization and poor prognosis in patients with intrahepatic cholangiocarcinoma. Front Genet 2023; 13:1127753. [PMID: 36699464 PMCID: PMC9870246 DOI: 10.3389/fgene.2022.1127753] [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: 12/20/2022] [Accepted: 12/27/2022] [Indexed: 01/11/2023] Open
Abstract
Introduction: Intrahepatic cholangiocarcinoma (ICC) is one of the most highly heterogeneous malignant solid tumors; it is generally insensitive to clinical treatment and has a poor prognosis. Evidence suggests that abnormal neovascularization in the tumor microenvironment is an important cause of treatment resistance as well as recurrence and metastasis, but the key regulatory molecules are still largely unknown and should be identified. Method: We assessed the novel extracellular matrix protein (ECM) Sushi, von Willebrand factor type A, EGF and pentraxin containing 1 (SVEP1) expression pattern in the ICC by using immunohistochemistry. Multiplex immunofluorescence and Kaplan-Meier analysis were applied to explore the correlation between the low expression of SVEP1 and abnormal blood vessels and the clinical prognosis of ICC. Results: Our study showed that the expression of SVEP1 in most ICC samples was relatively lower than in the adjacent tissues. Statistical analysis suggested that patients with decreased SVEP1 expression always had shorter overall survival (OS) and disease-free survival (DFS). Moreover, the expression of SVEP1 was negatively correlated with the proportion of abnormal neovascularization in the tumor microenvironment of the ICC. Consistently, the key molecule of promoting vascular normalization, Ang-1, is positively correlated with the SVEP1 expression and prognosis in the ICC. In addition, the proportion of high Ki-67 expression was higher in the ICC samples with low SVEP1 expression, suggesting that the SVEP1 low expressed sample is in a malignant phenotype with high proliferation. Conclusion: This study reveals that SVEP1 is a promising prognostic biomarker for ICC and provides fresh insight into the role and potential new mechanism of abnormal neovascularization in ICC progression.
Collapse
Affiliation(s)
- Liwei Chen
- Department of Hepatobiliary Cancer, Liver Cancer Research Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China,Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Yuchao He
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Zhiqiang Han
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China,Department of Anesthesiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Wenchen Gong
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China,Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Xiangdong Tian
- Department of Endoscopy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Lin Guo
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China,Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Hua Guo
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Tianqiang Song
- Department of Hepatobiliary Cancer, Liver Cancer Research Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China,*Correspondence: Lu Chen, ; Tianqiang Song,
| | - Lu Chen
- Department of Hepatobiliary Cancer, Liver Cancer Research Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China,*Correspondence: Lu Chen, ; Tianqiang Song,
| |
Collapse
|
6
|
Wang Z, An J, Zhu D, Chen H, Lin A, Kang J, Liu W, Kang X. Periostin: an emerging activator of multiple signaling pathways. J Cell Commun Signal 2022; 16:515-530. [PMID: 35412260 PMCID: PMC9733775 DOI: 10.1007/s12079-022-00674-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/07/2022] [Indexed: 12/13/2022] Open
Abstract
Matricellular proteins are responsible for regulating the microenvironment, the behaviors of surrounding cells, and the homeostasis of tissues. Periostin (POSTN), a non-structural matricellular protein, can bind to many extracellular matrix proteins through its different domains. POSTN usually presents at low levels in most adult tissues but is highly expressed in pathological sites such as in tumors and inflamed organs. POSTN can bind to diverse integrins to interact with multiple signaling pathways within cells, which is one of its core biological functions. Increasing evidence shows that POSTN can activate the TGF-β, the PI3K/Akt, the Wnt, the RhoA/ROCK, the NF-κB, the MAPK and the JAK pathways to promote the occurrence and development of many diseases, especially cancer and inflammatory diseases. Furthermore, POSTN can interact with some pathways in an upstream and downstream relationship, forming complicated crosstalk. This article focuses on the interactions between POSTN and different signaling pathways in diverse diseases, attempting to explain the mechanisms of interaction and provide novel guidelines for the development of targeted therapies.
Collapse
Affiliation(s)
- Zhaoheng Wang
- grid.411294.b0000 0004 1798 9345Lanzhou University Second Hospital, 82, Cuiyingmen, Lanzhou, 730030 People’s Republic of China ,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730030 People’s Republic of China
| | - Jiangdong An
- grid.411294.b0000 0004 1798 9345Lanzhou University Second Hospital, 82, Cuiyingmen, Lanzhou, 730030 People’s Republic of China
| | - Daxue Zhu
- grid.411294.b0000 0004 1798 9345Lanzhou University Second Hospital, 82, Cuiyingmen, Lanzhou, 730030 People’s Republic of China ,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730030 People’s Republic of China
| | - Haiwei Chen
- grid.411294.b0000 0004 1798 9345Lanzhou University Second Hospital, 82, Cuiyingmen, Lanzhou, 730030 People’s Republic of China ,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730030 People’s Republic of China
| | - Aixin Lin
- grid.411294.b0000 0004 1798 9345Lanzhou University Second Hospital, 82, Cuiyingmen, Lanzhou, 730030 People’s Republic of China ,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730030 People’s Republic of China
| | - Jihe Kang
- grid.411294.b0000 0004 1798 9345Lanzhou University Second Hospital, 82, Cuiyingmen, Lanzhou, 730030 People’s Republic of China ,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730030 People’s Republic of China
| | - Wenzhao Liu
- grid.411294.b0000 0004 1798 9345Lanzhou University Second Hospital, 82, Cuiyingmen, Lanzhou, 730030 People’s Republic of China ,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730030 People’s Republic of China
| | - Xuewen Kang
- grid.411294.b0000 0004 1798 9345Lanzhou University Second Hospital, 82, Cuiyingmen, Lanzhou, 730030 People’s Republic of China ,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730030 People’s Republic of China
| |
Collapse
|
7
|
Chu H, Sun X, Wang J, Lei K, Shan Z, Zhao C, Ning Y, Gong R, Ren H, Cui Z. Synergistic effects of sodium butyrate and cisplatin against cervical carcinoma in vitro and in vivo. Front Oncol 2022; 12:999667. [PMID: 36338704 PMCID: PMC9633845 DOI: 10.3389/fonc.2022.999667] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/07/2022] [Indexed: 08/25/2023] Open
Abstract
BACKGROUNDS Cisplatin-based chemotherapy has been considered as the pivotal option for treating cervical cancer. However, some patients may present a poor prognosis due to resistance to chemotherapy. As a metabolite of natural products, sodium butyrate (NaB) could inhibit the proliferation of several malignant cells, but little is known about its combination with cisplatin in the treatment of cervical cancer. MATERIALS AND METHODS Flow cytometry, CCK-8 assay, and Transwell assay were utilized to analyze the cellular apoptosis, viability, cellular migration and invasion upon treating with NaB and/or cisplatin. The allograft mice model was established, followed by evaluating the tumor volume and necrotic area in mice treated with NaB and/or cisplatin. Western blot was performed for detecting protein expression involved in epithelial-mesenchymal transition (EMT) and the expression of MMPs. Immunohistochemical staining was conducted with the tumor sections. The transcription, expression, and cellular translocation of β-catenin were determined using luciferase reporter gene assay, Real-Time PCR, Western blot, and confocal laser scanning microscope, respectively. RESULTS NaB combined with cisplatin inhibited cell viability by promoting apoptosis of cervical cancer cells. In vivo experiments indicated that NaB combined with cisplatin could inhibit tumor growth and induce cancer cell necrosis. Single application of NaB activated the Wnt signaling pathway and induced partial EMT. NaB alone up-regulated MMP2, MMP7 and MMP9 expression, and promoted the migration and invasion of cervical cancer cells. The combination of cisplatin and NaB inhibited cellular migration and invasion by abrogating the nuclear transition of β-catenin, reverse EMT and down-regulate MMP2, MMP7 and MMP9. Immunohistochemical staining indicated that NaB combined with cisplatin up-regulated the expression of E-cadherin and reverse the EMT phenotype in the mice model. CONCLUSIONS NaB serves as a sensitizer for cisplatin, which may be a promising treatment regimen for cervical cancer when combined both. NaB alone should be utilized with caution for treating cervical cancer as it may promote the invasion and migration of cervical cancer cells.
Collapse
Affiliation(s)
- Huijun Chu
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaoyuan Sun
- Center of Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jia Wang
- Center of Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ke Lei
- Center of Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhengyi Shan
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chenyang Zhao
- Center of Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ying Ning
- Graduate School, Medical College of Qingdao University, Qingdao, China
| | - Ruining Gong
- Center of Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - He Ren
- Center for Gastrointestinal (GI) Cancer Diagnosis and Treatment, Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhumei Cui
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
8
|
Wu Y, Rong X, Pan M, Wang T, Yang H, Chen X, Xiao Z, Zhao C. Integrated Analysis Reveals the Gut Microbial Metabolite TMAO Promotes Inflammatory Hepatocellular Carcinoma by Upregulating POSTN. Front Cell Dev Biol 2022; 10:840171. [PMID: 35676936 PMCID: PMC9167932 DOI: 10.3389/fcell.2022.840171] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/12/2022] [Indexed: 12/12/2022] Open
Abstract
Liver cancer has a high mortality rate. Chronic inflammation is one of the leading causes of hepatocellular carcinoma. Recent studies suggested high levels of trimethylamine N-oxide (TMAO) may correlate with increased risk of inflammatory-induced liver cancer. However, the mechanisms by which TMAO promotes liver cancer remain elusive. Here, we established a model of inflammatory-induced liver cancer by treating Hepa1-6 cells and Huh7 cells with TNF-α. TMAO synergistically increased the proliferation, migration and invasion of Hepa1-6 cells and Huh7 cells in the presence of TNF-α. We conducted bulk RNA-Seq of the TMAO-treated cell model of inflammatory Hepatocellular carcinoma (HCC) and evaluated the influence of the differentially expressed genes (DEGs) on clinical prognosis using Kaplan-Meier Plotter Database and Gene Expression Profiling Interactive Analysis (GEPIA) database. Univariate and multivariate Cox regression analyses of tumor microenvironment and DEGs were performed using Timer2.0. Upregulation of POSTN, LAYN and HTRA3 and downregulation of AANAT and AFM were positively related to poorer overall survival in human liver cancer. Moreover, higher expression of POSTN and HTRA3 positively correlated with infiltration of neutrophils, which can promote tumor progression. In vitro experiments showed TMAO activates ILK/AKT/mTOR signaling via POSTN, and knocking down POSTN significantly reduced ILK/AKT/mTOR signaling and the tumorigenicity of Hepa1-6 cells and Huh7 cells. Collectively, our results suggest the gut microbial metabolite TMAO and POSTN may represent potential therapeutic targets for liver cancer.
Collapse
Affiliation(s)
- Yonglin Wu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College and National Clinical Research Center for Aging and Medicine, Shanghai Medical College, Huashan Hospital, Fudan University, Shanghai, China
| | - Xingyu Rong
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College and National Clinical Research Center for Aging and Medicine, Shanghai Medical College, Huashan Hospital, Fudan University, Shanghai, China
| | - Miaomiao Pan
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College and National Clinical Research Center for Aging and Medicine, Shanghai Medical College, Huashan Hospital, Fudan University, Shanghai, China
| | - Tongyao Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College and National Clinical Research Center for Aging and Medicine, Shanghai Medical College, Huashan Hospital, Fudan University, Shanghai, China
| | - Hao Yang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College and National Clinical Research Center for Aging and Medicine, Shanghai Medical College, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiejiu Chen
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College and National Clinical Research Center for Aging and Medicine, Shanghai Medical College, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhenming Xiao
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College and National Clinical Research Center for Aging and Medicine, Shanghai Medical College, Huashan Hospital, Fudan University, Shanghai, China
| | - Chao Zhao
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College and National Clinical Research Center for Aging and Medicine, Shanghai Medical College, Huashan Hospital, Fudan University, Shanghai, China
- Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, Shanghai, China
- *Correspondence: Chao Zhao,
| |
Collapse
|
9
|
Xu C, Wang Z, Zhang L, Feng Y, Lv J, Wu Z, Yang R, Wu T, Li J, Zhou R, Tian Z, Bai J, Zhang H, Lan Y, Lv Z. Periostin promotes the proliferation and metastasis of osteosarcoma by increasing cell survival and activates the PI3K/Akt pathway. Cancer Cell Int 2022; 22:34. [PMID: 35057799 PMCID: PMC8780812 DOI: 10.1186/s12935-021-02441-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 12/30/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Silencing of the periostin gene (POSTN) can inhibit the biological process of several different cancers, and this inhibition may be related to down-regulation of PI3K/AKT signaling. However, the effect of POSTN on the progression, proliferation, and invasion of osteosarcoma (OS) remain unclear. METHODS We used the Gene Expression Omnibus (GEO) database to screen datasets on in situ OS and lung metastases to identify core genes and potential pathways. We used additional bioinformatics tools to identify protein-protein interactions (PPIs) and gene networks, and selected the top seven genes whose expression had the strongest correlations with other genes. RESULTS The results indicated that POSTN was a major hub gene. Subsequent analysis of gene expression profiles showed that POSTN was highly expressed in 262 cases with sarcoma and expression was closely related to poor prognosis. We also performed enrichment analysis to identify differentially expressed genes and used real-time PCR, western blotting, and immunohistochemistry analyses to measure POSTN expression in cells and tissues. Transfection of a POSTN-shRNA plasmid into cultured OS cells (Saos-2) effectively inhibited the proliferation, invasion, and migration of these cells. Taken together, our results suggest that POSTN may play a role in promoting the proliferation and metastasis of OS by activation of the PI3K/Akt signaling pathway. CONCLUSIONS Our results provide a preliminary characterization of the mechanism by which POSTN may regulate the migration and invasion of OS cells and also provide a theoretical basis for identifying biomarkers that have potential use for the diagnosis and treatment of OS.
Collapse
Affiliation(s)
- Chaojian Xu
- Shanxi Medical University, Taiyuan, China
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, No. 382 Wuyi Road, Taiyuan, 030000, Shanxi, China
| | - Ziyue Wang
- Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | | | - Yi Feng
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, No. 382 Wuyi Road, Taiyuan, 030000, Shanxi, China
| | - Jia Lv
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, No. 382 Wuyi Road, Taiyuan, 030000, Shanxi, China
| | - Zhuangzhuang Wu
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, No. 382 Wuyi Road, Taiyuan, 030000, Shanxi, China
| | - Rong Yang
- Shanxi Medical University, Taiyuan, China
| | - Taiyong Wu
- Shanxi Medical University, Taiyuan, China
| | - Jian Li
- Shanxi Medical University, Taiyuan, China
| | - Ruhao Zhou
- Shanxi Medical University, Taiyuan, China
| | - Zhi Tian
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, No. 382 Wuyi Road, Taiyuan, 030000, Shanxi, China
| | - Junjun Bai
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, No. 382 Wuyi Road, Taiyuan, 030000, Shanxi, China
| | - Huadong Zhang
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, No. 382 Wuyi Road, Taiyuan, 030000, Shanxi, China
| | - Yanping Lan
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, No. 382 Wuyi Road, Taiyuan, 030000, Shanxi, China
| | - Zhi Lv
- Shanxi Medical University, Taiyuan, China.
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, No. 382 Wuyi Road, Taiyuan, 030000, Shanxi, China.
| |
Collapse
|
10
|
Barros JS, Aguiar TFM, Costa SS, Rivas MP, Cypriano M, Toledo SRC, Novak EM, Odone V, Cristofani LM, Carraro DM, Werneck da Cunha I, Costa CML, Vianna-Morgante AM, Rosenberg C, Krepischi ACV. Copy Number Alterations in Hepatoblastoma: Literature Review and a Brazilian Cohort Analysis Highlight New Biological Pathways. Front Oncol 2021; 11:741526. [PMID: 34956867 PMCID: PMC8692715 DOI: 10.3389/fonc.2021.741526] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 11/10/2021] [Indexed: 12/19/2022] Open
Abstract
Hepatoblastoma (HB) is a rare embryonal tumor, although it is the most common pediatric liver cancer. The aim of this study was to provide an accurate cytogenomic profile of this type of cancer, for which information in cancer databases is lacking. We performed an extensive literature review of cytogenetic studies on HBs disclosing that the most frequent copy number alterations (CNAs) are gains of 1q, 2/2q, 8/8q, and 20; and losses at 1p and 4q. Furthermore, the CNA profile of a Brazilian cohort of 26 HBs was obtained by array-CGH; the most recurrent CNAs were the same as shown in the literature review. Importantly, HBs from female patients, high-risk stratification tumors, tumors who developed in older patients (> 3 years at diagnosis) or from patients with metastasis and/or deceased carried a higher diversity of chromosomal alterations, specifically chromosomal losses at 1p, 4, 11q and 18q. In addition, we distinguished three major CNA profiles: no detectable CNA, few CNAs and tumors with complex genomes. Tumors with simpler genomes exhibited a significant association with the epithelial fetal subtype of HBs; in contrast, the complex genome group included three cases with epithelial embryonal histology, as well as the only HB with HCC features. A significant association of complex HB genomes was observed with older patients who developed high-risk tumors, metastasis, and deceased. Moreover, two patients with HBs exhibiting complex genomes were born with congenital anomalies. Together, these findings suggest that a high load of CNAs, mainly chromosomal losses, particularly losses at 1p and 18, increases the tendency to HB aggressiveness. Additionally, we identified six hot-spot chromosome regions most frequently affected in the entire group: 1q31.3q42.3, 2q23.3q37.3, and 20p13p11.1 gains, besides a 5,3 Mb amplification at 2q24.2q24.3, and losses at 1p36.33p35.1, 4p14 and 4q21.22q25. An in-silico analysis using the genes mapped to these six regions revealed several enriched biological pathways such as ERK Signaling, MicroRNAs in Cancer, and the PI3K-Akt Signaling, in addition to the WNT Signaling pathway; further investigation is required to evaluate if disturbances of these pathways can contribute to HB tumorigenesis. The analyzed gene set was found to be associated with neoplasms, abnormalities of metabolism/homeostasis and liver morphology, as well as abnormal embryonic development and cytokine secretion. In conclusion, we have provided a comprehensive characterization of the spectrum of chromosomal alterations reported in HBs and identified specific genomic regions recurrently altered in a Brazilian HB group, pointing to new biological pathways, and relevant clinical associations.
Collapse
Affiliation(s)
- Juliana Sobral Barros
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | - Talita Ferreira Marques Aguiar
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil.,Department of Urology, New York University (NYU) Grossman School of Medicine, New York, NY, United States
| | - Silvia Souza Costa
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | - Maria Prates Rivas
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | - Monica Cypriano
- Department of Pediatrics, Institute of Pediatric Oncology, Support Group for Children and Adolescents with Cancer (IOP-GRAACC), Federal University of São Paulo, São Paulo, Brazil
| | - Silvia Regina Caminada Toledo
- Department of Pediatrics, Institute of Pediatric Oncology, Support Group for Children and Adolescents with Cancer (IOP-GRAACC), Federal University of São Paulo, São Paulo, Brazil
| | - Estela Maria Novak
- Department of Pediatrics, Institute of Childhood Cancer Treatment (ITACI), Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Vicente Odone
- Department of Pediatrics, Institute of Childhood Cancer Treatment (ITACI), Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Lilian Maria Cristofani
- Department of Pediatrics, Institute of Childhood Cancer Treatment (ITACI), Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Dirce Maria Carraro
- International Research Center, AC Camargo Cancer Center (ACCCC), São Paulo, Brazil
| | | | | | - Angela M Vianna-Morgante
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | - Carla Rosenberg
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | - Ana Cristina Victorino Krepischi
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
11
|
Zhang S, Li J, Gao H, Tong Y, Li P, Wang Y, Du L, Wang C. lncRNA Profiles Enable Prognosis Prediction and Subtyping for Esophageal Squamous Cell Carcinoma. Front Cell Dev Biol 2021; 9:656554. [PMID: 34127945 PMCID: PMC8196240 DOI: 10.3389/fcell.2021.656554] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 04/08/2021] [Indexed: 12/24/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) have emerged as useful prognostic markers in many tumors. In this study, we investigated the potential application of lncRNA markers for the prognostic prediction of esophageal squamous cell carcinoma (ESCC). We identified ESCC-associated lncRNAs by comparing ESCC tissues with normal tissues. Subsequently, Kaplan–Meier (KM) method in combination with the univariate Cox proportional hazards regression (UniCox) method was used to screen prognostic lncRNAs. By combining the differential and prognostic lncRNAs, we developed a prognostic model using cox stepwise regression analysis. The obtained prognostic prediction model could effectively predict the 3- and 5-year prognosis and survival of ESCC patients by time-dependent receiver operating characteristic (ROC) curves (area under curve = 0.87 and 0.89, respectively). Besides, a lncRNA-based classification of ESCC was generated using k-mean clustering method and we obtained two clusters of ESCC patients with association with race and Barrett’s esophagus (BE) (both P < 0.001). Finally, we found that lncRNA AC007128.1 was upregulated in both ESCC cells and tissues and associated with poor prognosis of ESCC patients. Furthermore, AC007128.1 could promote epithelial-mesenchymal transition (EMT) of ESCC cells by increasing the activation of MAPK/ERK and MAPK/p38 signaling pathways. Collectively, our findings indicated the potentials of lncRNA markers in the prognosis, molecular subtyping, and EMT of ESCC.
Collapse
Affiliation(s)
- Shujun Zhang
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Juan Li
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Huiru Gao
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yao Tong
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Peilong Li
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yunshan Wang
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lutao Du
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, China.,Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, China
| | - Chuanxin Wang
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, China.,Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, China
| |
Collapse
|
12
|
miR-876 Inhibits EMT and Liver Fibrosis via POSTN to Suppress Metastasis in Hepatocellular Carcinoma. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1964219. [PMID: 33083453 PMCID: PMC7559219 DOI: 10.1155/2020/1964219] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 07/19/2020] [Accepted: 08/14/2020] [Indexed: 02/06/2023]
Abstract
Background The asymptomatic onset, frequent recurrence, and poor prognosis of hepatocellular carcinoma (HCC) prompted us to identify new therapeutic targets or predictive markers of HCC diagnosis or prognosis. Methods In this study, bioinformatics analysis was used to screen for target miRNAs from the open-access TCGA database. Transwell assays, Western blotting, and qRT-PCR analyses were used to detect cellular functions and gene expression in HCC cells and samples. A nude mouse tumorigenesis model was established to facilitate the observation of HCC progression. Other assays included luciferase reporter assays, IHC, and survival analysis. Results We found that the identified miR-876 from TCGA was expressed at low levels in HCC cell lines and that low miR-876 expression was corrected with liver cirrhosis, tumor thrombus, and TNM stage. Further research revealed that miR-876 regulated cell invasion, EMT, and collagen expression by targeting POSTN expression. miR-876 and POSTN were inversely correlated in HCC samples and associated with EMT status and liver fibrosis in clinical HCC tissues. miR-876 inhibited the liver cancer progression in in vivo animal assays. Finally, both miR-876 and POSTN were risk factors for HCC survival, and HCC patients with combined low miR-876 and high POSTN expression had worse prognosis. Conclusions miR-876 inhibited HCC EMT and fibrosis by targeting POSTN, thus affecting HCC progression and prognosis. miR-876 and POSTN may be useful therapeutic targets or prognostic markers of HCC.
Collapse
|
13
|
Wang D, Zhang H, Fang X, Cao D, Liu H. Pan-cancer analysis reveals the role of long non-coding RNA LINC01614 as a highly cancer-dependent oncogene and biomarker. Oncol Lett 2020; 20:1383-1399. [PMID: 32724381 PMCID: PMC7377058 DOI: 10.3892/ol.2020.11648] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 04/16/2020] [Indexed: 12/24/2022] Open
Abstract
Long intergenic non-coding RNA 1614 (LINC01614) is highly expressed in several malignant tumor types, suggesting that it may act as an oncogene. However, the specific roles of LINC01614 in malignant tumors have remained elusive. To examine the expression pattern of LINC01614 in various malignancies, a comprehensive pan-cancer analysis was performed using public databases, including 53 normal tissue types and 32 cancer datasets with samples from 9,091 patients. The results were validated using reverse transcription-quantitative PCR analysis of tissue specimens from patients. LINC01614 expression was upregulated in most malignant tumors, thus demonstrating diagnostic potential. Furthermore, upregulation of LINC01614 was associated with poor overall survival in the majority of cases. However, the association with clinical outcome was highly cancer-dependent; LINC01614 appeared to be an oncogene and diagnostic/prognostic biomarker in cancers of the digestive, respiratory, nervous and endocrine systems, as well as breast and head and neck cancer, but not in the cancers of the reproductive system or some of the urinary system. High LINC01614 expression was also markedly associated with the epithelial-mesenchymal transition (EMT) and associated signaling pathways. Overall, the present results suggest that LINC01614 is an EMT-associated oncogene that influences the metastasis and prognosis of several cancers, thus highlighting its potential as a novel diagnostic and prognostic marker.
Collapse
Affiliation(s)
- Dingding Wang
- Department of Pathology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, P.R. China
| | - Hong Zhang
- Department of Pathology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, P.R. China
| | - Xiaolian Fang
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, P.R. China
| | - Dingfang Cao
- Department of Pathology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, P.R. China
| | - Honggang Liu
- Department of Pathology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, P.R. China
| |
Collapse
|
14
|
García P, Bizama C, Rosa L, Espinoza JA, Weber H, Cerda-Infante J, Sánchez M, Montecinos VP, Lorenzo-Bermejo J, Boekstegers F, Dávila-López M, Alfaro F, Leiva-Acevedo C, Parra Z, Romero D, Kato S, Leal P, Lagos M, Roa JC. Functional and genomic characterization of three novel cell lines derived from a metastatic gallbladder cancer tumor. Biol Res 2020; 53:13. [PMID: 32293552 PMCID: PMC7158131 DOI: 10.1186/s40659-020-00282-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 04/06/2020] [Indexed: 12/19/2022] Open
Abstract
Background Gallbladder cancer (GBC) is the most common tumor of the biliary tract. The incidence of GBC shows a large geographic variability, being particularly frequent in Native American populations. In Chile, GBC represents the second cause of cancer-related death among women. We describe here the establishment of three novel cell lines derived from the ascitic fluid of a Chilean GBC patient, who presented 46% European, 36% Mapuche, 12% Aymara and 6% African ancestry. Results After immunocytochemical staining of the primary cell culture, we isolated and comprehensively characterized three independent clones (PUC-GBC1, PUC-GBC2 and PUC-GBC3) by short tandem repeat DNA profiling and RNA sequencing as well as karyotype, doubling time, chemosensitivity, in vitro migration capability and in vivo tumorigenicity assay. Primary culture cells showed high expression of CK7, CK19, CA 19-9, MUC1 and MUC16, and negative expression of mesothelial markers. The three isolated clones displayed an epithelial phenotype and an abnormal structure and number of chromosomes. RNA sequencing confirmed the increased expression of cytokeratin and mucin genes, and also of TP53 and ERBB2 with some differences among the three cells lines, and revealed a novel exonic mutation in NF1. The PUC-GBC3 clone was the most aggressive according to histopathological features and the tumorigenic capacity in NSG mice. Conclusions The first cell lines established from a Chilean GBC patient represent a new model for studying GBC in patients of Native American descent.
Collapse
Affiliation(s)
- Patricia García
- Department of Pathology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carolina Bizama
- Department of Pathology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Lorena Rosa
- Department of Pathology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,Applied Molecular and Cellular Biology PhD Program, Universidad de La Frontera, Temuco, Chile
| | - Jaime A Espinoza
- Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Helga Weber
- Center of Excellence in Translational Medicine (CEMT) and Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco, Chile
| | - Javier Cerda-Infante
- Department of Hematology Oncology; Cellular and Molecular Biology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Marianela Sánchez
- Department of Hematology Oncology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Viviana P Montecinos
- Department of Hematology Oncology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Justo Lorenzo-Bermejo
- Statistical Genetics Research Group, Institute of Medical Biometry and Informatics, University of Heidelberg, Heidelberg, Germany
| | - Felix Boekstegers
- Statistical Genetics Research Group, Institute of Medical Biometry and Informatics, University of Heidelberg, Heidelberg, Germany
| | - Marcela Dávila-López
- Bioinformatics Core Facility, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Francisca Alfaro
- Department of Pathology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia Leiva-Acevedo
- Department of Pathology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Zasha Parra
- Cytogenetics Laboratory, Complejo Asistencial Dr. Sótero del Río, Santiago, Chile
| | - Diego Romero
- Department of Pathology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Sumie Kato
- Division of Obstetrics and Gynecology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pamela Leal
- Center of Excellence in Translational Medicine (CEMT) and Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco, Chile
| | - Marcela Lagos
- Department of Clinical Laboratory, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan Carlos Roa
- Department of Pathology, Faculty of Medicine, Millennium Institute of Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
15
|
Gerarduzzi C, Hartmann U, Leask A, Drobetsky E. The Matrix Revolution: Matricellular Proteins and Restructuring of the Cancer Microenvironment. Cancer Res 2020; 80:2705-2717. [PMID: 32193287 DOI: 10.1158/0008-5472.can-18-2098] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 12/04/2019] [Accepted: 03/17/2020] [Indexed: 11/16/2022]
Abstract
The extracellular matrix (ECM) surrounding cells is indispensable for regulating their behavior. The dynamics of ECM signaling are tightly controlled throughout growth and development. During tissue remodeling, matricellular proteins (MCP) are secreted into the ECM. These factors do not serve classical structural roles, but rather regulate matrix proteins and cell-matrix interactions to influence normal cellular functions. In the tumor microenvironment, it is becoming increasingly clear that aberrantly expressed MCPs can support multiple hallmarks of carcinogenesis by interacting with various cellular components that are coupled to an array of downstream signals. Moreover, MCPs also reorganize the biomechanical properties of the ECM to accommodate metastasis and tumor colonization. This realization is stimulating new research on MCPs as reliable and accessible biomarkers in cancer, as well as effective and selective therapeutic targets.
Collapse
Affiliation(s)
- Casimiro Gerarduzzi
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, Québec, Canada. .,Département de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Ursula Hartmann
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Andrew Leask
- College of Dentistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Elliot Drobetsky
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, Québec, Canada.,Département de Médecine, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
16
|
The Emerging Roles of Cancer Stem Cells and Wnt/Beta-Catenin Signaling in Hepatoblastoma. Cancers (Basel) 2019; 11:cancers11101406. [PMID: 31547062 PMCID: PMC6826653 DOI: 10.3390/cancers11101406] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 09/11/2019] [Accepted: 09/11/2019] [Indexed: 02/07/2023] Open
Abstract
Hepatoblastoma (HB) is the most common form of primary liver malignancy found in pediatric populations. HB is considered to be clonal and arises from hepatoblasts, or embryonic liver progenitor cells. These less differentiated tumor-initiating progenitor cells, or cancer stem cells (CSCs), may contribute to tumor recurrence and resistance to therapies, and have high metastatic abilities. Phenotypic heterogeneity, undesired genetic and epigenetic alterations, and dysregulated signaling pathways provide CSCs with a survival advantage over current therapies. The molecular and cellular basis of HB and the mechanism of CSC induction are not fully understood. The Wnt/beta-catenin pathway is one of the major developmental pathways and is believed to play an important role in the pathogenesis of HB and CSC formation. This review summarizes the cellular and molecular characteristics of HB with a specific emphasis on CSCs and Wnt/beta-catenin signaling.
Collapse
|