1
|
Zhu W, Qian H, Cao S, Xia W, Wang X, Jin J, Wang X, Zhang H, Liu D, Chen Y. A new platform of electrowetting-on-dielectric digital microfluidics for rapid detection of early-stage Hepatocellular Carcinoma(HCC) specific biomarker. Anal Chim Acta 2025; 1336:343533. [PMID: 39788685 DOI: 10.1016/j.aca.2024.343533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/06/2024] [Accepted: 12/06/2024] [Indexed: 01/30/2025]
Abstract
BACKGROUND The early detection of Hepatocellular Carcinoma (HCC) is crucial for improving patient survival rates.Early diagnosis of HCC can significantly enhance treatment outcomes and reduce disease progression. Antigen detection of tumor markers is one of the important diagnostic methods for HCC. However, Traditional antigen detection methods often rely on heavy detection equipment, involve lengthy turnaround times, and must be conducted in laboratory settings. Therefore, there is a clear need for a portable, low-skill, rapid sample-to-result detection method for early HCC biomarkers. RESULTS We propose a new platform based on electrowetting-on-dielectric digital microfluidic(EWOD-DMF) for the detection of early-stage HCC biomarkers, enabling the quantitative measurement of Alpha-Fetoprotein (AFP), the proportion of AFP-L3 in total AFP (AFP-L3%), and Des-Gamma-Carboxy Prothrombin (DCP). First, serum samples are processed through the microfluidic system, achieving the separation of AFP-L3 within 10 min. Next, immunoassays are performed within 15 min, using magnetic particles to capture biomarkers such as AFP, AFP-L3, and DCP, followed by enzymatic reactions that generate detectable signals. Each chip can simultaneously detect three biomarkers from five different samples, allowing for a total of fifteen targets to be tested, with only approximately 2.4 μL of serum required for each biomarker detection. Ultimately, data are analyzed with dedicated software to quantitatively measure the HCC biomarkers. The detection limits for AFP or AFP-L3 and for DCP are 0.24 ng/mL and 1.89 ng/mL, respectively. SIGNIFICANCE This study presents a EWOD-DMF platform for early-stage HCC diagnosis, capable of simultaneously detecting multiple samples and biomarkers, thus improving detection efficiency and diagnostic accuracy. Moreover, the platform has POCT capability, with advantages in portability and cost-effectiveness, providing clinicians and primary healthcare institutions with a fast and convenient solution for early-stage HCC diagnosis.
Collapse
Affiliation(s)
- Wenjie Zhu
- School of Medical Technology, Xuzhou Medical University, Xuzhou, 221004, China
| | - Hong Qian
- School of Medical Technology, Xuzhou Medical University, Xuzhou, 221004, China; Department of Laboratory Medicine, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, 223800, China
| | - Shengya Cao
- Department of Clinical Laboratory, Xuzhou Cancer Hospital, Xuzhou, 221004, China
| | - Wei Xia
- Nanjing RealMind Biotech Co., Ltd., Nanjing, 210046, China
| | - Xilong Wang
- Nanjing RealMind Biotech Co., Ltd., Nanjing, 210046, China
| | - Jing Jin
- Nanjing RealMind Biotech Co., Ltd., Nanjing, 210046, China
| | - Xin Wang
- Department of Molecular Diagnostics, Roche Diagnostics (Shanghai) Limited Company, Shanghai, 200131, China
| | - Hao Zhang
- Thoracic Surgery Laboratory, Xuzhou Medical University, Xuzhou, 221004, China; Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221000, China
| | - Dongsheng Liu
- Department of Laboratory Medicine, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, 223800, China
| | - Ying Chen
- School of Medical Technology, Xuzhou Medical University, Xuzhou, 221004, China.
| |
Collapse
|
2
|
Caltavituro A, Buonaiuto R, Salomone F, Pecoraro G, Martorana F, Lauro VD, Barchiesi G, Puglisi F, Del Mastro L, Montemurro F, Giuliano M, Arpino G, De Laurentiis M. Warming-up the immune cell engagers (ICEs) era in breast cancer: state of the art and future directions. Crit Rev Oncol Hematol 2024; 206:104577. [PMID: 39613237 DOI: 10.1016/j.critrevonc.2024.104577] [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: 07/22/2024] [Revised: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 12/01/2024] Open
Abstract
The advent of immune checkpoint inhibitors (ICIs) has deeply reshaped the therapeutic algorithm of triple-negative breast cancer (TNBC). However, there is considerable scope for better engagement of the immune system in other BC subtypes. ICIs have paved the way for investigations into emerging immunotherapeutic strategies, such as immune cell engagers (ICEs) that work by promoting efficient tumor cell killing through the redirection of immune system against cancer cells. Most ICEs are bispecific antibodies that simultaneously recognize and bind to both cancer and immune cells generating an artificial synapse. Major side effects are cytokine release syndrome, hepatotoxicity, and neurotoxicity related to inappropriate immune system activation. Here, we provide a comprehensive overview of this compounds, the available preclinical and clinical evidence supporting their investigation and development in BC also highlighting the challenges that have prevented their widespread use in oncology. Finally, major strategies are explored to broaden their use in BC.
Collapse
Affiliation(s)
- Aldo Caltavituro
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Via Sergio Pansini 5, Naples 80131, Italy; Clinical and Translational Oncology, Scuola Superiore Meridionale, Naples, Italy
| | - Roberto Buonaiuto
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Via Sergio Pansini 5, Naples 80131, Italy; Clinical and Translational Oncology, Scuola Superiore Meridionale, Naples, Italy
| | - Fabio Salomone
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Via Sergio Pansini 5, Naples 80131, Italy
| | - Giovanna Pecoraro
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Via Sergio Pansini 5, Naples 80131, Italy
| | - Federica Martorana
- Department of Clinical and Experimental Medicine, University of Catania, Catania 95123, Italy
| | - Vincenzo Di Lauro
- Department of Breast & Thoracic Oncology, Division of Breast Medical Oncology, Istituto Nazionale Tumori IRCCS 'Fondazione G. Pascale,' Naples, Italy.
| | - Giacomo Barchiesi
- Azienda Ospedaliera Universitaria Policlinico Umberto I, UOC Oncologia, Roma, Italy
| | - Fabio Puglisi
- Department of Medicine, University of Udine, Via Palladio 8, Udine 33100, Italy; Department of Medical Oncology, CRO Aviano, National Cancer Institute, IRCCS, Via Franco Gallini 2, Aviano, Pordenone 33081, Italy
| | - Lucia Del Mastro
- Department of Medical Oncology, UO Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, Genova 16132, Italy
| | - Filippo Montemurro
- Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142 -KM 3.95, Candiolo, Torino 10060, Italy
| | - Mario Giuliano
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Via Sergio Pansini 5, Naples 80131, Italy
| | - Grazia Arpino
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Via Sergio Pansini 5, Naples 80131, Italy
| | - Michelino De Laurentiis
- Department of Breast & Thoracic Oncology, Division of Breast Medical Oncology, Istituto Nazionale Tumori IRCCS 'Fondazione G. Pascale,' Naples, Italy
| |
Collapse
|
3
|
Zhou J, Cao C, Liu T, Wu L, Miao J. Association Between Free Triiodothyronine and Carcinoembryonic Antigen Levels in Type 2 Diabetes Mellitus Patients. Diabetes Metab Syndr Obes 2024; 17:4267-4275. [PMID: 39564449 PMCID: PMC11575458 DOI: 10.2147/dmso.s481433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 11/01/2024] [Indexed: 11/21/2024] Open
Abstract
Background Thyroid dysfunction is more common in other endocrine disorders such as diabetes mellitus (DM). Carcinoembryonic antigen (CEA), a common tumor biomarker, is found elevated in patients with thyroid dysfunction. However, the relationship between thyroid hormone levels and CEA levels remains unclear. Methods In total, 663 patients with type 2 diabetes at the Tongzhou Branch of Dongzhimen Hospital were enrolled in this retrospective study. Data were collected from inpatient electronic files between December 2011 and December 2019. Laboratory indices were statistically analyzed using logistic regression and Spearman correlation analyses. Results In our study, total triiodothyronine (TT3), free triiodothyronine (FT3), serum albumin(ALB), total protein (TP), and triglyceride (TG) levels were significantly higher in the T2DM patients with normal values of CEA than T2DM patients who had abnormal values of CEA, whereas alkaline phosphatase (ALP), Glucose (GLU), and HbA1c levels were significantly increased in the T2DM patients with abnormal CEA level. Binary logistic regression analysis demonstrated that FT3, GLU, HbA1c, and TG levels remained as independent risk factors for CEA in patients with T2DM (β=-0.907, P =0.004; β =-1.009, P=0.004; β =0.090, P = 0001; β= 0.336, P <0.001; β= -0.293, P =0.009, resp). Spearman correlation analysis showed that CEA level was significantly positively correlated with HbA1c and GLU (rs value: 0.265, P <0.001; rs value: 0.270, P <0.001, resp.) and negatively correlated with FT3 and TG levels (rs value: -0.139, P <0.001; rs value: -0.103 P =0.008, resp). Furthermore, multivariate logistic regression analysis indicated that the FT3 quartiles were significantly associated with CEA levels before and after adjusting for confounding factors. Conclusion Our study determined that FT3 remained an independent risk factor for CEA in patients with T2DM and was significantly negatively correlated with CEA levels.
Collapse
Affiliation(s)
- Jingxin Zhou
- Department of Nephrology and Endocrinology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 101121, People's Republic of China
| | - Can Cao
- Department of Nephrology and Endocrinology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 101121, People's Republic of China
| | - Tonghua Liu
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Lili Wu
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Juan Miao
- Department of Nephrology and Endocrinology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 101121, People's Republic of China
| |
Collapse
|
4
|
Akinrinde AS, Oyewole SO, Adekanmbi AO. Grape seed oil attenuates sodium arsenite-induced gastric, hepatic and colonic damage in Wistar rats. Biotech Histochem 2024; 99:414-425. [PMID: 39514780 DOI: 10.1080/10520295.2024.2426049] [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] [Indexed: 11/16/2024] Open
Abstract
Arsenic exposure is associated with numerous morbidities due to dysfunction of various organ systems including the gastrointestinal tract. We investigated the protective effect of grape seed oil (GSO) against sodium arsenite (NaAsO2)-induced gastric, hepatic and colonic injuries in rats. Twenty-four male Wistar rats were divided into four groups of six as follows: Group A (control) received saline; group B received NaAsO2 (2.5 mg/kg) orally for 7 days; group C were treated concurrently with NaAsO2 and GSO (2 ml/kg), while group D received only GSO. Administration of NaAsO2 induced significant (p < 0.05) increases in alanine aminotransferase (ALT) and aspartate aminotransferase (AST); increased periodic acid Schiff (PAS) staining for mucus and increased goblet cell numbers in the stomach and colon; inflammatory cell infiltration and vascular congestion and alterations in the fecal bacterial flora. GSO supplementation generally promoted a reversal of changes induced by NaAsO2 towards control levels. Additionally, there was increased immunohistochemically detected expression of colonic B-cell lymphoma-1 (Bcl-2) and cytokeratins AE1/AE3, but reduced expression of mucin 1 (MUC1) and carcinoembryonic antigen (CEA) in NaAsO2 + GSO and GSO treated rats when compared with the NaAsO2 group. These results suggest that GSO promoted anti-inflammatory processes in the liver, stomach and colon, as well as opposing apoptosis in the colon, resulting in significant attenuation of damage to these tissues.
Collapse
Affiliation(s)
- Akinleye Stephen Akinrinde
- Gastrointestinal and Environmental Toxicology Laboratory, Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Stephen Oluwasemiloore Oyewole
- Gastrointestinal and Environmental Toxicology Laboratory, Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Abimbola Olumide Adekanmbi
- Environmental Microbiology and Biotechnology Laboratory, Department of Microbiology, Faculty of Science, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
5
|
Yang X, Zhang Z, Bi X. A nomogram for predicting colorectal cancer liver metastasis using circulating tumor cells from the first drainage vein. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:108579. [PMID: 39121633 DOI: 10.1016/j.ejso.2024.108579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 03/05/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024]
Abstract
PURPOSE To use circulating tumor cells (CTC) from the first drainage vein (FDV) of the primary lesion and other clinically relevant parameters to construct a nomogram for predicting liver metastasis in colorectal cancer (CRC) patients, and to provide a theoretical basis for clinical diagnosis and treatment. METHODS Information from 343 CRC patients was collected and a database was established. Multivariate logistic analysis was used to identify independent factors for colorectal cancer liver metastasis(mCRC) and nomograms were constructed. Receiver operating characteristic curves(ROC), calibration plots, and decision curve analysis (DCA) were used to assess discrimination, agreement with actual risk, and the clinical utility of the prediction model, respectively. RESULT CTC levels in FDV were significantly higher in patients with liver metastasis than in those without liver metastasis. Logistic multivariate analysis showed that vascular invasion, T stage, carcinoembryonic antigen (CEA), CA19-9, and CTC could be used as predictors to construct nomograms. The nomograms showed good discriminatory ability in predicting mCRC, with area under the curve (AUC) values of 0.871 [95 % CI: 0.817-0.924) and 0.891 (95 % CI: 0.817-0.964) for the training and validation sets, respectively.] The calibration curves of both the training and validation sets showed that the model was effective in predicting the probability of mCRC. DCA was used to evaluate this predictive model and showed good net clinical benefit. CONCLUSION We developed and validated a nomogram model based on the combination of CTC in the FDV with other clinical parameters to better predict the occurrence of mCRC.
Collapse
Affiliation(s)
- Xiaoyu Yang
- Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, PR China
| | - Zhongguo Zhang
- Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, PR China.
| | - Xue Bi
- Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, PR China.
| |
Collapse
|
6
|
Wu K, Zhang G, Shen C, Zhu L, Yu C, Sartorius K, Ding W, Jiang Y, Lu Y. Role of T cells in liver metastasis. Cell Death Dis 2024; 15:341. [PMID: 38755133 PMCID: PMC11099083 DOI: 10.1038/s41419-024-06726-2] [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: 12/20/2023] [Revised: 04/24/2024] [Accepted: 05/07/2024] [Indexed: 05/18/2024]
Abstract
The liver is a major metastatic site (organ) for gastrointestinal cancers (such as colorectal, gastric, and pancreatic cancers) as well as non-gastrointestinal cancers (such as lung, breast, and melanoma cancers). Due to the innate anatomical position of the liver, the apoptosis of T cells in the liver, the unique metabolic regulation of hepatocytes and other potential mechanisms, the liver tends to form an immunosuppressive microenvironment and subsequently form a pre-metastatic niche (PMN), which can promote metastasis and colonization by various tumor cells(TCs). As a result, the critical role of immunoresponse in liver based metastasis has become increasingly appreciated. T cells, a centrally important member of adaptive immune response, play a significant role in liver based metastases and clarifying the different roles of the various T cells subsets is important to guide future clinical treatment. In this review, we first introduce the predisposing factors and related mechanisms of liver metastasis (LM) before introducing the PMN and its transition to LM. Finally, we detail the role of different subsets of T cells in LM and advances in the management of LM in order to identify potential therapeutic targets for patients with LM.
Collapse
Affiliation(s)
- Kejia Wu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Guozhu Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
- Department of Emergency Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Changbing Shen
- Department of Hepatobiliary and Pancreatic Surgery, Taizhou Second People's Hospital Affiliated with Yangzhou University, Taizhou, China
| | - Li Zhu
- Department of Hepatobiliary and Pancreatic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
- Department of Emergency Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Chongyuan Yu
- Department of Hepatobiliary and Pancreatic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Kurt Sartorius
- School of Laboratory Medicine and Molecular Sciences, University of Kwazulu-Natal, Durban, South Africa
- Africa Hepatopancreatobiliary Cancer Consortium, Mayo Clinic, Jacksonville, FL, USA
| | - Wei Ding
- Department of General Surgery, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China.
- Department of General Surgery, The Wujin Clinical College of Xuzhou Medical University, Changzhou, China.
- Changzhou Medical Center, Nanjing Medical University, Changzhou, China.
| | - Yong Jiang
- Department of Hepatobiliary and Pancreatic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, China.
| | - Yunjie Lu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China.
- Africa Hepatopancreatobiliary Cancer Consortium, Mayo Clinic, Jacksonville, FL, USA.
- Department of General Surgery, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China.
| |
Collapse
|
7
|
Lazzarato L, Bianchi L, Andolfo A, Granata A, Lombardi M, Sinelli M, Rolando B, Carini M, Corsini A, Fruttero R, Arnaboldi L. Proteomics Studies Suggest That Nitric Oxide Donor Furoxans Inhibit In Vitro Vascular Smooth Muscle Cell Proliferation by Nitric Oxide-Independent Mechanisms. Molecules 2023; 28:5724. [PMID: 37570694 PMCID: PMC10420201 DOI: 10.3390/molecules28155724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/19/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Physiologically, smooth muscle cells (SMC) and nitric oxide (NO) produced by endothelial cells strictly cooperate to maintain vasal homeostasis. In atherosclerosis, where this equilibrium is altered, molecules providing exogenous NO and able to inhibit SMC proliferation may represent valuable antiatherosclerotic agents. Searching for dual antiproliferative and NO-donor molecules, we found that furoxans significantly decreased SMC proliferation in vitro, albeit with different potencies. We therefore assessed whether this property is dependent on their thiol-induced ring opening. Indeed, while furazans (analogues unable to release NO) are not effective, furoxans' inhibitory potency parallels with the electron-attractor capacity of the group in 3 of the ring, making this effect tunable. To demonstrate whether their specific block on G1-S phase could be NO-dependent, we supplemented SMCs with furoxans and inhibitors of GMP- and/or of the polyamine pathway, which regulate NO-induced SMC proliferation, but they failed in preventing the antiproliferative effect. To find the real mechanism of this property, our proteomics studies revealed that eleven cellular proteins (with SUMO1 being central) and networks involved in cell homeostasis/proliferation are modulated by furoxans, probably by interaction with adducts generated after degradation. Altogether, thanks to their dual effect and pharmacological flexibility, furoxans may be evaluated in the future as antiatherosclerotic molecules.
Collapse
Affiliation(s)
- Loretta Lazzarato
- Department of Drug Science and Technology, Università degli Studi di Torino, Via Pietro Giuria 9, 10125 Torino, Italy; (L.L.); (B.R.); (R.F.)
| | - Laura Bianchi
- Functional Proteomics Laboratory, Department of Life Sciences, Università degli Studi di Siena, Via Aldo Moro 2, 53100 Siena, Italy;
| | - Annapaola Andolfo
- Proteomics and Metabolomics Facility (ProMeFa), Center for Omics Sciences (COSR), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milano, Italy;
| | - Agnese Granata
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy; (A.G.); (M.L.); (M.S.); (A.C.)
| | - Matteo Lombardi
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy; (A.G.); (M.L.); (M.S.); (A.C.)
| | - Matteo Sinelli
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy; (A.G.); (M.L.); (M.S.); (A.C.)
| | - Barbara Rolando
- Department of Drug Science and Technology, Università degli Studi di Torino, Via Pietro Giuria 9, 10125 Torino, Italy; (L.L.); (B.R.); (R.F.)
| | - Marina Carini
- Department of Pharmaceutical Sciences “Pietro Pratesi”, Università degli Studi di Milano, Via Mangiagalli 25, 20133 Milano, Italy;
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy; (A.G.); (M.L.); (M.S.); (A.C.)
| | - Roberta Fruttero
- Department of Drug Science and Technology, Università degli Studi di Torino, Via Pietro Giuria 9, 10125 Torino, Italy; (L.L.); (B.R.); (R.F.)
| | - Lorenzo Arnaboldi
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy; (A.G.); (M.L.); (M.S.); (A.C.)
| |
Collapse
|
8
|
CEACAMS 1, 5, and 6 in disease and cancer: interactions with pathogens. Genes Cancer 2023; 14:12-29. [PMID: 36741860 PMCID: PMC9891707 DOI: 10.18632/genesandcancer.230] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 01/20/2023] [Indexed: 02/04/2023] Open
Abstract
The CEA family comprises 18 genes and 11 pseudogenes located at chromosome 19q13.2 and is divided into two main groups: cell surface anchored CEA-related cell adhesion molecules (CEACAMs) and the secreted pregnancy-specific glycoproteins (PSGs). CEACAMs are highly glycosylated cell surface anchored, intracellular, and intercellular signaling molecules with diverse functions, from cell differentiation and transformation to modulating immune responses associated with infection, inflammation, and cancer. In this review, we explore current knowledge surrounding CEACAM1, CEACAM5, and CEACAM6, highlight their pathological significance in the areas of cancer biology, immunology, and inflammatory disease, and describe the utility of murine models in exploring questions related to these proteins.
Collapse
|
9
|
Plüss L, Peissert F, Elsayed A, Rotta G, Römer J, Dakhel Plaza S, Villa A, Puca E, De Luca R, Oxenius A, Neri D. Generation and in vivo characterization of a novel high-affinity human antibody targeting carcinoembryonic antigen. MAbs 2023; 15:2217964. [PMID: 37243574 DOI: 10.1080/19420862.2023.2217964] [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: 02/20/2023] [Revised: 05/09/2023] [Accepted: 05/22/2023] [Indexed: 05/29/2023] Open
Abstract
There are no effective treatment options for most patients with metastatic colorectal cancer (mCRC). mCRC remains a leading cause of tumor-related death, with a five-year survival rate of only 15%, highlighting the urgent need for novel pharmacological products. Current standard drugs are based on cytotoxic chemotherapy, VEGF inhibitors, EGFR antibodies, and multikinase inhibitors. The antibody-based delivery of pro-inflammatory cytokines provides a promising and differentiated strategy to improve the treatment outcome for mCRC patients. Here, we describe the generation of a novel fully human monoclonal antibody (termed F4) targeting the carcinoembryonic antigen (CEA), a tumor-associated antigen overexpressed in colorectal cancer and other malignancies. The F4 antibody was selected by antibody phage display technology after two rounds of affinity maturation. F4 in single-chain variable fragment format bound to CEA in surface plasmon resonance with an affinity of 7.7 nM. Flow cytometry and immunofluorescence on human cancer specimens confirmed binding to CEA-expressing cells. F4 selectively accumulated in CEA-positive tumors, as evidenced by two orthogonal in vivo biodistribution studies. Encouraged by these results, we genetically fused murine interleukin (IL) 12 to F4 in the single-chain diabody format. F4-IL12 exhibited potent antitumor activity in two murine models of colon cancer. Treatment with F4-IL12 led to an increased density of tumor-infiltrating lymphocytes and an upregulation of interferon γ expression by tumor-homing lymphocytes. These data suggest that the F4 antibody is an attractive delivery vehicle for targeted cancer therapy.
Collapse
Affiliation(s)
- Louis Plüss
- Philochem AG, Libernstrasse 3, Otelfingen, Switzerland
- Department of Biology, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | | | - Abdullah Elsayed
- Philochem AG, Libernstrasse 3, Otelfingen, Switzerland
- Department of Biology, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | - Giulia Rotta
- Philochem AG, Libernstrasse 3, Otelfingen, Switzerland
| | - Jonas Römer
- Department of Biology, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | | | | | - Emanuele Puca
- Philochem AG, Libernstrasse 3, Otelfingen, Switzerland
| | | | - Annette Oxenius
- Department of Biology, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | - Dario Neri
- Philochem AG, Libernstrasse 3, Otelfingen, Switzerland
- Department of Biology, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
- Philogen SpA, Località Bellaria, Sovicille, Italy
| |
Collapse
|
10
|
Roles of anoikis in colorectal cancer therapy and the assessment of anoikis-regulatory molecules as therapeutic targets. Pathol Res Pract 2023; 241:154256. [PMID: 36455367 DOI: 10.1016/j.prp.2022.154256] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 11/21/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022]
Abstract
Colorectal cancer (CRC) is a deadly malignancy and therapeutic approaches for CRC are evolving every day. Anoikis is a key mechanism for programmed cell death of cancer cells that undergo anchorage-independent growth at a different matrix than the one which is expected. Yet, anoikis is a less studied mechanism of cell death in comparison to other mechanisms such as apoptosis. Relating to this, resistance to anoikis among cancer cells remains critical for improved metastasis and survival in a new environment evading anoikis. Since CRC cells have the ability to metastasize from proximal sites to secondary organs such as liver and promote cancer in those distant sites, a clear knowledge of the mechanisms essential for anchorage-independent growth and subsequent metastasis is necessary to counteract CRC progression and spread. Therefore, the identification of novel drug candidates and studying the roles of anoikis in assisting CRC therapy using such drugs can prevent anchorage-independent cancer cell growth. Additionally, the identification of novel biomarkers or therapeutic targets seems essential for implementing superior therapy, impeding relapse among malignant cells and improving the survival rate of clinical patients. As there are no reviews published on this topic till date, anoikis as a mechanism of cell death and its therapeutic roles in CRC are discussed in this review. In addition, several molecules were identified as therapeutic targets for CRC.
Collapse
|
11
|
Bray AW, Duan R, Malalur P, Drusbosky LM, Gourdin TS, Hill EG, Lilly MB. Elevated serum CEA is associated with liver metastasis and distinctive circulating tumor DNA alterations in patients with castration-resistant prostate cancer. Prostate 2022; 82:1264-1272. [PMID: 35766303 PMCID: PMC9388585 DOI: 10.1002/pros.24400] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 02/14/2022] [Accepted: 06/01/2022] [Indexed: 11/21/2022]
Abstract
BACKGROUND Elevated serum carcinoembryonic antigen (CEA) is used to identify "treatment emergent" forms of castration-resistant prostate cancer (CRPC) such as aggressive variant prostate cancer (AVPC). However, its individual utility as a prognostic marker and the genetic alterations associated with its expression have not been extensively studied in CRPC. METHODS This study retrospectively analyzed clinical outcomes and circulating tumor DNA profiles in 163 patients with CRPC and elevated or normal serum CEA. These same patients were then classified as AVPC or non-AVPC and compared to determine the uniqueness of CEA-associated gene alterations. RESULTS Patients with elevated CEA demonstrated higher rates of liver metastasis (37.5% vs. 19.1%, p = 0.02) and decreased median overall survival from CRPC diagnosis (28.7 vs. 73.2 mo, p < 0.0001). In addition, patients with elevated CEA were more likely to harbor copy number amplifications (CNAs) in AR, PIK3CA, MYC, BRAF, CDK6, MET, CCNE1, KIT, RAF1, and KRAS. Based on variant allele frequency we also defined "clonal" single-nucleotide variants (SNVs) thought to be driving disease progression in each patient and found that CEA expression was negatively correlated with clonal AR SNVs and positively correlated with clonal TP53 SNVs. Of these genetic associations, only the increases in clonal TP53 SNVs and KRAS amplifications were recapitulated among patients with AVPC when compared to patients without AVPC. CONCLUSIONS Together these findings suggest that CEA expression in CRPC is associated with aggressive clinical behavior and gene alterations distinct from those in AVPC.
Collapse
Affiliation(s)
- Alexander W. Bray
- Department of MedicineMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Rong Duan
- Department of Public Health SciencesMedical University of South CarolinaCharlestonSouth CarolinaUSA
- Hollings Cancer CenterMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Pannaga Malalur
- The Ohio State University Wexner Medical CenterColumbusOhioUSA
| | | | - Theodore S. Gourdin
- Department of MedicineMedical University of South CarolinaCharlestonSouth CarolinaUSA
- Hollings Cancer CenterMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Elizabeth G. Hill
- Department of Public Health SciencesMedical University of South CarolinaCharlestonSouth CarolinaUSA
- Hollings Cancer CenterMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Michael B. Lilly
- Department of MedicineMedical University of South CarolinaCharlestonSouth CarolinaUSA
- Hollings Cancer CenterMedical University of South CarolinaCharlestonSouth CarolinaUSA
| |
Collapse
|
12
|
Ouyang ZM, Lin JZ, Tang AJ, Yang ZH, Yang LJ, Wei XN, Li QH, Liang JJ, Zheng DH, Guo BP, Zhao G, Han Q, Dai L, Mo YQ. A Matrix Prediction Model for the 6-Month Mortality Risk in Patients With Anti-Melanoma Differentiation-Associated Protein-5-Positive Dermatomyositis. Front Med (Lausanne) 2022; 9:860798. [PMID: 35433730 PMCID: PMC9010999 DOI: 10.3389/fmed.2022.860798] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 02/25/2022] [Indexed: 12/26/2022] Open
Abstract
Objectives The purpose of this study was to investigate the baseline independent risk factors for predicting 6-month mortality of patients with anti-melanoma differentiation-associated gene 5 (anti-MDA5)-positive dermatomyositis (DM) and develop a matrix prediction model formed by these risk factors. Methods The hospitalized patients with DM who completed at least 6-month follow-up were recruited as a derivation cohort. The primary exposure was defined as positive anti-MDA5 at the baseline. The primary outcome was all-cause 6-month mortality after enrollment. A matrix prediction model was developed in the derivation cohort, and another published cohort was used for external validation. Results In derivation cohort, 82 patients with DM were enrolled (mean age of onset 50 ± 11 years and 63% women), with 40 (49%) showing positive anti-MDA5. Gottron sign/papules (OR: 5.135, 95%CI: 1.489–17.708), arthritis (OR: 5.184, 95%CI: 1.455–18.467), interstitial lung disease (OR: 7.034, 95%CI: 1.157–42.785), and higher level of C4 (OR: 1.010, 95%CI: 1.002–1.017) were the independent associators with positive anti-MDA5 in patients with DM. Patients with anti-MDA5-positive DM had significant higher 6-month all-cause mortality than those with anti-MDA5-negative (30 vs. 0%). Among the patients with anti-MDA5-positive DM, compared to the survivors, non-survivors had significantly advanced age of onset (59 ± 6 years vs. 46 ± 9 years), higher rates of fever (75 vs. 18%), positive carcinoma embryonic antigen (CEA, 75 vs. 14%), higher level of ferritin (median 2,858 ug/L vs. 619 ug/L, all p < 0.05). A stepwise multivariate Cox regression showed that ferritin ≥1,250 μg/L (HR: 10.4, 95%CI: 1.8–59.9), fever (HR: 11.2, 95%CI: 2.5–49.9), and positive CEA (HR: 5.2, 95%CI: 1.0–25.7) were the independent risk factors of 6-month mortality. A matrix prediction model was built to stratify patients with anti-MDA5-positive DM into different subgroups with various probabilities of 6-month mortality risk. In an external validation cohort, the observed 6-month all-cause mortality was 78% in high-risk group, 43% in moderate-risk group, and 25% in low-risk group, which shows good accuracy of the model. Conclusion Baseline characteristics such as fever, ferritin ≥1,250 μg/L, and positive CEA are the independent risk factors for 6-month all-cause mortality in patients with anti-MDA5-positive DM. A novel matrix prediction model composed of these three clinical indicators is first proposed to provide a chance for the exploration of individual treatment strategies in anti-MDA5-positive DM subgroups with various probabilities of mortality risk.
Collapse
Affiliation(s)
- Zhi-Ming Ouyang
- Department of Rheumatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jian-Zi Lin
- Department of Rheumatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ao-Juan Tang
- Department of Rheumatology, Shenshan Medical Center, Memorial Hospital of Sun Yat-sen University, Shanwei, China
| | - Ze-Hong Yang
- Departments of Radiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Li-Juan Yang
- Department of Rheumatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiu-Ning Wei
- Department of Rheumatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qian-Hua Li
- Department of Rheumatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jin-Jian Liang
- Department of Rheumatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dong-Hui Zheng
- Department of Rheumatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Bing-Peng Guo
- State Key Laboratory of Respiratory Disease, National Clinical Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Gui Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qian Han
- State Key Laboratory of Respiratory Disease, National Clinical Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- *Correspondence: Qian Han
| | - Lie Dai
- Department of Rheumatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Lie Dai
| | - Ying-Qian Mo
- Department of Rheumatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Rheumatology, Shenshan Medical Center, Memorial Hospital of Sun Yat-sen University, Shanwei, China
- Ying-Qian Mo
| |
Collapse
|
13
|
Kuracha MR, Thomas P, Tobi M, McVicker BL. Role of cell-free network communication in alcohol-associated disorders and liver metastasis. World J Gastroenterol 2021; 27:7080-7099. [PMID: 34887629 PMCID: PMC8613644 DOI: 10.3748/wjg.v27.i41.7080] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 07/02/2021] [Accepted: 09/30/2021] [Indexed: 02/06/2023] Open
Abstract
The aberrant use of alcohol is a major factor in cancer progression and metastasis. Contributing mechanisms include the systemic effects of alcohol and the exchange of bioactive molecules between cancerous and non-cancerous cells along the brain-gut-liver axis. Such interplay leads to changes in molecular, cellular, and biological functions resulting in cancer progression. Recent investigations have examined the role of extracellular vesicles (EVs) in cancer mechanisms in addition to their contribution as diagnostic biomarkers. Also, EVs are emerging as novel cell-free mediators in pathophysiological scenarios including alcohol-mediated gut microbiome dysbiosis and the release of nanosized EVs into the circulatory system. Interestingly, EVs in cancer patients are enriched with oncogenes, miRNA, lipids, and glycoproteins whose delivery into the hepatic microenvironment may be enhanced by the detrimental effects of alcohol. Proof-of-concept studies indicate that alcohol-associated liver disease is impacted by the effects of exosomes, including altered immune responses, reprogramming of stromal cells, and remodeling of the extracellular matrix. Moreover, the culmination of alcohol-related changes in the liver likely contributes to enhanced hepatic metastases and poor outcomes for cancer patients. This review summarizes the numerous aspects of exosome communications between organs with emphasis on the relationship of EVs in alcohol-associated diseases and cancer metastasis. The potential impact of EV cargo and release along a multi-organ axis is highly relevant to the promotion of tumorigenic mechanisms and metastatic disease. It is hypothesized that EVs target recipient tissues to initiate the formation of prometastatic niches and cancer progression. The study of alcohol-associated mechanisms in metastatic cancers is expected to reveal a better understanding of factors involved in the growth of secondary malignancies as well as novel approaches for therapeutic interventions.
Collapse
Affiliation(s)
- Murali R Kuracha
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Peter Thomas
- Department of Surgery, Creighton University School of Medicine, Omaha, NE 68178, United States
| | - Martin Tobi
- Research and Development Service, Detroit VAMC, Detroit, MI 48201, United States
- Department of Medicine, Central Michigan University College of Medicine, Detroit, MI 48201, United States
| | - Benita L McVicker
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, United States
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105, United States
| |
Collapse
|
14
|
Zhu D, Qiao J, Tang S, Pan Y, Li S, Yang C, Fang H. Elevated carcinoembryonic antigen predicts rapidly progressive interstitial lung disease in clinically amyopathic dermatomyositis. Rheumatology (Oxford) 2021; 60:3896-3903. [PMID: 33398346 DOI: 10.1093/rheumatology/keaa819] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 11/16/2020] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES The present study aimed to determine the correlation between serum carcinoembryonic antigen (CEA) level and the severity of interstitial lung disease (ILD) in clinically amyopathic DM (CADM) patients. METHODS We performed a retrospective study including 41 Chinese CADM patients without malignancy. Serum CEA levels, clinical and laboratory findings were collected. Association tests between CEA levels and disease activity parameters were performed. RESULTS Among the 41 patients, 16 (39.0%) developed rapidly progressive (RP)-ILD; of them, 14 (87.5%) had elevated serum CEA levels. Multivariate logistic regression analysis indicated that an elevated serum CEA level was an independent risk factor for RP-ILD. The incidence of elevated CEA level was significantly higher in patients with RP-ILD than in those without RP-ILD (87.5 vs 16.0%, P < 0.001). Furthermore, CEA levels were higher in patients with CADM with RP-ILD [26.87 (6.71) μg/l] than in those without RP-ILD [3.23 (0.64) μg/l] (P < 0.001). CEA levels in CADM patients were associated with the ferritin, alanine aminotransferase, aspartate aminotransferase and lactate dehydrogenase levels, and CT scores of the lungs. Also, elevated CEA levels are related to the organizing pneumonia pattern and lower lung zone consolidation in high-resolution CT. Moreover, the cumulative survival rate was significantly lower (68.4 vs 31.6%, P < 0.001) in the group with a CEA level >8.75 μg/l than that in the group with a CEA level <8.75 μg/l. CONCLUSIONS An elevated serum CEA level is common in patients with CADM, and a higher serum CEA level is a powerful indicator of RP-ILD and poor prognosis in those patients.
Collapse
Affiliation(s)
- Dingxian Zhu
- Department of Dermatology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jianjun Qiao
- Department of Dermatology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Shunli Tang
- Department of Dermatology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yunlei Pan
- Department of Dermatology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Sheng Li
- Department of Dermatology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Changyi Yang
- Department of Dermatology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Hong Fang
- Department of Dermatology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
15
|
Stavenhagen K, Laan LC, Gao C, Mehta AY, Heimburg-Molinaro J, Glickman JN, van Die I, Cummings RD. Tumor cells express pauci- and oligomannosidic N-glycans in glycoproteins recognized by the mannose receptor (CD206). Cell Mol Life Sci 2021; 78:5569-5585. [PMID: 34089345 PMCID: PMC11072813 DOI: 10.1007/s00018-021-03863-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 05/07/2021] [Accepted: 05/22/2021] [Indexed: 01/21/2023]
Abstract
The macrophage mannose receptor (CD206, MR) is an endocytic lectin receptor which plays an important role in homeostasis and innate immunity, however, the endogenous glycan and glycoprotein ligands recognized by its C-type lectin domains (CTLD) have not been well studied. Here we used the murine MR CTLD4-7 coupled to the Fc-portion of human IgG (MR-Fc) to investigate the MR glycan and glycoprotein recognition. We probed 16 different cancer and control tissues using the MR-Fc, and observed cell- and tissue-specific binding with varying intensity. All cancer tissues and several control tissues exhibited MR-Fc ligands, intracellular and/or surface-located. We further confirmed the presence of ligands on the surface of cancer cells by flow cytometry. To characterize the fine specificity of the MR for glycans, we screened a panel of glycan microarrays. Remarkably, the results indicate that the CTLD4-7 of the MR is highly selective for specific types of pauci- and oligomannose N-glycans among hundreds of glycans tested. As lung cancer tissue and the lung cancer cell line A549 showed intense MR-Fc binding, we further investigated the MR glycoprotein ligands in those cells by immunoprecipitation and glycoproteomic analysis. All enriched glycoproteins, of which 42 were identified, contained pauci- or oligomannose N-glycans, confirming the microarray results. Our study demonstrates that the MR CTLD4-7 is highly selective for pauci- and oligomannosidic N-glycans, structures that are often elevated in tumor cells, and suggest a potential role for the MR in tumor biology.
Collapse
Affiliation(s)
- Kathrin Stavenhagen
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, CLS 11087 - 3 Blackfan Circle, Boston, MA, 02115, USA
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC (VU Medical Center), Amsterdam, The Netherlands
| | - Lisa C Laan
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC (VU Medical Center), Amsterdam, The Netherlands
| | - Chao Gao
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, CLS 11087 - 3 Blackfan Circle, Boston, MA, 02115, USA
| | - Akul Y Mehta
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, CLS 11087 - 3 Blackfan Circle, Boston, MA, 02115, USA
| | - Jamie Heimburg-Molinaro
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, CLS 11087 - 3 Blackfan Circle, Boston, MA, 02115, USA
| | - Jonathan N Glickman
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Irma van Die
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC (VU Medical Center), Amsterdam, The Netherlands
| | - Richard D Cummings
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, CLS 11087 - 3 Blackfan Circle, Boston, MA, 02115, USA.
| |
Collapse
|
16
|
Kamiar A, Yousefi K, Dunkley JC, Webster KA, Shehadeh LA. β 2-Adrenergic receptor agonism as a therapeutic strategy for kidney disease. Am J Physiol Regul Integr Comp Physiol 2021; 320:R575-R587. [PMID: 33565369 DOI: 10.1152/ajpregu.00287.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Approximately 14% of the general population suffer from chronic kidney disease that can lead to acute kidney injury (AKI), a condition with up to 50% mortality for which there is no effective treatment. Hypertension, diabetes, and cardiovascular disease are the main comorbidities, and more than 660,000 Americans have kidney failure. β2-Adrenergic receptors (β2ARs) have been extensively studied in association with lung and cardiovascular disease, but with limited scope in kidney and renal diseases. β2ARs are expressed in multiple parts of the kidney including proximal and distal convoluted tubules, glomeruli, and podocytes. Classical and noncanonical β2AR signaling pathways interface with other intracellular mechanisms in the kidney to regulate important cellular functions including renal blood flow, electrolyte balance and salt handling, and tubular function that in turn exert control over critical physiology and pathology such as blood pressure and inflammatory responses. Nephroprotection through activation of β2ARs has surfaced as a promising field of investigation; however, there is limited data on the pharmacology and potential side effects of renal β2AR modulation. Here, we provide updates on some of the major areas of preclinical kidney research involving β2AR signaling that have advanced to describe molecular pathways and identify potential drug targets some of which are currently under clinical development for the treatment of kidney-related diseases.
Collapse
Affiliation(s)
- Ali Kamiar
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida.,Department of Molecular and Cellular Pharmacology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida
| | - Keyvan Yousefi
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida.,Department of Molecular and Cellular Pharmacology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida
| | - Julian C Dunkley
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida.,Division of Cardiology, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, Florida
| | - Keith A Webster
- Vascular Biology Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida
| | - Lina A Shehadeh
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida.,Division of Cardiology, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, Florida.,Peggy and Harold Katz Family Drug Discovery Center, University of Miami Leonard M. Miller School of Medicine, Miami, Florida
| |
Collapse
|
17
|
Ma J, Liu X, Chen H, Abbas MK, Yang L, Sun H, Sun T, Wu B, Yang S, Zhou D. c-KIT-ERK1/2 signaling activated ELK1 and upregulated carcinoembryonic antigen expression to promote colorectal cancer progression. Cancer Sci 2020; 112:655-667. [PMID: 33247506 PMCID: PMC7894012 DOI: 10.1111/cas.14750] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 11/16/2020] [Accepted: 11/23/2020] [Indexed: 12/27/2022] Open
Abstract
Carcinoembryonic antigen (CEA) is highly expressed in embryo and colorectal cancer (CRC) and has been widely used as a marker for CRC. Emerging evidence has demonstrated that elevated CEA levels promote CRC progression. However, the mechanism of the increased CEA expression in patients with primary and recurrent CRC is still an open question. In this study, we showed that c‐KIT, ELK1, and CEA were hyperexpressed in patients with CRC, especially patients with recurrent disease. From bioinformatics analysis, we picked ELK1 as a candidate transcription factor (TF) for CEA; the binding site of ELK1 within the CEA promoter was confirmed by chromatin immunoprecipitation and dual luciferase reporter assays. Overexpression of ELK1 increased CEA expression in vitro, while knockdown of ELK1 decreased CEA. Upregulated ELK1 promoted the adhesion, migration, and invasion of CRC cells, however knockdown of CEA blocked the activities of ELK1‐overexpressed CRC cells. Furthermore, we explored the role of c‐KIT‐ERK1/2 signaling in activation of ELK1. Blocking c‐KIT signaling using Imatinib or ISCK03 reduced p‐ELK1 expression and consequently decreased CEA levels in CRC cells, as did blocking the ERK1/2 pathway by U0126. Compared with wild type littermates, the c‐kit loss‐of‐functional Wadsm/m mice showed lowered c‐KIT, ELK1, and CEA expression. In conclusion, our study revealed that ELK1, which was activated by c‐KIT‐ERK1/2 signaling, was a key TF for CEA expression. Blocking ELK1 or its upstream signaling could be an alternative way to decelerate CRC progression. Besides being a biomarker for CRC, CEA could be used for guiding targeted therapy.
Collapse
Affiliation(s)
- Jian Ma
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China
| | - Xiaohui Liu
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China
| | - Hong Chen
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China
| | - Muhammad Khawar Abbas
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China
| | - Liu Yang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China
| | - Haimei Sun
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China
| | - Tingyi Sun
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China
| | - Bo Wu
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China.,Cancer Institute of Capital Medical University, Beijing, China
| | - Shu Yang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China.,Cancer Institute of Capital Medical University, Beijing, China
| | - Deshan Zhou
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China.,Cancer Institute of Capital Medical University, Beijing, China
| |
Collapse
|
18
|
Talwar H, McVicker B, Tobi M. p38γ Activation and BGP (Biliary Glycoprotein) Induction in Primates at Risk for Inflammatory Bowel Disease and Colorectal Cancer-A Comparative Study with Humans. Vaccines (Basel) 2020; 8:E720. [PMID: 33276422 PMCID: PMC7712431 DOI: 10.3390/vaccines8040720] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/17/2020] [Accepted: 11/26/2020] [Indexed: 12/13/2022] Open
Abstract
Colorectal cancer (CRC) is a common cause of cancer-related deaths largely due to CRC liver metastasis (CRLM). Identification of targetable mechanisms continues and includes investigations into the role of inflammatory pathways. Of interest, MAPK is aberrantly expressed in CRC patients, yet the activation status is not defined. The present study assessed p38γ activation in CRC patients, cancer cells, and tissues of cotton top tamarin (CTT) and common marmoset (CM). The primate world is an overlooked resource as colitis-CRC-prone CTT are usually inure to liver metastasis while CM develop colitis but not CRC. The results demonstrate that p38γ protein and phosphorylation levels are significantly increased in CRC patients compared to normal subjects and CTT. Furthermore, p38γ phosphorylation is significantly elevated in human CRC cells and hepatoblastoma cells but not in CM colon. Additionally, carcinoembryonic antigen (CEA) and biliary glycoprotein (BGP) are induced in the CRC patients that showed p38γ phosphorylation. Inhibition of p38 MAPK in CRC cells showed a significant decline in cell growth with no effect on apoptosis or BGP level. Overall, p38γ is activated in CRC tumorigenesis and likely involves CEA antigens during CRLM in humans but not in the CTT or CM, that rarely develop CRLM.
Collapse
Affiliation(s)
- Harvinder Talwar
- Research and Development VA Medical Center and Internal Medicine, Wayne State University, Detroit, MI 48201, USA;
| | - Benita McVicker
- Research Service, VA Nebraska-Western Iowa Health Care System, The University of Nebraska Medical Center, Omaha, NE 68105, USA;
| | - Martin Tobi
- Research and Development Service, Department of Internal Medicine, Detroit VAMC, Detroit, MI 48201, USA
- Central Michigan University College of Medicine, Mount Pleasant, MI 48859, USA
| |
Collapse
|
19
|
Messaritakis I, Sfakianaki M, Vogiatzoglou K, Koulouridi A, Koutoulaki C, Mavroudis D, Tzardi M, Gouvas N, Tsiaoussis J, Souglakos J. Evaluation of the Role of Circulating Tumor Cells and Microsatellite Instability Status in Predicting Outcome of Advanced CRC Patients. J Pers Med 2020; 10:jpm10040235. [PMID: 33217974 PMCID: PMC7712177 DOI: 10.3390/jpm10040235] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 10/29/2020] [Accepted: 11/16/2020] [Indexed: 12/19/2022] Open
Abstract
Colorectal cancer (CRC) remains one of the leading causes of cancer-related death due to its high metastatic potential. This study aimed to investigate the detection and heterogeneity of circulating tumor cells (CTCs) and the microsatellite instability (MSI) status in advanced CRC patients prior to any systemic front-line treatment. Peripheral whole blood was obtained from 198 patients. CTCs were detected using double immunofluorescence and a real time-polymerase chain reaction assay; whereas MSI status was evaluated using fragment analysis. Median age of the patients was 66 years, 63.1% were males, 65.2% had a colon/sigmoid tumor location and 90.4% had a good performance status (PS). MSI-High status was detected in 4.9% of the patients; 33.3%, 56.1% and 8.6% patients had at least one detectable CEACAM5+/EpCAM+, CEACAM5+/EpCAM- and CEACAM5-/EpCAM+ CTC, respectively, and 9.1% of the patients had CEACAM5mRNA-positive CTCs. Following multivariate analysis, age, PS and MSI were confirmed as independent prognostic factors for decreased time to progression, whereas age, PS and CTC presence were confirmed as independent prognostic factors for decreased overall survival. In conclusion, our data support the use of CEACAM5 as a dynamic adverse prognostic CTC biomarker in patients with metastatic CRC and MSI-High is considered an unfavorable prognostic factor in metastatic CRC patient tumors.
Collapse
Affiliation(s)
- Ippokratis Messaritakis
- Laboratory of Translational Oncology, Medical School, University of Crete, 70013 Heraklion, Greece; (M.S.); (K.V.); (A.K.); (C.K.); (D.M.); (J.S.)
- Correspondence: ; Tel.: +30-2810-394926
| | - Maria Sfakianaki
- Laboratory of Translational Oncology, Medical School, University of Crete, 70013 Heraklion, Greece; (M.S.); (K.V.); (A.K.); (C.K.); (D.M.); (J.S.)
| | - Konstantinos Vogiatzoglou
- Laboratory of Translational Oncology, Medical School, University of Crete, 70013 Heraklion, Greece; (M.S.); (K.V.); (A.K.); (C.K.); (D.M.); (J.S.)
| | - Asimina Koulouridi
- Laboratory of Translational Oncology, Medical School, University of Crete, 70013 Heraklion, Greece; (M.S.); (K.V.); (A.K.); (C.K.); (D.M.); (J.S.)
| | - Chara Koutoulaki
- Laboratory of Translational Oncology, Medical School, University of Crete, 70013 Heraklion, Greece; (M.S.); (K.V.); (A.K.); (C.K.); (D.M.); (J.S.)
| | - Dimitrios Mavroudis
- Laboratory of Translational Oncology, Medical School, University of Crete, 70013 Heraklion, Greece; (M.S.); (K.V.); (A.K.); (C.K.); (D.M.); (J.S.)
- Department of Medical Oncology, University General Hospital of Heraklion, 71100 Heraklion, Greece
| | - Maria Tzardi
- Laboratory of Pathology, University General Hospital of Heraklion, 70013 Heraklion, Greece;
| | - Nikolaos Gouvas
- Medical School, University of Cyprus, 20537 Nicosia, Cyprus;
| | - John Tsiaoussis
- Department of Anatomy, School of Medicine, University of Crete, 70013 Heraklion, Greece;
| | - John Souglakos
- Laboratory of Translational Oncology, Medical School, University of Crete, 70013 Heraklion, Greece; (M.S.); (K.V.); (A.K.); (C.K.); (D.M.); (J.S.)
- Department of Medical Oncology, University General Hospital of Heraklion, 71100 Heraklion, Greece
| |
Collapse
|
20
|
Liu Y, Du Z, Ji J, Li J, Bi D, Tang F. Bidirectional association between serum carcinoembryonic antigen and metabolic syndrome among the Chinese male population: two cohort studies. Lipids Health Dis 2020; 19:233. [PMID: 33148263 PMCID: PMC7643476 DOI: 10.1186/s12944-020-01411-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/20/2020] [Indexed: 11/11/2022] Open
Abstract
Purpose Previous studies have shown that serum carcinoembryonic antigen (CEA) is independently associated with metabolic syndrome (MetS). However, these studies were mainly cross-sectional analyses, and cause was not clarified. In the present study, two bidirectional cohort studies were conducted to investigate the bidirectional associations between CEA and MetS using a Chinese male sample cohort. Methods The initial longitudinal cohort included 9629 Chinese males enrolled from January 2010 to December 2015. Two bidirectional cohorts were conducted in the study: subcohort A (from CEA to MetS, n = 6439) included participants without MetS at baseline to estimate the risk of developing incident MetS; subcohort B (from MetS to CEA, n = 8533) included participants without an elevated CEA level (Hyper-CEA) at baseline to examine the risk of developing incident Hyper-CEA. Hazard ratios (HRs) and 95% confidence intervals (CIs) were estimated using Cox proportional hazards models. Results In subcohort A, the incidence densities of MetS among participants with and without Hyper-CEA were 84.56 and 99.28 per 1000 person-years, respectively. No significant effects of Hyper-CEA on incident MetS were observed in subcohort A (HR, 0.89; 95% CI, 0.71 to 1.12; P = 0.326). In subcohort B, a higher incidence density of Hyper-CEA was found among participants with MetS (33.42 and 29.13 per 1000 person-years for those with and without MetS, respectively). For nonsmoking participants aged > 65 years, MetS increased the risk of incident Hyper-CEA (HR, 1.87; 95% CI, 1.09 to 3.20; P = 0.022). Conclusion For the direction of CEA on incident MetS, no significant association was observed. For the direction of MetS on incident Hyper-CEA, MetS in nonsmoking elderly men could increase the risk of incident Hyper-CEA, while this association was not found in other stratified participants. The clinical implications of the association between CEA and MetS should be interpreted with caution. Supplementary Information The online version contains supplementary material available at 10.1186/s12944-020-01411-7.
Collapse
Affiliation(s)
- Yafei Liu
- Center for Big Data Research in Health and Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China.,Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhaohui Du
- Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Jiadong Ji
- Department of Data Science, School of Statistics, Shandong University of Finance and Economics, Jinan, China
| | - Jingru Li
- School of Public Health, Weifang Medical University, Weifang, China
| | - Deming Bi
- Department of Surgery, Zhangqiu District Hospital of Traditional Chinese Medicine, Jinan, China.
| | - Fang Tang
- Center for Big Data Research in Health and Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China. .,Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
21
|
Kamal Y, Schmit SL, Frost HR, Amos CI. The tumor microenvironment of colorectal cancer metastases: opportunities in cancer immunotherapy. Immunotherapy 2020; 12:1083-1100. [PMID: 32787587 DOI: 10.2217/imt-2020-0026] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
About a fifth of individuals with colorectal cancer (CRC) present with disease metastasis at the time of diagnosis. While the role of the tumor microenvironment (TME) in governing CRC progression is undeniable, the role of the TME in either establishing or suppressing the formation of distant metastases of CRC is less well established. Despite advances in immunotherapy, many individuals with metastatic CRC do not respond to standard-of-care therapy. Therefore, understanding the role of the TME in establishing distant metastases is essential for developing new immunological agents. Here, we summarize our current understanding of the TME of CRC metastases, describe differences between the TME of primary tumors and their distant metastases, and discuss advances in the design and combinations of immunotherapeutic agents.
Collapse
Affiliation(s)
- Yasmin Kamal
- Department of Biomedical Data Sciences, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA.,Quantitative Biomedical Sciences, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Stephanie L Schmit
- Department of Cancer Epidemiology, H Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Hildreth Robert Frost
- Department of Biomedical Data Sciences, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA.,Quantitative Biomedical Sciences, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Christopher I Amos
- Department of Biomedical Data Sciences, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA.,Quantitative Biomedical Sciences, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA.,Dan L Duncan Comprehensive Cancer Center at Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
22
|
Chi X, Jiang Y, Chen Y, Lv L, Chen J, Yang F, Zhang X, Pan F, Cai Q. microR-505/heterogeneous nuclear ribonucleoprotein M inhibits hepatocellular carcinoma cell proliferation and induces cell apoptosis through the Wnt/β-catenin signaling pathway. Biomark Med 2020; 14:981-996. [PMID: 32940078 DOI: 10.2217/bmm-2019-0511] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 05/22/2020] [Indexed: 12/12/2022] Open
Abstract
Aim: This study aimed to investigate the expression of microRNA-505 (miR-505) and explore its clinical significance, biological function and mechanisms in hepatocellular carcinoma (HCC). Methods: Expression of miR-505 was measured in 128 paired HCC tissues and five cell lines by quantitative real-time polymerase chain reaction (qRT-PCR). MTT assay, Transwell migration, invasion assays and apoptosis assay were performed to explore the functional role of miR-505. The target gene of miR-505 was assessed using the bioinformatics assay and the related signaling pathway was confirmed using western blot. Results: Expression of miR-505 in HCC serum and tissues were downregulated. The overexpression of miR-505 in HCC cells inhibited cell proliferation and metastasis, as well as enhanced cell apoptosis by directly downregulating heterogeneous nuclear ribonucleoprotein M (HNRNPM). The activity of the Wnt/β-catenin signaling pathway was suppressed by the overexpression of miR-505 but was promoted by the upregulation of HNRNPM. Conclusion: The results suggest that the regulation of miR-505/HNRNPM may be a novel strategy to improve the targeted therapy of HCC.
Collapse
Affiliation(s)
- Xiaobin Chi
- Department of Hepatobiliary Surgery, 900 Hospital of The Joint Logistics Team, Fuzhou 350025, China
| | - Yi Jiang
- Department of Hepatobiliary Surgery, 900 Hospital of The Joint Logistics Team, Fuzhou 350025, China
| | - Yongbiao Chen
- Department of Hepatobiliary Surgery, 900 Hospital of The Joint Logistics Team, Fuzhou 350025, China
| | - Lizhi Lv
- Department of Hepatobiliary Surgery, 900 Hospital of The Joint Logistics Team, Fuzhou 350025, China
| | - Jianwei Chen
- Department of Hepatobiliary Surgery, 900 Hospital of The Joint Logistics Team, Fuzhou 350025, China
| | - Fang Yang
- Department of Hepatobiliary Surgery, 900 Hospital of The Joint Logistics Team, Fuzhou 350025, China
| | - Xiaojin Zhang
- Department of Hepatobiliary Surgery, 900 Hospital of The Joint Logistics Team, Fuzhou 350025, China
| | - Fan Pan
- Department of Hepatobiliary Surgery, 900 Hospital of The Joint Logistics Team, Fuzhou 350025, China
| | - Qiucheng Cai
- Department of Hepatobiliary Surgery, 900 Hospital of The Joint Logistics Team, Fuzhou 350025, China
| |
Collapse
|
23
|
Prognostic implications of EGFR protein expression in sporadic colorectal tumors: Correlation with copy number status, mRNA levels and miRNA regulation. Sci Rep 2020; 10:4662. [PMID: 32170146 PMCID: PMC7070091 DOI: 10.1038/s41598-020-61688-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 02/26/2020] [Indexed: 11/10/2022] Open
Abstract
Sporadic colorectal cancer (sCRC) is the third most frequent cancer worldwide and the second most common cause of cancer-related deaths (mainly due metastatic dissemination). We investigated the immunohistochemical expression of frequently altered proteins in primary tumors from 51 patients (25 liver metastatic and 26 non-metastatic cases) with a median 103 months follow-up (103 months). We evaluated EGFR copy number (using SNP arrays and FISH) and its expression and regulation (by mRNA and miRNA arrays). We found differences between metastatic and non-metastatic sCRCs for MLH1 (p = 0.05), PMS2 (p = 0.02), CEA (p < 0.001) and EGFR (p < 0.001) expression. EGFR expression was associated with lymph node metastases (p = 0.001), liver metastases at diagnosis (p < 0.001), and advanced stage (p < 0.001). There were associations between EGFR expression-, EGFR gene copy number- and EGFR mRNA levels. We found potential interactions of two miRNAs targeting EGFR expression, (miR-134 and miR-4328, in non-metastatic and metastatic tumors, respectively). EGFR expression was associated with a worse outcome (p = 0.005). Multivariate analysis of prognostic factors for overall survival identified that, the expression of EGFR expression (p = 0.047) and pTNM stage (p < 0.001) predicted an adverse outcome. EGFR expression could be regulated by amplification or polysomies (in metastatic tumors), or miRNAs (miRNA-134, in non-metastatic tumors). EGFR expression in sCRC appears to be related to metastases and poor outcome.
Collapse
|
24
|
West KO, Scott HM, Torres-Odio S, West AP, Patrick KL, Watson RO. The Splicing Factor hnRNP M Is a Critical Regulator of Innate Immune Gene Expression in Macrophages. Cell Rep 2019; 29:1594-1609.e5. [PMID: 31693898 PMCID: PMC6981299 DOI: 10.1016/j.celrep.2019.09.078] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 08/13/2019] [Accepted: 09/26/2019] [Indexed: 12/16/2022] Open
Abstract
While transcriptional control of innate immune gene expression is well characterized, almost nothing is known about how pre-mRNA splicing decisions influence, or are influenced by, macrophage activation. Here, we demonstrate that the splicing factor hnRNP M is a critical repressor of innate immune gene expression and that its function is regulated by pathogen sensing cascades. Loss of hnRNP M led to hyperinduction of a unique regulon of inflammatory and antimicrobial genes following diverse innate immune stimuli. While mutating specific serines on hnRNP M had little effect on its ability to control pre-mRNA splicing or transcript levels of housekeeping genes in resting macrophages, it greatly impacted the protein's ability to dampen induction of specific innate immune transcripts following pathogen sensing. These data reveal a previously unappreciated role for pattern recognition receptor signaling in controlling splicing factor phosphorylation and establish pre-mRNA splicing as a critical regulatory node in defining innate immune outcomes.
Collapse
Affiliation(s)
- Kelsi O West
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX 77807, USA
| | - Haley M Scott
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX 77807, USA
| | - Sylvia Torres-Odio
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX 77807, USA
| | - A Phillip West
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX 77807, USA
| | - Kristin L Patrick
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX 77807, USA.
| | - Robert O Watson
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX 77807, USA.
| |
Collapse
|
25
|
Cao P, Luo WW, Li C, Tong Z, Zheng ZQ, Zhou L, Xiong Y, Li S. The heterogeneous nuclear ribonucleoprotein hnRNPM inhibits RNA virus-triggered innate immunity by antagonizing RNA sensing of RIG-I-like receptors. PLoS Pathog 2019; 15:e1007983. [PMID: 31433824 PMCID: PMC6703689 DOI: 10.1371/journal.ppat.1007983] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 07/15/2019] [Indexed: 12/15/2022] Open
Abstract
Recognition of viral RNA by the retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs), including RIG-I and MDA5, initiates innate antiviral responses. Although regulation of RLR-mediated signal transduction has been extensively investigated, how the recognition of viral RNA by RLRs is regulated remains enigmatic. In this study, we identified heterogeneous nuclear ribonucleoprotein M (hnRNPM) as a negative regulator of RLR-mediated signaling. Overexpression of hnRNPM markedly inhibited RNA virus-triggered innate immune responses. Conversely, hnRNPM-deficiency increased viral RNA-triggered innate immune responses and inhibited replication of RNA viruses. Viral infection caused translocation of hnRNPM from the nucleus to the cytoplasm. hnRNPM interacted with RIG-I and MDA5, and impaired the binding of the RLRs to viral RNA, leading to inhibition of innate antiviral response. Our findings suggest that hnRNPM acts as an important decoy for excessive innate antiviral immune response.
Collapse
Affiliation(s)
- Pan Cao
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, College of Life Sciences, Medical Research Institute, Wuhan University, Wuhan, China
| | - Wei-Wei Luo
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- * E-mail: (WWL); (SL)
| | - Chen Li
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, College of Life Sciences, Medical Research Institute, Wuhan University, Wuhan, China
| | - Zhen Tong
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Zhou-Qin Zheng
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Lu Zhou
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, College of Life Sciences, Medical Research Institute, Wuhan University, Wuhan, China
| | - Yong Xiong
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, College of Life Sciences, Medical Research Institute, Wuhan University, Wuhan, China
| | - Shu Li
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, College of Life Sciences, Medical Research Institute, Wuhan University, Wuhan, China
- * E-mail: (WWL); (SL)
| |
Collapse
|
26
|
Kim H, Jung HI, Kwon SH, Bae SH, Kim HC, Baek MJ, Lee MS. Preoperative neutrophil-lymphocyte ratio and CEA is associated with poor prognosis in patients with synchronous colorectal cancer liver metastasis. Ann Surg Treat Res 2019; 96:191-200. [PMID: 30941323 PMCID: PMC6444047 DOI: 10.4174/astr.2019.96.4.191] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 07/18/2018] [Accepted: 08/03/2018] [Indexed: 12/15/2022] Open
Abstract
Purpose Recently, the neutrophil-to-lymphocyte ratio (NLR), an inflammatory response marker, has been reported to be associated with the prognosis in patients with various type of cancer. However, there have been no studies until now that have explored the prognostic role of combined detection of NLR and CEA in patients with synchronous liver-limited colorectal metastases (sCRLM). Methods Eighty-three patients who histologically diagnosed as sCRLM were selected. Their laboratory and clinical data were collected retrospectively. Using receiver operating characteristic curve analysis, the cutoff value of NLR was calculated based on which patients were assigned to a high NLR (more than 1.94) group and low NLR (less than 1.94) group. A cutoff value of 100 ng/mL for serum CEA level was used in light of the previous literature. Results CEA level and Poorly differentiated histology of colon cancer was significantly correlated with high NLR (P = 0.005 and P = 0.048, respectively). The multivariate analysis identified the high NLR as independent prognostic factors for OS and DFS in all patients (P = 0.010 and P = 0.006, respectively). Patients with both low levels of NLR and CEA had a significantly longer OS and DFS (P = 0.026 and P = 0.009, respectively). Conclusion In conclusion, elevated preoperative NLR is strongly correlated with both survival and recurrence in patients who have been diagnosed with resectable sCRLM. The combination of NLR and CEA level could be a more powerful prognostic marker than NLR alone.
Collapse
Affiliation(s)
- Hyunjung Kim
- Department of Surgery, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Hae Il Jung
- Department of Surgery, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Soon Ha Kwon
- Department of Surgery, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Sang Ho Bae
- Department of Surgery, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Hyung Chul Kim
- Department of Surgery, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Moo-Jun Baek
- Department of Surgery, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Moon Soo Lee
- Department of Surgery, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| |
Collapse
|
27
|
Abdul-Wahid A, Cydzik M, Fischer NW, Prodeus A, Shively JE, Martel A, Alminawi S, Ghorab Z, Berinstein NL, Gariépy J. Serum-derived carcinoembryonic antigen (CEA) activates fibroblasts to induce a local re-modeling of the extracellular matrix that favors the engraftment of CEA-expressing tumor cells. Int J Cancer 2018; 143:1963-1977. [PMID: 29756328 DOI: 10.1002/ijc.31586] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 04/04/2018] [Accepted: 04/18/2018] [Indexed: 12/21/2022]
Abstract
Elevated levels of the carcinoembryonic antigen (CEA; CEACAM5) in the serum of colorectal cancer (CRC) patients represent a clinical biomarker that correlates with disease recurrence. However, a mechanistic role for soluble CEA (sCEA) in tumor progression and metastasis remains to be established. In our study, we report that sCEA acts as a paracrine factor, activating human fibroblasts by signaling through both the STAT3 and AKT1-mTORC1 pathways, promoting their transition to a cancer-associated fibroblast (CaF) phenotype. sCEA-activated fibroblasts express and secrete higher levels of fibronectin, including cellular EDA+ -fibronectin (Fn-EDA) that selectively promote the implantation and adherence of CEA-expressing cancer cells. Immunohistochemical analyses of liver tissues derived from CRC patients with elevated levels of sCEA reveal that the expression of cellular Fn-EDA co-registers with CEA-expressing liver metastases. Taken together, these findings indicate a direct role for sCEA as a human fibroblast activation factor, in priming target tissues for the engraftment of CEA-expressing cancer cells, through the differentiation of tissue-resident fibroblasts, resulting in a local change in composition of the extracellular matrix.
Collapse
Affiliation(s)
- Aws Abdul-Wahid
- Departments of Medical Biophysics and Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada.,Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Marzena Cydzik
- Departments of Medical Biophysics and Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada.,Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Nicholas W Fischer
- Departments of Medical Biophysics and Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada.,Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Aaron Prodeus
- Departments of Medical Biophysics and Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada.,Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - John E Shively
- Department of Immunology, Beckman Research Institute, City of Hope, Duarte, CA
| | - Anne Martel
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Samira Alminawi
- Department of Anatomic Pathology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, CANADA
| | - Zeina Ghorab
- Department of Anatomic Pathology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, CANADA
| | | | - Jean Gariépy
- Departments of Medical Biophysics and Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada.,Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| |
Collapse
|
28
|
Keirsse J, Van Damme H, Geeraerts X, Beschin A, Raes G, Van Ginderachter JA. The role of hepatic macrophages in liver metastasis. Cell Immunol 2018; 330:202-215. [PMID: 29661474 DOI: 10.1016/j.cellimm.2018.03.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Revised: 03/12/2018] [Accepted: 03/29/2018] [Indexed: 12/21/2022]
Abstract
The liver is a major target organ for metastasis of both gastrointestinal and extra-gastrointestinal cancers. Due to its frequently inoperable nature, liver metastasis represents a leading cause of cancer-associated death worldwide. In the past years, the pivotal role of the immune system in this process is being increasingly recognised. In particular, the role of the hepatic macrophages, both recruited monocyte-derived macrophages (Mo-Mfs) and tissue-resident Kupffer cells (KCs), has been shown to be more versatile than initially imagined. However, the lack of tools to easily distinguish between these two macrophage populations has hampered the assignment of particular functionalities to specific hepatic macrophage subsets. In this Review, we highlight the most remarkable findings regarding the origin and functions of hepatic macrophage populations, and we provide a detailed description of their distinct roles in the different phases of the liver metastatic process.
Collapse
Affiliation(s)
- Jiri Keirsse
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Helena Van Damme
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Xenia Geeraerts
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Alain Beschin
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Geert Raes
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jo A Van Ginderachter
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
29
|
Nader JS, Abadie J, Deshayes S, Boissard A, Blandin S, Blanquart C, Boisgerault N, Coqueret O, Guette C, Grégoire M, Pouliquen DL. Characterization of increasing stages of invasiveness identifies stromal/cancer cell crosstalk in rat models of mesothelioma. Oncotarget 2018; 9:16311-16329. [PMID: 29662647 PMCID: PMC5893242 DOI: 10.18632/oncotarget.24632] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 02/25/2018] [Indexed: 12/11/2022] Open
Abstract
Sarcomatoid mesothelioma (SM) is a devastating cancer associated with one of the poorest outcome. Therefore, representative preclinical models reproducing different tumor microenvironments (TME) observed in patients would open up new prospects for the identification of markers and evaluation of innovative therapies. Histological analyses of four original models of rat SM revealed their increasing infiltrative and metastatic potential were associated with differences in Ki67 index, blood-vessel density, and T-lymphocyte and macrophage infiltration. In comparison with the noninvasive tumor M5-T2, proteomic analysis demonstrated the three invasive tumors F4-T2, F5-T1 and M5-T1 shared in common a very significant increase in the abundance of the multifunctional proteins galectin-3, prohibitin and annexin A5, and a decrease in proteins involved in cell adhesion, tumor suppression, or epithelial differentiation. The increased metastatic potential of the F5-T1 tumor, relative to F4-T2, was associated with an increased macrophage vs T-cell infiltrate, changes in the levels of expression of a panel of cytokine genes, an increased content of proteins involved in chromatin organization, ribosome structure, splicing, or presenting anti-adhesive properties, and a decreased content of proteins involved in protection against oxidative stress, normoxia and intracellular trafficking. The most invasive tumor, M5-T1, was characterized by a pattern of specific phenotypic and molecular features affecting the presentation of MHC class I-mediated antigens and immune cell infiltration, or involved in the reorganization of the cytoskeleton and composition of the extracellular matrix. These four preclinical models and data represent a new resource available to the cancer research community to catalyze further investigations on invasiveness.
Collapse
Affiliation(s)
- Joëlle S. Nader
- CRCINA, INSERM, Université d’Angers, Université de Nantes, Nantes, France
| | - Jérôme Abadie
- CRCINA, INSERM, Université d’Angers, Université de Nantes, Nantes, France
- ONIRIS, Nantes, France
| | - Sophie Deshayes
- CRCINA, INSERM, Université d’Angers, Université de Nantes, Nantes, France
| | - Alice Boissard
- CRCINA, INSERM, Université de Nantes, Université d’Angers, Angers, France
- ICO, Angers, France
| | - Stéphanie Blandin
- Plate-Forme MicroPICell, SFR François Bonamy, Université de Nantes, France
| | | | | | - Olivier Coqueret
- CRCINA, INSERM, Université de Nantes, Université d’Angers, Angers, France
- ICO, Angers, France
| | - Catherine Guette
- CRCINA, INSERM, Université de Nantes, Université d’Angers, Angers, France
- ICO, Angers, France
| | - Marc Grégoire
- CRCINA, INSERM, Université d’Angers, Université de Nantes, Nantes, France
| | | |
Collapse
|
30
|
Yoo BC, Yeo SG. Identification of CEA-interacting proteins in colon cancer cells and their changes in expression after irradiation. Radiat Oncol J 2017; 35:281-288. [PMID: 28881503 PMCID: PMC5654139 DOI: 10.3857/roj.2017.00255] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 07/16/2017] [Accepted: 07/31/2017] [Indexed: 01/16/2023] Open
Abstract
Purpose The serum carcinoembryonic antigen (CEA) level has been recognized as a prognostic factor in colorectal cancer, and associated with response of rectal cancer to radiotherapy. This study aimed to identify CEA-interacting proteins in colon cancer cells and observe post-irradiation changes in their expression. Materials and Methods CEA expression in colon cancer cells was examined by Western blot analysis. Using an anti-CEA antibody or IgG as a negative control, immunoprecipitation was performed in colon cancer cell lysates. CEA and IgG immunoprecipitates were used for liquid chromatography–tandem mass spectrometry (LC-MS/MS) analysis. Proteins identified in the CEA immunoprecipitates but not in the IgG immunoprecipitates were selected as CEA-interacting proteins. After radiation treatment, changes in expression of CEA-interacting proteins were monitored by Western blot analysis. Results CEA expression was higher in SNU-81 cells compared with LoVo cells. The membrane localization of CEA limited the immunoprecipitation results and thus the number of CEA-interacting proteins identified. Only the Ras-related protein Rab-6B and lysozyme C were identified as CEA-interacting proteins in LoVo and SNU-81 cells, respectively. Lysozyme C was detected only in SNU-81, and CEA expression was differently regulated in two cell lines; it was down-regulated in LoVo but up-regulated in SNU-81 in radiation dosage-dependent manner. Conclusion CEA-mediated radiation response appears to vary, depending on the characteristics of individual cancer cells. The lysozyme C and Rab subfamily proteins may play a role in the link between CEA and tumor response to radiation, although further studies are needed to clarify functional roles of the identified proteins.
Collapse
Affiliation(s)
- Byong Chul Yoo
- Colorectal Cancer Branch, Research Institute, National Cancer Center, Goyang, Korea
| | - Seung-Gu Yeo
- Department of Radiation Oncology, Soonchunhyang University College of Medicine, Soonchunhyang University Hospital, Cheonan, Korea
| |
Collapse
|
31
|
Bjersand K, Seidal T, Sundström-Poromaa I, Åkerud H, Skirnisdottir I. The clinical and prognostic correlation of HRNPM and SLC1A5 in pathogenesis and prognosis in epithelial ovarian cancer. PLoS One 2017; 12:e0179363. [PMID: 28609484 PMCID: PMC5469483 DOI: 10.1371/journal.pone.0179363] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 05/30/2017] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVES To evaluate the prognostic effect of the Heterogeneous nuclear ribonucleoprotein type M (HNRPM) and Solute carrier 1A5 (SLC1A5) in FIGO-stages I-II epithelial ovarian cancer. METHODS A retrospective cohort study was designed to investigate the prognostic effect of HNRPM and SLC1A5, and the association with clinical-pathologic characteristics in 131 patients with FIGO-stages I-II epithelial ovarian cancer. Tissue microarrays were constructed and protein levels were assessed by immunohistochemistry (IHC). RESULTS Positive HRNPM status was associated with positive staining for PUMA (P = 0.04), concomitant PUMA and p21 staining (P = 0.005), and VEGF-R2 (P = 0.003). Positive SLC1A5 staining was associated with positive staining of p27 (P = 0.030), PUMA (P = 0.039), concomitant PUMA and p27 staining, and VEGF-R2 (P = 0.039). In non-serous tumors (n = 72), the SLC1A5 positivity was associated with recurrent disease (P = 0.01). In a multivariable logistic regression analysis FIGO-stage (OR = 12.4), tumor grade (OR = 5.1) and SLC1A5 positivity (OR = 0.1) were independent predictive factors for recurrent disease. Disease-free survival (DFS) in women with SLC1A5-positive non-serous tumors was 92% compared with of 66% in patients with SLC1A5-negative non-serous tumors (Log-rank = 15.343; P = 0.008). In Cox analysis with DFS as endpoint, FIGO-stage (HR = 4.5) and SLC1A5 status (HR = 0.3) were prognostic factors. CONCLUSIONS As the proteins HRNPM and SLC1A5 are associated with the cell cycle regulators p21 or p27, the apoptosis regulators PTEN and PUMA, and the VEGF-R2 it is concluded that both proteins have role in the pathogenesis of ovarian cancer. In patients with non-serous ovarian cancer SLC1A5 protects from recurrent disease, presumably by means of biological mechanisms that are unrelated to cytotoxic drug sensitivity.
Collapse
Affiliation(s)
- Kathrine Bjersand
- Department of Women’s and Children’s Health, Uppsala University, Uppsala, Sweden
| | - Tomas Seidal
- Department of Pathology, Halmstad Medical Center Hospital, Halmstad, Sweden
| | | | - Helena Åkerud
- Department of Immunology, Genetics and Pathology, Uppsala, Sweden
| | | |
Collapse
|
32
|
The Roles of Carcinoembryonic Antigen in Liver Metastasis and Therapeutic Approaches. Gastroenterol Res Pract 2017; 2017:7521987. [PMID: 28588612 PMCID: PMC5447280 DOI: 10.1155/2017/7521987] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 04/16/2017] [Indexed: 12/22/2022] Open
Abstract
Metastasis is a highly complicated and sequential process in which primary cancer spreads to secondary organic sites. Liver is a well-known metastatic organ from colorectal cancer. Carcinoembryonic antigen (CEA) is expressed in most gastrointestinal, breast, and lung cancer cells. Overexpression of CEA is closely associated with liver metastasis, which is the main cause of death from colorectal cancer. CEA is widely used as a diagnostic and prognostic tumor marker in cancer patients. It affects many steps of liver metastasis from colorectal cancer cells. CEA inhibits circulating cancer cell death. CEA also binds to heterogeneous nuclear RNA binding protein M4 (hnRNP M4), a Kupffer cell receptor protein, and activates Kupffer cells to secrete various cytokines that change the microenvironments for the survival of colorectal cancer cells in the liver. CEA also activates cell adhesion-related molecules. The close correlation between CEA and cancer has spurred the exploration of many CEA-targeted approaches as anticancer therapeutics. Understanding the detailed functions and mechanisms of CEA in liver metastasis will provide great opportunities for the improvement of anticancer approaches against colorectal cancers. In this report, the roles of CEA in liver metastasis and CEA-targeting anticancer modalities are reviewed.
Collapse
|
33
|
Sun H, Liu T, Zhu D, Dong X, Liu F, Liang X, Chen C, Shao B, Wang M, Wang Y. HnRNPM and CD44s expression affects tumor aggressiveness and predicts poor prognosis in breast cancer with axillary lymph node metastases. Genes Chromosomes Cancer 2017; 56:598-607. [PMID: 28393427 DOI: 10.1002/gcc.22463] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/04/2017] [Accepted: 04/05/2017] [Indexed: 12/12/2022] Open
Abstract
HnRNPM is an essential splicing factor and its expression is closely correlated with invasion and metastasis of tumor cells. The CD44 cell adhesion molecule is aberrantly expressed in many breast tumors and CD44 splice variants have been implicated in specific oncogenic signaling pathways. To investigate the clinical significance and biological function of hnRNPM, immunohistochemistry, quantitative, and semiquantitative polymerase chain reaction, lentiviral transfection system and transwell invasion assays were performed. We found that hnRNPM expression was significantly upregulated in breast cancer tissues compared with benign breast lesions. Although there was no significant correlation between hnRNPM and total CD44 protein or mRNA level, there was a negative correlation between hnRNPM and CD44v6. HnRNPM and CD44s expression showed positive correlation and in particular, they were dually expressed in breast cancer tissues. Interestingly, cancer stem cells marker, ALDH1+ phenotype was positively associated with overexpression of CD44s or hnRNPM and negatively related to CD44v6. Patients with high hnRNPM tended to have higher levels of CD44s, shorter overall survival (OS) and higher rates of lymph node metastases (LNM). Remarkably, Kaplan-Meier and Cox regression analyses displayed that hnRNPM+ or CD44shigh was a poor prognostic factor for OS of patients with LNM. Upregulation of hnRNPM in MCF-7 cells caused a significant increase in cell invasion, and this effect may occur through the regulation of CD44s expression. In conclusion, overexpression of hnRNPM promotes breast cancer aggressiveness by regulating the level of CD44s, indicates a poor prognosis for patients with LNM, and has potential as therapeutic targets.
Collapse
Affiliation(s)
- Huizhi Sun
- Department of Pathology, Tianjin Medical University, 300070, China
| | - Tieju Liu
- Department of Pathology, Tianjin Medical University, 300070, China.,Department of Pathology, General Hospital of Tianjin Medical University, 300052, China
| | - Dongwang Zhu
- Stomatology Hospital of Tianjin Medical University, China
| | - Xueyi Dong
- Department of Pathology, Tianjin Medical University, 300070, China.,Department of Pathology, General Hospital of Tianjin Medical University, 300052, China
| | - Fang Liu
- Department of Pathology, Tianjin Medical University, 300070, China.,Department of Pathology, General Hospital of Tianjin Medical University, 300052, China
| | - Xiaohui Liang
- Department of Pathology, Tianjin Medical University, 300070, China.,Department of Pathology, General Hospital of Tianjin Medical University, 300052, China
| | - Chen Chen
- Department of Pathology, Tianjin Medical University, 300070, China
| | - Bing Shao
- Department of Pathology, Tianjin Medical University, 300070, China
| | - Meili Wang
- Department of Pathology, Tianjin Medical University, 300070, China
| | - Yi Wang
- Department of Pathology, Tianjin Medical University, 300070, China
| |
Collapse
|
34
|
Tian N, Li J, Shi J, Sui G. From General Aberrant Alternative Splicing in Cancers and Its Therapeutic Application to the Discovery of an Oncogenic DMTF1 Isoform. Int J Mol Sci 2017; 18:ijms18030191. [PMID: 28257090 PMCID: PMC5372486 DOI: 10.3390/ijms18030191] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 01/03/2017] [Accepted: 01/10/2017] [Indexed: 12/20/2022] Open
Abstract
Alternative pre-mRNA splicing is a crucial process that allows the generation of diversified RNA and protein products from a multi-exon gene. In tumor cells, this mechanism can facilitate cancer development and progression through both creating oncogenic isoforms and reducing the expression of normal or controllable protein species. We recently demonstrated that an alternative cyclin D-binding myb-like transcription factor 1 (DMTF1) pre-mRNA splicing isoform, DMTF1β, is increasingly expressed in breast cancer and promotes mammary tumorigenesis in a transgenic mouse model. Aberrant pre-mRNA splicing is a typical event occurring for many cancer-related functional proteins. In this review, we introduce general aberrant pre-mRNA splicing in cancers and discuss its therapeutic application using our recent discovery of the oncogenic DMTF1 isoform as an example. We also summarize new insights in designing novel targeting strategies of cancer therapies based on the understanding of deregulated pre-mRNA splicing mechanisms.
Collapse
Affiliation(s)
- Na Tian
- College of Life Science, Northeast Forestry University, Harbin 150040, China.
| | - Jialiang Li
- College of Life Science, Northeast Forestry University, Harbin 150040, China.
| | - Jinming Shi
- College of Life Science, Northeast Forestry University, Harbin 150040, China.
| | - Guangchao Sui
- College of Life Science, Northeast Forestry University, Harbin 150040, China.
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.
| |
Collapse
|
35
|
Huang T, Xiao Y, Yi L, Li L, Wang M, Tian C, Ma H, He K, Wang Y, Han B, Ye X, Li X. Coptisine from Rhizoma Coptidis Suppresses HCT-116 Cells-related Tumor Growth in vitro and in vivo. Sci Rep 2017; 7:38524. [PMID: 28165459 PMCID: PMC5292956 DOI: 10.1038/srep38524] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 11/11/2016] [Indexed: 12/21/2022] Open
Abstract
Colorectal cancer is one of the most common causes of cancer-related death in humans. Coptisine (COP) is a natural alkaloid from Coptidis Rhizoma with unclear antitumor mechanism. Human colon cancer cells (HCT-116) and xenograft mice were used to systematically explore the anti-tumor activity of COP in this study. The results indicated that COP exhibited remarkably cytotoxic activities against the HCT-116 cells by inducing G1-phase cell cycle arrest and increasing apoptosis, and preferentially inhibited the survival pathway and induced the activation of caspase proteases family of HCT-116 cells. Experimental results on male BALB/c nude mice confirmed that orally administration of COP at high-dose (150 mg/kg) could suppress tumor growth, and may reduce cancer metastasis risk by inhibiting the RAS-ERK pathway in vivo. Taken together, the results suggested that COP may be potential as a novel anti-tumor candidate in the HCT-116 cells-related colon cancer, further studies are still needed to suggest COP for the further use.
Collapse
Affiliation(s)
- Tao Huang
- School of Chinese Traditional Medicine, School of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Yubo Xiao
- School of Chinese Traditional Medicine, School of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
- Department of Clinical Laboratory, Hunan University of Medicine, Hunan 418000, China
| | - Lin Yi
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing Cancer Institute & Hospital & Cancer Center, Chongqing 400030, China
| | - Ling Li
- School of Chinese Traditional Medicine, School of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Meimei Wang
- School of Chinese Traditional Medicine, School of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Cheng Tian
- School of Chinese Traditional Medicine, School of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Hang Ma
- School of Chinese Traditional Medicine, School of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Kai He
- School of Chinese Traditional Medicine, School of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
- Department of Clinical Laboratory, Hunan University of Medicine, Hunan 418000, China
| | - Yue Wang
- School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Bing Han
- School of Chinese Traditional Medicine, School of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Xiaoli Ye
- School of Life Sciences, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center for Pharmaceutical Process and Quality Control, Chongqing 400716, China
| | - Xuegang Li
- School of Chinese Traditional Medicine, School of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
- Chongqing Engineering Research Center for Pharmaceutical Process and Quality Control, Chongqing 400716, China
| |
Collapse
|
36
|
Mohr AM, Gould JJ, Kubik JL, Talmon GA, Casey CA, Thomas P, Tuma DJ, McVicker BL. Enhanced colorectal cancer metastases in the alcohol-injured liver. Clin Exp Metastasis 2017; 34:171-184. [PMID: 28168393 DOI: 10.1007/s10585-017-9838-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 01/16/2017] [Indexed: 02/08/2023]
Abstract
Metastatic liver disease is a major cause of mortality in colorectal cancer (CRC) patients. Alcohol consumption is a noted risk factor for secondary cancers yet the role of alcoholic liver disease (ALD) in colorectal liver metastases (CRLM) is not defined. This work evaluated tumor cell colonization in the alcoholic host liver using a novel preclinical model of human CRC liver metastases. Immunocompromised Rag1-deficient mice were fed either ethanol (E) or isocaloric control (C) diets for 4 weeks prior to intrasplenic injection of LS174T human CRC cells. ALD and CRLM were evaluated 3 or 5 weeks post-LS174T cell injection with continued C/E diet administration. ALD was confirmed by increased serum transaminases, hepatic steatosis and expression of cytochrome P4502E1, a major ethanol-metabolizing enzyme. Alcohol-mediated liver dysfunction was validated by impaired endocytosis of asialoorosomucoid and carcinoembryonic antigen (CEA), indicators of hepatocellular injury and progressive CRC disease, respectively. Strikingly, the rate and burden of CRLM was distinctly enhanced in alcoholic livers with metastases observed earlier and more severely in E-fed mice. Further, alcohol-related increases (1.5-3.0 fold) were observed in the expression of hepatic cytokines (TNF-α, IL-1 beta, IL-6, IL-10) and other factors noted to be involved in the colonization of CRC cells including ICAM-1, CCL-2, CCL-7, MMP-2, and MMP-9. Also, alcoholic liver injury was associated with altered hepatic localization as well as increased circulating levels of CEA released from CRC cells. Altogether, these findings indicate that the alcoholic liver provides a permissive environment for the establishment of CRLM, possibly through CEA-related inflammatory mechanisms.
Collapse
Affiliation(s)
- Ashley M Mohr
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - John J Gould
- Research Service, VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Jacy L Kubik
- Research Service, VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA.,Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Geoffrey A Talmon
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Carol A Casey
- Research Service, VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA.,Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Peter Thomas
- Department of Surgery and Biomedical Sciences, Creighton University, Omaha, NE, USA
| | - Dean J Tuma
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Benita L McVicker
- Research Service, VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA. .,Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
37
|
Bajenova O, Gorbunova A, Evsyukov I, Rayko M, Gapon S, Bozhokina E, Shishkin A, O’Brien SJ. The Genome-Wide Analysis of Carcinoembryonic Antigen Signaling by Colorectal Cancer Cells Using RNA Sequencing. PLoS One 2016; 11:e0161256. [PMID: 27583792 PMCID: PMC5008809 DOI: 10.1371/journal.pone.0161256] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 08/02/2016] [Indexed: 02/06/2023] Open
Abstract
Сarcinoembryonic antigen (CEA, CEACAM5, CD66) is a promoter of metastasis in epithelial cancers that is widely used as a prognostic clinical marker of metastasis. The aim of this study is to identify the network of genes that are associated with CEA-induced colorectal cancer liver metastasis. We compared the genome-wide transcriptomic profiles of CEA positive (MIP101 clone 8) and CEA negative (MIP 101) colorectal cancer cell lines with different metastatic potential in vivo. The CEA-producing cells displayed quantitative changes in the level of expression for 100 genes (over-expressed or down-regulated). They were confirmed by quantitative RT-PCR. The KEGG pathway analysis identified 4 significantly enriched pathways: cytokine-cytokine receptor interaction, MAPK signaling pathway, TGF-beta signaling pathway and pyrimidine metabolism. Our results suggest that CEA production by colorectal cancer cells triggers colorectal cancer progression by inducing the epithelial- mesenchymal transition, increasing tumor cell invasiveness into the surrounding tissues and suppressing stress and apoptotic signaling. The novel gene expression distinctions establish the relationships between the existing cancer markers and implicate new potential biomarkers for colorectal cancer hepatic metastasis.
Collapse
Affiliation(s)
- Olga Bajenova
- Theodosius Dobzhansky Center for Genome Bioinformatics, St. Petersburg State University, St. Petersburg, Russia
- Department of Genetics and Biotechnology, St. Petersburg State University, St. Petersburg, Russia
- * E-mail:
| | - Anna Gorbunova
- Theodosius Dobzhansky Center for Genome Bioinformatics, St. Petersburg State University, St. Petersburg, Russia
| | - Igor Evsyukov
- Theodosius Dobzhansky Center for Genome Bioinformatics, St. Petersburg State University, St. Petersburg, Russia
| | - Michael Rayko
- Theodosius Dobzhansky Center for Genome Bioinformatics, St. Petersburg State University, St. Petersburg, Russia
| | - Svetlana Gapon
- Boston Children’s Hospital, Boston, MA, United States of America
| | | | - Alexander Shishkin
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, United States of America
| | - Stephen J. O’Brien
- Theodosius Dobzhansky Center for Genome Bioinformatics, St. Petersburg State University, St. Petersburg, Russia
- Oceanographic Center, 8000 N. Ocean Drive, Nova Southeastern University, Ft Lauderdale, Florida, 33004, United States of America
| |
Collapse
|
38
|
Tobi M, Thomas P, Ezekwudo D. Avoiding hepatic metastasis naturally: Lessons from the cotton top tamarin (Saguinus oedipus). World J Gastroenterol 2016; 22:5479-94. [PMID: 27350726 PMCID: PMC4917608 DOI: 10.3748/wjg.v22.i24.5479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 04/26/2016] [Accepted: 05/21/2016] [Indexed: 02/06/2023] Open
Abstract
Much has been written about hepatic metastasis and animal models abound. In terms of the human experience, progress in treating this final common pathway, a terminal event of many human malignancies has been relatively slow. The current thinking is that primary prevention is best served by early detection of cancer and eradication of early stage cancers by screening. Some cancers spread early in their course and the role of screening may be limited. Until relatively recently there has not been a pathfinder model that makes the evasion of this unfortunate event a reality. This review discusses such an animal model and attempts to relate it to human disease in terms of intervention. Concrete proposals are also offered on how scientists may be able to intervene to prevent this deadly progression of the cancer process.
Collapse
|
39
|
Saito G, Sadahiro S, Okada K, Tanaka A, Suzuki T, Kamijo A. Relation between Carcinoembryonic Antigen Levels in Colon Cancer Tissue and Serum Carcinoembryonic Antigen Levels at Initial Surgery and Recurrence. Oncology 2016; 91:85-9. [PMID: 27260164 DOI: 10.1159/000447062] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 05/23/2016] [Indexed: 11/19/2022]
Abstract
OBJECTIVES Carcinoembryonic antigen (CEA) is widely used for postoperative surveillance of colon cancer. Even if serum CEA is negative at initial surgery, it may turn positive at recurrence. We investigated the relation between serum CEA levels and the immunohistochemical staining status of CEA in the primary and resected metastatic tissues. METHODS Out of 224 patients with recurrent colon cancer between 1998 and 2012, we studied 46 patients in whom serum CEA levels were measured and immunohistochemical staining for CEA was possible in the primary and metastatic tissues. RESULTS The positive rate of serum CEA did not differ between initial surgery and recurrence, regardless of whether the cutoff value was set at 5 or 10 ng/ml (p = 0.829, p = 0.671). There was no relation between the CEA staining status and serum CEA level at initial surgery. However, the CEA staining status of metastatic tissue was significantly related to the serum CEA level at recurrence (p = 0.0046 and p = 0.0026). CONCLUSIONS The immunohistochemical staining status of CEA in metastatic tissue is closely related to the serum CEA level. This finding suggests that serum CEA levels are influenced not only by the CEA production capacity of cancer cells but also by the ability of the surrounding tissue to release CEA into the blood.
Collapse
Affiliation(s)
- Gota Saito
- Department of Surgery, Tokai University School of Medicine, Kanagawa, Japan
| | | | | | | | | | | |
Collapse
|
40
|
Haenisch F, Cooper JD, Reif A, Kittel-Schneider S, Steiner J, Leweke FM, Rothermundt M, van Beveren NJM, Crespo-Facorro B, Niebuhr DW, Cowan DN, Weber NS, Yolken RH, Penninx BWJH, Bahn S. Towards a blood-based diagnostic panel for bipolar disorder. Brain Behav Immun 2016; 52:49-57. [PMID: 26441135 DOI: 10.1016/j.bbi.2015.10.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 09/08/2015] [Accepted: 10/02/2015] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Bipolar disorder (BD) is a costly, devastating and life shortening mental disorder that is often misdiagnosed, especially on initial presentation. Misdiagnosis frequently results in ineffective treatment. We investigated the utility of a biomarker panel as a diagnostic test for BD. METHODS AND FINDINGS We performed a meta-analysis of eight case-control studies to define a diagnostic biomarker panel for BD. After validating the panel on established BD patients, we applied it to undiagnosed BD patients. We analysed 249 BD, 122 pre-diagnostic BD, 75 pre-diagnostic schizophrenia and 90 first onset major depression disorder (MDD) patients and 371 controls. The biomarker panel was identified using ten-fold cross-validation with lasso regression applied to the 87 analytes available across the meta-analysis studies. We identified 20 protein analytes with excellent predictive performance [area under the curve (AUC)⩾0.90]. Importantly, the panel had a good predictive performance (AUC 0.84) to differentiate 12 misdiagnosed BD patients from 90 first onset MDD patients, and a fair to good predictive performance (AUC 0.79) to differentiate between 110 pre-diagnostic BD patients and 184 controls. We also demonstrated the disease specificity of the panel. CONCLUSIONS An early and accurate diagnosis has the potential to delay or even prevent the onset of BD. This study demonstrates the potential utility of a biomarker panel as a diagnostic test for BD.
Collapse
Affiliation(s)
- Frieder Haenisch
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, United Kingdom
| | - Jason D Cooper
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, United Kingdom
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Goethe University Hospital Frankfurt, Germany
| | - Sarah Kittel-Schneider
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Goethe University Hospital Frankfurt, Germany
| | - Johann Steiner
- Department of Psychiatry, Psychotherapy and Psychosomatic Medicine, University Magdeburg, Magdeburg, Germany
| | - F Markus Leweke
- Central Institute of Mental Health, University of Heidelberg, Medical Faculty Mannheim, Mannheim, Germany
| | | | - Nico J M van Beveren
- Department of Psychiatry, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Benedicto Crespo-Facorro
- CIBERSAM, University Hospital Marqués de Valdecilla, University of Cantabria - IDIVAL, Department of Psychiatry, Santander, Spain
| | - David W Niebuhr
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - David N Cowan
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Natalya S Weber
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Robert H Yolken
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Brenda W J H Penninx
- Department of Psychiatry, EMGO Institute, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Sabine Bahn
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
41
|
Abstract
OBJECTIVES High serum level of interleukin 6 (IL-6) is associated with high degree of tumor progression and systemic weakness. Anti-IL-6 therapy possibly improves the deterioration of clinical characteristics in patients with high IL-6 level. However, IL-6-related factors in patients with treatment-naive advanced pancreatic cancer (PC) have not been established. The goal of this study was to identify IL-6-related factors in patients with advanced PC who were scheduled to undergo first-line chemotherapy. METHODS Patients with treatment-naive advanced PC were eligible for inclusion in this study. Patients who did not receive first-line chemotherapy were excluded. Serum IL-6 levels and clinical parameters were prospectively recorded. Analyses were performed to identify risk factors for high IL-6 levels. RESULTS Eighty patients were analyzed. IL-6-related factors were advanced age (P < 0.01), the presence of liver metastasis (P < 0.01), the large volume of liver metastasis (P < 0.01), severe fatigue (P = 0.02), high carcinoembryonic antigen levels (P = 0.02), anemia (P < 0.01), and high C-reactive protein levels (P = 0.02) in multivariate analyses. Decreased skeletal muscle mass tended to be associated with high IL-6 levels. CONCLUSIONS High serum IL-6 was related to advanced age, the presence of hepatic metastasis, large tumor burden in liver, severe fatigue, high carcinoembryonic antigen, high C-reactive protein, and anemia in patients with treatment-naive advanced PC.
Collapse
|
42
|
Lee KA, Bae EA, Song YC, Kim EK, Lee YS, Kim TG, Kang CY. A multimeric carcinoembryonic antigen signal inhibits the activation of human T cells by a SHP-independent mechanism: a potential mechanism for tumor-mediated suppression of T-cell immunity. Int J Cancer 2015; 136:2579-87. [PMID: 25379865 DOI: 10.1002/ijc.29314] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2013] [Accepted: 10/11/2014] [Indexed: 01/21/2023]
Abstract
Carcinoembryonic antigen (CEA) is a well-known tumor antigen that is found in the serum of patients with various cancers and is correlated with an increased risk of cancer recurrence and metastasis. To understand the tumor environment and to develop antitumor therapies, CEA has been studied as an antigen to activate/tolerate specific T cells. In this study, we show that CEA can function as a coinhibitory molecule and can inhibit the activation of human peripheral blood mononucleated cell-derived T cells. The addition of CEA-overexpressing tumor cells or immobilized CEA dampened both cell proliferation and the expression of IL-2 and CD69 expression in T cells after TCR stimulation. The phosphorylation of ERK and translocation of NFAT were hampered in these cells, whereas the phosphorylation of proximal TCR signaling molecules such as ZAP70 and phospholipase Cγ was not affected by immobilized CEA. To determine the relevance of carcinoembryonic antigen-related cell adhesion molecule-1 and Src homology region 2 domain-containing phosphatase (SHP) molecules to CEA-mediated suppression, we tested the effect of the SHP inhibitor, NSC-87877, on CEA-mediated suppression of T cells; however, it did not reverse the effect of CEA. Collectively, these results indicate that CEA can function as a modulator of T-cell responses suggesting a novel mechanism of tumor evasion.
Collapse
Affiliation(s)
- Kyoo-A Lee
- Laboratory of Immunology, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
43
|
Jensen-Jarolim E, Fazekas J, Singer J, Hofstetter G, Oida K, Matsuda H, Tanaka A. Crosstalk of carcinoembryonic antigen and transforming growth factor-β via their receptors: comparing human and canine cancer. Cancer Immunol Immunother 2015; 64:531-7. [PMID: 25832000 PMCID: PMC4412651 DOI: 10.1007/s00262-015-1684-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 03/19/2015] [Indexed: 01/27/2023]
Abstract
There is accumulating evidence that the transforming growth factor beta (TGF-β) and nuclear factor kappa-B (NFκB) pathways are tightly connected and play a key role in malignant transformation in cancer. Immune infiltration by regulatory T- and B-lymphocytes (Tregs, Bregs) has recently gained increased attention for being an important source of TGF-β. There is a plethora of studies examining the pro-tumorigenic functions of carcinoembryonic antigen (CEA), but its receptor CEAR is far less studied. So far, there is a single connecting report that TGF-β also may signal through CEAR. The crosstalk between cancer tissues is further complicated by the expression of CEAR and TGF-β receptors in stromal cells, and implications of TGF-β in epithelial–mesenchymal transition. Furthermore, tumor-infiltrating Tregs and Bregs may directly instruct cancer cells by secreting TGF-β binding to their CEAR. Therefore, both TGF-β and CEA may act synergistically in breast cancer and cause disease progression, and NFκB could be a common crossing point between their signaling. CEAR, TGF-β1–3, TGF-β-R types I–III and NFκB class I and II molecules have an outstanding human–canine sequence identity, and only a canine CEA homolog has not yet been identified. For these reasons, the dog may be a valid translational model patient for investigating the crosstalk of the interconnected CEA and TGF-β networks.
Collapse
Affiliation(s)
- Erika Jensen-Jarolim
- Department of Comparative Medicine, Comparative Immunology and Oncology, Messerli Research Institute of the University of Veterinary Medicine Vienna, c/o Institute of Pathophysiology and Allergy Research, AKH 4Q, Medical University Vienna and University Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria,
| | | | | | | | | | | | | |
Collapse
|
44
|
A549 cells adapted to high nitric oxide show reduced surface CEACAM expression and altered adhesion and migration properties. Tumour Biol 2014; 36:1871-9. [PMID: 25500969 DOI: 10.1007/s13277-014-2789-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 10/29/2014] [Indexed: 12/12/2022] Open
Abstract
The migration and adhesion properties of tumors affect their metastatic rate. In the present study, we investigated carcinoembryonic antigen-related cell adhesion molecule (CEACAM) 1, 5, and 6 expression in high nitric oxide (HNO)-adapted lung cancer cells compared to parent cells. We observed high transcript levels of CEACAM 1 (4S, 4L), CEACAM 5, and CEACAM 6 in HNO cells compared to parent cells. However, the surface expression was low in HNO cells. Interestingly, the intracellular protein levels were high for these three CEACAMs. We confirmed these results with immunohistochemical experiments. Further, the adhesion and migration assays showed reduced clumping in HNO-adapted A549 (A549-HNO) cells and faster migration rates, respectively. These results document the altered adhesion and migration properties of cells adapted to HNO. Further, our studies also indicate a dynamic regulation of CEACAM protein expression and surface transport in HNO cells.
Collapse
|
45
|
Choi M, Thakur A. Identifying Appropriate Colorectal Cancer-Associated Antigens for the Clinical Trials. CURRENT COLORECTAL CANCER REPORTS 2014. [DOI: 10.1007/s11888-014-0256-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
46
|
Fontana R, Herman P, Hermam P, Pugliese V, Perini MV, Coelho FF, Velho FF, Cecconello I. Surgical outcomes and prognostic factors in patients with synchronous colorectal liver metastases. ARQUIVOS DE GASTROENTEROLOGIA 2014; 51:4-9. [PMID: 24760056 DOI: 10.1590/s0004-28032014000100002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 10/25/2013] [Indexed: 02/06/2023]
Abstract
CONTEXT Colorectal cancer is the second most prevalent cancer worldwide, and the liver is the most common site of metastases. Surgical resection of colorectal liver metastases provides the sole possibility of cure and the best odds of long-term survival. Objectives To describe surgical outcomes and identify features associated with disease prognosis in patients submitted to synchronous colorectal cancer liver metastasis resection. METHODS Retrospective study of 59 patients who underwent surgery for synchronous colorectal cancer liver metastasis. Actuarial survival and disease-free survival were assessed, depending on the prognostic variable of interest. RESULTS Postoperative mortality and morbidity rates were 3.38% and 30.50% respectively. Five-year disease-free survival was estimated at 23.96%, and 5-year overall survival, at 38.45%. Carcinoembryonic antigen levels ≥ 50 ng/mL and presence of three or more liver metastasis were limiting factors for disease-free survival, but did not affect late survival. No patient with liver metastases and extrahepatic disease had disease-free interval longer than 20 months, but this had no significance or impact on long-term survival. None of the prognostic factors assessed had an impact on late survival, although no patients with more than three liver metastases survived beyond 40 months. CONCLUSIONS Although Carcinoembryonic antigen levels and number of metastases are prognostic factors that limit disease-free survival, they had no impact on 5-year survival and, therefore, should not determine exclusion from surgical treatment. Resection is the best treatment option for synchronous colorectal liver metastases, and even for patients with multiple metastases, large tumors and extrahepatic disease, it can provide long-term survival rates over 38%.
Collapse
Affiliation(s)
- Rafael Fontana
- Universidade de Caxias do Sul, Faculdade de Medicina, Caxias do Sul, RS, Brasil
| | | | - Paulo Hermam
- Universidade de São Paulo, Faculdade de Medicina, Departmento de Gastroenterologia, São Paulo, SP, Brasil
| | - Vincenzo Pugliese
- Universidade de São Paulo, Faculdade de Medicina, Departmento de Gastroenterologia, São Paulo, SP, Brasil
| | - Marcos Vinicius Perini
- Universidade de São Paulo, Faculdade de Medicina, Departmento de Gastroenterologia, São Paulo, SP, Brasil
| | | | - Fabricio Ferreira Velho
- Universidade de São Paulo, Faculdade de Medicina, Departmento de Gastroenterologia, São Paulo, SP, Brasil
| | - Ivan Cecconello
- Universidade de São Paulo, Faculdade de Medicina, Departmento de Gastroenterologia, São Paulo, SP, Brasil
| |
Collapse
|
47
|
A Monoclonal Antibody Against Neem Leaf Glycoprotein Recognizes Carcinoembryonic Antigen (CEA) and Restricts CEA Expressing Tumor Growth. J Immunother 2014; 37:394-406. [DOI: 10.1097/cji.0000000000000050] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
48
|
Berkovich L, Shpitz B, Ghinea R, Greemland I, Kravtsov V, Kidron D, Mishaeli M, Avital S. Evaluation of peritoneal CEA levels following colorectal cancer surgery. J Surg Oncol 2014; 110:458-62. [PMID: 24910092 DOI: 10.1002/jso.23676] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 05/15/2014] [Indexed: 12/22/2022]
Abstract
BACKGROUND Peritoneal carcinoembryonic antigen (pCEA) levels in the early postoperative period following a curative resection of colorectal cancer (CRC) have not been previously studied. METHODS Postoperative peritoneal fluids of 36 CRC patients followed by 24 benign colonic disease patients were evaluated for CEA levels and tumor cell presence. Serum CEA levels were also evaluated prior and after surgery. RESULTS Although high postoperative pCEA levels were observed in some benign patients, more CRC patients exhibited significant elevation of postoperative pCEA (>5 ng/ml) compared to benign patients (50% vs. 23%, P = 0.039). Postoperative median pCEA levels of CRC patients were significantly higher compared to benign patients (5.4 vs. 2 ng/ml, P = 0.011). Specifically, pCEA levels in CRC patients were significantly elevated when measured during the first 24 hr after surgery. Postoperative pCEA levels were associated with colon tumor location compared to rectal location. However, no correlation was found with known risk factors for cancer recurrence or with serum CEA levels. CONCLUSIONS Postoperative pCEA levels may be significantly elevated following a curative resection for CRC. Its significance within patient's prognostic evaluation remains to be studied. Inclusion of patient's follow-up data may reveal the significance of elevated pCEA levels following CRC resection.
Collapse
Affiliation(s)
- Liron Berkovich
- Department of Surgery B, Meir Medical Center and the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Xu Y, Gao XD, Lee JH, Huang H, Tan H, Ahn J, Reinke LM, Peter ME, Feng Y, Gius D, Siziopikou KP, Peng J, Xiao X, Cheng C. Cell type-restricted activity of hnRNPM promotes breast cancer metastasis via regulating alternative splicing. Genes Dev 2014; 28:1191-203. [PMID: 24840202 PMCID: PMC4052765 DOI: 10.1101/gad.241968.114] [Citation(s) in RCA: 183] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Tumor metastasis remains the major cause of cancer-related death, but its molecular basis is still not well understood. Here we uncovered a splicing-mediated pathway that is essential for breast cancer metastasis. We show that the RNA-binding protein heterogeneous nuclear ribonucleoprotein M (hnRNPM) promotes breast cancer metastasis by activating the switch of alternative splicing that occurs during epithelial-mesenchymal transition (EMT). Genome-wide deep sequencing analysis suggests that hnRNPM potentiates TGFβ signaling and identifies CD44 as a key downstream target of hnRNPM. hnRNPM ablation prevents TGFβ-induced EMT and inhibits breast cancer metastasis in mice, whereas enforced expression of the specific CD44 standard (CD44s) splice isoform overrides the loss of hnRNPM and permits EMT and metastasis. Mechanistically, we demonstrate that the ubiquitously expressed hnRNPM acts in a mesenchymal-specific manner to precisely control CD44 splice isoform switching during EMT. This restricted cell-type activity of hnRNPM is achieved by competition with ESRP1, an epithelial splicing regulator that binds to the same cis-regulatory RNA elements as hnRNPM and is repressed during EMT. Importantly, hnRNPM is associated with aggressive breast cancer and correlates with increased CD44s in patient specimens. These findings demonstrate a novel molecular mechanism through which tumor metastasis is endowed by the hnRNPM-mediated splicing program.
Collapse
Affiliation(s)
- Yilin Xu
- Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | - Xin D Gao
- Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | - Jae-Hyung Lee
- Department of Integrative Biology and Physiology, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Huilin Huang
- Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | - Haiyan Tan
- Department of Structural Biology, St. Jude Proteomics Facility, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA; Department of Developmental Neurobiology, St. Jude Proteomics Facility, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - Jaegyoon Ahn
- Department of Integrative Biology and Physiology, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Lauren M Reinke
- Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | - Marcus E Peter
- Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | - Yue Feng
- Department of Pharmacology, Emory University, Atlanta, Georgia 30322, USA
| | - David Gius
- Department of Radiation Oncology, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | - Kalliopi P Siziopikou
- Department of Pathology, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | - Junmin Peng
- Department of Structural Biology, St. Jude Proteomics Facility, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA; Department of Developmental Neurobiology, St. Jude Proteomics Facility, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - Xinshu Xiao
- Department of Integrative Biology and Physiology, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Chonghui Cheng
- Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| |
Collapse
|
50
|
Bajenova O, Chaika N, Tolkunova E, Davydov-Sinitsyn A, Gapon S, Thomas P, O'Brien S. Carcinoembryonic antigen promotes colorectal cancer progression by targeting adherens junction complexes. Exp Cell Res 2014; 324:115-23. [PMID: 24726916 DOI: 10.1016/j.yexcr.2014.04.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 03/25/2014] [Accepted: 04/04/2014] [Indexed: 12/18/2022]
Abstract
Oncomarkers play important roles in the detection and management of human malignancies. Carcinoembryonic antigen (CEA, CEACAM5) and epithelial cadherin (E-cadherin) are considered as independent tumor markers in monitoring metastatic colorectal cancer. They are both expressed by cancer cells and can be detected in the blood serum. We investigated the effect of CEA production by MIP101 colorectal carcinoma cell lines on E-cadherin adherens junction (AJ) protein complexes. No direct interaction between E-cadherin and CEA was detected; however, the functional relationships between E-cadherin and its AJ partners: α-, β- and p120 catenins were impaired. We discovered a novel interaction between CEA and beta-catenin protein in the CEA producing cells. It is shown in the current study that CEA overexpression alters the splicing of p120 catenin and triggers the release of soluble E-cadherin. The influence of CEA production by colorectal cancer cells on the function of E-cadherin junction complexes may explain the link between the elevated levels of CEA and the increase in soluble E-cadherin during the progression of colorectal cancer.
Collapse
Affiliation(s)
- Olga Bajenova
- Theodosius Dobzhansky Center for Genome Bioinformatics, St. Petersburg State University, St. Petersburg 199034, Russia; Department of Genetics and Biotechnology, St. Petersburg State University, St. Petersburg 199034, Russia; Department of Surgery and Biomedical Sciences, Creighton University, Omaha, NE 68178, USA.
| | - Nina Chaika
- Department of Surgery and Biomedical Sciences, Creighton University, Omaha, NE 68178, USA
| | - Elena Tolkunova
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg 194064, Russia
| | | | | | - Peter Thomas
- Department of Surgery and Biomedical Sciences, Creighton University, Omaha, NE 68178, USA
| | - Stephen O'Brien
- Theodosius Dobzhansky Center for Genome Bioinformatics, St. Petersburg State University, St. Petersburg 199034, Russia
| |
Collapse
|