1
|
Fang J, Wang J, Zhao X, Yang Y, Xiao Y. KLHDC8A knockdown in normal ovarian epithelial cells promoted the polarization of pro-tumoral macrophages via the C5a/C5aR/p65 NFκB signaling pathway. Cell Immunol 2025; 409-410:104913. [PMID: 39805213 DOI: 10.1016/j.cellimm.2024.104913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/13/2024] [Accepted: 12/22/2024] [Indexed: 01/16/2025]
Abstract
AIMS Tumor-associated macrophages (TAM) is related to Ovarian cancer (OC) pathogenesis, but the exact mechanism remains unclear. This study investigated the expression of Kelch Domain Containing 8 A (KLHDC8A) in OC and the mechanism associated with TAM. MAIN METHODS Bioinformatics analysis of differential expression genes between normal and OC tissues were analyzed based on the Tumor Genome Atlas (TCGA) databases. KLHDC8A mRNA expression was knocked down in normal epithelial cells (IOSE80), and then the effects of siKLHDC8A on the proliferation, invasion, migration and C5a secretion of IOSE80 cells were explored. THP1-derived macrophages were cultured with medium of NC-IOSE80 cells, siKLHDC8A-IOSE80 cells with or without C5aR antagonists. KEY FINDINGS KLHDC8A was lowly expressed in OC and negatively correlated with the infiltration of tumor-promoting macrophages, contributing to the survival of OC patients. Furthermore, siKLHDC8A promotes the proliferation, invasion and migration of IOSE80 cells and leads to polarization of pro-tumoral macrophages, which can be rescued by C5aR antagonists. SIGNIFICANCE Our results indicated that KLHDC8A knockdown could modulate the development of OC by affecting macrophage polarization to pro-tumoral type via the C5a/C5aR/p65 NFκB signaling pathway. It may play an essential role as the tumor suppressor genes in diagnosis and treatment of OC.
Collapse
Affiliation(s)
- Jie Fang
- Department of Gynecology, the Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu 212001, China.
| | - Jin Wang
- Department of Gynecology, the Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - Xinyue Zhao
- Department of Gynecology, the Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - Yaping Yang
- Department of Gynecology, the Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - Yujia Xiao
- Department of Gynecology, the Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu 212001, China
| |
Collapse
|
2
|
Hsieh C, Wu Y, Chen Y, Wang C, Li C, Liu I, Chou C, Lin Y, Huang P, Huang T, Chen C. SERPING1 Reduces Cell Migration via ERK-MMP2-MMP-9 Cascade in Sorafenib- Resistant Hepatocellular Carcinoma. ENVIRONMENTAL TOXICOLOGY 2025; 40:318-327. [PMID: 39474998 PMCID: PMC11726270 DOI: 10.1002/tox.24434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/20/2024] [Accepted: 10/17/2024] [Indexed: 01/04/2025]
Abstract
Hepatocellular carcinoma (HCC) is the most common primary hepatic malignant tumor, and it ranks 2nd in terms of mortality rate among all malignancies in Taiwan. Sorafenib is a multiple tyrosine kinase inhibitor that suppresses tumor cell proliferation and angiogenesis around tumors via different pathways. However, the survival outcome of advanced HCC patients treated with sorafenib is still unsatisfactory. Unfortunately, there are no clinically applicable biomarkers to predict sorafenib therapeutic efficiency in HCC thus far. We found that serpin peptidase inhibitor, clade G, member 1 (SERPING1) is highly associated with overall and recurrence-free survival rates in HCC patients and is also highly correlated with several clinical parameters. SERPING1 expression was increased with sorafenib in both the HCC cell extract and conditioned medium, which was also observed in sorafenib-resistant HepG2 and Huh7 cells. Sorafenib decreased cell viability and migration, which was similar to the effect of SERPING1 in HCC progression. Moreover, sorafenib inhibited both MMP-2 and MMP-9 activity and enhanced the expression of p-ERK in HCC cells. In summary, sorafenib reduces HCC cancer progression might through the p-ERK-MMP-2-MMP-9 cascade via upregulation of SERPING1. In the present study, the roles and molecular mechanisms of SERPING1 and its value as a marker for predicting sorafenib resistance and progression in HCC patients were examined. The results of the present study provide a deep understanding of the roles of SERPING1 in HCC sorafenib resistance, which can be applied to develop early diagnosis and prognosis evaluation methods and establish novel therapeutic targets for specifically treating HCC.
Collapse
Affiliation(s)
- Ching‐Chuan Hsieh
- Division of General Surgery, Chang Gung Memorial HospitalChiayiTaiwan
| | - Yuh‐Harn Wu
- Department of Cell Biology and Anatomy, College of MedicineNational Cheng Kung UniversityTainanTaiwan
| | - Yi‐Li Chen
- Department of Cell Biology and Anatomy, College of MedicineNational Cheng Kung UniversityTainanTaiwan
| | - Chun‐I Wang
- Department of Biochemistry, School of MedicineChina Medical UniversityTaichungTaiwan
| | - Chao‐Jen Li
- Department of General & Gastroenterological Surgery, An Nan HospitalChina Medical UniversityTainanTaiwan
| | - I‐Hsiu Liu
- Department of Cell Biology and Anatomy, College of MedicineNational Cheng Kung UniversityTainanTaiwan
| | - Chen‐Wei Chou
- Department of Cell Biology and Anatomy, College of MedicineNational Cheng Kung UniversityTainanTaiwan
| | - Yang‐Hsiang Lin
- Liver Research Center, Chang Gung Memorial HospitalTaoyuanTaiwan
| | - Po‐Shuan Huang
- Department of Biochemistry, College of MedicineChang Gung UniversityTaoyuanTaiwan
- Graduate Institute of Biochemical and Biomedical EngineeringChang Gung UniversityTaoyuanTaiwan
| | - Te‐Chia Huang
- Department of General & Gastroenterological Surgery, An Nan HospitalChina Medical UniversityTainanTaiwan
| | - Cheng‐Yi Chen
- Department of Cell Biology and Anatomy, College of MedicineNational Cheng Kung UniversityTainanTaiwan
| |
Collapse
|
3
|
Nissinen L, Riihilä P, Viiklepp K, Rajagopal V, Storek MJ, Kähäri VM. C1s targeting antibodies inhibit the growth of cutaneous squamous carcinoma cells. Sci Rep 2024; 14:13465. [PMID: 38866870 PMCID: PMC11169539 DOI: 10.1038/s41598-024-64088-3] [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: 02/13/2024] [Accepted: 06/05/2024] [Indexed: 06/14/2024] Open
Abstract
Cutaneous squamous cell carcinoma (cSCC) is the most common metastatic skin cancer. The incidence of cSCC is increasing globally and the prognosis of metastatic disease is poor. Currently there are no specific targeted therapies for advanced or metastatic cSCC. We have previously shown abundant expression of the complement classical pathway C1 complex components, serine proteases C1r and C1s in tumor cells in invasive cSCCs in vivo, whereas the expression of C1r and C1s was lower in cSCCs in situ, actinic keratoses and in normal skin. We have also shown that knockdown of C1s expression results in decreased viability and growth of cSCC cells by promoting apoptosis both in culture and in vivo. Here, we have studied the effect of specific IgG2a mouse monoclonal antibodies TNT003 and TNT005 targeting human C1s in five primary non-metastatic and three metastatic cSCC cell lines that show intracellular expression of C1s and secretion of C1s into the cell culture media. Treatment of cSCC cells with TNT003 and TNT005 significantly inhibited their growth and viability and promoted apoptosis of cSCC cells. These data indicate that TNT003 and TNT005 inhibit cSCC cell growth in culture and warrant further investigation of C1s targeted inhibition in additional in vitro and in vivo models of cSCC.
Collapse
Affiliation(s)
- Liisa Nissinen
- Department of Dermatology and FICAN West Cancer Centre Research Laboratory, University of Turku and Turku University Hospital, Hämeentie 11 TE6, 20520, Turku, Finland
| | - Pilvi Riihilä
- Department of Dermatology and FICAN West Cancer Centre Research Laboratory, University of Turku and Turku University Hospital, Hämeentie 11 TE6, 20520, Turku, Finland
| | - Kristina Viiklepp
- Department of Dermatology and FICAN West Cancer Centre Research Laboratory, University of Turku and Turku University Hospital, Hämeentie 11 TE6, 20520, Turku, Finland
| | | | | | - Veli-Matti Kähäri
- Department of Dermatology and FICAN West Cancer Centre Research Laboratory, University of Turku and Turku University Hospital, Hämeentie 11 TE6, 20520, Turku, Finland.
| |
Collapse
|
4
|
Maugeri S, Sibbitts J, Privitera A, Cardaci V, Di Pietro L, Leggio L, Iraci N, Lunte SM, Caruso G. The Anti-Cancer Activity of the Naturally Occurring Dipeptide Carnosine: Potential for Breast Cancer. Cells 2023; 12:2592. [PMID: 37998326 PMCID: PMC10670273 DOI: 10.3390/cells12222592] [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: 09/25/2023] [Revised: 10/27/2023] [Accepted: 11/06/2023] [Indexed: 11/25/2023] Open
Abstract
Carnosine is an endogenous dipeptide composed of β-alanine and L-histidine, possessing a multimodal pharmacodynamic profile that includes anti-inflammatory and anti-oxidant activities. Carnosine has also shown its ability to modulate cell proliferation, cell cycle arrest, apoptosis, and even glycolytic energy metabolism, all processes playing a key role in the context of cancer. Cancer is one of the most dreaded diseases of the 20th and 21st centuries. Among the different types of cancer, breast cancer represents the most common non-skin cancer among women, accounting for an estimated 15% of all cancer-related deaths in women. The main aim of the present review was to provide an overview of studies on the anti-cancer activity of carnosine, and in particular its activity against breast cancer. We also highlighted the possible advantages and limitations involved in the use of this dipeptide. The first part of the review entailed a brief description of carnosine's biological activities and the pathophysiology of cancer, with a focus on breast cancer. The second part of the review described the anti-tumoral activity of carnosine, for which numerous studies have been carried out, especially at the preclinical level, showing promising results. However, only a few studies have investigated the therapeutic potential of this dipeptide for breast cancer prevention or treatment. In this context, carnosine has shown to be able to decrease the size of cancer cells and their viability. It also reduces the levels of vascular endothelial growth factor (VEGF), cyclin D1, NAD+, and ATP, as well as cytochrome c oxidase activity in vitro. When tested in mice with induced breast cancer, carnosine proved to be non-toxic to healthy cells and exhibited chemopreventive activity by reducing tumor growth. Some evidence has also been reported at the clinical level. A randomized phase III prospective placebo-controlled trial showed the ability of Zn-carnosine to prevent dysphagia in breast cancer patients undergoing adjuvant radiotherapy. Despite this evidence, more preclinical and clinical studies are needed to better understand carnosine's anti-tumoral activity, especially in the context of breast cancer.
Collapse
Affiliation(s)
- Salvatore Maugeri
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy
| | - Jay Sibbitts
- Ralph N. Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, KS 66047, USA
- Department of Chemistry, University of Kansas, Lawrence, KS 66047, USA
| | - Anna Privitera
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Vincenzo Cardaci
- Scuola Superiore di Catania, University of Catania, 95123 Catania, Italy
- Vita-Salute San Raffaele University, 20132 Milano, Italy
| | - Lucia Di Pietro
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy
- Scuola Superiore di Catania, University of Catania, 95123 Catania, Italy
| | - Loredana Leggio
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Nunzio Iraci
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Susan M. Lunte
- Ralph N. Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, KS 66047, USA
- Department of Chemistry, University of Kansas, Lawrence, KS 66047, USA
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66047, USA
| | - Giuseppe Caruso
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy
- Unit of Neuropharmacology and Translational Neurosciences, Oasi Research Institute-IRCCS, 94018 Troina, Italy
| |
Collapse
|
5
|
Khan A, Hussain S, Iyer JK, Kaul A, Bonnewitz M, Kaul R. Human papillomavirus-mediated expression of complement regulatory proteins in human cervical cancer cells. Eur J Obstet Gynecol Reprod Biol 2023; 288:222-228. [PMID: 37572452 DOI: 10.1016/j.ejogrb.2023.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/18/2023] [Accepted: 07/24/2023] [Indexed: 08/14/2023]
Abstract
OBJECTIVES This study aimed to evaluate the expression pattern of complement regulatory proteins (CRPs) CD46, CD59, and CD55 in HPV-positive (HPV+) & negative (HPV-) cervical cancer cell lines in search of a reliable differential biomarker. STUDY DESIGN We analysed the expression of CRPs in HPV 16-positive SiHa cell line, HPV 18-positive HeLa cell line, and HPV-negative cell line C33a using RT-qPCR, Western blotting, flow cytometry, and confocal microscopy. RESULTS We observed a differential expression profile of CRPs in HPV+ and HPV- cervical cancer cell lines. The mRNA level of CD59 & CD55 showed a higher expression pattern in HPV+ cells when compared to HPV- cancer cells. However, flow cytometry-based experiments revealed that CD46 was preferentially expressed more in HPV 16-positive SiHa cells followed by HPV 18-positive HeLa cells when compared to HPV- C33a cells. Interestingly, confocal microscopy revealed a high level of CD59 expression in Hela cells and SiHa cells but low expression in HPV- C33a cells. In addition, HPV 18-positive HeLa cells expressed more CD55, which was lower in SiHa cells and very weak in C33a cells. CONCLUSION The study demonstrates the differential expression of CRPs in both HPV+ and HPV- cervical cancer cells for the first time, and their potential to serve as an early diagnostic marker for cervical carcinogenesis.
Collapse
Affiliation(s)
- Asiya Khan
- Dr. Babasaheb R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi 110029, India; Amity Institute of Biotechnology, Amity University, Noida 201303, India
| | - Showket Hussain
- Division of Molecular Oncology & Molecular Diagnostics, Indian Council of Medical Research-National Institute of Cancer Prevention and Research, Noida 201301, India
| | - Janaki K Iyer
- Department of Biochemistry and Microbiology, Oklahoma State University Centre for Health Sciences, 1111 West 17(th) Street, Tulsa, OK 74107, USA; Department of Natural Sciences, Northeastern State University, Broken Arrow, OK 74014, USA
| | - Anil Kaul
- Health Care Administration, Oklahoma State University Centre for Health Sciences, Tulsa, OK 74107, USA
| | - Mackenzie Bonnewitz
- Department of Natural Sciences, Northeastern State University, Broken Arrow, OK 74014, USA
| | - Rashmi Kaul
- Department of Biochemistry and Microbiology, Oklahoma State University Centre for Health Sciences, 1111 West 17(th) Street, Tulsa, OK 74107, USA.
| |
Collapse
|
6
|
Liljedahl E, Konradsson E, Gustafsson E, Jonsson KF, Olofsson JK, Osther K, Ceberg C, Redebrandt HN. Combined anti-C1-INH and radiotherapy against glioblastoma. BMC Cancer 2023; 23:106. [PMID: 36717781 PMCID: PMC9887755 DOI: 10.1186/s12885-023-10583-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 01/25/2023] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND A more effective immune response against glioblastoma is needed in order to achieve better tumor control. Radiotherapy can induce anti-tumor mediated immune reactions, in addition to its dose response effects. The complement system can function as a bridge between innate and adaptive immune responses. Combining radiotherapy and complement activating therapy is theoretically interesting. METHODS Radiotherapy at 8 Gy × 2 was combined with treatment against C1-inhibitor (C1-INH), a potent inhibitor of activation of the classical pathway of the complement system. Anti-C1-INH was delivered as intratumoral injections. Fully immunocompetent Fischer 344 rats with NS1 glioblastoma tumors were treated. Survival was monitored as primary outcome. Models with either intracranial or subcutaneous tumors were evaluated separately. RESULTS In the intracranial setting, irradiation could prolong survival, but there was no additional survival gain as a result of anti-C1-INH treatment. In animals with subcutaneous tumors, combined radio-immunotherapy with anti-C1-INH and irradiation at 8 Gy × 2 significantly prolonged survival compared to control animals, whereas irradiation or anti-C1-INH treatment as single therapies did not lead to significantly increased survival compared to control animals. CONCLUSIONS Anti-C1-INH treatment could improve the efficacy of irradiation delivered at sub-therapeutic doses and delay tumor growth in the subcutaneous tumor microenvironment. In the intracranial setting, the doses of anti-C1-INH were not enough to achieve any survival effect in the present setting.
Collapse
Affiliation(s)
- Emma Liljedahl
- grid.4514.40000 0001 0930 2361The Rausing Laboratory, Division of Neurosurgery, Department of Clinical Sciences, Lund University, BMC D10, 221 84 Lund, Sweden
| | - Elise Konradsson
- grid.4514.40000 0001 0930 2361Medical Radiation Physics, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Emma Gustafsson
- grid.4514.40000 0001 0930 2361The Rausing Laboratory, Division of Neurosurgery, Department of Clinical Sciences, Lund University, BMC D10, 221 84 Lund, Sweden
| | - Karolina Förnvik Jonsson
- grid.4514.40000 0001 0930 2361The Rausing Laboratory, Division of Neurosurgery, Department of Clinical Sciences, Lund University, BMC D10, 221 84 Lund, Sweden
| | - Jill K. Olofsson
- grid.5254.60000 0001 0674 042XDepartment for Geosciences and Natural Resource Management, University of Copenhagen, Copenhagen, Denmark
| | - Kurt Osther
- grid.4514.40000 0001 0930 2361The Rausing Laboratory, Division of Neurosurgery, Department of Clinical Sciences, Lund University, BMC D10, 221 84 Lund, Sweden
| | - Crister Ceberg
- grid.4514.40000 0001 0930 2361Medical Radiation Physics, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Henrietta Nittby Redebrandt
- grid.4514.40000 0001 0930 2361The Rausing Laboratory, Division of Neurosurgery, Department of Clinical Sciences, Lund University, BMC D10, 221 84 Lund, Sweden ,grid.411843.b0000 0004 0623 9987Department of Neurosurgery, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
7
|
Narvekar A, Pardeshi A, Jain R, Dandekar P. ADCC enhancement: A conundrum or a boon to mAb therapy? Biologicals 2022; 79:10-18. [PMID: 36085129 DOI: 10.1016/j.biologicals.2022.08.006] [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: 09/15/2021] [Revised: 06/27/2022] [Accepted: 08/28/2022] [Indexed: 11/25/2022] Open
Abstract
The ability of antibodies to distinctly identify the antigens is an important feature exploited by the scientific community for the treatment of various diseases. The therapeutic action of monoclonal antibodies (mAbs) is mediated along with the cells of the immune system, such as natural killer cells, T cells and macrophages. The two major mechanisms that govern the therapeutic efficacy of mAbs are the antibody dependent cell mediated cytotoxicity (ADCC) and the complement dependent cytotoxicity (CDC). Consequently, much of the research dedicated to improving their action is focussed on enhancing either of these mechanisms. This manuscript focuses on the strategies to enhance ADCC, for providing more efficacious mAb therapeutics. These approaches essentially bring about changes in the elements of ADCC mechanism, such as the effector cell or the antibody itself and thus favour an enhanced therapeutic response. Several technologies of ADCC enhancement have been developed, based on the success of various strategies advanced by the researchers. These technologies show success with a few antibody therapeutics while they do not work with others. This review presents a detailed overview on these strategies and presents perspectives regarding the same.
Collapse
Affiliation(s)
- Aditya Narvekar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Matunga, Mumbai, 400019, India
| | - Apurva Pardeshi
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Matunga, Mumbai, 400019, India
| | - Ratnesh Jain
- Department of Chemical Engineering, Institute of Chemical Technology, Matunga, Mumbai, 400019, India.
| | - Prajakta Dandekar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Matunga, Mumbai, 400019, India.
| |
Collapse
|
8
|
Innate Immunity: A Balance between Disease and Adaption to Stress. Biomolecules 2022; 12:biom12050737. [PMID: 35625664 PMCID: PMC9138980 DOI: 10.3390/biom12050737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/17/2022] [Accepted: 05/20/2022] [Indexed: 12/01/2022] Open
Abstract
Since first being documented in ancient times, the relation of inflammation with injury and disease has evolved in complexity and causality. Early observations supported a cause (injury) and effect (inflammation) relationship, but the number of pathologies linked to chronic inflammation suggests that inflammation itself acts as a potent promoter of injury and disease. Additionally, results from studies over the last 25 years point to chronic inflammation and innate immune signaling as a critical link between stress (exogenous and endogenous) and adaptation. This brief review looks to highlight the role of the innate immune response in disease pathology, and recent findings indicating the innate immune response to chronic stresses as an influence in driving adaptation.
Collapse
|
9
|
Zhuang K, Zhang Y, Mo P, Deng L, Jiang Y, Yu L, Mei F, Huang S, Chen X, Yan Y, Tang H, Li X, Xiong Y, Wu S, Ke H, Gui X, Lan K. Plasma proteomic analysis reveals altered protein abundances in HIV-infected patients with or without Non-Hodgkin Lymphoma. J Med Virol 2022; 94:3876-3889. [PMID: 35415847 DOI: 10.1002/jmv.27775] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/08/2022] [Accepted: 04/10/2022] [Indexed: 11/07/2022]
Abstract
The identification of circulating proteins associated with acquired immunodeficiency syndrome-related non-Hodgkin lymphoma (AIDS-NHL) may help in the development of promising biomarkers for screening, diagnosis, treatment and prognosis. Here, we used quantitative liquid chromatography-tandem mass spectrometry (LC-MS/MS) to identify differentially expressed proteins (DEPs) in plasma collected from patients with AIDS-NHL and human immunodeficiency virus (HIV)-infected patients without NHL (HIV+ ). Proteins with a log2 (fold change) in abundance >0.26 and p value less than 0.05 (p < 0.05) were considered differentially abundant. In total, 84 DEPs were identified, among which 20 were further validated as potential biomarkers, with immunoglobulin and complement components being the most common proteins. Some of the proteins were further verified in a retrospective analysis of the medical records of patients in a larger cohort. These markedly altered proteins were found to mediate pathophysiological pathways that likely contribute to AIDS-NHL pathogenesis, such as the humoral immune response, complement activation, and complement and coagulation cascades. Our findings provide a new molecular understanding of AIDS-NHL pathogenesis and provide new evidence supporting the identification of these proteins as possible biomarkers in AIDS-NHL. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Ke Zhuang
- ABSL-III Laboratory at the Center for Animal Experiment, State Key Laboratory of Virology, Wuhan University, Wuhan, Hubei, China
| | - Yongxi Zhang
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Pingzheng Mo
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Liping Deng
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yong Jiang
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, Hubei, P. R. China
| | - Lei Yu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, Hubei, P. R. China
| | - Fanghua Mei
- Hubei Provincial Center for Disease Control and Prevention, Wuhan, Hubei, China
| | - Shaoxin Huang
- SpecAlly Life Technology Co., Ltd., Wuhan, Hubei, China
| | - Xi Chen
- SpecAlly Life Technology Co., Ltd., Wuhan, Hubei, China
| | - Yajun Yan
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Hongbin Tang
- ABSL-III Laboratory at the Center for Animal Experiment, State Key Laboratory of Virology, Wuhan University, Wuhan, Hubei, China
| | - Xiangdong Li
- ABSL-III Laboratory at the Center for Animal Experiment, State Key Laboratory of Virology, Wuhan University, Wuhan, Hubei, China
| | - Yong Xiong
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Shuwen Wu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, Hubei, P. R. China
| | - Hengning Ke
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xien Gui
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ke Lan
- ABSL-III Laboratory at the Center for Animal Experiment, State Key Laboratory of Virology, Wuhan University, Wuhan, Hubei, China.,State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, Hubei, P. R. China
| |
Collapse
|
10
|
Cedzyński M, Świerzko AS. Components of the Lectin Pathway of Complement in Solid Tumour Cancers. Cancers (Basel) 2022; 14:cancers14061543. [PMID: 35326694 PMCID: PMC8946279 DOI: 10.3390/cancers14061543] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 03/14/2022] [Indexed: 02/04/2023] Open
Abstract
The complement system is an important branch of the humoral innate immune response that can be activated via three distinct pathways (classical, alternative, lectin), contributing to keeping/restoring homeostasis. It can also interact with cellular innate immunity and with components of acquired immunity. Cross-talk between the complement system and other enzyme-dependent cascades makes it a more influential defence system, but on the other hand, over- or chronic activation can be harmful. This short review is focused on the dual role of the lectin pathway of complement activation in human solid tumour cancers, including those of the female reproductive system, lung, and alimentary tract, with emphasis on the aforementioned cross-talk.
Collapse
|
11
|
Abstract
Tumorigenesis has long been linked to the evasion of the immune system and the uncontrolled proliferation of transformed cells. The complement system, a major arm of innate immunity, is a key factor in the progression of cancer because many of its components have critical regulatory roles in the tumor microenvironment. For example, complement anaphylatoxins directly and indirectly inhibit antitumor T-cell responses in primary and metastatic sites, enhance proliferation of tumor cells, and promote metastasis and tumor angiogenesis. Many recent studies have provided evidence that cancer is able to hijack the immunoregulatory components of the complement system which fundamentally are tasked with protecting the body against abnormal cells and pathogens. Indeed, recent evidence shows that many types of cancer use C1q receptors (C1qRs) to promote tumor growth and progression. More importantly, most cancer cells express both C1q and its major receptors (gC1qR and cC1qR) on their surface which are essential for cell proliferation and survival. In this review, we discuss the ability of cancer to control and manipulate the complement system in the tumor microenvironment and identify possible therapeutic targets, including C1q and gC1qR.
Collapse
Affiliation(s)
- Danyaal Ain
- The Department of Medicine, Stony Brook University, 100 Nicholls Road, Stony Brook, NY 11794-8161, USA
| | - Talha Shaikh
- The Department of Medicine, Stony Brook University, 100 Nicholls Road, Stony Brook, NY 11794-8161, USA
| | - Samantha Manimala
- The Department of Medicine, Stony Brook University, 100 Nicholls Road, Stony Brook, NY 11794-8161, USA
| | - Berhane Ghebrehiwet
- The Department of Medicine, Stony Brook University, 100 Nicholls Road, Stony Brook, NY 11794-8161, USA
| |
Collapse
|
12
|
Lee MJ, Na K, Shin H, Kim CY, Cho JY, Kang CM, Kim SH, Kim H, Choi HJ, Lee CK, Bae S, Son S, Paik YK. Early Diagnostic Ability of Human Complement Factor B in Pancreatic Cancer Is Partly Linked to Its Potential Tumor-Promoting Role. J Proteome Res 2021; 20:5315-5328. [PMID: 34766501 DOI: 10.1021/acs.jproteome.1c00805] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Although plasma complement factor B (CFB, NX_P00751), both alone and in combination with CA19-9 (i.e., the ComB-CAN), previously exhibited a reliable diagnostic ability for pancreatic cancer (PC), its detectability of the early stages and the cancer detection mechanism remained elusive. We first evaluated the diagnostic accuracy of ComB-CAN using plasma samples from healthy donors (HDs), patients with chronic pancreatitis (CP), and patients with different PC stages (I/II vs III/IV). An analysis of the area under the curve (AUC) by PanelComposer using logistic regression revealed that ComB-CAN has a superior diagnostic ability for early-stage PC (97.1.% [95% confidence interval (CI): (97.1-97.2)]) compared with CFB (94.3% [95% CI: 94.2-94.4]) or CA19-9 alone (34.3% [95% CI: 34.1-34.4]). In the comparisons of all stages of patients with PC vs CP and HDs, the AUC values of ComB-CAN, CFB, and CA19-9 were 0.983 (95% CI: 0.983-0.983), 0.950 (95% CI: 0.950-0.951), and 0.873 (95% CI: 0.873-0.874), respectively. We then investigated the molecular mechanism underlying the detection of early-stage PC by using stable cell lines of CFB knockdown and CFB overexpression. A global transcriptomic analysis coupled to cell invasion assays of both CFB-modulated cell lines suggested that CFB plays a tumor-promoting role in PC, which likely initiates the PI3K-AKT cancer signaling pathway. Thus our study establishes ComB-CAN as a reliable early diagnostic marker for PC that can be clinically applied for early PC screening in the general public.
Collapse
Affiliation(s)
- Min Jung Lee
- Yonsei Proteome Research Center, Yonsei University, 50 Yonsei-ro, Seodaemoon-ku, Seoul 03722, South Korea
| | - Keun Na
- Yonsei Proteome Research Center, Yonsei University, 50 Yonsei-ro, Seodaemoon-ku, Seoul 03722, South Korea
| | - Heon Shin
- Yonsei Proteome Research Center, Yonsei University, 50 Yonsei-ro, Seodaemoon-ku, Seoul 03722, South Korea
| | - Chae-Yeon Kim
- Yonsei Proteome Research Center, Yonsei University, 50 Yonsei-ro, Seodaemoon-ku, Seoul 03722, South Korea
| | - Jin-Young Cho
- Yonsei Proteome Research Center, Yonsei University, 50 Yonsei-ro, Seodaemoon-ku, Seoul 03722, South Korea
| | | | | | | | | | | | - Sumi Bae
- JW Bioscience Corp., 2477, Nambusunhwan-ro, Seocho-gu, Seoul 06725, South Korea
| | - Sunghwa Son
- JW Holdings Corp., 2477, Nambusunhwan-ro, Seocho-gu, Seoul 06725, South Korea
| | - Young-Ki Paik
- Yonsei Proteome Research Center, Yonsei University, 50 Yonsei-ro, Seodaemoon-ku, Seoul 03722, South Korea
| |
Collapse
|
13
|
Xiang M, Zhang H, Kou L, Chen J, Xu Z, He J. Low level of complement factor H increases the risk of cancer-related death in patients with small-cell lung cancer. Postgrad Med J 2021; 98:919-924. [PMID: 34725230 DOI: 10.1136/postgradmedj-2021-141186] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/08/2021] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Pulmonary cancer is a kind of deeply invasive tumour which is difficult to treat, and its mortality rate is high. Previous research has shown that activation of complement could contribute to the progression of non-small-cell lung cancer (SCLC). However, little research has been done on SCLC. METHODS Complement factor H (CFH), complements C3 as well as C4 were measured in patients, and the prognostic impact of different parameters was assessed by log-rank function analysis and Cox multifactor models. Besides, we constructed a predictive model based on complement fractions and validated the accuracy of the model. RESULTS Among these 242 patients, 200 (82.6%) died. The median survival time was 18.3 months. We found by multifactorial analysis that high levels of CFH decreased the risk of death (HR 0.23, 95% CI 0.10 to 0.57, p<0.001), while elevated complement C4 displayed poor prognosis (HR 2.28, 95% CI 1.66 to 3.13, p<0.001). We screened variables by Cox models and constructed CFH-based prediction models to plot a nomogram by internal validation. The nomogram showed excellent accuracy in assessing the probability of death, yielding an adjusted C-statistics of 0.905. CONCLUSIONS CFH can be recognised as a biomarker to predict the risk of death in SCLC. The prediction model established based on CFH, C3 and C4 levels has good accuracy in patients' prognostic assessment.
Collapse
Affiliation(s)
- Mengqi Xiang
- Department of Medical Oncology, Sichuan Cancer Hospital and Institute, Chengdu, Sichuan, China
| | - Huachuan Zhang
- Department of Thoracic Surgery, Sichuan Cancer Hospital and Institute, Chengdu, Sichuan, China
| | - Lingna Kou
- Department of Medical Oncology, Sichuan Cancer Hospital and Institute, Chengdu, Sichuan, China
| | - Jing Chen
- Department of Medical Oncology, Sichuan Cancer Hospital and Institute, Chengdu, Sichuan, China
| | - Zhihua Xu
- General Surgery, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jintao He
- Department of Thoracic Surgery, Sichuan Cancer Hospital and Institute, Chengdu, Sichuan, China
| |
Collapse
|
14
|
C1r Upregulates Production of Matrix Metalloproteinase-13 and Promotes Invasion of Cutaneous Squamous Cell Carcinoma. J Invest Dermatol 2021; 142:1478-1488.e9. [PMID: 34756877 DOI: 10.1016/j.jid.2021.10.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 10/01/2021] [Accepted: 10/04/2021] [Indexed: 12/14/2022]
Abstract
Cutaneous squamous cell carcinoma (cSCC) is the most common metastatic skin cancer with increasing incidence worldwide. Previous studies have demonstrated the role of complement system in cSCC progression. In this study we have investigated the mechanistic role of serine protease C1r, a component of the classical pathway of complement system, in cSCC. Knockout of C1r in cSCC cells using CRISPR/Cas9 resulted in significant decrease in their proliferation, migration, and invasion through collagen type I compared to wild type cSCC cells. Knockout of C1r suppressed growth and vascularization of cSCC xenograft tumors, and promoted apoptosis of tumor cells in vivo. mRNA-seq analysis after C1r knockdown revealed significantly regulated GO terms Cell-matrix adhesion, Extracellular matrix component, Basement membrane, Metalloendopeptidase activity and KEGG pathway Extracellular matrix-receptor interaction. Among the significantly regulated genes were invasion-associated matrix metalloproteinases MMP1, MMP13, MMP10, and MMP12. Knockout of C1r resulted in decreased production of MMP-1, MMP-13, MMP-10, and MMP-12 by cSCC cells in culture. Knockout of C1r inhibited expression of MMP-13 by tumor cells, suppressed invasion, and reduced the amount of degraded collagen in vivo in xenografts. These results provide evidence for the role of C1r in promoting the invasion of cSCC cells by increasing MMP production.
Collapse
|
15
|
Yang H, Zheng H, Pan Y, Zhang W, Yang M, Du H, Yu A, Li P, Chen X, Xie W, Ren K, Zhao Y, Wang T, He X, Zhou Z. Quantitative proteomic analysis of the effects of dietary deprivation of methionine and cystine on A549 xenograft and A549 xenograft-bearing mouse. Proteomics 2021; 21:e2100007. [PMID: 34482643 DOI: 10.1002/pmic.202100007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 08/18/2021] [Accepted: 08/20/2021] [Indexed: 11/11/2022]
Abstract
Methionine (Met) and cystine (CySS) are key sulfur donors in cell metabolism and are important nutrients for sustaining tumor growth; however, the molecular effects associated with their deprivation remain to be characterized. Here, we applied a xenograft mouse model to assess the impact of their deprivation on A549 xenografts and the xenograft-bearing animal. Results show that Met and CySS deprivation inhibits A549 growth in vitro, not in vivo. Deprivation was detrimental to the xenograft-bearing mouse, as demonstrated by weight loss and renal dysfunction. Differentially expressed proteins in A549 xenograft and mouse kidneys were characterized using quantitative proteomics. Functional annotation and protein-protein interaction network analysis revealed the enriched signaling pathways, including focal adhesion (Fn1) in the A549 xenograft, and xenobiotic metabolism (Cyp2e1) and glutathione metabolism (Ggt1) in the mouse kidney. Met and CySS deprivation inhibits the migratory and invasive properties of cancer cells, as evidenced by reduced expression of the epithelial to mesenchymal transition marker N-cadherin in A549 cells in vitro. Moreover, IGFBP1 protein expression was inhibited in both A549 xenograft and mouse kidneys. This study provides the first insights into changes within the proteome profile and biological processes upon Met and CySS deprivation in a A549 xenograft mouse model.
Collapse
Affiliation(s)
- Hao Yang
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China.,School of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Haoyang Zheng
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China.,School of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Yue Pan
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China.,School of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Weiguo Zhang
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Mengjing Yang
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Huiling Du
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China.,School of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Anan Yu
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China.,School of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Ping Li
- School of Medical Instrument, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Xiaoyan Chen
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China.,School of Medical Technology, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Wei Xie
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China.,School of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Kaiming Ren
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ying Zhao
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Tianjiao Wang
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Xiaoyan He
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China.,School of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Zhaoli Zhou
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China.,School of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai, China
| |
Collapse
|
16
|
Zhao Z, Liu N, Wang C, Cheng J, Guo M. Proteomic analysis of differentially expressed whey proteins in Saanen goat milk from different provinces in China using a data-independent acquisition technique. J Dairy Sci 2021; 104:10513-10527. [PMID: 34419278 DOI: 10.3168/jds.2020-19877] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 05/17/2021] [Indexed: 11/19/2022]
Abstract
Whey proteins of Saanen goat milk samples from 3 provinces in China (Guangdong, GD; Inner Mongolia, IM; Shaanxi, SX) were characterized and compared using data-independent acquisition quantitative proteomics technique. A total of 550 proteins were quantified in all 3 samples. There were 44, 44, and 33 differentially expressed proteins (DEP) for GD versus IM, GD versus SX, and IM versus SX, respectively. Gene ontology annotation analysis showed that the largest number of DEP for the 3 comparisons were as follows: for biological processes: response to progesterone, glyceraldehyde-3-phosphate metabolic process, and negative regulation of megakaryocyte differentiation; for molecular functions: antioxidant activity, binding, and peroxiredoxin activity; and for cellular components: the same category of extracellular regions for the 3 comparisons, respectively. Pathways for the DEP of 3 comparisons were (1) disease; (2) synthesis and metabolism; and (3) synthesis, degradation, and metabolism. Protein-protein interaction network analysis showed that DEP for GD versus SX had the most interactions.
Collapse
Affiliation(s)
- Zixuan Zhao
- Key Laboratory of Dairy Science, Northeast Agricultural University, Harbin, Heilongjiang, 150030, China
| | - Ning Liu
- Key Laboratory of Dairy Science, Northeast Agricultural University, Harbin, Heilongjiang, 150030, China
| | - Cuina Wang
- Key Laboratory of Dairy Science, Northeast Agricultural University, Harbin, Heilongjiang, 150030, China; Department of Food Science, Jilin University, Changchun, 130062, China.
| | - Jianjun Cheng
- Key Laboratory of Dairy Science, Northeast Agricultural University, Harbin, Heilongjiang, 150030, China.
| | - Mingruo Guo
- Department of Nutrition and Food Sciences, College of Agriculture and Life Sciences, University of Vermont, Burlington 05405
| |
Collapse
|
17
|
Zaitsev VG, Zheltova AA, Martynova SA, Tibirkova EV. Can conventional clinical chemistry tests help doctors in the monitoring of oncology patients? RUSSIAN OPEN MEDICAL JOURNAL 2021. [DOI: 10.15275/rusomj.2021.0103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The use of laboratory assays in the diagnostic care of oncology patients can markedly increase the efficacy of cancer treatments. Many cancer-specific biomarker assays have been developed. However, the use of these has some limitations due to their cost. Moreover, not every diagnostic laboratory can perform a complete set of these assays. On the other hand, the smart use of conventional clinical chemistry tests could improve the management of cancer. They could be especially valuable tools in the long-term care of patients with a verified diagnosis. In this review, we discuss the utilization of the conventional clinical chemistry assays for the diagnosis, monitoring and prognosis of various oncological diseases. The use of conventional blood tests to assess the levels of chemical elements, metabolites and proteins (including enzymatic activity measurements) in the care of oncology patients is discussed. We have shown that some clinical chemistry assays could be used in the management of distinct kinds of cancer.
Collapse
|
18
|
Anti-Cancer Effects of Carnosine-A Dipeptide Molecule. Molecules 2021; 26:molecules26061644. [PMID: 33809496 PMCID: PMC8002160 DOI: 10.3390/molecules26061644] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 03/04/2021] [Accepted: 03/10/2021] [Indexed: 12/25/2022] Open
Abstract
Background: Carnosine is a dipeptide molecule (β-alanyl-l-histidine) with anti-inflammatory, antioxidant, anti-glycation, and chelating properties. It is used in exercise physiology as a food supplement to increase performance; however, in vitro evidence suggests that carnosine may exhibit anti-cancer properties. Methods: In this study, we investigated the effect of carnosine on breast, ovarian, colon, and leukemic cancer cell proliferation. We further examined U937 promonocytic, human myeloid leukemia cell phenotype, gene expression, and cytokine secretion to determine if these are linked to carnosine’s anti-proliferative properties. Results: Carnosine (1) inhibits breast, ovarian, colon, and leukemic cancer cell proliferation; (2) upregulates expression of pro-inflammatory molecules; (3) modulates cytokine secretion; and (4) alters U937 differentiation and phenotype. Conclusion: These effects may have implications for a role for carnosine in anti-cancer therapy.
Collapse
|
19
|
O’Brien RM, Cannon A, Reynolds JV, Lysaght J, Lynam-Lennon N. Complement in Tumourigenesis and the Response to Cancer Therapy. Cancers (Basel) 2021; 13:1209. [PMID: 33802004 PMCID: PMC7998562 DOI: 10.3390/cancers13061209] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/04/2021] [Accepted: 03/05/2021] [Indexed: 12/16/2022] Open
Abstract
In recent years, our knowledge of the complement system beyond innate immunity has progressed significantly. A modern understanding is that the complement system has a multifaceted role in malignancy, impacting carcinogenesis, the acquisition of a metastatic phenotype and response to therapies. The ability of local immune cells to produce and respond to complement components has provided valuable insights into their regulation, and the subsequent remodeling of the tumour microenvironment. These novel discoveries have advanced our understanding of the immunosuppressive mechanisms supporting tumour growth and uncovered potential therapeutic targets. This review discusses the current understanding of complement in cancer, outlining both direct and immune cell-mediated roles. The role of complement in response to therapies such as chemotherapy, radiation and immunotherapy is also presented. While complement activities are largely context and cancer type-dependent, it is evident that promising therapeutic avenues have been identified, in particular in combination therapies.
Collapse
Affiliation(s)
- Rebecca M. O’Brien
- Department of Surgery, Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Trinity College Dublin and St. James’s Hospital, Dublin 8, Ireland; (R.M.O.); (A.C.); (J.V.R.); (J.L.)
- Cancer Immunology and Immunotherapy Group, Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Trinity College Dublin and St. James’s Hospital, Dublin 8, Ireland
| | - Aoife Cannon
- Department of Surgery, Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Trinity College Dublin and St. James’s Hospital, Dublin 8, Ireland; (R.M.O.); (A.C.); (J.V.R.); (J.L.)
| | - John V. Reynolds
- Department of Surgery, Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Trinity College Dublin and St. James’s Hospital, Dublin 8, Ireland; (R.M.O.); (A.C.); (J.V.R.); (J.L.)
| | - Joanne Lysaght
- Department of Surgery, Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Trinity College Dublin and St. James’s Hospital, Dublin 8, Ireland; (R.M.O.); (A.C.); (J.V.R.); (J.L.)
- Cancer Immunology and Immunotherapy Group, Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Trinity College Dublin and St. James’s Hospital, Dublin 8, Ireland
| | - Niamh Lynam-Lennon
- Department of Surgery, Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Trinity College Dublin and St. James’s Hospital, Dublin 8, Ireland; (R.M.O.); (A.C.); (J.V.R.); (J.L.)
| |
Collapse
|
20
|
Popeda M, Markiewicz A, Stokowy T, Szade J, Niemira M, Kretowski A, Bednarz-Knoll N, Zaczek AJ. Reduced expression of innate immunity-related genes in lymph node metastases of luminal breast cancer patients. Sci Rep 2021; 11:5097. [PMID: 33658651 PMCID: PMC7930267 DOI: 10.1038/s41598-021-84568-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 02/18/2021] [Indexed: 01/31/2023] Open
Abstract
Immune system plays a dual role in cancer by either targeting or supporting neoplastic cells at various stages of disease, including metastasis. Yet, the exact immune-related transcriptome profiles of primary tumours (PT) and lymph node metastases (LNM) and their evolution during luminal breast cancer (BCa) dissemination remain undiscovered. In order to identify the immune-related transcriptome changes that accompany lymphatic spread, we analysed PT-LNM pairs of luminal BCa using NanoString technology. Decrease in complement C3-one of the top-downregulated genes, in LNM was validated at the protein level using immunohistochemistry. Thirty-three of 360 analysed genes were downregulated (9%), whereas only 3 (0.8%) upregulated in LNM when compared to the corresponding PT. In LNM, reduced expression was observed in genes related to innate immunity, particularly to the complement system (C1QB, C1S, C1R, C4B, CFB, C3, SERPING1 and C3AR1). In validation cohort, complement C3 protein was less frequently expressed in LNM than in PT and it was associated with worse prognosis. To conclude, local expression of the complement system components declines during lymphatic spread of non-metastatic luminal BCa, whilst further reduction of tumoral complement C3 in LNM is indicative for poor survival. This points to context-dependent role of complement C3 in BCa dissemination.
Collapse
Affiliation(s)
- Marta Popeda
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, Medical University of Gdansk, 80-211, Gdansk, Poland
| | - Aleksandra Markiewicz
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, Medical University of Gdansk, 80-211, Gdansk, Poland
| | - Tomasz Stokowy
- Department of Clinical Science, University of Bergen, 5021, Bergen, Norway
| | - Jolanta Szade
- Department of Pathomorphology, Medical University of Gdansk, 80-211, Gdansk, Poland
| | - Magdalena Niemira
- Clinical Research Centre, Medical University of Bialystok, 15-276, Bialystok, Poland
| | - Adam Kretowski
- Clinical Research Centre, Medical University of Bialystok, 15-276, Bialystok, Poland
| | - Natalia Bednarz-Knoll
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, Medical University of Gdansk, 80-211, Gdansk, Poland
| | - Anna J Zaczek
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, Medical University of Gdansk, 80-211, Gdansk, Poland.
| |
Collapse
|
21
|
de Boer ECW, van Mourik AG, Jongerius I. Therapeutic Lessons to be Learned From the Role of Complement Regulators as Double-Edged Sword in Health and Disease. Front Immunol 2020; 11:578069. [PMID: 33362763 PMCID: PMC7758290 DOI: 10.3389/fimmu.2020.578069] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/12/2020] [Indexed: 12/22/2022] Open
Abstract
The complement system is an important part of the innate immune system, providing a strong defense against pathogens and removing apoptotic cells and immune complexes. Due to its strength, it is important that healthy human cells are protected against damage induced by the complement system. To be protected from complement, each cell type relies on a specific combination of both soluble and membrane-bound regulators. Their importance is indicated by the amount of pathologies associated with abnormalities in these complement regulators. Here, we will discuss the current knowledge on complement regulatory protein polymorphisms and expression levels together with their link to disease. These diseases often result in red blood cell destruction or occur in the eye, kidney or brain, which are tissues known for aberrant complement activity or regulation. In addition, complement regulators have also been associated with different types of cancer, although their mechanisms here have not been elucidated yet. In most of these pathologies, treatments are limited and do not prevent the complement system from attacking host cells, but rather fight the consequences of the complement-mediated damage, using for example blood transfusions in anemic patients. Currently only few drugs targeting the complement system are used in the clinic. With further demand for therapeutics rising linked to the wide range of complement-mediated disease we should broaden our horizon towards treatments that can actually protect the host cells against complement. Here, we will discuss the latest insights on how complement regulators can benefit therapeutics. Such therapeutics are currently being developed extensively, and can be categorized into full-length complement regulators, engineered complement system regulators and antibodies targeting complement regulators. In conclusion, this review provides an overview of the complement regulatory proteins and their links to disease, together with their potential in the development of novel therapeutics.
Collapse
Affiliation(s)
- Esther C W de Boer
- Sanquin Research, Department of Immunopathology, and Landsteiner Laboratory, Amsterdam University Medical Centre, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands.,Department of Pediatric Immunology, Rheumatology, and Infectious Diseases, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam, Netherlands
| | - Anouk G van Mourik
- Sanquin Research, Department of Immunopathology, and Landsteiner Laboratory, Amsterdam University Medical Centre, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Ilse Jongerius
- Sanquin Research, Department of Immunopathology, and Landsteiner Laboratory, Amsterdam University Medical Centre, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands.,Department of Pediatric Immunology, Rheumatology, and Infectious Diseases, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam, Netherlands
| |
Collapse
|
22
|
Shi C, Han W, Zhang M, Zang R, Du K, Li L, Xu X, Li C, Wang S, Qiu P, Guan H, Yang J, Xiao S, Wang X. Sulfated polymannuroguluronate TGC161 ameliorates leukopenia by inhibiting CD4 + T cell apoptosis. Carbohydr Polym 2020; 247:116728. [PMID: 32829850 PMCID: PMC7336955 DOI: 10.1016/j.carbpol.2020.116728] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/23/2020] [Accepted: 07/02/2020] [Indexed: 02/07/2023]
Abstract
Polysaccharides have aroused considerable interest due to their diverse biological activities and low toxicity. In this study, we evaluated the effect of marine polysaccharide sulfated polymannuroguluronate (TGC161) on the leukopenia induced by chemotherapy. It is found that TGC161 ameliorates the leukopenia. Besides, TGC161 would promote CD4+ T cell differentiation and maturation in the thymus, but does not have a significant effect on precursor cells in bone marrow. Furthermore, TGC161 inhibits CD4+ T cell apoptosis in vitro. Collectively, our findings offer a natural and harmless polysaccharide to ameliorate leukopenia.
Collapse
Affiliation(s)
- Chuanqin Shi
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China
| | - Wenwei Han
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China
| | - Meifang Zhang
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China
| | - Ruochen Zang
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China
| | - Kaixin Du
- Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China; Marine Biomedical Research Institute of Qingdao, Qingdao, 266071, China
| | - Li Li
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China; Marine Biomedical Research Institute of Qingdao, Qingdao, 266071, China
| | - Ximing Xu
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China; Marine Biomedical Research Institute of Qingdao, Qingdao, 266071, China
| | - Chunxia Li
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Marine Biomedical Research Institute of Qingdao, Qingdao, 266071, China
| | - Shixin Wang
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Marine Biomedical Research Institute of Qingdao, Qingdao, 266071, China
| | - Peiju Qiu
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China; Marine Biomedical Research Institute of Qingdao, Qingdao, 266071, China
| | - Huashi Guan
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China; Marine Biomedical Research Institute of Qingdao, Qingdao, 266071, China
| | - Jinbo Yang
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China; Marine Biomedical Research Institute of Qingdao, Qingdao, 266071, China
| | - Shuai Xiao
- Department of Gastrointestinal Surgery and Institute of Clinical Medicine, the First Affiliated Hospital, University of South China, Hengyang, 421001, China.
| | - Xin Wang
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China; Marine Biomedical Research Institute of Qingdao, Qingdao, 266071, China.
| |
Collapse
|
23
|
Anti gC1qR/p32/HABP1 Antibody Therapy Decreases Tumor Growth in an Orthotopic Murine Xenotransplant Model of Triple Negative Breast Cancer. Antibodies (Basel) 2020; 9:antib9040051. [PMID: 33036212 PMCID: PMC7709104 DOI: 10.3390/antib9040051] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 08/18/2020] [Accepted: 09/08/2020] [Indexed: 01/09/2023] Open
Abstract
gC1qR is highly expressed in breast cancer and plays a role in cancer cell proliferation. This study explored therapy with gC1qR monoclonal antibody 60.11, directed against the C1q binding domain of gC1qR, in a murine orthotopic xenotransplant model of triple negative breast cancer. MDA231 breast cancer cells were injected into the mammary fat pad of athymic nu/nu female mice. Mice were segregated into three groups (n = 5, each) and treated with the vehicle (group 1) or gC1qR antibody 60.11 (100 mg/kg) twice weekly, starting at day 3 post-implantation (group 2) or when the tumor volume reached 100 mm3 (group 3). At study termination (d = 35), the average tumor volume in the control group measured 895 ± 143 mm3, compared to 401 ± 48 mm3 and 701 ± 100 mm3 in groups 2 and 3, respectively (p < 0.05). Immunohistochemical staining of excised tumors revealed increased apoptosis (caspase 3 and TUNEL staining) in 60.11-treated mice compared to controls, and decreased angiogenesis (CD31 staining). Slightly decreased white blood cell counts were noted in 60.11-treated mice. Otherwise, no overt toxicities were observed. These data are the first to demonstrate an in vivo anti-tumor effect of 60.11 therapy in a mouse model of triple negative breast cancer.
Collapse
|
24
|
Peerschke E, Stier K, Li X, Kandov E, de Stanchina E, Chang Q, Xiong Y, Manova-Todorova K, Fan N, Barlas A, Ghebrehiwet B, Adusumilli PS. gC1qR/HABP1/p32 Is a Potential New Therapeutic Target Against Mesothelioma. Front Oncol 2020; 10:1413. [PMID: 32903438 PMCID: PMC7435067 DOI: 10.3389/fonc.2020.01413] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 07/06/2020] [Indexed: 02/05/2023] Open
Abstract
Mesothelioma is an aggressive cancer of the serous membranes with poor prognosis despite combination therapy consisting of surgery, radiotherapy, and platinum-based chemotherapy. Targeted therapies, including immunotherapies, have reported limited success, suggesting the need for additional therapeutic targets. This study investigates a potential new therapeutic target, gC1qR/HABP1/p32 (gC1qR), which is overexpressed in all morphologic subtypes of mesothelioma. gC1qR is a complement receptor that is associated with several cellular functions, including cell proliferation and angiogenesis. In vitro and in vivo experiments were conducted to test the hypothesis that targeting gC1qR with a specific gC1qR monoclonal antibody 60.11 reduces mesothelioma tumor growth, using the biphasic mesothelioma cell line MSTO-211H (MSTO). In vitro studies demonstrate cell surface and extracellular gC1qR expression by MSTO cells, and a modest 25.3 ± 1.8% (n = 4) reduction in cell proliferation by the gC1qR blocking 60.11 antibody. This inhibition was specific for targeting the C1q binding domain of gC1qR at aa 76–93, as a separate monoclonal antibody 74.5.2, directed against amino acids 204–218, had no discernable effect. In vivo studies, using a murine orthotopic xenotransplant model, demonstrated an even greater reduction in MSTO tumor growth (50% inhibition) in mice treated with the 60.11 antibody compared to controls. Immunohistochemical studies of resected tumors revealed increased cellular apoptosis by caspase 3 and TUNEL staining, in 60.11 treated tumors compared to controls, as well as impaired angiogenesis by decreased CD31 staining. Taken together, these data identify gC1qR as a potential new therapeutic target against mesothelioma with both antiproliferative and antiangiogenic properties.
Collapse
Affiliation(s)
- Ellinor Peerschke
- Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Kenneth Stier
- Departments of Medicine and Pathology, Stony Brook University, Stony Brook, New York, NY, United States
| | - Xiaoyu Li
- Department of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China.,Department of Surgery, Thoracic Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Evelyn Kandov
- Departments of Medicine and Pathology, Stony Brook University, Stony Brook, New York, NY, United States
| | - Elisa de Stanchina
- Memorial Sloan Kettering Cancer Center, Sloan Kettering Institute, New York, NY, United States
| | - Qing Chang
- Memorial Sloan Kettering Cancer Center, Sloan Kettering Institute, New York, NY, United States
| | - Yuquan Xiong
- Department of Surgery, Thoracic Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Katia Manova-Todorova
- Molecular Cytology Core Facility, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Ning Fan
- Molecular Cytology Core Facility, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Afsar Barlas
- Molecular Cytology Core Facility, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Berhane Ghebrehiwet
- Departments of Medicine and Pathology, Stony Brook University, Stony Brook, New York, NY, United States
| | - Prasad S Adusumilli
- Department of Surgery, Thoracic Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
25
|
Deciphering the Intricate Roles of Radiation Therapy and Complement Activation in Cancer. Int J Radiat Oncol Biol Phys 2020; 108:46-55. [PMID: 32629082 DOI: 10.1016/j.ijrobp.2020.06.067] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 12/22/2022]
Abstract
The complement system consists of a collection of serum proteins that act as the main frontline effector arm of the innate immune system. Activation of complement can occur through 3 individual induction pathways: the classical, mannose-binding lectin, and alternative pathways. Activation results in opsonization, recruitment of effector cells through potent immune mediators known as anaphylatoxins, and cell lysis via the formation of the membrane attack complex. Stringent regulation of complement is required to protect against inappropriate activation of the complement cascade. Complement activation within the tumor microenvironment does not increase antitumoral action; instead, it enhances tumor growth and disease progression. Radiation therapy (RT) is a staple in the treatment of malignancies and controls tumor growth through direct DNA damage and the influx of immune cells, reshaping the makeup of the tumor microenvironment. The relationship between RT and complement activity in the tumor microenvironment is uncertain at best. The following review will focus on the complex interaction of complement activation and the immune-modulating effects of RT and the overall effect on tumor progression. The clinical implications of complement activation in cancer and the use of therapeutics and potential biomarkers will also be covered.
Collapse
|
26
|
Franzin R, Stasi A, Fiorentino M, Stallone G, Cantaluppi V, Gesualdo L, Castellano G. Inflammaging and Complement System: A Link Between Acute Kidney Injury and Chronic Graft Damage. Front Immunol 2020; 11:734. [PMID: 32457738 PMCID: PMC7221190 DOI: 10.3389/fimmu.2020.00734] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 03/31/2020] [Indexed: 12/13/2022] Open
Abstract
The aberrant activation of complement system in several kidney diseases suggests that this pillar of innate immunity has a critical role in the pathophysiology of renal damage of different etiologies. A growing body of experimental evidence indicates that complement activation contributes to the pathogenesis of acute kidney injury (AKI) such as delayed graft function (DGF) in transplant patients. AKI is characterized by the rapid loss of the kidney's excretory function and is a complex syndrome currently lacking a specific medical treatment to arrest or attenuate progression in chronic kidney disease (CKD). Recent evidence suggests that independently from the initial trigger (i.e., sepsis or ischemia/reperfusions injury), an episode of AKI is strongly associated with an increased risk of subsequent CKD. The AKI-to-CKD transition may involve a wide range of mechanisms including scar-forming myofibroblasts generated from different sources, microvascular rarefaction, mitochondrial dysfunction, or cell cycle arrest by the involvement of epigenetic, gene, and protein alterations leading to common final signaling pathways [i.e., transforming growth factor beta (TGF-β), p16 ink4a , Wnt/β-catenin pathway] involved in renal aging. Research in recent years has revealed that several stressors or complications such as rejection after renal transplantation can lead to accelerated renal aging with detrimental effects with the establishment of chronic proinflammatory cellular phenotypes within the kidney. Despite a greater understanding of these mechanisms, the role of complement system in the context of the AKI-to-CKD transition and renal inflammaging is still poorly explored. The purpose of this review is to summarize recent findings describing the role of complement in AKI-to-CKD transition. We will also address how and when complement inhibitors might be used to prevent AKI and CKD progression, therefore improving graft function.
Collapse
Affiliation(s)
- Rossana Franzin
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
- Department Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Alessandra Stasi
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Marco Fiorentino
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Giovanni Stallone
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Vincenzo Cantaluppi
- Department Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Loreto Gesualdo
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Giuseppe Castellano
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| |
Collapse
|
27
|
Zhang J, Chen M, Zhao Y, Xiong H, Sneh T, Fan Y, Wang J, Zhou X, Gong C. Complement and coagulation cascades pathway correlates with chemosensitivity and overall survival in patients with soft tissue sarcoma. Eur J Pharmacol 2020; 879:173121. [PMID: 32339514 DOI: 10.1016/j.ejphar.2020.173121] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/30/2020] [Accepted: 04/15/2020] [Indexed: 10/24/2022]
Abstract
Chemotherapy is an indispensable method in treatment of Soft tissue sarcomas (STS), but variability in sensitivity as a result of tumor heterogeneity is a key factor in determining patient outcome. Several studies have investigated the phenomenon of chemotherapy resistance in STS, while its precise complex mechanism is still unknown. This study aims to identify potential biomarkers for predicting the STS chemosensitivity, with the goal of both aiding patient treatment determination in the clinic and providing insight into key parts of the underlying mechanism. Gene expression profiles of 265 patients were obtained from The Cancer Genome Atlas dataset and differentially expressed genes (DEGs) associated with chemosensitivity were identified in groups of varying chemosensitivity, including 177 upregulated and 21 downregulated genes (P < 0.05). Then, DEGs were found to be enriched in complement and coagulation cascades and the osteoclast differentiation pathway. Protein-protein interaction analysis showed 15 genes (52 edges) enriched in the complement and coagulation cascades while 11 genes (28 edges) enriched in the osteoclast differentiation pathway. Notably, all the genes that significantly correlated to disease-free survival (DFS) and overall survival (OS), such as C1QC, C3AR1, C7, CFI and SERPINE1, are enriched in complement and coagulation cascades pathway. The differential expression of these genes was further verified by using the GSE database. Our findings support that C1QC, C3AR1, C7, CFI and SERPINE1 in the complement and coagulation cascade pathway are potential biomarkers for chemotherapy resistance and survival of STS patients.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Maoshan Chen
- Myeloma Research Group, Australian Centre for Blood Diseases (ACBD), Clinical Central School, Monash University, Melbourne, 3004, Australia
| | - Yuanyuan Zhao
- Department of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Huihua Xiong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Tal Sneh
- Center for Molecular Design and Biomimetics, The Biodesign Institute, Arizona State University, Tempe, AZ, 85281, USA
| | - Yang Fan
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Jianhua Wang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Xiao Zhou
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Chen Gong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China.
| |
Collapse
|
28
|
Riihilä P, Viiklepp K, Nissinen L, Farshchian M, Kallajoki M, Kivisaari A, Meri S, Peltonen J, Peltonen S, Kähäri V. Tumour-cell-derived complement components C1r and C1s promote growth of cutaneous squamous cell carcinoma. Br J Dermatol 2020; 182:658-670. [PMID: 31049937 PMCID: PMC7065064 DOI: 10.1111/bjd.18095] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND The incidence of epidermal keratinocyte-derived cutaneous squamous cell carcinoma (cSCC) is increasing worldwide. OBJECTIVES To study the role of the complement classical pathway components C1q, C1r and C1s in the progression of cSCC. METHODS The mRNA levels of C1Q subunits and C1R and C1S in cSCC cell lines, normal human epidermal keratinocytes, cSCC tumours in vivo and normal skin were analysed with quantitative real-time polymerase chain reaction. The production of C1r and C1s was determined with Western blotting. The expression of C1r and C1s in tissue samples in vivo was analysed with immunohistochemistry and further investigated in human cSCC xenografts by knocking down C1r and C1s. RESULTS Significantly elevated C1R and C1S mRNA levels and production of C1r and C1s were detected in cSCC cells, compared with normal human epidermal keratinocytes. The mRNA levels of C1R and C1S were markedly elevated in cSCC tumours in vivo compared with normal skin. Abundant expression of C1r and C1s by tumour cells was detected in invasive sporadic cSCCs and recessive dystrophic epidermolysis bullosa-associated cSCCs, whereas the expression of C1r and C1s was lower in cSCC in situ, actinic keratosis and normal skin. Knockdown of C1r and C1s expression in cSCC cells inhibited activation of extracellular signal-related kinase 1/2 and Akt, promoted apoptosis of cSCC cells and significantly suppressed growth and vascularization of human cSCC xenograft tumours in vivo. CONCLUSIONS These results provide evidence for the role of tumour-cell-derived C1r and C1s in the progression of cSCC and identify them as biomarkers and putative therapeutic targets in cSCC. What's already known about this topic? The incidences of actinic keratosis, cutaneous squamous cell carcinoma (cSCC) in situ and invasive cSCC are increasing globally. Few specific biomarkers for progression of cSCC have been identified, and no biological markers are in clinical use to predict the aggressiveness of actinic keratosis, cSCC in situ and invasive cSCC. What does this study add? Our results provide novel evidence for the role of complement classical pathway components C1r and C1s in the progression of cSCC. What is the translational message? Our results identify complement classical pathway components C1r and C1s as biomarkers and putative therapeutic targets in cSCC.
Collapse
Affiliation(s)
- P. Riihilä
- Department of DermatologyUniversity of Turku and Turku University HospitalHämeentie 11 TE6FI‐20520TurkuFinland
- MediCity Research LaboratoryUniversity of TurkuTurkuFinland
- The Western Cancer Centre of the Cancer Center Finland (FICAN West)University of Turku and Turku University HospitalHämeentie 11 TE6FI‐20520TurkuFinland
| | - K. Viiklepp
- Department of DermatologyUniversity of Turku and Turku University HospitalHämeentie 11 TE6FI‐20520TurkuFinland
- MediCity Research LaboratoryUniversity of TurkuTurkuFinland
- The Western Cancer Centre of the Cancer Center Finland (FICAN West)University of Turku and Turku University HospitalHämeentie 11 TE6FI‐20520TurkuFinland
| | - L. Nissinen
- Department of DermatologyUniversity of Turku and Turku University HospitalHämeentie 11 TE6FI‐20520TurkuFinland
- MediCity Research LaboratoryUniversity of TurkuTurkuFinland
- The Western Cancer Centre of the Cancer Center Finland (FICAN West)University of Turku and Turku University HospitalHämeentie 11 TE6FI‐20520TurkuFinland
| | - M. Farshchian
- Department of DermatologyUniversity of Turku and Turku University HospitalHämeentie 11 TE6FI‐20520TurkuFinland
- MediCity Research LaboratoryUniversity of TurkuTurkuFinland
| | - M. Kallajoki
- Department of PathologyTurku University HospitalTurkuFinland
| | - A. Kivisaari
- Department of DermatologyUniversity of Turku and Turku University HospitalHämeentie 11 TE6FI‐20520TurkuFinland
- MediCity Research LaboratoryUniversity of TurkuTurkuFinland
| | - S. Meri
- Haartman InstituteUniversity of HelsinkiHelsinkiFinland
| | - J. Peltonen
- Department of Anatomy and Cell BiologyUniversity of TurkuTurkuFinland
| | - S. Peltonen
- Department of DermatologyUniversity of Turku and Turku University HospitalHämeentie 11 TE6FI‐20520TurkuFinland
| | - V.‐M. Kähäri
- Department of DermatologyUniversity of Turku and Turku University HospitalHämeentie 11 TE6FI‐20520TurkuFinland
- MediCity Research LaboratoryUniversity of TurkuTurkuFinland
- The Western Cancer Centre of the Cancer Center Finland (FICAN West)University of Turku and Turku University HospitalHämeentie 11 TE6FI‐20520TurkuFinland
| |
Collapse
|
29
|
Moschetta MG, Leonel C, Maschio-Signorini LB, Borin TF, Gelaleti GB, Jardim-Perassi BV, Ferreira LC, Sonehara NM, Carvalho LGS, Hellmén E, de Campos Zuccari DAP. Evaluation of Angiogenesis Process after Metformin and LY294002 Treatment in Mammary Tumor. Anticancer Agents Med Chem 2020; 19:655-666. [PMID: 30569877 DOI: 10.2174/1871520619666181218164050] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 09/24/2018] [Accepted: 12/11/2018] [Indexed: 12/24/2022]
Abstract
BACKGROUND The angiogenesis process is regulated by many factors, such as Hypoxia-Inducible Factor-1 (HIF-1) and Vascular Endothelial Growth Factor (VEGF). Metformin has demonstrated its ability to inhibit cell growth and the LY294002 is the major inhibitor of PI3K/AKT/mTOR pathway that has antiangiogenic properties. METHODS Canine mammary tumor cell lines CMT-U229 and CF41 were treated with metformin and LY294002. Cell viability, protein and gene expression of VEGF and HIF-1 were determined in vitro. For the in vivo study, CF41 cells were inoculated in female athymic nude mice treated with either metformin or LY294002. The microvessel density by immunohistochemistry for CD31 as well as the gene and protein expression of HIF-1 and VEGF were evaluated. RESULTS The treatment with metformin and LY294002 was able to reduce the cellular viability after 24 hours. The protein and gene expression of HIF-1 and VEGF decreased after treatment with metformin and LY294002. In the in vivo study, there was a decrease in tumor size, protein and gene expression of HIF-1 and VEGFA, in addition to the decreasing of CD31 expression after all treatments. CONCLUSION Our results demonstrate the effectiveness of metformin and LY294002 in controlling the angiogenesis process in mammary tumors by VEGF and HIF-1, the most important angiogenic markers.
Collapse
Affiliation(s)
- Marina G Moschetta
- Faculdade de Medicina de Sao Jose do Rio Preto (FAMERP), Departament of Molecular Biology, Laboratorio de Investigacao Molecular no Cancer (LIMC), Sao Jose do Rio Preto, SP, Brazil
| | - Camila Leonel
- Faculdade de Medicina de Sao Jose do Rio Preto (FAMERP), Departament of Molecular Biology, Laboratorio de Investigacao Molecular no Cancer (LIMC), Sao Jose do Rio Preto, SP, Brazil
| | - Larissa B Maschio-Signorini
- Faculdade de Medicina de Sao Jose do Rio Preto (FAMERP), Departament of Molecular Biology, Laboratorio de Investigacao Molecular no Cancer (LIMC), Sao Jose do Rio Preto, SP, Brazil
| | - Thaiz F Borin
- Augusta University, Department of Biochemistry and Molecular Biology, Tumor Imaging Angiogenesis Laboratory, Augusta, GA, United States
| | - Gabriela B Gelaleti
- Faculdade de Medicina de Sao Jose do Rio Preto (FAMERP), Departament of Molecular Biology, Laboratorio de Investigacao Molecular no Cancer (LIMC), Sao Jose do Rio Preto, SP, Brazil
| | | | - Lívia C Ferreira
- Faculdade de Medicina de Sao Jose do Rio Preto (FAMERP), Departament of Molecular Biology, Laboratorio de Investigacao Molecular no Cancer (LIMC), Sao Jose do Rio Preto, SP, Brazil
| | - Nathália M Sonehara
- Faculdade de Medicina de Sao Jose do Rio Preto (FAMERP), Departament of Molecular Biology, Laboratorio de Investigacao Molecular no Cancer (LIMC), Sao Jose do Rio Preto, SP, Brazil
| | - Livia G S Carvalho
- Faculdade de Medicina de Sao Jose do Rio Preto (FAMERP), Departament of Molecular Biology, Laboratorio de Investigacao Molecular no Cancer (LIMC), Sao Jose do Rio Preto, SP, Brazil
| | - Eva Hellmén
- Swedish University of Agricultural Sciences (SLU), Department of Anatomy, Physiology and Biochemistry, Uppsala, Sweden
| | - Debora A P de Campos Zuccari
- Faculdade de Medicina de Sao Jose do Rio Preto (FAMERP), Departament of Molecular Biology, Laboratorio de Investigacao Molecular no Cancer (LIMC), Sao Jose do Rio Preto, SP, Brazil
| |
Collapse
|
30
|
Wan Q, Jin L, Su Y, liu Y, Li C, Wang Z. Development and validation of autophagy‐related‐gene biomarker and nomogram for predicting the survival of cutaneous melanoma. IUBMB Life 2020; 72:1364-1378. [DOI: 10.1002/iub.2258] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 02/10/2020] [Indexed: 12/18/2022]
Affiliation(s)
- Qi Wan
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic Center, Sun Yat‐Sen University Guangzhou China
| | - Lin Jin
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic Center, Sun Yat‐Sen University Guangzhou China
| | - Yaru Su
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic Center, Sun Yat‐Sen University Guangzhou China
| | - Ying liu
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic Center, Sun Yat‐Sen University Guangzhou China
| | - Chaoyang Li
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic Center, Sun Yat‐Sen University Guangzhou China
| | - Zhichong Wang
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic Center, Sun Yat‐Sen University Guangzhou China
| |
Collapse
|
31
|
Loveridge CJ, Slater S, Campbell KJ, Nam NA, Knight J, Ahmad I, Hedley A, Lilla S, Repiscak P, Patel R, Salji M, Fleming J, Mitchell L, Nixon C, Strathdee D, Neilson M, Ntala C, Bryson S, Zanivan S, Edwards J, Robson CN, Goodyear CS, Blyth K, Leung HY. BRF1 accelerates prostate tumourigenesis and perturbs immune infiltration. Oncogene 2020; 39:1797-1806. [PMID: 31740786 PMCID: PMC7033044 DOI: 10.1038/s41388-019-1106-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 10/22/2019] [Accepted: 11/06/2019] [Indexed: 01/10/2023]
Abstract
BRF1 is a rate-limiting factor for RNA Polymerase III-mediated transcription and is elevated in numerous cancers. Here, we report that elevated levels of BRF1 associate with poor prognosis in human prostate cancer. In vitro studies in human prostate cancer cell lines demonstrated that transient overexpression of BRF1 increased cell proliferation whereas the transient downregulation of BRF1 reduced proliferation and mediated cell cycle arrest. Consistent with our clinical observations, BRF1 overexpression in a Pten-deficient mouse (PtenΔ/Δ BRF1Tg) prostate cancer model accelerated prostate carcinogenesis and shortened survival. In PtenΔ/Δ BRF1Tg tumours, immune and inflammatory processes were altered, with reduced tumoral infiltration of neutrophils and CD4 positive T cells, which can be explained by decreased levels of complement factor D (CFD) and C7 components of the complement cascade, an innate immune pathway that influences the adaptive immune response. We tested if the secretome was involved in BRF1-driven tumorigenesis. Unbiased proteomic analysis on BRF1-overexpresing PC3 cells confirmed reduced levels of CFD in the secretome, implicating the complement system in prostate carcinogenesis. We further identify that expression of C7 significantly correlates with expression of CD4 and has the potential to alter clinical outcome in human prostate cancer, where low levels of C7 associate with poorer prognosis.
Collapse
Affiliation(s)
- Carolyn J Loveridge
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden, Glasgow, G61 1QH, UK
- CRUK Beatson Institute, Bearsden, Glasgow, G61 1BD, UK
| | - Sarah Slater
- CRUK Beatson Institute, Bearsden, Glasgow, G61 1BD, UK
| | - Kirsteen J Campbell
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden, Glasgow, G61 1QH, UK
- CRUK Beatson Institute, Bearsden, Glasgow, G61 1BD, UK
| | - Noor A Nam
- CRUK Beatson Institute, Bearsden, Glasgow, G61 1BD, UK
- Department of Basic Sciences and Oral Biology, Faculty of Dentistry, Universiti Sains Islam Malaysia, Kuala Lumpur, Malaysia
| | - John Knight
- CRUK Beatson Institute, Bearsden, Glasgow, G61 1BD, UK
| | - Imran Ahmad
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden, Glasgow, G61 1QH, UK
- CRUK Beatson Institute, Bearsden, Glasgow, G61 1BD, UK
| | - Ann Hedley
- CRUK Beatson Institute, Bearsden, Glasgow, G61 1BD, UK
| | - Sergio Lilla
- CRUK Beatson Institute, Bearsden, Glasgow, G61 1BD, UK
| | | | - Rachana Patel
- CRUK Beatson Institute, Bearsden, Glasgow, G61 1BD, UK
| | - Mark Salji
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden, Glasgow, G61 1QH, UK
- CRUK Beatson Institute, Bearsden, Glasgow, G61 1BD, UK
| | - Janis Fleming
- CRUK Beatson Institute, Bearsden, Glasgow, G61 1BD, UK
| | | | - Colin Nixon
- CRUK Beatson Institute, Bearsden, Glasgow, G61 1BD, UK
| | | | | | - Chara Ntala
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden, Glasgow, G61 1QH, UK
- CRUK Beatson Institute, Bearsden, Glasgow, G61 1BD, UK
| | - Sheila Bryson
- CRUK Beatson Institute, Bearsden, Glasgow, G61 1BD, UK
| | - Sara Zanivan
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden, Glasgow, G61 1QH, UK
- CRUK Beatson Institute, Bearsden, Glasgow, G61 1BD, UK
| | - Joanne Edwards
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden, Glasgow, G61 1QH, UK
| | - Craig N Robson
- Northern Institute for Cancer Research, The Medical School, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Carl S Goodyear
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - Karen Blyth
- CRUK Beatson Institute, Bearsden, Glasgow, G61 1BD, UK
| | - Hing Y Leung
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden, Glasgow, G61 1QH, UK.
- CRUK Beatson Institute, Bearsden, Glasgow, G61 1BD, UK.
| |
Collapse
|
32
|
Petrie JL, Swan C, Ingram RM, Frame FM, Collins AT, Dumay-Odelot H, Teichmann M, Maitland NJ, White RJ. Effects on prostate cancer cells of targeting RNA polymerase III. Nucleic Acids Res 2019; 47:3937-3956. [PMID: 30820548 PMCID: PMC6486637 DOI: 10.1093/nar/gkz128] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 02/13/2019] [Accepted: 02/19/2019] [Indexed: 12/12/2022] Open
Abstract
RNA polymerase (pol) III occurs in two forms, containing either the POLR3G subunit or the related paralogue POLR3GL. Whereas POLR3GL is ubiquitous, POLR3G is enriched in undifferentiated cells. Depletion of POLR3G selectively triggers proliferative arrest and differentiation of prostate cancer cells, responses not elicited when POLR3GL is depleted. A small molecule pol III inhibitor can cause POLR3G depletion, induce similar differentiation and suppress proliferation and viability of cancer cells. This response involves control of the fate-determining factor NANOG by small RNAs derived from Alu short interspersed nuclear elements. Tumour initiating activity in vivo can be reduced by transient exposure to the pol III inhibitor. Untransformed prostate cells appear less sensitive than cancer cells to pol III depletion or inhibition, raising the possibility of a therapeutic window.
Collapse
Affiliation(s)
- John L Petrie
- Department of Biology, University of York, Heslington, York YO10 5DD, UK
| | - Caroline Swan
- Department of Biology, University of York, Heslington, York YO10 5DD, UK
| | - Richard M Ingram
- Department of Biology, University of York, Heslington, York YO10 5DD, UK
| | - Fiona M Frame
- Department of Biology, University of York, Heslington, York YO10 5DD, UK
| | - Anne T Collins
- Department of Biology, University of York, Heslington, York YO10 5DD, UK
| | - Hélène Dumay-Odelot
- Université de Bordeaux, ARNA Laboratory, F-33076 Bordeaux, France INSERM, U1212 - CNRS UMR 5320, ARNA Laboratory, F-33000 Bordeaux, France
| | - Martin Teichmann
- Université de Bordeaux, ARNA Laboratory, F-33076 Bordeaux, France INSERM, U1212 - CNRS UMR 5320, ARNA Laboratory, F-33000 Bordeaux, France
| | - Norman J Maitland
- Department of Biology, University of York, Heslington, York YO10 5DD, UK
| | - Robert J White
- Department of Biology, University of York, Heslington, York YO10 5DD, UK
| |
Collapse
|
33
|
Osther K, Förnvik K, Liljedahl E, Salford LG, Redebrandt HN. Upregulation of C1-inhibitor in pancreatic cancer. Oncotarget 2019; 10:5703-5712. [PMID: 31620245 PMCID: PMC6779287 DOI: 10.18632/oncotarget.27191] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 08/16/2019] [Indexed: 12/17/2022] Open
Abstract
Purpose The complement system has recently sparked more interest in cancer research. The classical pathway is initiated by activation of the C1 complex, which irreversibly can be bound to and inhibited by C1-INH. We have previously shown that C1-INH is upregulated in human glioblastoma (astrocytoma grade IV) on both gene and protein level. We here examine whether the complement system seems to play a role also in pancreatic cancer. Technique and results We performed an expression analysis of complement associated genes in 36 pancreatic ductal adenocarcinoma tumors and matching normal pancreatic tissue samples from pancreatic cancer patients (data from the publicly available database GSE15471). C1-INH was significantly upregulated in the pancreatic cancer tissue. None of the downstream components of the cascade were significantly upregulated in the cancer samples as compared to the control samples, which is the same pattern as we found in glioblastoma. GO analysis showed that membrane attack complex came up as the second most significantly associated cellular component. Analyzing gene expression of C1-INH in the pancreatic cancer cell lines from primary tumors versus metastatic tumor revealed no difference for the two mRNA transcripts (GSE59357). Interpretation Analysis of gene expression of complement related genes shows an upregulation of C1-INH and a downregulation of downstream components. This could suggest that C1-INH plays a role also in pancreatic cancer.
Collapse
Affiliation(s)
- Kurt Osther
- The Rausing Laboratory, Division of Neurosurgery, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Karolina Förnvik
- The Rausing Laboratory, Division of Neurosurgery, Department of Clinical Sciences, Lund University, Lund, Sweden.,Department of Clinical Chemistry, Skåne University Hospital, Scania, Sweden
| | - Emma Liljedahl
- The Rausing Laboratory, Division of Neurosurgery, Department of Clinical Sciences, Lund University, Lund, Sweden.,Department of Neurosurgery, Skåne University Hospital, Scania, Sweden
| | - Leif G Salford
- The Rausing Laboratory, Division of Neurosurgery, Department of Clinical Sciences, Lund University, Lund, Sweden.,Department of Neurosurgery, Skåne University Hospital, Scania, Sweden
| | - Henrietta Nittby Redebrandt
- The Rausing Laboratory, Division of Neurosurgery, Department of Clinical Sciences, Lund University, Lund, Sweden.,Department of Neurosurgery, Skåne University Hospital, Scania, Sweden
| |
Collapse
|
34
|
Uher O, Caisova V, Hansen P, Kopecky J, Chmelar J, Zhuang Z, Zenka J, Pacak K. Coley's immunotherapy revived: Innate immunity as a link in priming cancer cells for an attack by adaptive immunity. Semin Oncol 2019; 46:385-392. [PMID: 31739997 PMCID: PMC6904499 DOI: 10.1053/j.seminoncol.2019.10.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 10/31/2019] [Indexed: 12/11/2022]
Abstract
There is no doubt that immunotherapy lies in the spotlight of current cancer research and clinical trials. However, there are still limitations in the treatment response in certain types of tumors largely due to the presence of the complex network of immunomodulatory and immunosuppressive pathways. These limitations are not likely to be overcome by current immunotherapeutic options, which often target isolated steps in immune pathways preferentially involved in adaptive immunity. Recently, we have developed an innovative anti-cancer immunotherapeutic strategy that initially elicits a strong innate immune response with subsequent activation of adaptive immunity in mouse models. Robust primary innate immune response against tumor cells is induced by toll-like receptor ligands and anti-CD40 agonistic antibodies combined with the phagocytosis-stimulating ligand mannan, anchored to a tumor cell membrane by biocompatible anchor for membrane. This immunotherapeutic approach results in a dramatic therapeutic response in large established murine subcutaneous tumors including melanoma, sarcoma, pancreatic adenocarcinoma, and pheochromocytoma. Additionally, eradication of metastases and/or long-lasting resistance to subsequent re-challenge with tumor cells was also accomplished. Current and future advantages of this immunotherapeutic approach and its possible combinations with other available therapies are discussed in this review.
Collapse
Affiliation(s)
- Ondrej Uher
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, MD 20814, USA; Department of Medical Biology, Faculty of Science, University of South Bohemia, Ceske Budejovice 37005, Czech Republic
| | - Veronika Caisova
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, MD 20814, USA
| | - Per Hansen
- Immunoaction LLC, Charlotte, Vermont, VT 05445, USA
| | - Jan Kopecky
- Department of Medical Biology, Faculty of Science, University of South Bohemia, Ceske Budejovice 37005, Czech Republic
| | - Jindrich Chmelar
- Department of Medical Biology, Faculty of Science, University of South Bohemia, Ceske Budejovice 37005, Czech Republic
| | - Zhengping Zhuang
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, MD 20814, USA
| | - Jan Zenka
- Department of Medical Biology, Faculty of Science, University of South Bohemia, Ceske Budejovice 37005, Czech Republic
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, MD 20814, USA.
| |
Collapse
|
35
|
Comparative transcriptome analysis of peripheral blood mononuclear cells in renal transplant recipients in everolimus- and tacrolimus-based immunosuppressive therapy. Eur J Pharmacol 2019; 859:172494. [PMID: 31238062 DOI: 10.1016/j.ejphar.2019.172494] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 06/21/2019] [Accepted: 06/21/2019] [Indexed: 12/19/2022]
Abstract
To better define the biological impact of immunosuppression on peripheral blood mononuclear cells (PBMC), we employed RNASeq analysis to compare the whole transcriptomic profile of a group of renal transplant recipients undergoing maintenance treatment with Everolimus (EVE) with those treated with Tacrolimus (TAC). Then, obtained results were validated by classical biomolecular methodologies. The statistical analysis allowed the identification of four genes discriminating the 2 study groups: Sushi Domain Containing 4 (SUSD4, P = 0.02), T Cell Leukemia/Lymphoma 1A (TCL1A, P = 0.02), adhesion G protein-coupled receptor E3 (ADGRE3, P = 0.01), Immunoglobulin Heavy Constant Gamma 3 (IGHG3, P = 0.03). All of them were significantly down-regulated in patients treated with EVE compared to TAC. The Area under Receiver Operating Characteristic (AUROC) of the final model based on these 4 genes was 73.1% demonstrating its good discriminative power. RT-PCR and ELISA validated transcriptomic results. Additionally, an in vitro model confirmed that EVE significantly down-regulates (P<0.001) TCL1A, SUSD4, ADGRE3 and IgHG3 in PBMCs as well as in T cells and monocytes isolated from healthy subjects. Taken together, our data, revealed, for the first time, a new four gene-based transcriptomic fingerprint down-regulated by EVE in PBMCs of renal transplant patients that could improve the available knowledge regarding some of the biological/cellular effects of the mTOR-Is (including their antineoplastic and immune-regulatory properties).
Collapse
|
36
|
Kleczko EK, Kwak JW, Schenk EL, Nemenoff RA. Targeting the Complement Pathway as a Therapeutic Strategy in Lung Cancer. Front Immunol 2019; 10:954. [PMID: 31134065 PMCID: PMC6522855 DOI: 10.3389/fimmu.2019.00954] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 04/15/2019] [Indexed: 12/20/2022] Open
Abstract
Lung cancer is the leading cause of cancer death in men and women. Lung adenocarcinoma (LUAD), represents approximately 40% of all lung cancer cases. Advances in recent years, such as the identification of oncogenes and the use of immunotherapies, have changed the treatment of LUAD. Yet survival rates still remain low. Additionally, there is still a gap in understanding the molecular and cellular interactions between cancer cells and the immune tumor microenvironment (TME). Defining how cancer cells with distinct oncogenic drivers interact with the TME and new strategies for enhancing anti-tumor immunity are greatly needed. The complement cascade, a central part of the innate immune system, plays an important role in regulation of adaptive immunity. Initially it was proposed that complement activation on the surface of cancer cells would inhibit cancer progression via membrane attack complex (MAC)-dependent killing. However, data from several groups have shown that complement activation promotes cancer progression, probably through the actions of anaphylatoxins (C3a and C5a) on the TME and engagement of immunoevasive pathways. While originally shown to be produced in the liver, recent studies show localized complement production in numerous cell types including immune cells and tumor cells. These results suggest that complement inhibitory drugs may represent a powerful new approach for treatment of NSCLC, and numerous new anti-complement drugs are in clinical development. However, the mechanisms by which complement is activated and affects tumor progression are not well understood. Furthermore, the role of local complement production vs. systemic activation has not been carefully examined. This review will focus on our current understanding of complement action in LUAD, and describe gaps in our knowledge critical for advancing complement therapy into the clinic.
Collapse
Affiliation(s)
- Emily K Kleczko
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Jeff W Kwak
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Erin L Schenk
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Raphael A Nemenoff
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
37
|
Zhang B, Wu H. Decreased expression of COLEC10 predicts poor overall survival in patients with hepatocellular carcinoma. Cancer Manag Res 2018; 10:2369-2375. [PMID: 30122986 PMCID: PMC6078074 DOI: 10.2147/cmar.s161210] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Purpose Collectin subfamily member 10 (COLEC10) encodes for collectin liver 1 (CL-L1), which is highly expressed in normal liver. Nevertheless, the association between COLEC10 and the prognosis of patients with hepatocellular carcinoma (HCC) remains unclear. To address this question, the prognostic value of COLEC10 expression in HCC was explored in this study. Patients and methods Data from The Cancer Genome Atlas were used to compared transcriptional levels of COLEC10 in HCC samples and samples from healthy controls. In addition, COLEC10 mRNA and protein expression levels were analyzed in HCC tissue samples from Chinese patients and matched adjacent nontumorous tissue samples, by quantitative reverse-transcription polymerase chain reaction and immunohistochemistry, respectively. The prognostic value of COLEC10 was further examined using the Gene Expression Profiling Interactive Analysis online tool. Results Both the mRNA and protein levels of COLEC10 were found to be downregulated in HCC tissues compared with normal controls. Survival analysis indicated that decreased mRNA and protein levels of COLEC10 were related with shorter overall survival in patients with HCC. In addition, univariate and multivariate analysis demonstrated that COLEC10 is an independent prognostic factor for overall survival of HCC patients. Conclusion Together, the results suggest that decreased expression of COLEC10 may predict poor overall survival in patients with HCC.
Collapse
Affiliation(s)
- Baozhu Zhang
- Department of Oncology, The People's Hospital of Baoan Shenzhen, The Affiliated Baoan Hospital of Southern Medical University, Shenzhen, Guangdong 518101, People's Republic of China,
| | - Haibo Wu
- Department of Medical Administration, The People's Hospital of Baoan Shenzhen, The Affiliated Baoan Hospital of Southern Medical University, Shenzhen, Guangdong 518101, People's Republic of China
| |
Collapse
|
38
|
Peng S, Du T, Wu W, Chen X, Lai Y, Zhu D, Wang Q, Ma X, Lin C, Li Z, Guo Z, Huang H. Decreased expression of serine protease inhibitor family G1 (SERPING1) in prostate cancer can help distinguish high-risk prostate cancer and predicts malignant progression. Urol Oncol 2018; 36:366.e1-366.e9. [DOI: 10.1016/j.urolonc.2018.05.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 04/01/2018] [Accepted: 05/15/2018] [Indexed: 10/28/2022]
|
39
|
Ceylan K, Jahns LJ, Lode BN, Ehlert K, Kietz S, Troschke-Meurer S, Siebert N, Lode HN. Inflammatory response and treatment tolerance of long-term infusion of the anti-GD 2 antibody ch14.18/CHO in combination with interleukin-2 in patients with high-risk neuroblastoma. Pediatr Blood Cancer 2018; 65:e26967. [PMID: 29350486 DOI: 10.1002/pbc.26967] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 12/18/2017] [Accepted: 12/19/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND The monoclonal anti-GD2 antibody ch14.18/CHO in combination with IL-2 is active and effective in high-risk neuroblastoma (NB) patients. Here, we investigated the inflammatory response and treatment tolerance of long-term infusion (LTI) of ch14.18/CHO (10 × 10 mg/m2 ; 24 hr) in combination with subcutaneous (s.c.) IL-2 in a single center program. METHODS Fifty-three NB patients received up to six cycles of 100 mg/m2 ch14.18/CHO (d8-18, where d represents day(s)) as LTI combined with 6 × 106 IU/m2 s.c. IL-2 (d1-5; 8-12) and 160 mg/m2 oral 13-cis retinoic acid (RA) (d19-32). Side effects of ch14.18/CHO and IL-2 treatment require hospitalization of patients on d8. Treatment tolerance was evaluated daily with clinical parameters (body temperature, vital signs, Lansky performance status, requirement of i.v. concomitant medication) to define an outpatient candidate status. sIL-2-R and C-reactive protein values were determined to assess the inflammatory response. RESULTS LTI of ch14.18/CHO (d8-18) in combination with s.c.IL-2 (d8-12) showed an acceptable treatment tolerance that allowed all patients to receive part of the treatment as an outpatient (median time point of discharge: d15 for all cycles). The treatment tolerance improved from cycle to cycle and the time to become an outpatient candidate decreased from d15 to d13 in subsequent cycles. Clinical and laboratory parameters indicate a maximum inflammatory response at d11 of each cycle. Interestingly, the soluble IL-2 receptor remained increased at baseline of the next cycle indicating immune activation over the entire treatment period of 6 months. CONCLUSIONS LTI of ch14.18/CHO combined with s.c.IL-2 shows an improved tolerance in subsequent cycles allowing outpatient treatment.
Collapse
Affiliation(s)
- Kiraz Ceylan
- Pediatric Hematology and Oncology, University Medicine Greifswald, Greifswald, Germany
| | - Luciana J Jahns
- Pediatric Hematology and Oncology, University Medicine Greifswald, Greifswald, Germany
| | - Bjoern N Lode
- Pediatric Hematology and Oncology, University Medicine Greifswald, Greifswald, Germany
| | - Karoline Ehlert
- Pediatric Hematology and Oncology, University Medicine Greifswald, Greifswald, Germany
| | - Silke Kietz
- Pediatric Hematology and Oncology, University Medicine Greifswald, Greifswald, Germany
| | | | - Nikolai Siebert
- Pediatric Hematology and Oncology, University Medicine Greifswald, Greifswald, Germany
| | - Holger N Lode
- Pediatric Hematology and Oncology, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
40
|
Yu Y, Ma X, Gong R, Zhu J, Wei L, Yao J. Recent advances in CD8 + regulatory T cell research. Oncol Lett 2018; 15:8187-8194. [PMID: 29805553 DOI: 10.3892/ol.2018.8378] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Accepted: 02/01/2018] [Indexed: 11/05/2022] Open
Abstract
Various subgroups of CD8+ T lymphocytes do not only demonstrate cytotoxic effects, but also serve important regulatory roles in the body's immune response. In particular, CD8+ regulatory T cells (CD8+ Tregs), which possess important immunosuppressive functions, are able to effectively block the overreacting immune response and maintain the body's immune homeostasis. In recent years, studies have identified a small set of special CD8+ Tregs that can recognize major histocompatibility complex class Ib molecules, more specifically Qa-1 in mice and HLA-E in humans, and target the self-reactive CD4+ T ce lls. These findings have generated broad implications in the scientific community and attracted general interest to CD8+ Tregs. The present study reviews the recent research progress on CD8+ Tregs, including their origin, functional classification, molecular markers and underlying mechanisms of action.
Collapse
Affiliation(s)
- Yating Yu
- Department of Medical School, Guangxi University of Science and Technology, Liuzhou, Guangxi 545005, P.R. China
| | - Xinbo Ma
- Department of Medical School, Guangxi University of Science and Technology, Liuzhou, Guangxi 545005, P.R. China
| | - Rufei Gong
- Department of Medical School, Guangxi University of Science and Technology, Liuzhou, Guangxi 545005, P.R. China
| | - Jianmeng Zhu
- Department of Chunan First People's Hospital, Hangzhou, Zhejiang 310000, P.R. China
| | - Lihua Wei
- Department of Medical School, Guangxi University of Science and Technology, Liuzhou, Guangxi 545005, P.R. China
| | - Jinguang Yao
- Department of Medical School, Guangxi University of Science and Technology, Liuzhou, Guangxi 545005, P.R. China
| |
Collapse
|
41
|
Wang Y, Liang H. When MicroRNAs Meet RNA Editing in Cancer: A Nucleotide Change Can Make a Difference. Bioessays 2018; 40:10.1002/bies.201700188. [PMID: 29280160 PMCID: PMC5828010 DOI: 10.1002/bies.201700188] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 11/22/2017] [Indexed: 12/20/2022]
Abstract
RNA editing is a major post-transcriptional mechanism that changes specific nucleotides at the RNA level. The most common RNA editing type in humans is adenosine (A) to inosine (I) editing, which is mediated by ADAR enzymes. RNA editing events can not only change amino acids in proteins, but also affect the functions of non-coding RNAs such as miRNAs. Recent studies have characterized thousands of miRNA RNA editing events across different cancer types. Importantly, individual cases of miRNA editing have been reported to play a role in cancer development. In this review, we summarize the current knowledge of miRNA editing in cancer, and discuss the mechanisms on how miRNA-related editing events modulate the initiation and progression of human cancer. Finally, we discuss the challenges and future directions of studying miRNA editing in cancer.
Collapse
Affiliation(s)
- Yumeng Wang
- Graduate Program in Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Han Liang
- Graduate Program in Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
42
|
李 捷, 朱 玲, 左 大, 张 丽, 卢 晓, 陈 政, 周 嘉. [Plasma mannan?binding lectin and MBL?associated serine protease 2 in patients with hepatocellular carcinoma]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2017; 37:1667-1672. [PMID: 29292263 PMCID: PMC6744007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Indexed: 07/30/2024]
Abstract
OBJECTIVE To detect the plasma levels of mannan?binding lectin (MBL) and MBL?associated serine protease?2 (MASP-2) in patients with hepatocellular carcinoma (HCC) and explore their role in the tumorigenesis and progression of HCC. METHODS The plasma levels of MBL and MASP?2 were detected by enzyme?linked immunosorbent assay in 64 HCC patients and 30 healthy control subjects. The correlation of MBL and MASP?2 with the clinical parameters of HCC patients were analyzed. RESULTS The plasma levels of MBL (P=0.014) and MASP?2 (P=0.002) were significantly higher in HCC patients than in the healthy controls, but the MBL?to?MASP?2 ratio did not differ significantly between the two groups. In HCC patients, plasma MBL level was positively correlated with vascular invasion (r=0.253, P=0.047) and total bilirubin level (r=0.283, P=0.024). The plasma level of MASP?2 was positively correlated with TNM stage (r=0.276, P=0.027) and negatively correlated with plasma albumin level (r=0.?0.317, P=0.015). ROC curve analysis revealed an area under curve of 0.665 for MBL (P=0.010) and 0.694 for MASP?2 (P=0.003). The sensitivities of MBL and MASP?2 were 50% and 89.1% in the diagnosis of HCC, respectively. CONCLUSION MBL and MASP?2 are associated with the inflammatory state and disease progression in patients with HCC.
Collapse
Affiliation(s)
- 捷 李
- 南方医科大学基础医学院免疫教研室,广东 广州 510515Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- 南方医科大学第三附属医院(广东省骨科研究院),广东 广州 510000Third Affiliated Hospital of Southern Medical University, Guangdong Academy of Orthopaedics, Guangzhou 510000, China
| | - 玲燕 朱
- 中山大学生命科学学院,广东 广州 5102750School of Life Science, Sun Yat-sen University, Guangzhou 510275, China
| | - 大明 左
- 南方医科大学基础医学院免疫教研室,广东 广州 510515Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - 丽芸 张
- 南方医科大学基础医学院免疫教研室,广东 广州 510515Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - 晓 卢
- 南方医科大学基础医学院免疫教研室,广东 广州 510515Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - 政良 陈
- 南方医科大学基础医学院免疫教研室,广东 广州 510515Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - 嘉 周
- 南方医科大学基础医学院免疫教研室,广东 广州 510515Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
43
|
Gallenkamp J, Spanier G, Wörle E, Englbrecht M, Kirschfink M, Greslechner R, Braun R, Schäfer N, Bauer RJ, Pauly D. A novel multiplex detection array revealed systemic complement activation in oral squamous cell carcinoma. Oncotarget 2017; 9:3001-3013. [PMID: 29423024 PMCID: PMC5790441 DOI: 10.18632/oncotarget.22963] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 11/11/2017] [Indexed: 11/25/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is one of the most common tumors within the oral cavity. Early diagnosis and prognosis tools are urgently needed. This study aimed to investigate the activation of the complement system in OSCC patients as potential biomarker. Therefore, an innovative complement activation array was developed. Characterized antibodies detecting the complement activation specific epitopes C3a, C5a and sC5b-9 along with control antibodies were implemented into a suspension bead array. Human serum from a healthy (n = 46) and OSCC patient (n = 57) cohort were used to investigate the role of complement activation in oral tumor progression. The novel multiplex assay detected C3a, C5a and sC5b-9 from a minimal sample volume of human tears, aqueous humor and blood samples. Limits of detection were 0.04 ng/mL for C3a, 0.03 ng/mL for C5a and 18.9 ng/mL for sC5b-9, respectively. Biological cut-off levels guaranteed specific detections from serum. The mean serum concentration of a healthy control cohort was 680 ng/mL C3a, 70 ng/mL C5a and 2247 ng/mL sC5b-9, respectively. The assay showed an intra-assay precision of 2.9-6.4% and an inter-assay precision of 9.2-18.2%. Increased systemic C5a (p < 0.0001) and sC5b-9 (p = 0.01) concentrations in OSCC patients were determined using the validated multiplex complement assay. Higher C5a concentrations correlated with tumor differentiation and OSCC extension state. Systemic sC5b-9 determination provided a novel biomarker for infiltrating tumor growth and C3a levels were associated with local tumor spreading. Our study suggests that systemic complement activation levels in OSCC patients may be useful to assess disease progression.
Collapse
Affiliation(s)
- Juliane Gallenkamp
- University Hospital Regensburg, Department of Oral and Maxillofacial Surgery, Regensburg, Germany
| | - Gerrit Spanier
- University Hospital Regensburg, Department of Oral and Maxillofacial Surgery, Regensburg, Germany
| | - Elisabeth Wörle
- University Hospital Regensburg, Department of Ophthalmology, Regensburg, Germany
| | - Markus Englbrecht
- University Hospital Regensburg, Department of Ophthalmology, Regensburg, Germany
| | | | - Roman Greslechner
- University Hospital Regensburg, Department of Ophthalmology, Regensburg, Germany
| | - Regine Braun
- University Hospital Regensburg, Department of Ophthalmology, Regensburg, Germany
| | - Nicole Schäfer
- University Hospital Regensburg, Department of Ophthalmology, Regensburg, Germany
| | - Richard J Bauer
- University Hospital Regensburg, Department of Oral and Maxillofacial Surgery, Regensburg, Germany.,Center for Medical Biotechnology, Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Diana Pauly
- University Hospital Regensburg, Department of Ophthalmology, Regensburg, Germany
| |
Collapse
|
44
|
Wilson AL, Moffitt LR, Duffield N, Rainczuk A, Jobling TW, Plebanski M, Stephens AN. Autoantibodies against HSF1 and CCDC155 as Biomarkers of Early-Stage, High-Grade Serous Ovarian Cancer. Cancer Epidemiol Biomarkers Prev 2017; 27:183-192. [PMID: 29141850 DOI: 10.1158/1055-9965.epi-17-0752] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 10/23/2017] [Accepted: 11/09/2017] [Indexed: 11/16/2022] Open
Abstract
Background: Tumor-directed circulating autoantibodies (AAb) are a well-established feature of many solid tumor types, and are often observed prior to clinical disease manifestation. As such, they may provide a good indicator of early disease development. We have conducted a pilot study to identify novel AAbs as markers of early-stage HGSOCs.Methods: A rare cohort of patients with early (FIGO stage Ia-c) HGSOCs for IgG, IgA, and IgM-mediated AAb reactivity using high-content protein arrays (containing 9,184 individual proteins). AAb reactivity against selected antigens was validated by ELISA in a second, independent cohort of individual patients.Results: A total of 184 antigens were differentially detected in early-stage HGSOC patients compared with all other patient groups assessed. Among the six most highly detected "early-stage" antigens, anti-IgA AAbs against HSF1 and anti-IgG AAbs CCDC155 (KASH5; nesprin 5) were significantly elevated in patients with early-stage malignancy. Receiver operating characteristic (ROC) analysis suggested that AAbs against HSF1 provided better detection of early-stage malignancy than CA125 alone. Combined measurement of anti-HSF1, anti-CCDC155, and CA125 also improved efficacy at higher sensitivity.Conclusions: The combined measurement of anti-HSF1, anti-CCDC155, and CA125 may be useful for early-stage HGSOC detection.Impact: This is the first study to specifically identify AAbs associated with early-stage HGSOC. The presence and high frequency of specific AAbs in early-stage cancer patients warrants a larger scale examination to define their value for early disease detection at primary diagnosis and/or recurrence. Cancer Epidemiol Biomarkers Prev; 27(2); 183-92. ©2017 AACR.
Collapse
Affiliation(s)
- Amy L Wilson
- Department of Molecular and Translational Sciences, Monash University, Victoria, Australia.,Centre for Cancer Research, Hudson Institute of Medical Research, Victoria, Australia
| | - Laura R Moffitt
- Department of Molecular and Translational Sciences, Monash University, Victoria, Australia.,Centre for Cancer Research, Hudson Institute of Medical Research, Victoria, Australia
| | - Nadine Duffield
- Department of Molecular and Translational Sciences, Monash University, Victoria, Australia.,Centre for Cancer Research, Hudson Institute of Medical Research, Victoria, Australia
| | - Adam Rainczuk
- Department of Molecular and Translational Sciences, Monash University, Victoria, Australia.,Centre for Cancer Research, Hudson Institute of Medical Research, Victoria, Australia
| | - Tom W Jobling
- Obstetrics and Gynaecology, Monash Medical Centre, Clayton, Victoria, Australia.,Epworth Research Institute, Epworth HealthCare, Richmond, Victoria, Australia
| | - Magdalena Plebanski
- Department of Immunology and Pathology, Monash University, Melbourne, Australia.,School of Health and Biomedical Sciences, RMIT, Bundoora, Victoria, Australia
| | - Andrew N Stephens
- Department of Molecular and Translational Sciences, Monash University, Victoria, Australia. .,Centre for Cancer Research, Hudson Institute of Medical Research, Victoria, Australia.,Epworth Research Institute, Epworth HealthCare, Richmond, Victoria, Australia
| |
Collapse
|
45
|
Mirabelli P, Mukwaya A, Lennikov A, Xeroudaki M, Peebo B, Schaupper M, Lagali N. Genome-wide expression differences in anti-Vegf and dexamethasone treatment of inflammatory angiogenesis in the rat cornea. Sci Rep 2017; 7:7616. [PMID: 28811496 PMCID: PMC5557983 DOI: 10.1038/s41598-017-07129-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 06/22/2017] [Indexed: 12/14/2022] Open
Abstract
Angiogenesis as a pathological process in the eye can lead to blindness. In the cornea, suppression of angiogenesis by anti-VEGF treatment is only partially effective while steroids, although effective in treating inflammation and angiogenesis, have broad activity leading to undesirable side effects. In this study, genome-wide expression was investigated in a suture-induced corneal neovascularization model in rats, to investigate factors differentially targeted by dexamethasone and anti-Vegf. Topical treatment with either rat-specific anti-Vegf, dexamethasone, or normal goat IgG (sham) was given to sutured corneas for 48 hours, after which in vivo imaging, tissue processing for RNA microarray, and immunofluorescence were performed. Dexamethasone suppressed limbal vasodilation (P < 0.01) and genes in PI3K-Akt, focal adhesion, and chemokine signaling pathways more effectively than anti-Vegf. The most differentially expressed genes were confirmed by immunofluorescence, qRTPCR and Western blot. Strong suppression of Reg3g and the inflammatory chemokines Ccl2 and Cxcl5 and activation of classical complement pathway factors C1r, C1s, C2, and C3 occurred with dexamethasone treatment, effects absent with anti-Vegf treatment. The genome-wide results obtained in this study provide numerous potential targets for specific blockade of inflammation and angiogenesis in the cornea not addressed by anti-Vegf treatment, as possible alternatives to broad-acting immunosuppressive therapy.
Collapse
Affiliation(s)
- Pierfrancesco Mirabelli
- Department of Ophthalmology, Institute for Clinical and Experimental Medicine,Faculty of Health Sciences, Linkoping University, 58183, Linköping, Sweden
| | - Anthony Mukwaya
- Department of Ophthalmology, Institute for Clinical and Experimental Medicine,Faculty of Health Sciences, Linkoping University, 58183, Linköping, Sweden
| | - Anton Lennikov
- Department of Ophthalmology, Institute for Clinical and Experimental Medicine,Faculty of Health Sciences, Linkoping University, 58183, Linköping, Sweden
| | - Maria Xeroudaki
- Department of Ophthalmology, Institute for Clinical and Experimental Medicine,Faculty of Health Sciences, Linkoping University, 58183, Linköping, Sweden
| | - Beatrice Peebo
- Department of Ophthalmology, Institute for Clinical and Experimental Medicine,Faculty of Health Sciences, Linkoping University, 58183, Linköping, Sweden
| | - Mira Schaupper
- Department of Ophthalmology, Institute for Clinical and Experimental Medicine,Faculty of Health Sciences, Linkoping University, 58183, Linköping, Sweden
| | - Neil Lagali
- Department of Ophthalmology, Institute for Clinical and Experimental Medicine,Faculty of Health Sciences, Linkoping University, 58183, Linköping, Sweden.
| |
Collapse
|
46
|
Huang SJ, Cheng CL, Chen JR, Gong HY, Liu W, Wu JL. Inducible liver-specific overexpression of gankyrin in zebrafish results in spontaneous intrahepatic cholangiocarcinoma and hepatocellular carcinoma formation. Biochem Biophys Res Commun 2017; 490:1052-1058. [PMID: 28668389 DOI: 10.1016/j.bbrc.2017.06.164] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 06/27/2017] [Indexed: 12/30/2022]
Abstract
Liver cancer is the second leading cause of death worldwide. As such, establishing animal models of the disease is important for both basic and translational studies that move toward developing new therapies. Gankyrin is a critical oncoprotein in the genetic control of liver pathology. In order to evaluate the oncogenic role of gankyrin without cancer cell inoculation and drug treatment, we overexpressed gankyrin under the control of the fabp10a promoter. A Tet-Off system was used to drive expression in hepatocytes. At seven to twelve months of age, gankyrin transgenic fish spontaneously incurred persistent hepatocyte damage, steatosis, cholestasis, cholangitis, fibrosis and hepatic tumors. The tumors were both hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC). ICC is the second most frequent primary liver cancer in human patients and the first to develop in this tumor model. We further investigated the role of complement C3, a central molecule of the complement system, and found the expression levels of both in mRNA and protein are decreased during tumorigenesis. Together, these findings suggest that gankyrin can promote malignant transformation of liver cells in the context of persistent liver injury. This transformation may be related to compensatory proliferation and the inflammatory microenvironment. The observed decrease in complement C3 may allow transforming cells to escape coordinated induction of the immune response. Herein, we demonstrate an excellent zebrafish model for liver cancers that will be useful for studying the molecular mechanisms of tumorgenesis.
Collapse
Affiliation(s)
- Shin-Jie Huang
- Institute of Fisheries Science, National Taiwan University, Taipei 106, Taiwan
| | - Chih-Lun Cheng
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan
| | - Jim-Ray Chen
- Department of Pathology, Chang Gung Memorial Hospital, Keelung 204, Taiwan; College of Medicine, Chang Gung Univeristy, Taoyuan 333, Taiwan
| | - Hong-Yi Gong
- Department of Aquaculture, National Taiwan Ocean University, Keelung 202, Taiwan
| | - Wangta Liu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Center for Infectious Disease and Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Jen-Leih Wu
- Institute of Fisheries Science, National Taiwan University, Taipei 106, Taiwan; Department of Aquaculture, National Taiwan Ocean University, Keelung 202, Taiwan; College of Life Sciences, National Taiwan Ocean University, Keelung 202, Taiwan; Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 115, Taiwan.
| |
Collapse
|
47
|
Kotoula V, Fountzilas G. Overview of advances in cancer immunotherapy. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:260. [PMID: 27563647 PMCID: PMC4971370 DOI: 10.21037/atm.2016.07.09] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|