1
|
Li W, Liu J, Yu T, Lu F, Miao Q, Meng X, Xiao W, Yang H, Zhang X. ZDHHC9-mediated Bip/GRP78 S-palmitoylation inhibits unfolded protein response and promotes bladder cancer progression. Cancer Lett 2024; 598:217118. [PMID: 39002690 DOI: 10.1016/j.canlet.2024.217118] [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: 01/09/2024] [Revised: 07/07/2024] [Accepted: 07/09/2024] [Indexed: 07/15/2024]
Abstract
Recent studies have highlighted palmitoylation, a novel protein post-translational modification, as a key player in various signaling pathways that contribute to tumorigenesis and drug resistance. Despite this, its role in bladder cancer (BCa) development remains inadequately understood. In this study, ZDHHC9 emerged as a significantly upregulated oncogene in BCa. Functionally, ZDHHC9 knockdown markedly inhibited tumor proliferation, promoted tumor cell apoptosis, and enhanced the efficacy of gemcitabine (GEM) and cisplatin (CDDP). Mechanistically, SP1 was found to transcriptionally activate ZDHHC9 expression. ZDHHC9 subsequently bound to and palmitoylated the Bip protein at cysteine 420 (Cys420), thereby inhibiting the unfolded protein response (UPR). This palmitoylation at Cys420 enhanced Bip's protein stability and preserved its localization within the endoplasmic reticulum (ER). ZDHHC9 might become a novel therapeutic target for BCa and could also contribute to combination therapy with GEM and CDDP.
Collapse
Affiliation(s)
- Weiquan Li
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China
| | - Jingchong Liu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China
| | - Tiexi Yu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China
| | - Feiyi Lu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China
| | - Qi Miao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China
| | - Xiangui Meng
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China.
| | - Wen Xiao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China.
| | - Hongmei Yang
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Xiaoping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China.
| |
Collapse
|
2
|
Cagnin S, Pontisso P, Martini A. SerpinB3: A Multifaceted Player in Health and Disease-Review and Future Perspectives. Cancers (Basel) 2024; 16:2579. [PMID: 39061218 PMCID: PMC11274807 DOI: 10.3390/cancers16142579] [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: 06/05/2024] [Revised: 07/10/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
SerpinB3, a member of the serine-protease inhibitor family, has emerged as a crucial player in various physiological and pathological processes. Initially identified as an oncogenic factor in squamous cell carcinomas, SerpinB3's intricate involvement extends from fibrosis progression and cancer to cell protection in acute oxidative stress conditions. This review explores the multifaceted roles of SerpinB3, focusing on its implications in fibrosis, metabolic syndrome, carcinogenesis and immune system impairment. Furthermore, its involvement in tissue protection from oxidative stress and wound healing underscores its potential as diagnostic and therapeutic tool. Recent studies have described the therapeutic potential of targeting SerpinB3 through its upstream regulators, offering novel strategies for cancer treatment development. Overall, this review underscores the importance of further research to fully elucidate the mechanisms of action of SerpinB3 and to exploit its therapeutic potential across various medical conditions.
Collapse
Affiliation(s)
| | - Patrizia Pontisso
- Department of Medicine, University of Padova, 35123 Padova, Italy; (S.C.); (A.M.)
| | | |
Collapse
|
3
|
Martini A, Prasai K, Zemla TJ, Ahmed FY, Elnagar MB, Giama NH, Guzzardo V, Biasiolo A, Fassan M, Yin J, Pontisso P, Roberts LR. SerpinB3/4 Expression Is Associated with Poor Prognosis in Patients with Cholangiocarcinoma. Cancers (Basel) 2024; 16:225. [PMID: 38201652 PMCID: PMC10778206 DOI: 10.3390/cancers16010225] [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: 11/28/2023] [Revised: 12/30/2023] [Accepted: 01/01/2024] [Indexed: 01/12/2024] Open
Abstract
Cholangiocarcinoma (CCA), the second most common primary liver tumor, is associated with a dismal outcome, and useful prognostic markers are not currently available in clinical practice. SerpinB3, a serine protease inhibitor, was recently found to play a relevant role in malignant transformation in different cancers. The aim of the present study was to determine the expression of SerpinB3/4 in tissue and serum samples of patients with CCA in relation to clinical outcomes. SerpinB3/4 was assessed in the tissue microarrays (TMAs) of 123 surgically resected CCAs. ELISA assays were carried out in 188 patients with CCA to detect the free and IgM-linked forms of SerpinB3/4. Overall survival was analyzed in relation to SerpinB3/4 expression, and Cox models were used to identify the variables associated with survival. High levels of SerpinB3/4 (TMA score 2+/3+) were detected in 15 tumors (12.2%), characterized by a more advanced TNM stage (III/IV: 64.3% vs. 31.3%; p = 0.031) and lower overall patient survival, independently of CCA subclass (intrahepatic CCA: median 1.1 (0.8-Not Estimable, NE) vs. 2.4 (1.8-3.4) years; p = 0.0007; extrahepatic CCA: median 0.8 (0.2-NE) vs. 2.2 (1.5-5.4) years; p = 0.011). Vascular invasion (p = 0.027) and SerpinB3/4 scores (p = 0.0016) were independently associated with mortality in multivariate analysis. Patients who had detectable free or IgM-linked SerpinB3/4 in their serum showed poorer survival (1 vs. 2.4 years, p = 0.015, for free SerpinB3/4, and 1 vs. 2.6 years, p = 0.0026, for SerpinB3/4-IgM). In conclusion, high levels of SerpinB3/4 in tissue and serum in CCA are associated with poor outcomes after surgery, regardless of tumor subclass.
Collapse
Affiliation(s)
- Andrea Martini
- Department of Medicine, University of Padua, via Giustiniani 2, 35128 Padua, Italy; (A.M.); (V.G.); (A.B.); (M.F.)
| | - Kritika Prasai
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, 200 First Street SW, Rochester, MN 55905, USA; (K.P.); (F.Y.A.); (M.B.E.); (N.H.G.); (L.R.R.)
| | - Tyler J. Zemla
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic College of Medicine and Science, 200 First Street SW, Rochester, MN 55905, USA; (T.J.Z.); (J.Y.)
| | - Fowsiyo Y. Ahmed
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, 200 First Street SW, Rochester, MN 55905, USA; (K.P.); (F.Y.A.); (M.B.E.); (N.H.G.); (L.R.R.)
| | - Mamoun B. Elnagar
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, 200 First Street SW, Rochester, MN 55905, USA; (K.P.); (F.Y.A.); (M.B.E.); (N.H.G.); (L.R.R.)
| | - Nasra H. Giama
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, 200 First Street SW, Rochester, MN 55905, USA; (K.P.); (F.Y.A.); (M.B.E.); (N.H.G.); (L.R.R.)
| | - Vincenza Guzzardo
- Department of Medicine, University of Padua, via Giustiniani 2, 35128 Padua, Italy; (A.M.); (V.G.); (A.B.); (M.F.)
| | - Alessandra Biasiolo
- Department of Medicine, University of Padua, via Giustiniani 2, 35128 Padua, Italy; (A.M.); (V.G.); (A.B.); (M.F.)
| | - Matteo Fassan
- Department of Medicine, University of Padua, via Giustiniani 2, 35128 Padua, Italy; (A.M.); (V.G.); (A.B.); (M.F.)
- Veneto Institute of Oncology, (IOV-IRCCS), via Gattamelata 64, 35128 Padua, Italy
| | - Jun Yin
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic College of Medicine and Science, 200 First Street SW, Rochester, MN 55905, USA; (T.J.Z.); (J.Y.)
| | - Patrizia Pontisso
- Department of Medicine, University of Padua, via Giustiniani 2, 35128 Padua, Italy; (A.M.); (V.G.); (A.B.); (M.F.)
- European Reference Network—ERN RARE-LIVER, 72076 Tübingen, Germany
| | - Lewis R. Roberts
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, 200 First Street SW, Rochester, MN 55905, USA; (K.P.); (F.Y.A.); (M.B.E.); (N.H.G.); (L.R.R.)
| |
Collapse
|
4
|
Ohara Y, Tang W, Liu H, Yang S, Dorsey TH, Cawley H, Moreno P, Chari R, Guest MR, Azizian A, Gaedcke J, Ghadimi M, Hanna N, Ambs S, Hussain SP. SERPINB3-MYC axis induces the basal-like/squamous subtype and enhances disease progression in pancreatic cancer. Cell Rep 2023; 42:113434. [PMID: 37980563 PMCID: PMC10842852 DOI: 10.1016/j.celrep.2023.113434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 09/12/2023] [Accepted: 10/30/2023] [Indexed: 11/21/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) exhibits distinct molecular subtypes: classical/progenitor and basal-like/squamous. Our study aimed to identify genes contributing to the development of the basal-like/squamous subtype, known for its aggressiveness. Transcriptome analyses revealed consistent upregulation of SERPINB3 in basal-like/squamous PDAC, correlating with reduced patient survival. SERPINB3 transgene expression in PDAC cells enhanced in vitro invasion and promoted lung metastasis in a mouse PDAC xenograft model. Metabolome analyses unveiled a metabolic signature linked to both SERPINB3 and the basal-like/squamous subtype, characterized by heightened carnitine/acylcarnitine and amino acid metabolism, associated with poor prognosis in patients with PDAC and elevated cellular invasiveness. Further analysis uncovered that SERPINB3 inhibited the cysteine protease calpain, a key enzyme in the MYC degradation pathway, and drove basal-like/squamous subtype and associated metabolic reprogramming through MYC activation. Our findings indicate that the SERPINB3-MYC axis induces the basal-like/squamous subtype, proposing SERPINB3 as a potential diagnostic and therapeutic target for this variant.
Collapse
Affiliation(s)
- Yuuki Ohara
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Wei Tang
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Data Science & Artificial Intelligence, R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Huaitian Liu
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Shouhui Yang
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tiffany H Dorsey
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Helen Cawley
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Paloma Moreno
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Raj Chari
- Genome Modification Core, Laboratory Animal Sciences Program, Frederick National Lab for Cancer Research, Frederick, MD 21701, USA
| | - Mary R Guest
- Genome Modification Core, Laboratory Animal Sciences Program, Frederick National Lab for Cancer Research, Frederick, MD 21701, USA
| | - Azadeh Azizian
- Städtisches Klinikum Karlsruhe, Moltkestraße 90, 76133 Karlsruhe, Germany
| | - Jochen Gaedcke
- Städtisches Klinikum Karlsruhe, Moltkestraße 90, 76133 Karlsruhe, Germany
| | - Michael Ghadimi
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - Nader Hanna
- Division of General & Oncologic Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Stefan Ambs
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - S Perwez Hussain
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
5
|
Guyot B, Clément F, Drouet Y, Schmidt X, Lefort S, Delay E, Treilleux I, Foy JP, Jeanpierre S, Thomas E, Kielbassa J, Tonon L, Zhu HH, Saintigny P, Gao WQ, de la Fouchardiere A, Tirode F, Viari A, Blay JY, Maguer-Satta V. An Early Neoplasia Index (ENI10), Based on Molecular Identity of CD10 Cells and Associated Stemness Biomarkers, is a Predictor of Patient Outcome in Many Cancers. CANCER RESEARCH COMMUNICATIONS 2023; 3:1966-1980. [PMID: 37707389 PMCID: PMC10540743 DOI: 10.1158/2767-9764.crc-23-0196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/01/2023] [Accepted: 09/08/2023] [Indexed: 09/15/2023]
Abstract
An accurate estimate of patient survival at diagnosis is critical to plan efficient therapeutic options. A simple and multiapplication tool is needed to move forward the precision medicine era. Taking advantage of the broad and high CD10 expression in stem and cancers cells, we evaluated the molecular identity of aggressive cancer cells. We used epithelial primary cells and developed a breast cancer stem cell–based progressive model. The superiority of the early-transformed isolated molecular index was evaluated by large-scale analysis in solid cancers. BMP2-driven cell transformation increases CD10 expression which preserves stemness properties. Our model identified a unique set of 159 genes enriched in G2–M cell-cycle phases and spindle assembly complex. Using samples predisposed to transformation, we confirmed the value of an early neoplasia index associated to CD10 (ENI10) to discriminate premalignant status of a human tissue. Using a stratified Cox model, a large-scale analysis (>10,000 samples, The Cancer Genome Atlas Pan-Cancer) validated a strong risk gradient (HRs reaching HR = 5.15; 95% confidence interval: 4.00–6.64) for high ENI10 levels. Through different databases, Cox regression model analyses highlighted an association between ENI10 and poor progression-free intervals for more than 50% of cancer subtypes tested, and the potential of ENI10 to predict drug efficacy. The ENI10 index constitutes a robust tool to detect pretransformed tissues and identify high-risk patients at diagnosis. Owing to its biological link with refractory cancer stem cells, the ENI10 index constitutes a unique way of identifying effective treatments to improve clinical care. SIGNIFICANCE We identified a molecular signature called ENI10 which, owing to its biological link with stem cell properties, predicts patient outcome and drugs efficiency in breast and several other cancers. ENI10 should allow early and optimized clinical management of a broad number of cancers, regardless of the stage of tumor progression.
Collapse
Affiliation(s)
- Boris Guyot
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Department of Cancer Initiation and Tumor cell Identity, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Universite Claude Bernard Lyon 1, CRCL, Lyon, France
| | - Flora Clément
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Department of Cancer Initiation and Tumor cell Identity, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Universite Claude Bernard Lyon 1, CRCL, Lyon, France
| | | | - Xenia Schmidt
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Department of Cancer Initiation and Tumor cell Identity, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Universite Claude Bernard Lyon 1, CRCL, Lyon, France
| | - Sylvain Lefort
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Department of Cancer Initiation and Tumor cell Identity, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Universite Claude Bernard Lyon 1, CRCL, Lyon, France
| | - Emmanuel Delay
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Department of Cancer Initiation and Tumor cell Identity, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Universite Claude Bernard Lyon 1, CRCL, Lyon, France
- Centre Léon Bérard, Lyon, France
| | | | - Jean-Philippe Foy
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Department of Cancer Initiation and Tumor cell Identity, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Department of Tumor Escape Resistance and Immunity, CRCL, Lyon, France
| | - Sandrine Jeanpierre
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Department of Cancer Initiation and Tumor cell Identity, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Universite Claude Bernard Lyon 1, CRCL, Lyon, France
- Centre Léon Bérard, Lyon, France
| | - Emilie Thomas
- Bioinformatics Platform, Synergie Lyon Cancer Foundation, Lyon, France
| | - Janice Kielbassa
- Bioinformatics Platform, Synergie Lyon Cancer Foundation, Lyon, France
| | - Laurie Tonon
- Bioinformatics Platform, Synergie Lyon Cancer Foundation, Lyon, France
| | - Helen He Zhu
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Stem Cell Research Center, Shanghai Cancer Institute and Department of Urology, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Pierre Saintigny
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Department of Cancer Initiation and Tumor cell Identity, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Centre Léon Bérard, Lyon, France
- Department of Tumor Escape Resistance and Immunity, CRCL, Lyon, France
| | - Wei-Qiang Gao
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Stem Cell Research Center, Shanghai Cancer Institute and Department of Urology, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, P.R. China
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Arnaud de la Fouchardiere
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Department of Cancer Initiation and Tumor cell Identity, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Centre Léon Bérard, Lyon, France
- Department of Tumor Escape Resistance and Immunity, CRCL, Lyon, France
| | - Franck Tirode
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Department of Cancer Initiation and Tumor cell Identity, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Universite Claude Bernard Lyon 1, CRCL, Lyon, France
- Centre Léon Bérard, Lyon, France
| | - Alain Viari
- Bioinformatics Platform, Synergie Lyon Cancer Foundation, Lyon, France
| | - Jean-Yves Blay
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Department of Cancer Initiation and Tumor cell Identity, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Centre Léon Bérard, Lyon, France
- Department of Tumor Escape Resistance and Immunity, CRCL, Lyon, France
| | - Véronique Maguer-Satta
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Department of Cancer Initiation and Tumor cell Identity, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Universite Claude Bernard Lyon 1, CRCL, Lyon, France
- Centre Léon Bérard, Lyon, France
| |
Collapse
|
6
|
Gringeri E, Villano G, Brocco S, Polacco M, Calabrese F, Sacerdoti D, Cillo U, Pontisso P. SerpinB3 as hepatic marker of post-resective shear stress. Updates Surg 2023; 75:1541-1548. [PMID: 37204659 PMCID: PMC10435418 DOI: 10.1007/s13304-023-01531-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 05/10/2023] [Indexed: 05/20/2023]
Abstract
Post-resective liver failure is a frequent complication of liver surgery and it is due to portal hyperperfusion of the remnant liver and to arterial vasoconstriction, as buffer response of the hepatic artery. In this context, splenectomy allows a reduction of portal flow and increases the survival chance in preclinical models. SerpinB3 is over-expressed in the liver in oxidative stress conditions, as a mechanism of cell defense to provide survival by apoptosis inhibition and cell proliferation. In this study, the expression of SerpinB3 was assessed as predictor of liver damage in in vivo models of major hepatic resection with or without splenectomy. Wistar male rats were divided into 4 groups: group A received 30% hepatic resection, group B > 60% resection, group C > 60% resection with splenectomy and group D sham-operated. Before and after surgery liver function tests, echo Doppler ultrasound and gene expression were assessed. Transaminase values and ammonium were significantly higher in groups that underwent major hepatic resection. Echo Doppler ultrasound showed the highest portal flow and resistance of the hepatic artery in the group with > 60% hepatectomy without splenectomy, while the association of splenectomy determined no increase in portal flow and hepatic artery resistance. Only the group of rats without splenectomy showed higher shear-stress conditions, reflected by higher levels of HO-1, Nox1 and of Serpinb3, the latter associated with an increase of IL-6. In conclusion, splenectomy controls inflammation and oxidative damage, preventing the expression of Serpinb3. Therefore, SerpinB3 can be considered as a marker of post-resective shear stress.
Collapse
Affiliation(s)
- Enrico Gringeri
- Unit of Hepatobiliary Surgery and Liver Transplantation, University of Padova, Via Giustiniani 2, 35128 Padua, Italy
- Department of Surgical, Oncological and Gastroenterological Sciences-DISCOG, University of Padova, Via Giustiniani 2, 35128 Padua, Italy
| | - Gianmarco Villano
- Interdepartmental Center of Experimental Surgery, University of Padova, Via Giustiniani 2, 35128 Padua, Italy
- Department of Surgical, Oncological and Gastroenterological Sciences-DISCOG, University of Padova, Via Giustiniani 2, 35128 Padua, Italy
| | - Silvia Brocco
- Department of Medicine-DIMED, University of Padova, Via Giustiniani 2, 35128 Padua, Italy
| | - Marina Polacco
- Unit of Hepatobiliary Surgery and Liver Transplantation, University of Padova, Via Giustiniani 2, 35128 Padua, Italy
- Department of Surgical, Oncological and Gastroenterological Sciences-DISCOG, University of Padova, Via Giustiniani 2, 35128 Padua, Italy
| | - Fiorella Calabrese
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padua, Italy
| | - David Sacerdoti
- Department of Medicine-DIMED, University of Padova, Via Giustiniani 2, 35128 Padua, Italy
| | - Umberto Cillo
- Unit of Hepatobiliary Surgery and Liver Transplantation, University of Padova, Via Giustiniani 2, 35128 Padua, Italy
- Department of Surgical, Oncological and Gastroenterological Sciences-DISCOG, University of Padova, Via Giustiniani 2, 35128 Padua, Italy
| | - Patrizia Pontisso
- Department of Medicine-DIMED, University of Padova, Via Giustiniani 2, 35128 Padua, Italy
| |
Collapse
|
7
|
Lin X, Dong Y, Gu Y, Kapoor A, Peng J, Su Y, Wei F, Wang Y, Yang C, Gill A, Neira SV, Tang D. Taxifolin Inhibits Breast Cancer Growth by Facilitating CD8+ T Cell Infiltration and Inducing a Novel Set of Genes including Potential Tumor Suppressor Genes in 1q21.3. Cancers (Basel) 2023; 15:3203. [PMID: 37370814 DOI: 10.3390/cancers15123203] [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: 04/24/2023] [Revised: 05/27/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Taxifolin inhibits breast cancer (BC) via novel mechanisms. In a syngeneic mouse BC model, taxifolin suppressed 4T-1 cell-derived allografts. RNA-seq of 4T-1 tumors identified 36 differentially expressed genes (DEGs) upregulated by taxifolin. Among their human homologues, 19, 7, and 2 genes were downregulated in BCs, high-proliferative BCs, and BCs with high-fatality risks, respectively. Three genes were established as tumor suppressors and eight were novel to BC, including HNRN, KPRP, CRCT1, and FLG2. These four genes exhibit tumor suppressive actions and reside in 1q21.3, a locus amplified in 70% recurrent BCs, revealing a unique vulnerability of primary and recurrent BCs with 1q21.3 amplification with respect to taxifolin. Furthermore, the 36 DEGs formed a multiple gene panel (DEG36) that effectively stratified the fatality risk in luminal, HER2+, and triple-negative (TN) equivalent BCs in two large cohorts: the METABRIC and TCGA datasets. 4T-1 cells model human TNBC cells. The DEG36 most robustly predicted the poor prognosis of TNBCs and associated it with the infiltration of CD8+ T, NK, macrophages, and Th2 cells. Of note, taxifolin increased the CD8+ T cell content in 4T-1 tumors. The DEG36 is a novel and effective prognostic biomarker of BCs, particularly TNBCs, and can be used to assess the BC-associated immunosuppressive microenvironment.
Collapse
Affiliation(s)
- Xiaozeng Lin
- Department of Surgery, McMaster University, Hamilton, ON L8S 4K1, Canada
- Urological Cancer Center for Research and Innovation (UCCRI), St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada
- The Research Institute of St Joe's Hamilton, St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada
| | - Ying Dong
- Department of Surgery, McMaster University, Hamilton, ON L8S 4K1, Canada
- Urological Cancer Center for Research and Innovation (UCCRI), St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada
- The Research Institute of St Joe's Hamilton, St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada
| | - Yan Gu
- Department of Surgery, McMaster University, Hamilton, ON L8S 4K1, Canada
- Urological Cancer Center for Research and Innovation (UCCRI), St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada
- The Research Institute of St Joe's Hamilton, St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada
| | - Anil Kapoor
- Department of Surgery, McMaster University, Hamilton, ON L8S 4K1, Canada
- Urological Cancer Center for Research and Innovation (UCCRI), St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada
- The Research Institute of St Joe's Hamilton, St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada
| | - Jingyi Peng
- Department of Surgery, McMaster University, Hamilton, ON L8S 4K1, Canada
- Urological Cancer Center for Research and Innovation (UCCRI), St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada
- The Research Institute of St Joe's Hamilton, St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada
| | - Yingying Su
- Department of Surgery, McMaster University, Hamilton, ON L8S 4K1, Canada
- Urological Cancer Center for Research and Innovation (UCCRI), St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada
- The Research Institute of St Joe's Hamilton, St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada
| | - Fengxiang Wei
- The Genetics Laboratory, Longgang District Maternity and Child Healthcare Hospital of Shenzhen City, Shenzhen 518174, China
| | - Yanjun Wang
- Jilin Jianwei Songkou Biotechnology Co., Ltd., Changchun 510664, China
| | - Chengzhi Yang
- Benda International INC., Ottawa, ON K1X 0C1, Canada
| | - Armaan Gill
- Department of Surgery, McMaster University, Hamilton, ON L8S 4K1, Canada
- Urological Cancer Center for Research and Innovation (UCCRI), St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada
- The Research Institute of St Joe's Hamilton, St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada
| | - Sandra Vega Neira
- Department of Surgery, McMaster University, Hamilton, ON L8S 4K1, Canada
- Urological Cancer Center for Research and Innovation (UCCRI), St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada
- The Research Institute of St Joe's Hamilton, St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada
| | - Damu Tang
- Department of Surgery, McMaster University, Hamilton, ON L8S 4K1, Canada
- Urological Cancer Center for Research and Innovation (UCCRI), St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada
- The Research Institute of St Joe's Hamilton, St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada
| |
Collapse
|
8
|
Biasiolo A, Sandre M, Ferro S, Quarta S, Ruvoletto M, Villano G, Turato C, Guido M, Marin O, Pontisso P. Epitope-Specific Anti-SerpinB3 Antibodies for SerpinB3 Recognition and Biological Activity Inhibition. Biomolecules 2023; 13:biom13050739. [PMID: 37238609 DOI: 10.3390/biom13050739] [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: 02/21/2023] [Revised: 04/13/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
SerpinB3 is a serine protease inhibitor that plays a relevant role in disease progression and cancer by increasing fibrosis, cell proliferation, and invasion, besides conferring resistance to apoptosis. The mechanisms underlying these biological activities are not yet fully understood. The aim of this study was to generate antibodies directed against different SerpinB3 epitopes to better investigate their biological role. Five exposed epitopes were identified using the software DNASTAR Lasergene and the corresponding synthetic peptides were used for NZW rabbit immunization. Anti-P#2 and anti-P#4 antibodies were able to recognize both SerpinB3 and SerpinB4 by ELISA. Anti-P#5 antibody, produced against the reactive site loop of SerpinB3, showed the greatest specific reactivity for human SerpinB3. This antibody was able to recognize SerpinB3 at nuclear level, while anti-P#3 antibody recognized SerpinB3 only at cytoplasmic level, both by immunofluorescence and by immunohistochemistry. The biological activity of each antibody preparation was assessed in HepG2 cells overexpressing SerpinB3 and anti-P#5 antibody reduced proliferation by 12% cell and cell invasion by 75%, while trivial results were obtained with the other antibody preparations. These findings indicate that the reactive site loop of SerpinB3 is essential for the invasiveness features induced by this serpin and it could become a novel druggable target.
Collapse
Affiliation(s)
- Alessandra Biasiolo
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Michele Sandre
- Department of Biomedical Sciences, University of Padova, Via Colombo 3, 35131 Padova, Italy
| | - Stefania Ferro
- Department of Biomedical Sciences, University of Padova, Via Colombo 3, 35131 Padova, Italy
| | - Santina Quarta
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Mariagrazia Ruvoletto
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Gianmarco Villano
- Department of Surgical, Oncological and Gastroenterological Sciences, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Cristian Turato
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Maria Guido
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Oriano Marin
- Department of Biomedical Sciences, University of Padova, Via Colombo 3, 35131 Padova, Italy
| | - Patrizia Pontisso
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| |
Collapse
|
9
|
Chen L, Shi V, Wang S, Freeman R, Ruiz F, Jayachandran K, Zhang J, Cosper P, Sun L, Luke CJ, Spina C, Grigsby PW, Schwarz JK, Markovina S. SCCA1/SERPINB3 promotes suppressive immune environment via STAT-dependent chemokine production, blunting the therapy-induced T cell responses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.01.526675. [PMID: 36778224 PMCID: PMC9915608 DOI: 10.1101/2023.02.01.526675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Radiotherapy is a commonly used cancer treatment; however, patients with high serum squamous cell carcinoma antigen (SCCA1/SERPINB3) are associated with resistance and poor prognosis. Despite being a strong clinical biomarker, the modulation of SERPINB3 in tumor immunity is poorly understood. We investigated the microenvironment of SERPINB3 high tumors through RNAseq of primary cervix tumors and found that SERPINB3 was positively correlated with CXCL1/8, S100A8/A9 and myeloid cell infiltration. Induction of SERPINB3 in vitro resulted in increased CXCL1/8 and S100A8/A9 production, and supernatants from SERPINB3-expressing cultures attracted monocytes and MDSCs. In murine tumors, the orthologue mSerpinB3a promoted MDSC, TAM, and M2 macrophage infiltration contributing to an immunosuppressive phenotype, which was further augmented upon radiation. Radiation-enhanced T cell response was muted in SERPINB3 tumors, whereas Treg expansion was observed. A STAT-dependent mechanism was implicated, whereby inhibiting STAT signaling with ruxolitinib abrogated suppressive chemokine production. Patients with elevated pre-treatment serum SCCA and high pSTAT3 had increased intratumoral CD11b+ myeloid cell compared to patients with low SCCA and pSTAT3 cohort that had overall improved cancer specific survival after radiotherapy. These findings provide a preclinical rationale for targeting STAT signaling in tumors with high SERPINB3 to counteract the immunosuppressive microenvironment and improve response to radiation.
Collapse
|
10
|
Miñoza JMA, Rico JA, Zamora PRF, Bacolod M, Laubenbacher R, Dumancas GG, de Castro R. Biomarker Discovery for Meta-Classification of Melanoma Metastatic Progression Using Transfer Learning. Genes (Basel) 2022; 13:2303. [PMID: 36553569 PMCID: PMC9777873 DOI: 10.3390/genes13122303] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Melanoma is considered to be the most serious and aggressive type of skin cancer, and metastasis appears to be the most important factor in its prognosis. Herein, we developed a transfer learning-based biomarker discovery model that could aid in the diagnosis and prognosis of this disease. After applying it to the ensemble machine learning model, results revealed that the genes found were consistent with those found using other methodologies previously applied to the same TCGA (The Cancer Genome Atlas) data set. Further novel biomarkers were also found. Our ensemble model achieved an AUC of 0.9861, an accuracy of 91.05, and an F1 score of 90.60 using an independent validation data set. This study was able to identify potential genes for diagnostic classification (C7 and GRIK5) and diagnostic and prognostic biomarkers (S100A7, S100A7, KRT14, KRT17, KRT6B, KRTDAP, SERPINB4, TSHR, PVRL4, WFDC5, IL20RB) in melanoma. The results show the utility of a transfer learning approach for biomarker discovery in melanoma.
Collapse
Affiliation(s)
- Jose Marie Antonio Miñoza
- System Modeling and Simulation Laboratory, Department of Computer Science, University of the Philippines Diliman, Quezon City 1101, Philippines
| | - Jonathan Adam Rico
- Center for Informatics, University of San Agustin, Iloilo City 5000, Philippines
| | | | - Manny Bacolod
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY 10065, USA
| | | | - Gerard G. Dumancas
- Center for Informatics, University of San Agustin, Iloilo City 5000, Philippines
- Loyola Science Center, Department of Chemistry, The University of Scranton, Scranton, PA 18510, USA
| | - Romulo de Castro
- Center for Informatics, University of San Agustin, Iloilo City 5000, Philippines
- 3R Biosystems, Long Beach, CA 90840, USA
| |
Collapse
|
11
|
Turato C, Vairetti M, Cagna M, Biasiolo A, Ferrigno A, Quarta S, Ruvoletto M, De Siervi S, Pontisso P, Di Pasqua LG. SerpinB3 administration protects liver against ischemia-reperfusion injury. Eur J Histochem 2022; 66. [DOI: 10.4081/ejh.2022.3561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 10/12/2022] [Indexed: 11/07/2022] Open
Abstract
We have investigated the change in SerpinB3 during hepatic ischemia and the potential role of its antiprotease activity in cell protection by the administration of wild-type SerpinB3 (SerpinB3-WT) or active loop-deleted recombinant SerpinB3 protein (SerpinB3-D) in a rat model of ischemia (60 min)/reperfusion (60 min) (I/R). A time-dependent increase of SerpinB3, both at transcription and protein level, was found in ischemic livers after 60, 120 and 180 min. SerpinB3-WT decreased polymorphonuclear cell infiltration and serum enzymes and increased ATP when compared with I/R group. These events were not obtained using SerpinB3-D. No significant changes in both liver SerpinB3 mRNA and protein were found in all I/R groups considered. The present data show that the administration of SerpinB3-WT reduced the I/R injury and this effect appears to be dependent on its anti-protease activity.
Collapse
|
12
|
Lauko A, Volovetz J, Turaga SM, Bayik D, Silver DJ, Mitchell K, Mulkearns-Hubert EE, Watson DC, Desai K, Midha M, Hao J, McCortney K, Steffens A, Naik U, Ahluwalia MS, Bao S, Horbinski C, Yu JS, Lathia JD. SerpinB3 drives cancer stem cell survival in glioblastoma. Cell Rep 2022; 40:111348. [PMID: 36103817 PMCID: PMC9513382 DOI: 10.1016/j.celrep.2022.111348] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 06/22/2022] [Accepted: 08/22/2022] [Indexed: 12/11/2022] Open
Abstract
Despite therapeutic interventions for glioblastoma (GBM), cancer stem cells (CSCs) drive recurrence. The precise mechanisms underlying CSC resistance, namely inhibition of cell death, are unclear. We built on previous observations that the high cell surface expression of junctional adhesion molecule-A drives CSC maintenance and identified downstream signaling networks, including the cysteine protease inhibitor SerpinB3. Using genetic depletion approaches, we found that SerpinB3 is necessary for CSC maintenance, survival, and tumor growth, as well as CSC pathway activation. Knockdown of SerpinB3 also increased apoptosis and susceptibility to radiation therapy. SerpinB3 was essential to buffer cathepsin L-mediated cell death, which was enhanced with radiation. Finally, we found that SerpinB3 knockdown increased the efficacy of radiation in pre-clinical models. Taken together, our findings identify a GBM CSC-specific survival mechanism involving a cysteine protease inhibitor, SerpinB3, and provide a potential target to improve the efficacy of GBM therapies against therapeutically resistant CSCs.
Collapse
Affiliation(s)
- Adam Lauko
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA; Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44106, USA; Medical Scientist Training Program, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Josephine Volovetz
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA; Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44106, USA
| | - Soumya M Turaga
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA; Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, OH 44115, USA
| | - Defne Bayik
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA; Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| | - Daniel J Silver
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA; Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| | - Kelly Mitchell
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA; Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| | - Erin E Mulkearns-Hubert
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA; Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44106, USA
| | - Dionysios C Watson
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA; Case Comprehensive Cancer Center, Cleveland, OH 44106, USA; Division of Hematology/Oncology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Kiran Desai
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA; Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| | - Manav Midha
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA; Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| | - Jing Hao
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Kathleen McCortney
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Alicia Steffens
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Ulhas Naik
- Department of Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | - Shideng Bao
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44106, USA; Case Comprehensive Cancer Center, Cleveland, OH 44106, USA; Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Craig Horbinski
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA; Department of Neurosurgery, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jennifer S Yu
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44106, USA; Case Comprehensive Cancer Center, Cleveland, OH 44106, USA; Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA; Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, OH 44106, USA; Department of Radiation Oncology, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Justin D Lathia
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA; Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44106, USA; Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, OH 44115, USA; Case Comprehensive Cancer Center, Cleveland, OH 44106, USA; Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, OH 44106, USA.
| |
Collapse
|
13
|
Squamous Cell Carcinoma Antigen: Clinical Application and Research Status. Diagnostics (Basel) 2022; 12:diagnostics12051065. [PMID: 35626221 PMCID: PMC9139199 DOI: 10.3390/diagnostics12051065] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 04/21/2022] [Accepted: 04/22/2022] [Indexed: 02/04/2023] Open
Abstract
The squamous cell carcinoma antigen (SCCA) is a tumor marker that has gained increasing attention for its biological functions and significance in normal physiological and pathological processes. Not only SCCA but also circulating immune complexes of SCCA and immunoglobulin M (IgM) are involved in normal physiological and pathological processes, providing a background for numerous clinical studies aimed at assessing the potential role of SCCA, SCCA–IgM, and SCCA isoform complexes in clinical practice. Previous studies support the clinical value of SCCA as a tumor marker for either diagnosing squamous cancers or monitoring the response to radiotherapy or chemotherapy, tumor relapse, and treatment failure. However, these studies show contrasting results, making the diagnostic or prognostic value of SCCA controversial. To reduce clinical heterogeneity across studies and achieve a more accurate and reliable comparison of results, a standardized detection method, scoring system, and cutoff level need to be established. Moreover, despite the fact that performances of different methods are comparable, the dynamic observation of tumor marker kinetics should be conducted under the same method.
Collapse
|
14
|
Lin F, Liao C, Zhang J, Sun Y, Lu W, Bai Y, Liao Y, Li M, Qi Y, Chen Y. Hydrogen Sulfide Inhibits Bronchial Epithelial Cell Epithelial Mesenchymal Transition Through Regulating Endoplasm Reticulum Stress. Front Mol Biosci 2022; 9:828766. [PMID: 35495633 PMCID: PMC9039047 DOI: 10.3389/fmolb.2022.828766] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 03/14/2022] [Indexed: 02/02/2023] Open
Abstract
Epithelial mesenchymal transition (EMT) is a contributing factor in remodeling events of chronic obstructive pulmonary disease (COPD). Hydrogen sulfide (H2S) has been implicated in the pathogenesis of COPD, but the effect of H2S in regulating EMT and the underlying mechanisms is not clear. In this study, we assessed endoplasmic reticulum (ER) stress markers, EMT markers and associated signal molecules in rat lungs, bronchial epithelial cells, and human peripheral lung tissues to investigate the effect of H2S in regulating EMT and the underlying mechanisms. We found that EMT and ER stress occurred in lung epithelial cells, especially in the bronchial epithelial cells of smokers and COPD patients. In cigarette smoke (CS)-exposed rats, intraperitoneal injection of NaHS significantly alleviated CS-induced lung tissue damage, small airway fibrosis, ER stress, and EMT, while intraperitoneal injection of propargylglycine (cystathionine-gamma-lyase inhibitor) aggravated these effects induced by CS. In the nicotine-exposed 16HBE cells, an appropriate concentration of H2S donor not only inhibited nicotine-induced ER stress, but also inhibited nicotine-induced enhancement of cell migration ability and EMT. ER stress nonspecific inhibitors taurine and 4-phenyl butyric acid also inhibited nicotine-induced enhancement of cell migration ability and EMT. Both H2S and inositol-requiring enzyme 1 (IRE1) activation inhibitor 4μ8C inhibited nicotine-induced activation of IRE1, Smad2/3 and EMT. These results suggest that H2S inhibits CS- or nicotine-induced ER stress and EMT in bronchial epithelial cells and alleviates CS-induced lung tissue damage and small airway fibrosis. The IRE1 signal pathway and Smad2/3 may be responsible for the inhibitory effect of H2S.
Collapse
Affiliation(s)
- Fan Lin
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China
- Geriatric Medicine Center, Department of Pulmonary and Critical Care Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Chengcheng Liao
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Jinsheng Zhang
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, China
| | - Yun Sun
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Weiwei Lu
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, China
| | - Yu Bai
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Yixuan Liao
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Minxia Li
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Yongfen Qi
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, China
- *Correspondence: Yahong Chen, ; Yongfen Qi,
| | - Yahong Chen
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China
- *Correspondence: Yahong Chen, ; Yongfen Qi,
| |
Collapse
|
15
|
Rodríguez-Garzotto A, Iglesias-Docampo L, Díaz-García CV, Ruppen I, Ximénez-Embún P, Gómez C, Rodríguez-Peralto JL, de Frutos JO, Lopez-Martin JA, Grávalos C, Cortés-Funes H, Agulló-Ortuño MT. Topical heparin as an effective and safe treatment for patients with capecitabine-induced hand-foot syndrome: results of a phase IIA trial supported by proteomic profiling of skin biopsies. Ther Adv Med Oncol 2022; 14:17588359221086911. [PMID: 35356259 PMCID: PMC8958526 DOI: 10.1177/17588359221086911] [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: 09/20/2021] [Accepted: 02/23/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Hand-foot syndrome (HFS) is a common adverse reaction associated with capecitabine chemotherapy that significantly affects the quality of life of patients. This study evaluates the safety and effectiveness of a topical heparin (TH) treatment on the clinical manifestations and anatomopathological alterations of capecitabine-induced HFS. In addition, we performed proteome profiling of skin biopsies obtained from patients with HFS at baseline and after heparin treatment. Methods: Patients with grade ⩽ 2 HFS associated with capecitabine were included in this study. The primary end point was the effectiveness of TH in reducing HFS of any grade. Clinical improvement was evaluated by clinicians, and an improvement was perceived by patients who performed a weekly visual analog scale questionnaire. Secondary end points included a comparative histological analysis and protein expression in skin biopsies at baseline and after 3 weeks of HT treatment. Proteomic profiling was carried out using quantitative isobaric labelling and subsequently validated by a T-array. Results: Twenty-one patients were included in the study. The median TH treatment time was 7.6 weeks (range = 3.6–41.6 weeks), and the median response time was 3.01 weeks (95% CI = 2.15–3.97). At the end of treatment, 19 of 21 patients (90.48%) responded to treatment with a decrease in one or more grades of HFS. None of the patients experienced adverse effects related to TH usage, nor did they suspend chemotherapy treatment. The main findings observed in skin biopsies after treatment were a decrease in hyperkeratosis and lymphocytic infiltrates. The proteomic analysis showed altered expression of 34 proteins that were mainly related to wound healing, cell growth, and the immune response. Conclusion: Based on our results, topical heparin is an effective and safe treatment for clinical manifestations of HFS, probably due to the restauration of skin homeostasis after heparin treatment, as supported by our proteomics-derived data. Trial registration: EudraCT 2009-018171-13
Collapse
Affiliation(s)
- Analia Rodríguez-Garzotto
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
- Laboratory of Thoracic and Clinical-Translational Oncology, Instituto de Investigación Sanitaria Hospital 12 de Octubre (i + 12), Madrid, Spain
- Roche Farma España, Madrid, Spain
| | - Lara Iglesias-Docampo
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
- Laboratory of Thoracic and Clinical-Translational Oncology, Instituto de Investigación Sanitaria Hospital 12 de Octubre (i + 12), Madrid, Spain
- Lung Cancer Group, Clinical Research Program, CNIO- H12O, Madrid, Spain
| | - C. Vanesa Díaz-García
- Laboratory of Thoracic and Clinical-Translational Oncology, Instituto de Investigación Sanitaria Hospital 12 de Octubre (i + 12), Madrid, Spain
| | - Isabel Ruppen
- Proteomics Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Pilar Ximénez-Embún
- Proteomics Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Carlos Gómez
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | | | - Jose A. Lopez-Martin
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
- Laboratory of Thoracic and Clinical-Translational Oncology, Instituto de Investigación Sanitaria Hospital 12 de Octubre (i + 12), Madrid, Spain
| | - Cristina Grávalos
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Hernán Cortés-Funes
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - M. Teresa Agulló-Ortuño
- Laboratory of Thoracic and Clinical-Translational Oncology, Instituto de Investigación Sanitaria Hospital 12 de Octubre (i + 12), Avda de Córdoba, s/n, 28041 Madrid, Spain
- Lung Cancer Group, Clinical Research Program, CNIO- H12O, Madrid, Spain
- Biomedical Research Networking Centre: Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
- Department of Nursing, Physiotherapy and Occupational Therapy, Facultad de Fisioterapia y Enfermería, Universidad de Castilla-La Mancha (UCLM), Toledo, Spain
| |
Collapse
|
16
|
Jiang W, de Jong JM, van Hillegersberg R, Read M. Predicting Response to Neoadjuvant Therapy in Oesophageal Adenocarcinoma. Cancers (Basel) 2022; 14:cancers14040996. [PMID: 35205743 PMCID: PMC8869950 DOI: 10.3390/cancers14040996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/07/2022] [Accepted: 02/12/2022] [Indexed: 12/20/2022] Open
Abstract
(1) Background: Oesophageal cancers are often late-presenting and have a poor 5-year survival rate. The standard treatment of oesophageal adenocarcinomas involves neoadjuvant chemotherapy with or without radiotherapy followed by surgery. However, less than one third of patients respond to neoadjuvant therapy, thereby unnecessarily exposing patients to toxicity and deconditioning. Hence, there is an urgent need for biomarkers to predict response to neoadjuvant therapy. This review explores the current biomarker landscape. (2) Methods: MEDLINE, EMBASE and ClinicalTrial databases were searched with key words relating to “predictive biomarker”, “neoadjuvant therapy” and “oesophageal adenocarcinoma” and screened as per the inclusion and exclusion criteria. All peer-reviewed full-text articles and conference abstracts were included. (3) Results: The search yielded 548 results of which 71 full-texts, conference abstracts and clinical trials were eligible for review. A total of 242 duplicates were removed, 191 articles were screened out, and 44 articles were excluded. (4) Discussion: Biomarkers were discussed in seven categories including imaging, epigenetic, genetic, protein, immunologic, blood and serum-based with remaining studies grouped in a miscellaneous category. (5) Conclusion: Although promising markers and novel methods have emerged, current biomarkers lack sufficient evidence to support clinical application. Novel approaches have been recommended to assess predictive potential more efficiently.
Collapse
Affiliation(s)
- William Jiang
- Upper Gastrointestinal Surgery Department, St Vincent’s Hospital Melbourne, 41 Victoria Parade, Fitzroy, VIC 3065, Australia
- Correspondence: (W.J.); (M.R.)
| | - Jelske M. de Jong
- Gastrointestinal Oncology Department, The University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (J.M.d.J.); (R.v.H.)
| | - Richard van Hillegersberg
- Gastrointestinal Oncology Department, The University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (J.M.d.J.); (R.v.H.)
| | - Matthew Read
- Upper Gastrointestinal Surgery Department, St Vincent’s Hospital Melbourne, 41 Victoria Parade, Fitzroy, VIC 3065, Australia
- Correspondence: (W.J.); (M.R.)
| |
Collapse
|
17
|
Correnti M, Cappon A, Pastore M, Piombanti B, Lori G, Oliveira DVPN, Munoz‐Garrido P, Lewinska M, Andersen JB, Coulouarn C, Sulpice L, Peraldo Neia C, Cavalloni G, Quarta S, Biasiolo A, Fassan M, Ramazzotti M, Parri M, Recalcati S, di Tommaso L, Campani C, Invernizzi P, Torzilli G, Marra F, Pontisso P, Raggi C. The protease-inhibitor SerpinB3 as a critical modulator of the stem-like subset in human cholangiocarcinoma. Liver Int 2022; 42:233-248. [PMID: 34478594 PMCID: PMC9290104 DOI: 10.1111/liv.15049] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 08/07/2021] [Accepted: 08/25/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND AIMS Cholangiocarcinoma (CCA) is a form of primary liver cancer with limited therapeutic options. Recently, cancer stem cells (CSCs) have been proposed as a driving force of tumour initiation and dissemination, thus representing a crucial therapeutic target. The protease inhibitor SerpinB3 (SB3) has been identified in several malignancies including hepatocellular carcinoma. SB3 has been involved in the early events of hepatocarcinogenesis and is highly expressed in hepatic progenitor cells and in a mouse model of liver progenitor cell activation. However, only limited information on the possible role of SB3 in CCA stem-like compartment is available. METHODS Enrichment of CCA stem-like subset was performed by sphere culture (SPH) in CCA cell lines (CCLP1, HUCCT1, MTCHC01 and SG231). Quantitative RT-PCR and Western blotting were used to detect SB3 in both SPH and parental monolayer (MON) cells. Acquired CSC-like features were analysed using an endogenous and a paracrine in vitro model, with transfection of SB3 gene or addition of recombinant SB3 to cell medium respectively. SB3 tumorigenic role was explored in an in vivo mouse model of CCA by subcutaneous injection of SB3-transfected MON (MONSB3+ ) cells in immune-deficient NOD-SCID/IL2Rgnull (NSG) mice. SB3 expression in human CCA sections was investigated by immunohistochemistry. Overall survival (OS) and time to recurrence (TTR) analyses were carried out from a transcriptome database of 104 CCA patients. RESULTS SB3, barely detected in parental MON cells, was overexpressed in the same CCA cells grown as 3D SPH. Notably, MONSB3+ showed significant overexpression of genes associated with stemness (CD24, CD44, CD133), pluripotency (c-MYC, NOTCH1, STAT3, YAP, NANOG, BMI1, KLF4, OCT4, SOX2), epithelial mesenchymal transition (β-catenin, SLUG) and extracellular matrix remodelling (MMP1, MMP7, MMP9, ADAM9, ADAM10, ADAM17, ITGB3). SB3-overexpressing cells showed superior spherogenic capacity and invasion ability compared to control. Importantly, MONSB3+ exhibited activation of MAP kinases (ERK1/2, p38, JNK) as well as phosphorylation of NFκB (p65) in addition to up-regulation of the proto-oncogene β-catenin. All these effects were reversed after transient silencing of SB3. According to the in vitro finding, MONSB3+ cells retained high tumorigenic potential in NSG mice. SB3 overexpression was observed in human CCA tissues and analysis of OS as well as TTR indicated a worse prognosis in SB3+ CCA patients. CONCLUSION These findings indicate a SB3 role in mediating malignant phenotype of CCA and identify a new therapeutic target.
Collapse
Affiliation(s)
- Margherita Correnti
- Center for Autoimmune Liver DiseasesHumanitas Clinical and Research CenterRozzanoItaly
- Department of Biomedical Sciences for HealthUniversity of MilanMilanItaly
| | - Andrea Cappon
- Animal Care‐Polo Vallisneri University of PaduaPaduaItaly
| | - Mirella Pastore
- Department of Experimental and Clinical MedicineUniversity of FlorenceFlorenceItaly
| | - Benedetta Piombanti
- Department of Experimental and Clinical MedicineUniversity of FlorenceFlorenceItaly
| | - Giulia Lori
- Department of Experimental and Clinical MedicineUniversity of FlorenceFlorenceItaly
| | | | | | - Monika Lewinska
- Biotech Research and Innovation CentreUniversity of CopenhagenCopenhagenDenmark
| | - Jesper B. Andersen
- Biotech Research and Innovation CentreUniversity of CopenhagenCopenhagenDenmark
| | - Cédric Coulouarn
- CHU RennesService de Chirurgie Hépatobiliaire et DigestiveInsermUniv RennesCOSS (Chemistry, Oncogenesis Stress Signaling)UMR_S 1242Centre de Lutte contre le Cancer Eugène MarquisRennesFrance
| | - Laurent Sulpice
- CHU RennesService de Chirurgie Hépatobiliaire et DigestiveINSERM 1241Université de RennesRennesFrance
| | | | - Giuliana Cavalloni
- Division of Medical OncologyCandiolo Cancer InstituteFPO‐IRCCSCandiolo, TorinoItaly
| | - Santina Quarta
- Department of Medicine‐DIMEDUniversity of PaduaPaduaItaly
| | | | - Matteo Fassan
- Department of Medicine‐DIMEDUniversity of PaduaPaduaItaly
| | - Matteo Ramazzotti
- Department of Experimental and Clinical Biomedical SciencesUniversity of FlorenceFlorenceItaly
| | - Matteo Parri
- Department of Experimental and Clinical Biomedical SciencesUniversity of FlorenceFlorenceItaly
| | - Stefania Recalcati
- Department of Biomedical Sciences for HealthUniversity of MilanMilanItaly
| | - Luca di Tommaso
- Department of PathologyHumanitas Clinical and Research CenterRozzanoItaly
- Department of Biomedical SciencesHumanitas UniversityRozzanoItaly
| | - Claudia Campani
- Department of Experimental and Clinical MedicineUniversity of FlorenceFlorenceItaly
| | - Pietro Invernizzi
- Division of Gastroenterology and Center for Autoimmune Liver DiseasesDepartment of Medicine and SurgeryUniversity of Milano‐BicoccaMonzaItaly
- European Reference Network on Hepatological Diseases (ERN RARE‐LIVER)San Gerardo HospitalMonzaItaly
| | - Guido Torzilli
- Department of Hepatobiliary and General SurgeryHumanitas UniversityHumanitas Clinical and Research CenterIRCCS, RozzanoMilanItaly
| | - Fabio Marra
- Department of Experimental and Clinical MedicineUniversity of FlorenceFlorenceItaly
| | | | - Chiara Raggi
- Department of Experimental and Clinical MedicineUniversity of FlorenceFlorenceItaly
| |
Collapse
|
18
|
Bian E, Chen X, Cheng L, Cheng M, Chen Z, Yue X, Zhang Z, Chen J, Sun L, Huang K, Huang C, Fang Z, Zhao B, Li J. Super-enhancer-associated TMEM44-AS1 aggravated glioma progression by forming a positive feedback loop with Myc. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:337. [PMID: 34696771 PMCID: PMC8543865 DOI: 10.1186/s13046-021-02129-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/05/2021] [Indexed: 01/05/2023]
Abstract
Background Long non-coding RNAs (lncRNAs) have been considered as one type of gene expression regulator for cancer development, but it is not clear how these are regulated. This study aimed to identify a specific lncRNA that promotes glioma progression. Methods RNA sequencing (RNA-seq) and quantitative real-time PCR were performed to screen differentially expressed genes. CCK-8, transwell migration, invasion assays, and a mouse xenograft model were performed to determine the functions of TMEM44-AS1. Co-IP, ChIP, Dual-luciferase reporter assays, RNA pulldown, and RNA immunoprecipitation assays were performed to study the molecular mechanism of TMEM44-AS1 and the downstream target. Results We identified a novel lncRNA TMEM44-AS1, which was aberrantly expressed in glioma tissues, and that increased TMEM44-AS1 expression was correlated with malignant progression and poor survival for patients with glioma. Expression of TMEM44-AS1 increased the proliferation, colony formation, migration, and invasion of glioma cells. Knockdown of TMEM44-AS1 in glioma cells reduced cell proliferation, colony formation, migration and invasion, and tumor growth in a nude mouse xenograft model. Mechanistically, TMEM44-AS1 is directly bound to the SerpinB3, and sequentially activated Myc and EGR1/IL-6 signaling; Myc transcriptionally induced TMEM44-AS1 and directly bound to the promoter and super-enhancer of TMEM44-AS1, thus forming a positive feedback loop with TMEM44-AS. Further studies demonstrated that Myc interacts with MED1 regulates the super-enhancer of TMEM44-AS1. More importantly, a novel small-molecule Myc inhibitor, Myci975, alleviated TMEM44-AS1-promoted the growth of glioma cells. Conclusions Our study implicates a crucial role of the TMEM44-AS1-Myc axis in glioma progression and provides a possible anti-glioma therapeutic agent. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-02129-9.
Collapse
Affiliation(s)
- Erbao Bian
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China. .,Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, 230601, China.
| | - Xueran Chen
- Department of Laboratory Medicine, Hefei Cancer Hospital, Chinese Academy of Sciences, No. 350, Shushan Hu Road, Hefei, 230031, Anhui, China.,Anhui Province Key Laboratory of Medical Physics and Technology; Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, No. 350, Shushan Hu Road, Hefei, 230031, Anhui, China
| | - Li Cheng
- School of pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Meng Cheng
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.,Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, 230601, China
| | - Zhigang Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.,Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, 230601, China
| | - Xiaoyu Yue
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.,Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, 230601, China
| | - Zhengwei Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.,Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, 230601, China
| | - Jie Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.,Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, 230601, China
| | - Libo Sun
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.,Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, 230601, China
| | - Kebing Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.,Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, 230601, China
| | - Cheng Huang
- School of pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Zhiyou Fang
- Department of Laboratory Medicine, Hefei Cancer Hospital, Chinese Academy of Sciences, No. 350, Shushan Hu Road, Hefei, 230031, Anhui, China. .,Anhui Province Key Laboratory of Medical Physics and Technology; Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, No. 350, Shushan Hu Road, Hefei, 230031, Anhui, China.
| | - Bing Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China. .,Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, 230601, China.
| | - Jun Li
- School of pharmacy, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
19
|
Gong J, Song Y, Xu L, Che X, Hou K, Guo T, Cheng Y, Liu Y, Qu X. Upregulation of Serine Proteinase Inhibitor Clade B Member 3 (SERPINB3) Expression by Stromal Cell-Derived Factor (SDF-1)/CXCR4/Nuclear Factor kappa B (NF-κB) Promotes Migration and Invasion of Gastric Cancer Cells. Med Sci Monit 2020; 26:e927411. [PMID: 33110054 PMCID: PMC7604976 DOI: 10.12659/msm.927411] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 09/25/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Serine proteinase inhibitor clade B member 3 (SERPINB3) is a neutral glycoprotein. Its overexpression is related to the promotion of cell proliferation and activation via the nuclear factor kappa B (NF-kappaB) pathway in several tumors. Whether it can participate in stromal cell-derived factor (SDF-1)/NF-kappaB-induced metastasis of gastric cancer has not been reported. MATERIAL AND METHODS We analyzed the ability of SDF-1 to induce migration and invasion in vitro by knocking down the expression of SERPINB3 with siRNAs in gastric cancer cells. We also explored the effects of a CXCR4 antagonist and NF-kappaB inhibitor on SERPINB3 expression. We verified the effect of SERPINB3 on prognosis in gastric cancer specimens by immunohistochemistry. RESULTS In vitro experiments confirmed that SDF-1 upregulated the expression of SERPINB3 and promoted metastasis in gastric cancer cells. This phenomenon was reversed by knockdown of SERPINB3, a chemokine receptor 4 (CXCR4) antagonist, and an NF-kappaB inhibitor, which downregulated the expression of SERPINB3. In patients with gastric cancer, a significant positive correlation was observed between CXCR4 and SERPINB3 expression (r=0.222, P=0.029). Moreover, double positivity for SERPINB3 and CXCR4 was certified to be an independent prognostic factor (HR=3.332, P<0.001). CXCR4-positive patients who also expressed SERPINB3 were inclined to suffer from lymph node metastasis, confirming that SERPINB3 is a downstream molecule of CXCR4. CONCLUSIONS In vitro and pathological results showed that SDF-1/CXCR4 activated the NF-kappaB pathway and upregulated SERPINB3 to facilitate the migration and invasion of gastric cancer cells.
Collapse
Affiliation(s)
- Jing Gong
- Department of Geriatrics, The First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Yongxi Song
- Department of Surgical Oncology and General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Ling Xu
- Department of Medical Oncology, Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Xiaofang Che
- Department of Medical Oncology, Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Kezuo Hou
- Department of Medical Oncology, Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Tianshu Guo
- Department of Medical Oncology, Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Yu Cheng
- Department of Medical Oncology, Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Yunpeng Liu
- Department of Medical Oncology, Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Xiujuan Qu
- Department of Medical Oncology, Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| |
Collapse
|
20
|
Kim BH, Yu K, Lee PCW. Cancer classification of single-cell gene expression data by neural network. Bioinformatics 2020; 36:1360-1366. [PMID: 31603465 DOI: 10.1093/bioinformatics/btz772] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 08/13/2019] [Accepted: 10/08/2019] [Indexed: 01/16/2023] Open
Abstract
MOTIVATION Cancer classification based on gene expression profiles has provided insight on the causes of cancer and cancer treatment. Recently, machine learning-based approaches have been attempted in downstream cancer analysis to address the large differences in gene expression values, as determined by single-cell RNA sequencing (scRNA-seq). RESULTS We designed cancer classifiers that can identify 21 types of cancers and normal tissues based on bulk RNA-seq as well as scRNA-seq data. Training was performed with 7398 cancer samples and 640 normal samples from 21 tumors and normal tissues in TCGA based on the 300 most significant genes expressed in each cancer. Then, we compared neural network (NN), support vector machine (SVM), k-nearest neighbors (kNN) and random forest (RF) methods. The NN performed consistently better than other methods. We further applied our approach to scRNA-seq transformed by kNN smoothing and found that our model successfully classified cancer types and normal samples. AVAILABILITY AND IMPLEMENTATION Cancer classification by neural network. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Bong-Hyun Kim
- Department of Biomedical Sciences, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, Korea.,Advanced Bio Computing Center, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Kijin Yu
- Department of Biomedical Sciences, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, Korea
| | - Peter C W Lee
- Department of Biomedical Sciences, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, Korea
| |
Collapse
|
21
|
Autophagy Is Deficient and May be Negatively Regulated by SERPINB3 in Middle Ear Cholesteatoma. Otol Neurotol 2020; 41:e881-e888. [PMID: 32569142 DOI: 10.1097/mao.0000000000002690] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
HYPOTHESIS Whereas autophagy has been linked to various human diseases, whether it also plays a role in cholesteatoma is virtually unknown. This study aimed to investigate the activity and regulation of autophagy in cholesteatoma. BACKGROUND The treatment of middle ear cholesteatoma has been challenging due to an insufficient understanding of the underlying disease mechanism. METHODS Expression of microtubule-associated protein 1A/1B-light chain 3 (LC3), the autophagy protein marker, and phosphorylated Akt (p-Akt), and mammalian target of rapamycin (p-mTOR), the known autophagy regulators, in fresh retroauricular skin and cholesteatoma tissue samples was analyzed by immunoblotting. The results were further confirmed by immunohistochemistry and statistical analyses. Cell proliferation of primary retroauricular skin- and cholesteatoma-derived fibroblasts was evaluated by methyl thiazol tetrazolium (MTT) assay. Ectopic expression of serine proteinase inhibitor, clade B, member 3 (SERPINB3) in the fibroblasts was achieved by electroporation and the expression was detected by immunoblotting. RESULTS LC3 expression was significantly decreased in cholesteatoma in most of the 15 paired retroauricular skin/cholesteatoma tissue samples. However, p-Akt and p-mTOR expression in the cholesteatoma samples was not significantly different from that in the control subjects. Immunohistochemical studies further demonstrated an inverse correlation between LC3 expression and cholesteatoma. The cholesteatoma fibroblasts proliferated faster than the retroauricular skin fibroblasts, and had higher SERPINB3 but lower LC3 expression. Furthermore, overexpression of SERPINB3 in the retroauricular skin fibroblasts enhanced cell proliferation and downregulated LC3 expression. CONCLUSION Autophagy is significantly suppressed in cholesteatoma tissues, which may not involve the Akt/mTOR signaling pathway. More importantly, SERPINB3 may promote cell proliferation and negatively regulate autophagy in cholesteatoma fibroblasts. Together, these findings warrant further investigation into the pathogenic mechanism of cholesteatoma.
Collapse
|
22
|
Tunçer S, Sade-Memişoğlu A, Keşküş AG, Sheraj I, Güner G, Akyol A, Banerjee S. Enhanced expression of HNF4α during intestinal epithelial differentiation is involved in the activation of ER stress. FEBS J 2019; 287:2504-2523. [PMID: 31762160 DOI: 10.1111/febs.15152] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 10/17/2019] [Accepted: 11/21/2019] [Indexed: 01/19/2023]
Abstract
Intestinal epithelial cells are derived from stem cells at the crypts that undergo differentiation into transit-amplifying cells, which in turn form terminally differentiated enterocytes as these cells reach the villus. Extensive alterations in both transcriptional and translational programs occur during differentiation, which can induce the activation of cellular stress responses such as ER stress-related unfolded protein response (UPR) and autophagy, particularly in the cells that are already committed to becoming absorptive cells. Using an epithelial cell model of enterocyte differentiation, we report a mechanistic study connecting enterocyte differentiation to UPR and autophagy. We report that differentiated colon epithelial cells showed increased cytosolic Ca2+ levels and activation of all three pathways of UPR: inositol-requiring enzyme 1 (IRE1), protein kinase RNA-like ER kinase, and activating transcription factor 6 (ATF6) compared to the undifferentiated cells. Enhanced UPR in the differentiated cells was accompanied by the induction of autophagy as evidenced by increased ratio of light chain 3 II/I, upregulation of Beclin-1, and downregulation of p62. We show for the first time that mechanistically, the upregulation of hepatocyte nuclear factor 4α (HNF4α) during differentiation led to increased promoter binding and transcriptional upregulation of two major proteins of UPR: X-box binding protein-1 and ATF6, implicating HNF4α as a key regulator of UPR response during differentiation. Integrating wet-lab with in silico analyses, the present study links differentiation to cellular stress responses, and highlights the importance of transcription factor signaling and cross-talk between the cellular events in the regulation of intestinal cell differentiation.
Collapse
Affiliation(s)
- Sinem Tunçer
- Department of Biological Sciences, Orta Dogu Teknik Universitesi, Ankara, Turkey
| | - Aslı Sade-Memişoğlu
- Department of Biological Sciences, Orta Dogu Teknik Universitesi, Ankara, Turkey
| | - Ayşe Gökçe Keşküş
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Ilir Sheraj
- Department of Biological Sciences, Orta Dogu Teknik Universitesi, Ankara, Turkey
| | - Güneş Güner
- Department of Pathology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Aytekin Akyol
- Department of Pathology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Sreeparna Banerjee
- Department of Biological Sciences, Orta Dogu Teknik Universitesi, Ankara, Turkey.,Department of Biological Sciences and Cancer Systems Biology Laboratory (CanSyl), Orta Dogu Teknik Universitesi, Ankara, Turkey
| |
Collapse
|
23
|
Cannito S, Foglia B, Villano G, Turato C, C Delgado T, Morello E, Pin F, Novo E, Napione L, Quarta S, Ruvoletto M, Fasolato S, Zanus G, Colombatto S, Lopitz-Otsoa F, Fernández-Ramos D, Bussolino F, Sutti S, Albano E, Martínez-Chantar ML, Pontisso P, Parola M. SerpinB3 Differently Up-Regulates Hypoxia Inducible Factors -1α and -2α in Hepatocellular Carcinoma: Mechanisms Revealing Novel Potential Therapeutic Targets. Cancers (Basel) 2019; 11:1933. [PMID: 31817100 PMCID: PMC6966556 DOI: 10.3390/cancers11121933] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 11/29/2019] [Accepted: 12/02/2019] [Indexed: 12/15/2022] Open
Abstract
SerpinB3 (SB3) is a hypoxia and hypoxia-inducible factor (HIF)-2α-dependent cysteine-protease inhibitor up-regulated in hepatocellular carcinoma (HCC), released by cancer cells and able to stimulate proliferation and epithelial-to-mesenchymal-transition. Methods: In the study we employed transgenic and knock out SerpinB3 mice, liver cancer cell line, human HCC specimens, and mice receiving diethyl-nitrosamine (DEN) administration plus choline-deficient L-amino acid refined (CDAA) diet (DEN/CDAA protocol). Results: We provide detailed and mechanistic evidence that SB3 can act as a paracrine mediator able to affect the behavior of surrounding cells by differentially up-regulating, in normoxic conditions, HIF-1α and HIF-2α. SB3 acts by (i) up-regulating HIF-1α transcription, facilitating cell survival in a harsh microenvironment and promoting angiogenesis, (ii) increasing HIF-2α stabilization via direct/selective NEDDylation, promoting proliferation of liver cancer cells, and favoring HCC progression. Moreover (iii) the highest levels of NEDD8-E1 activating enzyme (NAE1) mRNA were detected in a subclass of HCC patients expressing the highest levels of HIF-2α transcripts; (iv) mice undergoing DEN/CDAA carcinogenic protocol showed a positive correlation between SB3 and HIF-2α transcripts with the highest levels of NAE1 mRNA detected in nodules expressing the highest levels of HIF-2α transcripts. Conclusions: These data outline either HIF-2α and NEDDylation as two novel putative therapeutic targets to interfere with the procarcinogenic role of SerpinB3 in the development of HCC.
Collapse
Affiliation(s)
- Stefania Cannito
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine & Clinical Pathology, University of Torino, 10125 Torino, Italy; (S.C.); (B.F.); (E.M.); (F.P.); (E.N.)
| | - Beatrice Foglia
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine & Clinical Pathology, University of Torino, 10125 Torino, Italy; (S.C.); (B.F.); (E.M.); (F.P.); (E.N.)
| | - Gianmarco Villano
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy; (G.V.); (M.R.); (S.F.)
| | - Cristian Turato
- Veneto Institute of Oncology IOV—IRCCS, 35128 Padova, Italy;
| | - Teresa C Delgado
- Liver Disease and Metabolism Laboratory, CIC bioGUNE, Centro de Investigacion Biomedica en Red de Enfermedades Hepaticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain; (T.C.D.); (F.L.-O.); (D.F.-R.); (M.L.M.-C.)
| | - Elisabetta Morello
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine & Clinical Pathology, University of Torino, 10125 Torino, Italy; (S.C.); (B.F.); (E.M.); (F.P.); (E.N.)
| | - Fabrizio Pin
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine & Clinical Pathology, University of Torino, 10125 Torino, Italy; (S.C.); (B.F.); (E.M.); (F.P.); (E.N.)
| | - Erica Novo
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine & Clinical Pathology, University of Torino, 10125 Torino, Italy; (S.C.); (B.F.); (E.M.); (F.P.); (E.N.)
| | - Lucia Napione
- Department of Applied Science and Technology, Politecnico di Torino, 10129 Torino, Italy;
- Laboratory of Vascular Oncology Candiolo Cancer Institute—FPO IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico), 10060 Candiolo, Italy;
| | - Santina Quarta
- Department of Medicine, University of Padova, 35128 Padova, Italy; (S.Q.); (P.P.)
| | - Mariagrazia Ruvoletto
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy; (G.V.); (M.R.); (S.F.)
| | - Silvano Fasolato
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy; (G.V.); (M.R.); (S.F.)
| | - Giacomo Zanus
- Hepatobiliary Surgery, University of Padova, 35128 Padova, Italy;
| | | | - Fernando Lopitz-Otsoa
- Liver Disease and Metabolism Laboratory, CIC bioGUNE, Centro de Investigacion Biomedica en Red de Enfermedades Hepaticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain; (T.C.D.); (F.L.-O.); (D.F.-R.); (M.L.M.-C.)
| | - David Fernández-Ramos
- Liver Disease and Metabolism Laboratory, CIC bioGUNE, Centro de Investigacion Biomedica en Red de Enfermedades Hepaticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain; (T.C.D.); (F.L.-O.); (D.F.-R.); (M.L.M.-C.)
| | - Federico Bussolino
- Laboratory of Vascular Oncology Candiolo Cancer Institute—FPO IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico), 10060 Candiolo, Italy;
- Department of Oncology, University of Torino, 10125 Torino, Italy;
| | - Salvatore Sutti
- Department of Health Sciences and Interdisciplinary Research Center for Autoimmune Diseases, University Amedeo Avogadro of East Piedmont, 28100 Novara, Italy; (S.S.); (E.A.)
| | - Emanuele Albano
- Department of Health Sciences and Interdisciplinary Research Center for Autoimmune Diseases, University Amedeo Avogadro of East Piedmont, 28100 Novara, Italy; (S.S.); (E.A.)
| | - Maria Luz Martínez-Chantar
- Liver Disease and Metabolism Laboratory, CIC bioGUNE, Centro de Investigacion Biomedica en Red de Enfermedades Hepaticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain; (T.C.D.); (F.L.-O.); (D.F.-R.); (M.L.M.-C.)
| | - Patrizia Pontisso
- Department of Medicine, University of Padova, 35128 Padova, Italy; (S.Q.); (P.P.)
| | - Maurizio Parola
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine & Clinical Pathology, University of Torino, 10125 Torino, Italy; (S.C.); (B.F.); (E.M.); (F.P.); (E.N.)
| |
Collapse
|
24
|
Sun D, Luo T, Dong P, Zhang N, Chen J, Zhang S, Dong L, Janssen HLA, Zhang S. M2-polarized tumor-associated macrophages promote epithelial-mesenchymal transition via activation of the AKT3/PRAS40 signaling pathway in intrahepatic cholangiocarcinoma. J Cell Biochem 2019; 121:2828-2838. [PMID: 31692069 DOI: 10.1002/jcb.29514] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 10/10/2019] [Indexed: 12/15/2022]
Abstract
Tumor-associated macrophages (TAMs) have been considered as a major component of the tumor microenvironment. However, the crosstalk between M2-polarized tumor-associated macrophages (M2-TAMs) and intrahepatic cholangiocarcinoma (ICC) remains undetermined. In the present study, we aimed to clarify the role of M2-TAMs in ICC and the underlying mechanism. The in vitro assay demonstrated M2-TAMs promoted epithelial-mesenchymal transition (EMT) of ICC cells, resulting in enhanced cell invasion and metastasis ability. Moreover, M2-TAMs modulated the microenvironment of ICC by increasing the secretion of cytokines (GM-CSF, tumor necrosis factor-α [TNF-α], ICAM-1, interleukin-6 [IL-6], etc) and chemokines (CCL1, CCL3, etc). In addition, p-AKT (Ser473) and p-PRAS40 (Thr246) were upregulated in ICC cells when cocultured with M2-TAMs or treated with M2-TAMs secreted core cytokines (GM-CSF, TNF-α, ICAM-1, and IL-6). Consistently, AKT3 silencing (but not AKT1 silencing and AKT2 silencing) markedly inhibited phosphorylation of AKT and PRAS40 of ICC cells and inhibited the EMT process when cocultured with M2-TAMs. Taken together, the current data indicated that M2-TAMs promoted ICC cells EMT, partially through increasing secretion of cytokines and chemokines, thus modulating the microenvironment and activating the AKT3/PRAS40 signaling pathway.
Collapse
Affiliation(s)
- Dalong Sun
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Liver Disease, Shanghai, China.,Department of Gastroenterology and Hepatology, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, China
| | - Tiancheng Luo
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Liver Disease, Shanghai, China
| | - Pingping Dong
- Department of Surgery, Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ningping Zhang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Liver Disease, Shanghai, China
| | - Jing Chen
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shuncai Zhang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Liver Disease, Shanghai, China
| | - Ling Dong
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Liver Disease, Shanghai, China
| | - Harry L A Janssen
- Toronto Center for Liver Disease, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Canada
| | - Si Zhang
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
25
|
Pluquet O, Galmiche A. Impact and Relevance of the Unfolded Protein Response in HNSCC. Int J Mol Sci 2019; 20:ijms20112654. [PMID: 31151143 PMCID: PMC6601021 DOI: 10.3390/ijms20112654] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/27/2019] [Accepted: 05/27/2019] [Indexed: 12/12/2022] Open
Abstract
Head and neck squamous cell carcinomas (HNSCC) encompass a heterogeneous group of solid tumors that arise from the upper aerodigestive tract. The tumor cells face multiple challenges including an acute demand of protein synthesis often driven by oncogene activation, limited nutrient and oxygen supply and exposure to chemo/radiotherapy, which forces them to develop adaptive mechanisms such as the Unfolded Protein Response (UPR). It is now well documented that the UPR, a homeostatic mechanism, is induced at different stages of cancer progression in response to intrinsic (oncogenic activation) or extrinsic (microenvironment) perturbations. This review will discuss the role of the UPR in HNSCC as well as in the key processes that characterize the physiology of HNSCC. The role of the UPR in the clinical context of HNSCC will also be addressed.
Collapse
Affiliation(s)
- Olivier Pluquet
- Institut Pasteur de Lille, Université de Lille, CNRS, UMR8161-M3T-Mechanisms of Tumorigenesis and Targeted Therapies, F-59000 Lille, France.
| | - Antoine Galmiche
- Service de Biochimie, Centre de Biologie Humaine (CBH), CHU Sud, 80054 Amiens, France.
- EA7516, Université de Picardie Jules Verne (UPJV), 80054 Amiens, France.
| |
Collapse
|
26
|
UPR: An Upstream Signal to EMT Induction in Cancer. J Clin Med 2019; 8:jcm8050624. [PMID: 31071975 PMCID: PMC6572589 DOI: 10.3390/jcm8050624] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 04/30/2019] [Accepted: 05/03/2019] [Indexed: 12/21/2022] Open
Abstract
The endoplasmic reticulum (ER) is the organelle where newly synthesized proteins enter the secretory pathway. Different physiological and pathological conditions may perturb the secretory capacity of cells and lead to the accumulation of misfolded and unfolded proteins. To relieve the produced stress, cells evoke an adaptive signalling network, the unfolded protein response (UPR), aimed at recovering protein homeostasis. Tumour cells must confront intrinsic and extrinsic pressures during cancer progression that produce a proteostasis imbalance and ER stress. To overcome this situation, tumour cells activate the UPR as a pro-survival mechanism. UPR activation has been documented in most types of human tumours and accumulating evidence supports a crucial role for UPR in the establishment, progression, metastasis and chemoresistance of tumours as well as its involvement in the acquisition of other hallmarks of cancer. In this review, we will analyse the role of UPR in cancer development highlighting the ability of tumours to exploit UPR signalling to promote epithelial-mesenchymal transition (EMT).
Collapse
|
27
|
Serum SCCA levels in patients suffering cancers or other diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 162:165-175. [PMID: 30905447 DOI: 10.1016/bs.pmbts.2018.12.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Increased serum squamous cell carcinoma antigen (SCCA) levels are clinically used diagnostic or prognostic biomarker for squamous cell carcinomas. According to recently published studies, increased serum SCCA levels are also observed in adenocarcinomas, hepatocarcinomas, kidney, and other inflammatory diseases, indicating squamous cell carcinoma is not the production source of serum SCCA in these diseases. However, serum SCCA levels in patients suffering different types of diseases have not been systematically measured and compared. Thus, in our current study, serum SCCA levels from 21,608 patients with 39 clinically defined diseases were collected and measured by the clinical laboratory in the Affiliated Hospital of Qingdao University over the past 5 years in addition to 232 serum samples from individuals who attend their annual physical examination as the healthy controls. According to the median, mean, and -log10p values, we found that patients with uremia, azotemia, diabetic nephropathy, and nephritic syndrome had the highest serum SCCA levels among all 39 different types of diseases including patients suffering squamous cell carcinomas. Moreover, patients suffering lung cancer, cervical cancer, esophagus cancer, or chronic pulmonary disease had lower median and interquartile range values but higher or comparable mean values and significantly higher SD values than that of the healthy controls. Furthermore, patients with endometrial cancer, pancreatitis, osteoporosis, and some other diseases had lower serum SCCA levels than that of the healthy controls. These results demonstrated that serum SCCA can not only be used in diagnosis and prognosis of squamous cell carcinomas but also as biomarkers for uremia, azotemia, diabetic nephropathy, and nephritic syndrome.
Collapse
|
28
|
Sebastião MJ, Serra M, Pereira R, Palacios I, Gomes-Alves P, Alves PM. Human cardiac progenitor cell activation and regeneration mechanisms: exploring a novel myocardial ischemia/reperfusion in vitro model. Stem Cell Res Ther 2019; 10:77. [PMID: 30845956 PMCID: PMC6407246 DOI: 10.1186/s13287-019-1174-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 02/12/2019] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Numerous studies from different labs around the world report human cardiac progenitor cells (hCPCs) as having a role in myocardial repair upon ischemia/reperfusion (I/R) injury, mainly through auto/paracrine signaling. Even though these cell populations are already being investigated in cell transplantation-based clinical trials, the mechanisms underlying their response are still poorly understood. METHODS To further investigate hCPC regenerative process, we established the first in vitro human heterotypic model of myocardial I/R injury using hCPCs and human-induced pluripotent cell-derived cardiomyocytes (hiPSC-CMs). The co-culture model was established using transwell inserts and evaluated in both ischemia and reperfusion phases regarding secretion of key cytokines, hiPSC-CM viability, and hCPC proliferation. hCPC proteome in response to I/R was further characterized using advanced liquid chromatography mass spectrometry tools. RESULTS This model recapitulates hallmarks of I/R, namely hiPSC-CM death upon insult, protective effect of hCPCs on hiPSC-CM viability (37.6% higher vs hiPSC-CM mono-culture), and hCPC proliferation (approximately threefold increase vs hCPCs mono-culture), emphasizing the importance of paracrine communication between these two populations. In particular, in co-culture supernatant upon injury, we report higher angiogenic functionality as well as a significant increase in the CXCL6 secretion rate, suggesting an important role of this chemokine in myocardial regeneration. hCPC whole proteome analysis allowed us to propose new pathways in the hCPC-mediated regenerative process, including cell cycle regulation, proliferation through EGF signaling, and reactive oxygen species detoxification. CONCLUSION This work contributes with new insights into hCPC biology in response to I/R, and the model established constitutes an important tool to study the molecular mechanisms involved in the myocardial regenerative process.
Collapse
Affiliation(s)
- Maria J. Sebastião
- Animal Cell Technology Unit, iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- ITQB-NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Margarida Serra
- Animal Cell Technology Unit, iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- ITQB-NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Rute Pereira
- Animal Cell Technology Unit, iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- ITQB-NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Itziar Palacios
- Coretherapix, S.L.U (Tigenix Group, Takeda), Parque Tecnológico de Madrid, Madrid, Spain
| | - Patrícia Gomes-Alves
- Animal Cell Technology Unit, iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- ITQB-NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Paula M. Alves
- Animal Cell Technology Unit, iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- ITQB-NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| |
Collapse
|
29
|
The Role of the ER-Induced UPR Pathway and the Efficacy of Its Inhibitors and Inducers in the Inhibition of Tumor Progression. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:5729710. [PMID: 30863482 PMCID: PMC6378054 DOI: 10.1155/2019/5729710] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 12/08/2018] [Accepted: 12/18/2018] [Indexed: 12/12/2022]
Abstract
Cancer is the second most frequent cause of death worldwide. It is considered to be one of the most dangerous diseases, and there is still no effective treatment for many types of cancer. Since cancerous cells have a high proliferation rate, it is pivotal for their proper functioning to have the well-functioning protein machinery. Correct protein processing and folding are crucial to maintain tumor homeostasis. Endoplasmic reticulum (ER) stress is one of the leading factors that cause disturbances in these processes. It is induced by impaired function of the ER and accumulation of unfolded proteins. Induction of ER stress affects many molecular pathways that cause the unfolded protein response (UPR). This is the way in which cells can adapt to the new conditions, but when ER stress cannot be resolved, the UPR induces cell death. The molecular mechanisms of this double-edged sword process are involved in the transition of the UPR either in a cell protection mechanism or in apoptosis. However, this process remains poorly understood but seems to be crucial in the treatment of many diseases that are related to ER stress. Hence, understanding the ER stress response, especially in the aspect of pathological consequences of UPR, has the potential to allow us to develop novel therapies and new diagnostic and prognostic markers for cancer.
Collapse
|
30
|
Gurel B, Cansev M, Sevinc C, Kelestemur S, Ocalan B, Cakir A, Aydin S, Kahveci N, Ozansoy M, Taskapilioglu O, Ulus IH, Başar MK, Sahin B, Tuzuner MB, Baykal AT. Early Stage Alterations in CA1 Extracellular Region Proteins Indicate Dysregulation of IL6 and Iron Homeostasis in the 5XFAD Alzheimer's Disease Mouse Model. J Alzheimers Dis 2019; 61:1399-1410. [PMID: 29376847 DOI: 10.3233/jad-170329] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
In recent years, an increasing number of research papers revealed that the compositional and volumetric alterations in the extracellular matrix are the consequences of aging and may be related to Alzheimer's disease (AD). In this study, we aimed to demonstrate the alterations in hippocampal extracellular fluid proteins in vivo using the 5XFAD mouse model. Samples were obtained from hippocampi of 5XFAD mice (n = 6) and their non-transgenic littermates by intracerebral push-pull perfusion technique at 3 months of age, representing the pre-pathological stage of the AD. Proteins in the hippocampal perfusates were analyzed by Ultra Performance Liquid Chromatography-Electrospray Ionization Quadrupole Time-of-Flight Mass Spectrometry (UPLC-ESI-qTOF-MS/MS). 178 proteins were identified and 19 proteins of them were found to be statistically significantly altered (p≤0.05, fold change ≥40%, unique peptide count ≥3) in the hippocampal CA1 extracellular fluid of the 5XFAD mouse model. Ingenuity pathway analysis of the protein expression results identified IL6 as an upstream regulator. The upregulation of IL6 was validated by immunohistochemical staining of the hippocampus and cortex of the 5XFAD mice prior to Aβ plaque formation. Furthermore, the iron level in the hippocampus was measured by inductively coupled plasma-mass spectrometry as IL6 is mentioned in several studies to take part in iron homeostasis and inflammation and found to be increased in 5XFAD mice hippocampus. Alterations in extracellular matrix proteins in addition to increasing amount of hippocampal IL6 and iron in the early stages of AD may reveal inflammation-mediated iron dyshomeostasis in the early stages of neurodegeneration.
Collapse
Affiliation(s)
- Busra Gurel
- Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Istanbul, Turkey.,Department of Medical Biochemistry, Faculty of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Mehmet Cansev
- Department of Pharmacology, Faculty of Medicine, Uludag University, Bursa, Turkey
| | - Cansu Sevinc
- Department of Pharmacology, Faculty of Medicine, Uludag University, Bursa, Turkey
| | - Seda Kelestemur
- Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Istanbul, Turkey
| | - Busra Ocalan
- Department of Physiology, Faculty of Medicine, Uludag University, Bursa, Turkey
| | - Aysen Cakir
- Department of Physiology, Faculty of Medicine, Uludag University, Bursa, Turkey
| | - Sami Aydin
- Department of Pharmacology, Faculty of Medicine, Uludag University, Bursa, Turkey
| | - Nevzat Kahveci
- Department of Physiology, Faculty of Medicine, Uludag University, Bursa, Turkey
| | - Mehmet Ozansoy
- Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Istanbul, Turkey.,Department of Physiology, Faculty of Medicine, Medipol University, Istanbul, Turkey
| | - Ozlem Taskapilioglu
- Department of Neurology, Faculty of Medicine, Uludag University, Bursa, Turkey
| | - Ismail Hakki Ulus
- Department of Pharmacology, Faculty of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Merve Karayel Başar
- Department of Medical Biochemistry, Faculty of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Betul Sahin
- Acibadem Labmed R&D Laboratory, Istanbul, Turkey
| | | | - Ahmet Tarik Baykal
- Department of Medical Biochemistry, Faculty of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| |
Collapse
|
31
|
Dynamic matrisome: ECM remodeling factors licensing cancer progression and metastasis. Biochim Biophys Acta Rev Cancer 2018; 1870:207-228. [DOI: 10.1016/j.bbcan.2018.09.002] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 09/07/2018] [Accepted: 09/30/2018] [Indexed: 01/04/2023]
|
32
|
Khowal S, Naqvi SH, Monga S, Jain SK, Wajid S. Assessment of cellular and serum proteome from tongue squamous cell carcinoma patient lacking addictive proclivities for tobacco, betel nut, and alcohol: Case study. J Cell Biochem 2018; 119:5186-5221. [PMID: 29236289 DOI: 10.1002/jcb.26554] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 11/30/2017] [Indexed: 02/06/2023]
Abstract
The intriguing molecular pathways involved in oral carcinogenesis are still ambiguous. The oral squamous cell carcinoma (OSCC) ranks as the most common type constituting more than 90% of the globally diagnosed oral cancers cases. The elevation in the OSCC incidence rate during past 10 years has an alarming impression on human healthcare. The major challenges associated with OSCC include delayed diagnosis, high metastatic rates, and low 5-year survival rates. The present work foundations on reverse genetic strategy and involves the identification of genes showing expressional variability in an OSCC case lacking addictive proclivities for tobacco, betel nut, and/or alcohol, major etiologies. The expression modulations in the identified genes were analyzed in 16 patients comprising oral pre-cancer and cancer histo-pathologies. The genes SCCA1 and KRT1 were found to down regulate while DNAJC13, GIPC2, MRPL17, IG-Vreg, SSFA2, and UPF0415 upregulated in the oral pre-cancer and cancer pathologies, implicating the genes as crucial players in oral carcinogenesis.
Collapse
Affiliation(s)
- Sapna Khowal
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Samar H Naqvi
- Molecular Diagnostics, Genetix Biotech Asia (P) Ltd., New Delhi, India
| | - Seema Monga
- Department of ENT, Hamdard Institute of Medical Sciences and Research, Jamia Hamdard, New Delhi, India
| | - Swatantra K Jain
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
- Department of Biochemistry, Hamdard Institute of Medical Sciences and Research, Jamia Hamdard, New Delhi, India
| | - Saima Wajid
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| |
Collapse
|
33
|
Ly T, Endo A, Brenes A, Gierlinski M, Afzal V, Pawellek A, Lamond AI. Proteome-wide analysis of protein abundance and turnover remodelling during oncogenic transformation of human breast epithelial cells. Wellcome Open Res 2018; 3:51. [PMID: 29904729 PMCID: PMC5989152 DOI: 10.12688/wellcomeopenres.14392.1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2018] [Indexed: 01/07/2023] Open
Abstract
Background: Viral oncogenes and mutated proto-oncogenes are potent drivers of cancer malignancy. Downstream of the oncogenic trigger are alterations in protein properties that give rise to cellular transformation and the acquisition of malignant cellular phenotypes. Developments in mass spectrometry enable large-scale, multidimensional characterisation of proteomes. Such techniques could provide an unprecedented, unbiased view of how oncogene activation remodels a human cell proteome. Methods: Using quantitative MS-based proteomics and cellular assays, we analysed how transformation induced by activating v-Src kinase remodels the proteome and cellular phenotypes of breast epithelial (MCF10A) cells. SILAC MS was used to comprehensively characterise the MCF10A proteome and to measure v-Src-induced changes in protein abundance across seven time-points (1-72 hrs). We used pulse-SILAC MS ( Boisvert et al., 2012), to compare protein synthesis and turnover in control and transformed cells. Follow-on experiments employed a combination of cellular and functional assays to characterise the roles of selected Src-responsive proteins. Results: Src-induced transformation changed the expression and/or turnover levels of ~3% of proteins, affecting ~1.5% of the total protein molecules in the cell. Transformation increased the average rate of proteome turnover and disrupted protein homeostasis. We identify distinct classes of protein kinetics in response to Src activation. We demonstrate that members of the polycomb repressive complex 1 (PRC1) are important regulators of invasion and migration in MCF10A cells. Many Src-regulated proteins are present in low abundance and some are regulated post-transcriptionally. The signature of Src-responsive proteins is highly predictive of poor patient survival across multiple cancer types. Open access to search and interactively explore all these proteomic data is provided via the EPD database ( www.peptracker.com/epd). Conclusions: We present the first comprehensive analysis measuring how protein expression and protein turnover is affected by cell transformation, providing a detailed picture at the protein level of the consequences of activation of an oncogene.
Collapse
Affiliation(s)
- Tony Ly
- Centre for Gene Regulation and Expression, University of Dundee, Dundee, UK
- Laboratory for Quantitative Proteomics, University of Dundee, Dundee, UK
- Wellcome Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Aki Endo
- Centre for Gene Regulation and Expression, University of Dundee, Dundee, UK
- Laboratory for Quantitative Proteomics, University of Dundee, Dundee, UK
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Tokyo, Japan
| | - Alejandro Brenes
- Centre for Gene Regulation and Expression, University of Dundee, Dundee, UK
- Laboratory for Quantitative Proteomics, University of Dundee, Dundee, UK
| | - Marek Gierlinski
- Centre for Gene Regulation and Expression, University of Dundee, Dundee, UK
- Laboratory for Quantitative Proteomics, University of Dundee, Dundee, UK
| | - Vackar Afzal
- Centre for Gene Regulation and Expression, University of Dundee, Dundee, UK
- Laboratory for Quantitative Proteomics, University of Dundee, Dundee, UK
| | - Andrea Pawellek
- Centre for Gene Regulation and Expression, University of Dundee, Dundee, UK
- Laboratory for Quantitative Proteomics, University of Dundee, Dundee, UK
| | - Angus I. Lamond
- Centre for Gene Regulation and Expression, University of Dundee, Dundee, UK
- Laboratory for Quantitative Proteomics, University of Dundee, Dundee, UK
| |
Collapse
|
34
|
Hogervorst M, Rietveld M, de Gruijl F, El Ghalbzouri A. A shift from papillary to reticular fibroblasts enables tumour-stroma interaction and invasion. Br J Cancer 2018; 118:1089-1097. [PMID: 29551776 PMCID: PMC5931114 DOI: 10.1038/s41416-018-0024-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 12/26/2017] [Accepted: 01/16/2018] [Indexed: 12/23/2022] Open
Abstract
Background Tumour stroma consists of a heterogeneous population of fibroblasts and related mesenchymal cells, collectively dubbed cancer-associated fibroblasts (CAFs). These CAFs are key players in cancer invasion of cutaneous squamous cell carcinoma (SCC). As we have shown earlier, papillary and reticular fibroblasts (Pfs and Rfs, respectively) have distinct effects on epidermal and dermal homeostasis, but their role in cancer invasion and epithelial-to-mesenchymal transition (EMT) remains to be determined. Methods We used 3D cultures of human skin equivalents (HSEs) to analyse the effects of Pfs and Rfs on the invasive behaviour of SCC and EMT. Results We reveal for the first time the importance of Pfs versus Rfs in SCC invasion and EMT. Cell lines from different stages of SCC showed significantly more extensive invasion into a dermis composed of Rfs than of Pfs. In addition, Rfs-based HSEs showed increased cell activation and stained positive for CAF biomarkers α-SMA and vimentin. Further analysis revealed that invasively growing cancer cells in Rf-HSEs express markers of EMT transition, like SNAIL2, N-cadherin and ZEB1. Conclusions Conversely, our results show that Pfs contain cancer cells more within the epidermis. Rfs are clearly predisposed to differentiate into CAFs upon SCC signals, assisting invasion and EMT.
Collapse
Affiliation(s)
- Marieke Hogervorst
- Department of Dermatology, Leiden University Medical Centre, 2333 ZA, Leiden, The Netherlands.,Biomimiq, J.H. Oortweg 19, 2333 CH, Leiden, The Netherlands
| | - Marion Rietveld
- Department of Dermatology, Leiden University Medical Centre, 2333 ZA, Leiden, The Netherlands
| | - Frank de Gruijl
- Department of Dermatology, Leiden University Medical Centre, 2333 ZA, Leiden, The Netherlands
| | | |
Collapse
|
35
|
Turato C, Kent P, Sebastiani G, Cannito S, Morello E, Terrin L, Biasiolo A, Simonato D, Parola M, Pantopoulos K, Pontisso P. Serpinb3 is overexpressed in the liver in presence of iron overload. J Investig Med 2017; 66:32-38. [PMID: 28935635 DOI: 10.1136/jim-2017-000473] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/02/2017] [Indexed: 11/04/2022]
Abstract
Iron overload results in cellular toxicity, tissue injury, organ fibrosis and increased risk of neoplastic transformation. SerpinB3 is a serine protease inhibitor overexpressed in the liver in oxidative stress conditions, able to induce fibrosis and increased risk of malignant transformation. Aim of the present study was to assess the effect of iron overload on SerpinB3 expression in the liver using in vivo and in vitro models.The expression of Serpinb3 was assessed in the liver of hemojuvelin knockout mice (Hjv-/-), an established model of hereditary hemochromatosis, and of wild type control mice, following dietary or pharmacological iron manipulation. To assess the direct effect of iron in vitro, cell lines were treated with different concentration of hemin or with an iron chelator.Hepatic Serpinb3 mRNA and protein were highly expressed in Hjv-/- mice, but not in wild type controls fed with a standard diet. Serpinb3 became detectable in wild type mice fed with a high iron diet or injected with iron dextran; these treatments further induced Serpinb3 expression in Hjv-/- mice. Livers expressing Serpinb3 showed a positive staining also for HIF-2α in the same areas. Hemin promoted induction of SerpinB3 mRNA in HeLa and HA22T/VGH cells, but a mild stimulation of SerpinB3 promoter activity in HeLa and Huh7 cells. In conclusion, Serpinb3 is strongly induced by iron in the mouse liver. The molecular link between iron, ROS and SerpinB3 seems to be HIF-2α, which is induced by iron overload and was previously found capable of up-regulating SerpinB3 at the transcriptional level.
Collapse
Affiliation(s)
| | | | | | - Stefania Cannito
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Elisabetta Morello
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Liliana Terrin
- Department of Medicine, University of Padova, Padova, Italy
| | | | | | - Maurizio Parola
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | | | | |
Collapse
|
36
|
Rivera C, Oliveira AK, Costa RAP, De Rossi T, Paes Leme AF. Prognostic biomarkers in oral squamous cell carcinoma: A systematic review. Oral Oncol 2017; 72:38-47. [DOI: 10.1016/j.oraloncology.2017.07.003] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 06/21/2017] [Accepted: 07/04/2017] [Indexed: 12/11/2022]
|
37
|
Avril T, Vauléon E, Chevet E. Endoplasmic reticulum stress signaling and chemotherapy resistance in solid cancers. Oncogenesis 2017; 6:e373. [PMID: 28846078 PMCID: PMC5608920 DOI: 10.1038/oncsis.2017.72] [Citation(s) in RCA: 193] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 06/01/2017] [Accepted: 07/07/2017] [Indexed: 02/07/2023] Open
Abstract
The unfolded protein response (UPR) is an adaptive cellular program used by eukaryotic cells to cope with protein misfolding stress. During tumor development, cancer cells are facing intrinsic (oncogene activation) and extrinsic (limiting nutrient or oxygen supply) challenges, with which they must cope to survive. Moreover, chemotherapy represents an additional extrinsic challenge that cancer cells are facing and to which they adapt in the case of resistance. As of today, resistance to chemotherapy and targeted therapies is one of the important issues that oncologists have to deal with for treating cancer patients. In this review, we first describe the key molecular mechanisms controlling the UPR and their implication in solid cancers. Then, we review the literature that connects cancer chemotherapy resistance mechanisms and activation of the UPR. Finally, we discuss the possible applications of targeting the UPR to bypass drug resistance.
Collapse
Affiliation(s)
- T Avril
- INSERM U1242, 'Chemistry, Oncogenesis, Stress, Signaling', Université de Rennes 1, Rennes, France.,Centre de Lutte contre le Cancer Eugène Marquis, Rennes, France
| | - E Vauléon
- INSERM U1242, 'Chemistry, Oncogenesis, Stress, Signaling', Université de Rennes 1, Rennes, France.,Centre de Lutte contre le Cancer Eugène Marquis, Rennes, France
| | - E Chevet
- INSERM U1242, 'Chemistry, Oncogenesis, Stress, Signaling', Université de Rennes 1, Rennes, France.,Centre de Lutte contre le Cancer Eugène Marquis, Rennes, France
| |
Collapse
|
38
|
Terrin L, Agostini M, Ruvoletto M, Martini A, Pucciarelli S, Bedin C, Nitti D, Pontisso P. SerpinB3 upregulates the Cyclooxygenase-2 / β-Catenin positive loop in colorectal cancer. Oncotarget 2017; 8:15732-15743. [PMID: 28178650 PMCID: PMC5362519 DOI: 10.18632/oncotarget.14997] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 01/03/2017] [Indexed: 12/30/2022] Open
Abstract
Colorectal cancer is characterized by aberrant Cyclooxigenase-2 (COX-2) and β-Catenin pathways. Recently, the protease inhibitor SerpinB3 has been described overexpressed in more advanced stages of this tumor. Aim of the study was to explore the possible relationship between these molecules in this setting. We evaluated colorectal cancer specimens from 105 patients and a positive correlation between SerpinB3, COX-2 and β-Catenin expression was observed, with higher levels in tumor than in adjacent tissue. The highest levels were associated with pathologic parameters of poor prognosis, including vascular invasion, lymph node metastasis and perineural invasion. The molecular and protein profiles of COX-2 and β-Catenin were analyzed in cell lines with different expression of SerpinB3. In those with high expression of SerpinB3, COX-2 and β-Catenin were higher than in controls. Cells with high levels of SerpinB3 showed higher proliferation and invasion compared to controls. In conclusion, in colorectal cancer SerpinB3, COX-2 and β-Catenin are positively correlated and associated with more advanced tumor stage. The in vitro experimental results support a driving role of SerpinB3 in the upregulation of COX-2/ β-Catenin positive loop, associated with a more aggressive cellular phenotype.
Collapse
Affiliation(s)
| | - Marco Agostini
- Surgery Branch, Department of Surgery Oncology and Gastroenterology, University of Padua, Italy.,Nano-Inspired Biomedicine Laboratory, Istituto di Ricerca Pediatrica - Città della Speranza, Padua, Italy.,Department of Nanomedicine, The Methodist Hospital Research Institute, Houston, TX, USA
| | | | | | - Salvatore Pucciarelli
- Surgery Branch, Department of Surgery Oncology and Gastroenterology, University of Padua, Italy
| | - Chiara Bedin
- Nano-Inspired Biomedicine Laboratory, Istituto di Ricerca Pediatrica - Città della Speranza, Padua, Italy
| | - Donato Nitti
- Surgery Branch, Department of Surgery Oncology and Gastroenterology, University of Padua, Italy
| | | |
Collapse
|
39
|
Papaioannou A, Chevet E. Driving Cancer Tumorigenesis and Metastasis Through UPR Signaling. Curr Top Microbiol Immunol 2017; 414:159-192. [PMID: 28710693 DOI: 10.1007/82_2017_36] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In the tumor microenvironment, cancer cells encounter both external and internal factors that can lead to the accumulation of improperly folded proteins in the Endoplasmic Reticulum (ER) lumen, thus causing ER stress. When this happens, an adaptive mechanism named the Unfolded Protein Response (UPR) is triggered to help the cell cope with this change and restore protein homeostasis in the ER. Sequentially, one would expect that the activation of the three UPR branches, driven namely by IRE1, PERK, and ATF6, are crucial for the adaptation of cancer cells to the changing environment and thus for their survival and further propagation. Indeed, in the last few years, an increasing amount of studies has shown the implication of UPR signaling in different aspects of carcinogenesis and tumor progression. Features such as sustaining proliferation and resistance to cell death, genomic instability, altered metabolism, increased inflammation and tumor-immune infiltration, invasion and metastasis, and angiogenesis, defined as "the hallmarks of cancer", can be regulated by the UPR machinery. At the same time, new potential therapeutic interventions applicable to different kinds of cancers are being revealed. In order to describe the emerging role of UPR in cancer biology, these are the points that will be discussed in this chapter.
Collapse
Affiliation(s)
- Alexandra Papaioannou
- Inserm U1242 «Chemistry, Oncogenesis, Stress and Signaling», University of Rennes 1, Rennes, France.,Centre de Lutte contre le Cancer Eugène Marquis, Avenue de la bataille Flandres Dunkerque, 35000, Rennes, France
| | - Eric Chevet
- Inserm U1242 «Chemistry, Oncogenesis, Stress and Signaling», University of Rennes 1, Rennes, France.
| |
Collapse
|
40
|
Oakes SA. Endoplasmic reticulum proteostasis: a key checkpoint in cancer. Am J Physiol Cell Physiol 2016; 312:C93-C102. [PMID: 27856431 DOI: 10.1152/ajpcell.00266.2016] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 11/14/2016] [Accepted: 11/15/2016] [Indexed: 01/24/2023]
Abstract
The unfolded protein response (UPR) is an intracellular signaling network largely controlled by three endoplasmic reticulum (ER) transmembrane proteins, inositol-requiring enzyme 1α, PRK-like ER kinase, and activating transcription factor 6, that monitor the protein-folding status of the ER and initiate corrective measures to maintain ER homeostasis. Hypoxia, nutrient deprivation, proteasome dysfunction, sustained demands on the secretory pathway or somatic mutations in its client proteins, conditions often encountered by cancer cells, can lead to the accumulation of misfolded proteins in the ER and cause "ER stress." Under remediable levels of ER stress, the homeostatic UPR outputs activate transcriptional and translational changes that promote cellular adaptation. However, if the ER stress is irreversible despite these measures, a terminal UPR program supersedes that actively signals cell destruction. In addition to its prosurvival and prodeath outputs, the UPR is now recognized to play a major role in the differentiation and activation of specific immune cells, as well as proinflammatory cytokine production in many cell types. Given the numerous intrinsic and extrinsic factors that threaten the fidelity of the secretory pathway in cancer cells, it is not surprising that ER stress is documented in many solid and hematopoietic malignancies, but whether ongoing UPR signaling is beneficial or detrimental to tumor growth remains hotly debated. Here I review recent evidence that cancer cells are prone to loss of proteostasis within the ER, and hence may be susceptible to targeted interventions that either reduce homeostatic UPR outputs or alternatively trigger the terminal UPR.
Collapse
Affiliation(s)
- Scott A Oakes
- Department of Pathology, Diabetes Center, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| |
Collapse
|
41
|
Xu YM, Wang HJ, Chen F, Guo WH, Wang YY, Li HY, Tang JH, Ding Y, Shen YC, Li M, Xuan WY, Liu LH, Wang J, Wang XR, Gao ZJ, Liang XB, Su DM. HRD1 suppresses the growth and metastasis of breast cancer cells by promoting IGF-1R degradation. Oncotarget 2016; 6:42854-67. [PMID: 26536657 PMCID: PMC4767476 DOI: 10.18632/oncotarget.5733] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Accepted: 10/09/2015] [Indexed: 12/22/2022] Open
Abstract
HRD1 (3-hydroxy-3-methylglutaryl reductase degradation) is an E3 ubiquitin ligase. We found that HRD1 was significantly downregulated in 170 breast cancer tissues. Low tumoral HRD1 expression was correlated with clinicopathological characteristics and a shorter survival in breast cancer patients. P65 specifically bound to the HRD1 promoter and inhibited HRD1 expression. Suppression of NF-κB activity reversed IL-6-induced downregulation of HRD1 expression. HRD1 interacted with IGF-1R and promoted its ubiquitination and degradation by the proteasome. Overexpression of HRD1 resulted in the inhibition of growth, migration and invasion of breast cancer cells in vitro and in vivo. Furthermore, HRD1 attenuated IL-6-induced epithelial-mesenchymal transition in MCF10A cells. These findings uncover a novel role for HRD1 in breast cancer.
Collapse
Affiliation(s)
- Yue-Mei Xu
- State Key Laboratory of Reproductive Medicine, Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Hong-Jiang Wang
- Department of Breast Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Fang Chen
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, China
| | - Wan-Hua Guo
- Department of Nuclear Medicine, The Affiliated Drum Tower Hospital of Nanjing University, Nanjing, China
| | - Yan-Yang Wang
- Department of Nuclear Medicine, The Affiliated Drum Tower Hospital of Nanjing University, Nanjing, China
| | - Hang-Yu Li
- Department of General Surgery, 4th Affiliated Hospital, China Medical University, Shenyang, China
| | - Jin-Hai Tang
- Department of General Surgery, The Affiliated Jiangsu Cancer Hospital, Nanjing Medical University, Nanjing, China
| | - Ying Ding
- State Key Laboratory of Reproductive Medicine, Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Ya-Chen Shen
- Center of Metabolic Research, Nanjing Medical University, Nanjing, China
| | - Min Li
- State Key Laboratory of Reproductive Medicine, Department of Pathology, Nanjing Medical University, Nanjing, China.,Center of Metabolic Research, Nanjing Medical University, Nanjing, China
| | - Wen-Ying Xuan
- State Key Laboratory of Reproductive Medicine, Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Lin-Hui Liu
- State Key Laboratory of Reproductive Medicine, Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Jia Wang
- Department of Breast Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xue-Rong Wang
- State Key Laboratory of Reproductive Medicine, Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Ze-Jun Gao
- State Key Laboratory of Reproductive Medicine, Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Xiu-Bin Liang
- Center of Metabolic Research, Nanjing Medical University, Nanjing, China
| | - Dong-Ming Su
- State Key Laboratory of Reproductive Medicine, Department of Pathology, Nanjing Medical University, Nanjing, China.,Center of Metabolic Research, Nanjing Medical University, Nanjing, China.,Center of Cellular therapy, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
42
|
Sun Y, Sheshadri N, Zong WX. SERPINB3 and B4: From biochemistry to biology. Semin Cell Dev Biol 2016; 62:170-177. [PMID: 27637160 DOI: 10.1016/j.semcdb.2016.09.005] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 09/08/2016] [Accepted: 09/12/2016] [Indexed: 12/15/2022]
Abstract
Human SERPINB3 and SERPINB4 are evolutionary duplicated serine/cysteine protease inhibitors. Genomic analysis indicates that these paralogous genes were encoded from independent loci arising from tandem gene duplication. Although the two molecules share 92% identity of their amino acid sequences, they are distinct in the Reactive Center Loop (RCL) including a hinge region and catalytic sequences which accounts for altered substrate specificity. Elevated expression of the two molecules has been reported to contribute to numerous pathological conditions such as inflammatory diseases and cancer. In this review, we focus on summarizing the biochemical features of SERPINB3/B4 and discussing the mechanistic basis for their biological functions and implications in human diseases.
Collapse
Affiliation(s)
- Yu Sun
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, United States
| | - Namratha Sheshadri
- National Cancer Institute, Center for Cancer Research, Frederick, MD 21702, United States
| | - Wei-Xing Zong
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, United States; Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, United States.
| |
Collapse
|
43
|
Le Reste PJ, Avril T, Quillien V, Morandi X, Chevet E. Reprint of: Signaling the Unfolded Protein Response in primary brain cancers. Brain Res 2016; 1648:542-552. [PMID: 27362469 DOI: 10.1016/j.brainres.2016.06.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 03/16/2016] [Accepted: 03/17/2016] [Indexed: 11/15/2022]
Abstract
The Unfolded Protein Response (UPR) is an adaptive cellular program used by eukaryotic cells to cope with protein misfolding stress in the Endoplasmic Reticulum (ER). During tumor development, cancer cells are facing intrinsic (oncogene activation) and extrinsic (limiting nutrient or oxygen supply; exposure to chemotherapies) challenges, with which they must cope to survive. Primary brain tumors are relatively rare but deadly and present a significant challenge in the determination of risk factors in the population. These tumors are inherently difficult to cure because of their protected location in the brain. As such surgery, radiation and chemotherapy options carry potentially lasting patient morbidity and incomplete tumor cure. Some of these tumors, such as glioblastoma, were reported to present features of ER stress and to depend on UPR activation to sustain growth, but to date there is no clear general representation of the ER stress status in primary brain tumors. In this review, we describe the key molecular mechanisms controlling the UPR and their implication in cancers. Then we extensively review the literature reporting the status of ER stress in various primary brain tumors and discuss the potential impact of such observation on patient stratification and on the possibility of developing appropriate targeted therapies using the UPR as therapeutic target. This article is part of a Special Issue entitled SI:ER stress.
Collapse
Affiliation(s)
- Pierre-Jean Le Reste
- Department of Neurosurgery, University Hospital Pontchaillou, Rennes, France; Inserm ERL440 "Oncogenesis, Stress, Signaling", Université de Rennes 1, Rennes, France
| | - Tony Avril
- Inserm ERL440 "Oncogenesis, Stress, Signaling", Université de Rennes 1, Rennes, France; Centre de Lutte Contre le Cancer Eugène Marquis, Rennes, France
| | - Véronique Quillien
- Inserm ERL440 "Oncogenesis, Stress, Signaling", Université de Rennes 1, Rennes, France; Centre de Lutte Contre le Cancer Eugène Marquis, Rennes, France
| | - Xavier Morandi
- Department of Neurosurgery, University Hospital Pontchaillou, Rennes, France
| | - Eric Chevet
- Inserm ERL440 "Oncogenesis, Stress, Signaling", Université de Rennes 1, Rennes, France; Centre de Lutte Contre le Cancer Eugène Marquis, Rennes, France.
| |
Collapse
|
44
|
Zuo M, Rashid A, Wang Y, Jain A, Li D, Behari A, Kapoor VK, Koay EJ, Chang P, Vauthey JN, Li Y, Espinoza JA, Roa JC, Javle M. RNA sequencing-based analysis of gallbladder cancer reveals the importance of the liver X receptor and lipid metabolism in gallbladder cancer. Oncotarget 2016; 7:35302-12. [PMID: 27167107 PMCID: PMC5085230 DOI: 10.18632/oncotarget.9181] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 04/16/2016] [Indexed: 12/24/2022] Open
Abstract
Gallbladder cancer (GBC) is an aggressive malignancy. Although surgical resection may be curable, most patients are diagnosed at an advanced unresectable disease stage. Cholelithiasis is the major risk factor; however the pathogenesis of the disease, from gallstone cholecystitis to cancer, is still not understood. To understand the molecular genetic underpinnings of this cancer and explore novel therapeutic targets for GBC, we examined the key genes and pathways involved in GBC using RNA sequencing. We performed gene expression analysis of 32 cases of surgically-resected GBC along with normal gallbladder tissue controls. We observed that 519 genes were differentially expressed between GBC and normal GB mucosal controls. The liver X receptor (LXR)/retinoid X receptor (RXR) and farnesoid X receptor (FXR) /RXR pathways were the top canonical pathways involved in GBC. Key genes in these pathways, including SERPINB3 and KLK1, were overexpressed in GBC, especially in female GBC patients. Additionally, ApoA1 gene expression suppressed in GBC as compared with normal control tissues. LXR and FXR genes, known to be important in lipid metabolism also function as tumor suppressors and their down regulation appears to be critical for GBC pathogenesis. LXR agonists may have therapeutic value and as potential therapeutic targets.
Collapse
Affiliation(s)
- Mingxin Zuo
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Asif Rashid
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ying Wang
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Apurva Jain
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Donghui Li
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Anu Behari
- Department of Surgical Gastroenterology, Sanjay Gandhi Post-Graduate Institute of Medical Sciences (SGPGIMS), Lucknow, UP, India
| | - Vinay Kumar Kapoor
- Department of Surgical Gastroenterology, Sanjay Gandhi Post-Graduate Institute of Medical Sciences (SGPGIMS), Lucknow, UP, India
| | - Eugene J. Koay
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ping Chang
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jean Nicholas Vauthey
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yanan Li
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jaime A. Espinoza
- SciLifeLab, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Stockholm, Sweden
| | - Juan Carlos Roa
- Department of Pathology, Advanced Center for Chronic Diseases (ACCDiS), UC-Center for Investigational Oncology (CITO), School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Milind Javle
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
45
|
Urra H, Dufey E, Avril T, Chevet E, Hetz C. Endoplasmic Reticulum Stress and the Hallmarks of Cancer. Trends Cancer 2016; 2:252-262. [PMID: 28741511 DOI: 10.1016/j.trecan.2016.03.007] [Citation(s) in RCA: 398] [Impact Index Per Article: 44.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 03/28/2016] [Accepted: 03/29/2016] [Indexed: 12/13/2022]
Abstract
Tumor cells are often exposed to intrinsic and external factors that alter protein homeostasis, thus producing endoplasmic reticulum (ER) stress. To cope with this, cells evoke an adaptive mechanism to restore ER proteostasis known as the unfolded protein response (UPR). The three main UPR signaling branches initiated by IRE1α, PERK, and ATF6 are crucial for tumor growth and aggressiveness as well as for microenvironment remodeling or resistance to treatment. We provide a comprehensive overview of the contribution of the UPR to cancer biology and the acquisition of malignant characteristics, thus highlighting novel aspects including inflammation, invasion and metastasis, genome instability, resistance to chemo/radiotherapy, and angiogenesis. The therapeutic potential of targeting ER stress signaling in cancer is also discussed.
Collapse
Affiliation(s)
- Hery Urra
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Estefanie Dufey
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Tony Avril
- Institut National de la Santé et de la Recherche Médicale (INSERM) Equipe de Recherche Labellisée (ERL) 440-Oncogenesis, Stress, and Signaling, University of Rennes 1, 35000 Rennes, France; Centre de Lutte Contre le Cancer Eugène Marquis, Rennes, France
| | - Eric Chevet
- Institut National de la Santé et de la Recherche Médicale (INSERM) Equipe de Recherche Labellisée (ERL) 440-Oncogenesis, Stress, and Signaling, University of Rennes 1, 35000 Rennes, France; Centre de Lutte Contre le Cancer Eugène Marquis, Rennes, France
| | - Claudio Hetz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile; Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA 02115, USA; Buck Institute for Research on Aging, Novato, CA 94945, USA.
| |
Collapse
|
46
|
Adel M, Tsao CK, Wei FC, Chien HT, Lai CH, Liao CT, Wang HM, Fan KH, Kang CJ, Chang JTC, Huang SF. Preoperative SCC Antigen, CRP Serum Levels, and Lymph Node Density in Oral Squamous Cell Carcinoma. Medicine (Baltimore) 2016; 95:e3149. [PMID: 27057838 PMCID: PMC4998754 DOI: 10.1097/md.0000000000003149] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 02/24/2016] [Accepted: 02/24/2016] [Indexed: 01/29/2023] Open
Abstract
The prognostic significance of squamous cell carcinoma antigen (SCC-Ag) and C-reactive protein (CRP) levels and lymph node density (LND) has been individually recognized in oral squamous cell carcinoma (OSCC). We investigated the relationship between preoperative serum markers (SCC-Ag and CRP) and postoperative prognostic marker (LND) in this study. We retrospectively analyzed 277 OSCC patients who underwent primary curative resection and neck dissection with/or without adjuvant therapy between March 2008 and November 2013. Serum SCC-Ag and CRP levels were measured preoperatively. Distant metastasis, overall survival (OS), and disease-free survival (DFS) were used to evaluate the prognostic significance of preoperative SCC-Ag and CRP levels in relation to LND. LND (cutoff point ≥0.06) correlated with the pathologic tumor status, pathologic nodal metastasis, degree of differentiation, tumor stage, tumor depth (≥10 mm vs <10 mm), and perineural invasion (all P values were <0.001). LND was significantly associated with development of distant metastasis, DFS, and OS (all P values were <0.001). Preoperative elevated CRP and SCC-Ag levels were significantly associated with LND (P = 0.006), DFS (P < 0.001), and OS (P < 0.001). LND patients were further stratified into prognostic groups according to their SCC-Ag and CRP levels (DFS: P = 0.010; OS: P = 0.003). LND correlated with the incidence of DM, DFS, and OS in patients with OSCC. Concurrent elevated preoperative SCC-Ag and CRP levels are predictors for LND. In addition, SCC-Ag and CRP are markers for classifying high-risk LND patients with OSCC into subgroups.
Collapse
Affiliation(s)
- Mohamad Adel
- From the Department of Surgery (MA), Division of Surgical Oncology, Al-Azhar University Hospitals, Al-Azhar Faculty of Medicine, Cairo, Egypt, Department of Otolaryngology (MA, CTL, CJK, SFH), Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital at Linkou and Chang Gung University (CKT, FCW), Department of Epidemiology (HTC, CHL), Department of Medical Oncology (HMW), and Department of Radiation Oncology (KHF, JTC), Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Le Reste PJ, Avril T, Quillien V, Morandi X, Chevet E. Signaling the Unfolded Protein Response in primary brain cancers. Brain Res 2016; 1642:59-69. [PMID: 27016056 DOI: 10.1016/j.brainres.2016.03.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 03/16/2016] [Accepted: 03/17/2016] [Indexed: 12/14/2022]
Abstract
The Unfolded Protein Response (UPR) is an adaptive cellular program used by eukaryotic cells to cope with protein misfolding stress in the Endoplasmic Reticulum (ER). During tumor development, cancer cells are facing intrinsic (oncogene activation) and extrinsic (limiting nutrient or oxygen supply; exposure to chemotherapies) challenges, with which they must cope to survive. Primary brain tumors are relatively rare but deadly and present a significant challenge in the determination of risk factors in the population. These tumors are inherently difficult to cure because of their protected location in the brain. As such surgery, radiation and chemotherapy options carry potentially lasting patient morbidity and incomplete tumor cure. Some of these tumors, such as glioblastoma, were reported to present features of ER stress and to depend on UPR activation to sustain growth, but to date there is no clear general representation of the ER stress status in primary brain tumors. In this review, we describe the key molecular mechanisms controlling the UPR and their implication in cancers. Then we extensively review the literature reporting the status of ER stress in various primary brain tumors and discuss the potential impact of such observation on patient stratification and on the possibility of developing appropriate targeted therapies using the UPR as therapeutic target.
Collapse
Affiliation(s)
- Pierre-Jean Le Reste
- Department of Neurosurgery, University Hospital Pontchaillou, Rennes, France; Inserm ERL440 "Oncogenesis, Stress, Signaling", Université de Rennes 1, Rennes, France
| | - Tony Avril
- Inserm ERL440 "Oncogenesis, Stress, Signaling", Université de Rennes 1, Rennes, France; Centre de Lutte Contre le Cancer Eugène Marquis, Rennes, France
| | - Véronique Quillien
- Inserm ERL440 "Oncogenesis, Stress, Signaling", Université de Rennes 1, Rennes, France; Centre de Lutte Contre le Cancer Eugène Marquis, Rennes, France
| | - Xavier Morandi
- Department of Neurosurgery, University Hospital Pontchaillou, Rennes, France
| | - Eric Chevet
- Inserm ERL440 "Oncogenesis, Stress, Signaling", Université de Rennes 1, Rennes, France; Centre de Lutte Contre le Cancer Eugène Marquis, Rennes, France.
| |
Collapse
|
48
|
Bharti R, Dey G, Mandal M. Cancer development, chemoresistance, epithelial to mesenchymal transition and stem cells: A snapshot of IL-6 mediated involvement. Cancer Lett 2016; 375:51-61. [PMID: 26945971 DOI: 10.1016/j.canlet.2016.02.048] [Citation(s) in RCA: 174] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Revised: 02/24/2016] [Accepted: 02/24/2016] [Indexed: 12/12/2022]
Abstract
Interleukin-6 (IL-6) is a cytokine present in tumor microenvironment. Elevated level of IL-6 is associated with cancer cell proliferation, angiogenesis and metastasis through fueling STAT3, MAPK and Akt signaling. It promotes epithelial to mesenchymal transition (EMT) through altered expression of N-cadherin, vimentin, snail, twist and E-cadherin leading to cancer metastasis. IL-6 boosts mammosphere formation, self-renewal of stem cells, stemness properties of cancer cells and recruitment of mesenchymal stem cells. IL-6 is also a contributing factor for multidrug resistance in cancer due to gp130/MAPK/STAT3 mediated activation of transcription factors C/EBPβ/δ, overexpression of p-glycoprotein, EMT transition and expansion of stem cells. The in-depth investigation of IL-6 mediated cellular effects and its signaling pathway can provide the new window for future research and clinical development of IL-6 targeted therapy in cancer. Thus, an overview is delivered in this review deciphering the emerging aspect of the predominant influence of IL-6 in malignant transformation, EMT, cancer-associated stem cells and chemoresistance.
Collapse
Affiliation(s)
- Rashmi Bharti
- School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur 721302, West Bengal, India
| | - Goutam Dey
- School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur 721302, West Bengal, India
| | - Mahitosh Mandal
- School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur 721302, West Bengal, India.
| |
Collapse
|
49
|
Biasiolo A, Trotta E, Fasolato S, Ruvoletto M, Martini A, Gallotta A, Fassina G, Angeli P, Gatta A, Pontisso P. Squamous cell carcinoma antigen-IgM is associated with hepatocellular carcinoma in patients with cirrhosis: A prospective study. Dig Liver Dis 2016; 48:197-202. [PMID: 26614642 DOI: 10.1016/j.dld.2015.10.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 09/29/2015] [Accepted: 10/23/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND Squamous cell carcinoma antigen (SCCA)-IgM complex has been described as a promising tool to identify patients with progressive liver disease at higher risk of hepatocellular carcinoma (HCC) development in retrospective studies. AIM To assess the clinical value of this biomarker in patients with cirrhosis in a prospective study. METHODS Patients with overt cirrhosis were prospectively evaluated at 6-month intervals for HCC development and decompensation with clinical examination, liver ultrasound, α-fetoprotein measurement. SCCA-IgM was measured in serum by immunoenzymatic assay. Median follow-up duration was 52 months (range 12-68 months). RESULTS 70 patients (26% male; mean age 56±10 years) were enrolled. The main aetiological factors were alcohol (44%) and hepatitis C (34%). Baseline values of SCCA-IgM were significantly higher in patients who developed HCC. Positivity of the biomarker at baseline was associated with a significantly shorter HCC-free survival, while α-fetoprotein (cut off >20 ng/ml) was not significant. SCCA-IgM positivity and hepatitis C were significant prognostic factors for HCC development. The biomarker was not associated with the development of clinical complications of cirrhosis. CONCLUSION This prospective study demonstrates that in patients with cirrhosis SCCA-IgM is associated with HCC development and may be useful for clinical management of cirrhotic patients at higher risk of HCC development.
Collapse
Affiliation(s)
| | - Elisa Trotta
- Department of Medicine, University of Padua, Italy
| | | | | | | | | | | | - Paolo Angeli
- Department of Medicine, University of Padua, Italy
| | - Angelo Gatta
- Department of Medicine, University of Padua, Italy
| | | |
Collapse
|
50
|
Turato C, Cannito S, Simonato D, Villano G, Morello E, Terrin L, Quarta S, Biasiolo A, Ruvoletto M, Martini A, Fasolato S, Zanus G, Cillo U, Gatta A, Parola M, Pontisso P. SerpinB3 and Yap Interplay Increases Myc Oncogenic Activity. Sci Rep 2015; 5:17701. [PMID: 26634820 PMCID: PMC4669520 DOI: 10.1038/srep17701] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 10/29/2015] [Indexed: 01/03/2023] Open
Abstract
SerpinB3 has been recently described as an early marker of liver carcinogenesis, but the potential mechanistic role of this serpin in tumor development is still poorly understood. Overexpression of Myc often correlates with more aggressive tumour forms, supporting its involvement in carcinogenesis. Yes-associated protein (Yap), the main effector of the Hippo pathway, is a central regulator of proliferation and it has been found up-regulated in hepatocellular carcinomas. The study has been designed to investigate and characterize the interplay and functional modulation of Myc by SerpinB3 in liver cancer. Results from this study indicate that Myc was up-regulated by SerpinB3 through calpain and Hippo-dependent molecular mechanisms in transgenic mice and hepatoma cells overexpressing human SerpinB3, and also in human hepatocellular carcinomas. Human recombinant SerpinB3 was capable to inhibit the activity of Calpain in vitro, likely reducing its ability to cleave Myc in its non oncogenic Myc-nick cytoplasmic form. SerpinB3 indirectly increased the transcription of Myc through the induction of Yap pathway. These findings provide for the first time evidence that SerpinB3 can improve the production of Myc through direct and indirect mechanisms that include the inhibition of generation of its cytoplasmic form and the activation of Yap pathway.
Collapse
Affiliation(s)
| | - Stefania Cannito
- Dept. of Clinical and Biological Sciences, Unit of Experimental Medicine and Interuniversity Center for Liver Pathophysiology, University of Torino, Italy
| | | | | | - Elisabetta Morello
- Dept. of Clinical and Biological Sciences, Unit of Experimental Medicine and Interuniversity Center for Liver Pathophysiology, University of Torino, Italy
| | | | | | | | | | | | | | - Giacomo Zanus
- Unit of Hepatobiliary Surgery and Liver Transplantation, University of Padova, Italy
| | - Umberto Cillo
- Unit of Hepatobiliary Surgery and Liver Transplantation, University of Padova, Italy
| | | | - Maurizio Parola
- Dept. of Clinical and Biological Sciences, Unit of Experimental Medicine and Interuniversity Center for Liver Pathophysiology, University of Torino, Italy
| | | |
Collapse
|