51
|
Kawahara R, Ortega F, Rosa-Fernandes L, Guimarães V, Quina D, Nahas W, Schwämmle V, Srougi M, Leite KRM, Thaysen-Andersen M, Larsen MR, Palmisano G. Distinct urinary glycoprotein signatures in prostate cancer patients. Oncotarget 2018; 9:33077-33097. [PMID: 30237853 PMCID: PMC6145689 DOI: 10.18632/oncotarget.26005] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 07/31/2018] [Indexed: 12/14/2022] Open
Abstract
Novel biomarkers are needed to complement prostate specific antigen (PSA) in prostate cancer (PCa) diagnostic screening programs. Glycoproteins represent a hitherto largely untapped resource with a great potential as specific and sensitive tumor biomarkers due to their abundance in bodily fluids and their dynamic and cancer-associated glycosylation. However, quantitative glycoproteomics strategies to detect potential glycoprotein cancer markers from complex biospecimen are only just emerging. Here, we describe a glycoproteomics strategy for deep quantitative mapping of N- and O-glycoproteins in urine with a view to investigate the diagnostic value of the glycoproteome to discriminate PCa from benign prostatic hyperplasia (BPH), two conditions that remain difficult to clinically stratify. Total protein extracts were obtained, concentrated and digested from urine of six PCa patients (Gleason score 7) and six BPH patients. The resulting peptide mixtures were TMT-labeled and mixed prior to a multi-faceted sample processing including hydrophilic interaction liquid chromatography (HILIC) and titanium dioxide SPE based enrichment, endo-/exoglycosidase treatment and HILIC-HPLC pre-fractionation. The isolated N- and O-glycopeptides were detected and quantified using high resolution mass spectrometry. We accurately quantified 729 N-glycoproteins spanning 1,310 unique N-glycosylation sites and observed 954 and 965 unique intact N- and O-glycopeptides, respectively, across the two disease conditions. Importantly, a panel of 56 intact N-glycopeptides perfectly discriminated PCa and BPH (ROC: AUC = 1). This study has generated a panel of intact glycopeptides that has a potential for PCa detection.
Collapse
Affiliation(s)
- Rebeca Kawahara
- Instituto de Ciências Biomédicas, Departamento de Parasitologia, Universidade de São Paulo, USP, São Paulo, Brazil
| | - Fabio Ortega
- Laboratório de Investigação Médica da Disciplina de Urologia da Faculdade de Medicina da USP, LIM55, São Paulo, Brazil
| | - Livia Rosa-Fernandes
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Vanessa Guimarães
- Laboratório de Investigação Médica da Disciplina de Urologia da Faculdade de Medicina da USP, LIM55, São Paulo, Brazil
| | - Daniel Quina
- Instituto de Ciências Biomédicas, Departamento de Parasitologia, Universidade de São Paulo, USP, São Paulo, Brazil
| | - Willian Nahas
- Instituto do Câncer do Estado de São Paulo, ICESP, São Paulo, Brazil
| | - Veit Schwämmle
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Miguel Srougi
- Laboratório de Investigação Médica da Disciplina de Urologia da Faculdade de Medicina da USP, LIM55, São Paulo, Brazil
| | - Katia R M Leite
- Laboratório de Investigação Médica da Disciplina de Urologia da Faculdade de Medicina da USP, LIM55, São Paulo, Brazil
| | | | - Martin R Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Giuseppe Palmisano
- Instituto de Ciências Biomédicas, Departamento de Parasitologia, Universidade de São Paulo, USP, São Paulo, Brazil
| |
Collapse
|
52
|
Zhang L, Fedorov Y, Adams D, Lin F. Identification of complement inhibitory activities of two chemotherapeutic agents using a high-throughput cell imaging-based screening assay. Mol Immunol 2018; 101:86-91. [PMID: 29909366 DOI: 10.1016/j.molimm.2018.06.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 05/30/2018] [Accepted: 06/07/2018] [Indexed: 11/25/2022]
Abstract
Excessive complement activation contributes significantly to the pathogeneses of various diseases. Currently, significant developmental research efforts aim to identify complement inhibitors with therapeutic uses have led to the approval of one inhibitor for clinical use. However, most existing complement inhibitors are based on monoclonal antibodies, which have many drawbacks such as high costs and limited administration options. With this report, we establish an inexpensive, cell imaging-based high-throughput assay for the large-scale screening of potential small molecule complement inhibitors. Using this assay, we screened a library containing 3115 bioactive chemical compounds and identified cisplatin and pyridostatin as two new complement inhibitors in addition to nafamostat mesylate, a compound with known complement inhibitory activity. We further demonstrated that cisplatin and pyridostatin inhibit C5 convertases in the classical pathway of complement activation but have no effects on the alternative pathway of complement activation. In summary, this work has established a simple, large-scale, high-throughput assay for screening novel complement inhibitors and discovered previously unknown complement activation inhibitory activities for cisplatin and pyridostatin.
Collapse
Affiliation(s)
- Lingjun Zhang
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Yuriy Fedorov
- Small Molecules Drug Development Core Facility, Office of Research Administration, Case Western Reserve University, Cleveland, OH, United States
| | - Drew Adams
- Department of Genetics, Case Western Reserve University, Cleveland, OH, United States
| | - Feng Lin
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States.
| |
Collapse
|
53
|
Bareke H, Akbuga J. Complement system's role in cancer and its therapeutic potential in ovarian cancer. Scand J Immunol 2018; 88:e12672. [PMID: 29734524 DOI: 10.1111/sji.12672] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 04/30/2018] [Indexed: 12/31/2022]
Abstract
Cancer immunotherapy is a strong candidate for the long-awaited new edition to standard cancer therapies. For an effective immunotherapy, it is imperative to delineate the players of antitumour immune response. As an important innate immune system effector mechanism, complement is highly likely to play a substantial role in cancer immunity. Studies suggest that there may be two different "states of complement" that show opposing effects on cancer cells; a complement profile that has antitumour effects with low expression of membrane-bound complement regulator proteins (mCRPs), lytic membrane attack complex (MAC) concentration and moderate C5a concentration, and a complement profile that has protumour effects with high expression of mCRPs, sublytic MAC and high concentrations of C5a. One of the cancers that urgently require innovative therapeutic approaches is ovarian cancer, and complement has a potential to be a good target for this purpose. A combinatorial approach where the complement cascade is fine-tuned by inhibiting some of its activities while promoting the others can prove to be a fruitful approach. Herein, we will briefly discuss the cancer-immune system interaction and then present a discussion of complement system's role in tumour immunity and its therapeutic potential for ovarian cancer immunotherapy.
Collapse
Affiliation(s)
- H Bareke
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Marmara University, Istanbul, Turkey.,Faculty of Pharmacy, Girne American University, Kyrenia, North Cyprus, Turkey
| | - J Akbuga
- Faculty of Pharmacy, Girne American University, Kyrenia, North Cyprus, Turkey
| |
Collapse
|
54
|
Olcina MM, Kim RK, Melemenidis S, Graves EE, Giaccia AJ. The tumour microenvironment links complement system dysregulation and hypoxic signalling. Br J Radiol 2018; 92:20180069. [PMID: 29544344 PMCID: PMC6435069 DOI: 10.1259/bjr.20180069] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The complement system is an innate immune pathway typically thought of as part of the first line of defence against “non-self” species. In the context of cancer, complement has been described to have an active role in facilitating cancer-associated processes such as increased proliferation, angiogenesis and migration. Several cellular members of the tumour microenvironment express and/or produce complement proteins locally, including tumour cells. Dysregulation of the complement system has been reported in numerous tumours and increased expression of complement activation fragments in cancer patient specimens correlates with poor patient prognosis. Importantly, genetic or pharmacological targeting of complement has been shown to reduce tumour growth in several cancer preclinical models, suggesting that complement could be an attractive therapeutic target. Hypoxia (low oxygen) is frequently found in solid tumours and has a profound biological impact on cellular and non-cellular components of the tumour microenvironment. In this review, we focus on hypoxia since this is a prevailing feature of the tumour microenvironment that, like increased complement, is typically associated with poor prognosis. Furthermore, interesting links between hypoxia and complement have been recently proposed but never collectively reviewed. Here, we explore how hypoxia alters regulation of complement proteins in different cellular components of the tumour microenvironment, as well as the downstream biological consequences of this regulation.
Collapse
Affiliation(s)
- Monica M Olcina
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Ryan K Kim
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | | | - Edward E Graves
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Amato J Giaccia
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| |
Collapse
|
55
|
Pleiotropic Effects of Risk Factors in Age-Related Macular Degeneration and Seemingly Unrelated Complex Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1074:247-255. [PMID: 29721950 DOI: 10.1007/978-3-319-75402-4_30] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
Age-related macular degeneration (AMD) is a complex disease with both environmental and genetic factors influencing disease risk. Genome-wide case-control association studies, candidate gene analyses, and epidemiological studies reinforced the notion that AMD is predominantly a disease of an impaired complement system and an altered high-density lipoprotein (HDL) metabolism. Recent reports demonstrated the pleiotropic role of the complement system and HDL in complex diseases such as cardiovascular disease, autoimmune disorders, cancer, and Alzheimer's disease. In light of these findings, we explore current evidence for a shared genetic and environmental risk of AMD and unrelated complex diseases based on epidemiological studies. Shared risk factors may indicate common pathways in disease pathology and thus may have implications for novel treatment options of AMD pathology.
Collapse
|
56
|
Vorobyova O, Deryabina O, Malygina D, Plotnikova N, Solovyeva A, Belyaeva K, Melnikova N. Betulin-3,28-diphosphate as a Component of Combination Cytostatic Drugs for the Treatment of Ehrlich Ascites Carcinoma In Vitro and In Vivo Experiments. Sci Pharm 2018; 86:E17. [PMID: 29690651 PMCID: PMC6027669 DOI: 10.3390/scipharm86020017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/17/2018] [Accepted: 04/18/2018] [Indexed: 12/18/2022] Open
Abstract
The activity of betulin-3,28-diphosphate (BDP) in combination with the cytostatics such as 5-fluorouracil (5-FU) and hydrazine sulfate (HS) was demonstrated by using the transplanted Ehrlich ascites carcinoma (EAC) in mice. The dose-dependent effect of combination drugs BDP + HS and BDP + 5-FU was revealed by in vitro experiments on rats. The synergetic effect of HS and BDP on oxidative stress and energy metabolism was established. The malonic dialdehyde (MDA) level both in plasma and erythrocytes decreased by 87 ± 2%, and the superoxide dismutase (SOD) activity increased by 105 ± 7% in comparison with the control. The combination of BDP + HS promoted the increase of lactate dehydrogenase (LDH) activity in the reverse reaction by 195 ± 21% compared to the control. The combination drug of 5-FU with BDP caused the synergetic decrease of the lipid peroxidation (LPO) intensity estimated by the MDA level decrease up to 14 ± 4% compared to pure compounds. Betulin-3,28-diphosphate in combination with cytostatics for EAC treatment improved the animal health status, as well as decreased the cytostatics dose that can be used in palliative therapy.
Collapse
Affiliation(s)
- Olga Vorobyova
- Department of Pharmaceutical Chemistry, Federal State Budgetary Educational Institution of Higher Education "Privolzhsky Research Medical University", Ministry of Health of the Russian Federation, Minin sq., 10/1, 603005 Nizhny Novgorod, Russia.
| | - Olga Deryabina
- Department of Chemistry, N.P. Ogarev Mordovian State University, Bolshevistskaya St. 68, 430005 Saransk, Russia.
| | - Darina Malygina
- Department of Pharmaceutical Chemistry, Federal State Budgetary Educational Institution of Higher Education "Privolzhsky Research Medical University", Ministry of Health of the Russian Federation, Minin sq., 10/1, 603005 Nizhny Novgorod, Russia.
| | - Nadezhda Plotnikova
- Department of Chemistry, N.P. Ogarev Mordovian State University, Bolshevistskaya St. 68, 430005 Saransk, Russia.
| | - Anna Solovyeva
- Department of Pharmaceutical Chemistry, Federal State Budgetary Educational Institution of Higher Education "Privolzhsky Research Medical University", Ministry of Health of the Russian Federation, Minin sq., 10/1, 603005 Nizhny Novgorod, Russia.
| | - Kseniya Belyaeva
- Department of Pharmaceutical Chemistry, Federal State Budgetary Educational Institution of Higher Education "Privolzhsky Research Medical University", Ministry of Health of the Russian Federation, Minin sq., 10/1, 603005 Nizhny Novgorod, Russia.
| | - Nina Melnikova
- Department of Pharmaceutical Chemistry, Federal State Budgetary Educational Institution of Higher Education "Privolzhsky Research Medical University", Ministry of Health of the Russian Federation, Minin sq., 10/1, 603005 Nizhny Novgorod, Russia.
| |
Collapse
|
57
|
García-Heredia JM, Carnero A. Dr. Jekyll and Mr. Hyde: MAP17's up-regulation, a crosspoint in cancer and inflammatory diseases. Mol Cancer 2018; 17:80. [PMID: 29650022 PMCID: PMC5896160 DOI: 10.1186/s12943-018-0828-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 03/28/2018] [Indexed: 12/14/2022] Open
Affiliation(s)
- José M García-Heredia
- Instituto de Biomedicina de Sevilla, IBIS/Hospital Universitario Virgen del Rocío/ Universidad de Sevilla/Consejo Superior de Investigaciones Científicas, Avda. Manuel Siurot s/n, 41013, Sevilla, Spain.,Department of Vegetal Biochemistry and Molecular Biology, University of Seville, Seville, Spain.,CIBER de Cáncer, Instituto de Salud Carlos III, Pabellón 11, Madrid, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla, IBIS/Hospital Universitario Virgen del Rocío/ Universidad de Sevilla/Consejo Superior de Investigaciones Científicas, Avda. Manuel Siurot s/n, 41013, Sevilla, Spain. .,CIBER de Cáncer, Instituto de Salud Carlos III, Pabellón 11, Madrid, Spain.
| |
Collapse
|
58
|
Park JW, Kim MS, Voon DC, Kim SJ, Bae J, Mun DG, Ko SI, Kim HK, Lee SW, Kim DY. Multi-omics analysis identifies pathways and genes involved in diffuse-type gastric carcinogenesis induced by E-cadherin, p53, and Smad4 loss in mice. Mol Carcinog 2018. [PMID: 29528141 DOI: 10.1002/mc.22803] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The molecular mechanisms underlying the pathogenesis of diffuse-type gastric cancer (DGC) have not been adequately explored due to a scarcity of appropriate animal models. A recently developed tool well suited for this line of investigation is the Pdx-1-Cre;Cdh1F/+ ;Trp53F/F ;Smad4F/F (pChe PS) mouse model that spontaneously develops metastatic DGC showing nearly complete E-cadherin loss. Here, we performed a proteogenomic analysis to uncover the molecular changes induced by the concurrent targeting of E-cadherin, p53, and Smad4 loss. The gene expression profiles of mouse DGCs and in vivo gastric phenotypes from various combinations of gene knockout demonstrated that these mutations collaborate to activate cancer-associated pathways to generate aggressive DGC. Of note, WNT-mediated epithelial-to-mesenchymal transition (EMT) and extracellular matrix (ECM)-cytokine receptor interactions were prominently featured. In particular, the WNT target gene osteopontin (OPN) that functions as an ECM cytokine is highly upregulated. In validation experiments, OPN contributed to DGC stemness by promoting cancer stem cell (CSC) survival and chemoresistance. It was further found that Bcl-xL acts as a targetable downstream effector of OPN in DGC CSC survival. In addition, Zeb2 and thymosin-β4 (Tβ4) were identified as prime candidates as suppressors of E-cadherin expression from the remaining Cdh1 allele during DGC development. Specifically, Tβ4 suppressed E-cadherin expression and anoikis while promoting cancer cell growth and migration. Collectively, these proteogenomic analyses broaden and deepen our understanding of the contribution of key driver mutations in the stepwise carcinogenesis of DGC through novel effectors, namely OPN and Tβ4.
Collapse
Affiliation(s)
- Jun Won Park
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.,National Cancer Center, Goyang, Gyeonggi, Republic of Korea
| | - Min-Sik Kim
- Department of Applied Chemistry, College of Applied Science, Kyung Hee University, Yongin-si, Gyeonggi-do, Republic of Korea.,Department of Biomedical Science and Technology, Kyung Hee Medical Science Research Institute, Kyung Hee University, Yongin-si, Gyeonggi-do, Republic of Korea
| | - Dominic C Voon
- Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Su-Jin Kim
- Department of Chemistry, Research Institute for Natural Sciences, Korea University, Seoul, Republic of Korea
| | - Jingi Bae
- Department of Chemistry, Center for Proteogenome Research, Korea University, Seoul, Republic of Korea
| | - Dong-Gi Mun
- Department of Chemistry, Center for Proteogenome Research, Korea University, Seoul, Republic of Korea
| | - Seung-Ik Ko
- Department of Chemistry, Center for Proteogenome Research, Korea University, Seoul, Republic of Korea
| | - Hark K Kim
- National Cancer Center, Goyang, Gyeonggi, Republic of Korea
| | - Sang-Won Lee
- Department of Chemistry, Center for Proteogenome Research, Korea University, Seoul, Republic of Korea
| | - Dae-Yong Kim
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
59
|
Möginger U, Grunewald S, Hennig R, Kuo CW, Schirmeister F, Voth H, Rapp E, Khoo KH, Seeberger PH, Simon JC, Kolarich D. Alterations of the Human Skin N- and O-Glycome in Basal Cell Carcinoma and Squamous Cell Carcinoma. Front Oncol 2018; 8:70. [PMID: 29619343 PMCID: PMC5871710 DOI: 10.3389/fonc.2018.00070] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 03/01/2018] [Indexed: 12/20/2022] Open
Abstract
The glycome of one of the largest and most exposed human organs, the skin, as well as glycan changes associated with non-melanoma skin cancers have not been studied in detail to date. Skin cancers such as basal cell carcinoma (BCC) and squamous cell carcinoma (SCC) are among the most frequent types of cancers with rising incidence rates in the aging population. We investigated the healthy human skin N- and O-glycome and its changes associated with BCC and SCC. Matched patient samples were obtained from frozen biopsy and formalin-fixed paraffin-embedded tissue samples for glycomics analyses using two complementary glycomics approaches: porous graphitized carbon nano-liquid chromatography electro spray ionization tandem mass spectrometry and capillary gel electrophoresis with laser induced fluorescence detection. The human skin N-glycome is dominated by complex type N-glycans that exhibit almost similar levels of α2-3 and α2-6 sialylation. Fucose is attached exclusively to the N-glycan core. Core 1 and core 2 type O-glycans carried up to three sialic acid residues. An increase of oligomannose type N-glycans and core 2 type O-glycans was observed in BCC and SCC, while α2-3 sialylation levels were decreased in SCC but not in BCC. Furthermore, glycopeptide analyses provided insights into the glycoprotein candidates possibly associated with the observed N-glycan changes, with glycoproteins associated with binding events being the most frequently identified class.
Collapse
Affiliation(s)
- Uwe Möginger
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany.,Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Sonja Grunewald
- Department of Dermatology, Venerology and Allergology, Leipzig University Medical Center, Leipzig, Germany
| | - René Hennig
- Department of Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany.,glyXera GmbH, Magdeburg, Germany
| | - Chu-Wei Kuo
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Falko Schirmeister
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany.,Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Harald Voth
- Department of Dermatology, Venerology and Allergology, Leipzig University Medical Center, Leipzig, Germany
| | - Erdmann Rapp
- Department of Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany.,glyXera GmbH, Magdeburg, Germany
| | - Kay-Hooi Khoo
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Peter H Seeberger
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany.,Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Jan C Simon
- Department of Dermatology, Venerology and Allergology, Leipzig University Medical Center, Leipzig, Germany
| | - Daniel Kolarich
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany.,Institute for Glycomics, Griffith University, Southport, QLD, Australia
| |
Collapse
|
60
|
Lu ZL, Chen YJ, Jing XY, Wang NN, Zhang T, Hu CJ. Detection and Identification of Serum Peptides Biomarker in Papillary Thyroid Cancer. Med Sci Monit 2018; 24:1581-1587. [PMID: 29549708 PMCID: PMC5870111 DOI: 10.12659/msm.907768] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background Papillary thyroid cancer (PTC) is currently the most commonly diagnosed endocrine malignancy. In addition, the sex- and age-adjusted incidence of PTC has exhibited a greater increase over the last 2 decades than in many other malignancies. Thus, discovering noninvasive specific serum biomarker to distinguish PTC from cancer-free controls in its early stages remains an important goal. Material/Methods Serum samples from 88 PTC patients and 80 cancer-free controls were randomly allocated into training or validation sets. Serum peptide profiling was performed by matrix-assisted laser desorption/ionization-time of flight mass spectrometry (MALDI-TOF-MS) after using weak cation exchange magnetic beads (WCX-MB), and the results were evaluated by use of ClinProTools™ Software. To distinguish PTC from cancer-free controls, quick classifier (QC), supervised neural network (SNN), and genetic algorithm (GA) models were established. The models were blindly validated to verify their diagnostic capabilities. The most discriminative peaks were subsequently identified with a nano-liquid chromatography-electrospray ionization-tandem mass spectrometry system. Results Six peptide ions were identified as the most discriminative peaks between the PTC and cancer-free control samples. The QC model exhibited satisfactory sensitivity and specificity among the 3 models that were validated. Two peaks, at m/z 2671.17 and m/z 1464.68, were identified as fragments of the alpha chain of fibrinogen, while a peak at m/z 1738.92 was a fragment of complement component 4A/B. Conclusions MS combined with ClinProTools™ software was able to detect peptide biomarkers in PTC patients. In addition, the constructed classification models provided a serum peptidome pattern for distinguishing PTC from cancer-free controls. Both fibrinogen α and complement C4A/B were identified as potential markers for diagnosis of PTC.
Collapse
Affiliation(s)
- Zhao-Lian Lu
- School of Graduate, Second Military Medicinal University, Shanghai, China (mainland).,Department of Laboratory Medicine, General Hospital of Jinan Military Command Region, Jinan, Shandong, China (mainland)
| | - Ying-Jian Chen
- Department of Laboratory Medicine, General Hospital of Jinan Military Command Region, Jinan, Shandong, China (mainland)
| | - Xin-Yan Jing
- Department of Laboratory Medicine, General Hospital of Jinan Military Command Region, Jinan, Shandong, China (mainland)
| | - Na-Na Wang
- Department of Laboratory Medicine, General Hospital of Jinan Military Command Region, Jinan, Shandong, China (mainland)
| | - Ting Zhang
- Department of Laboratory Medicine, General Hospital of Jinan Military Command Region, Jinan, Shandong, China (mainland)
| | - Cheng-Jin Hu
- Department of Laboratory Medicine, General Hospital of Jinan Military Command Region, Jinan, Shandong, China (mainland)
| |
Collapse
|
61
|
Dho SH, Lim JC, Kim LK. Beyond the Role of CD55 as a Complement Component. Immune Netw 2018; 18:e11. [PMID: 29503741 PMCID: PMC5833118 DOI: 10.4110/in.2018.18.e11] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 02/09/2018] [Accepted: 02/11/2018] [Indexed: 01/28/2023] Open
Abstract
The complement is a part of the immune system that plays several roles in removing pathogens. Despite the importance of the complement system, the exact role of each component has been overlooked because the complement system was thought to be a nonspecific humoral immune mechanism that worked against pathogens. Decay-accelerating factor (DAF or CD55) is a known inhibitor of the complement system and has recently attracted substantial attention due to its role in various diseases, such as cancer, protein-losing enteropathy, and malaria. Some protein-losing enteropathy cases are caused by CD55 deficiency, which leads to complement hyperactivation, malabsorption, and angiopathic thrombosis. In addition, CD55 has been reported to be an essential host receptor for infection by the malaria parasite. Moreover, CD55 is a ligand of the seven-span transmembrane receptor CD97. Since CD55 is present in various cells, the functional role of CD55 has been expanded by showing that CD55 is associated with a variety of diseases, including cancer, malaria, protein-losing enteropathy, paroxysmal nocturnal hemoglobinuria, and autoimmune diseases. This review summarizes the current understanding of CD55 and the role of CD55 in these diseases. It also provides insight into the development of novel drugs for the diagnosis and treatment of diseases associated with CD55.
Collapse
Affiliation(s)
- So Hee Dho
- Radioisotope Research Division, Department of Research Reactor Utilization, Korea Atomic Energy Research Institute, Daejeon 34057, Korea
| | - Jae Cheong Lim
- Radioisotope Research Division, Department of Research Reactor Utilization, Korea Atomic Energy Research Institute, Daejeon 34057, Korea
| | - Lark Kyun Kim
- Severance Biomedical Science Institute and BK21 PLUS Project to Medical Sciences, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06230, Korea
| |
Collapse
|
62
|
Reis ES, Mastellos DC, Ricklin D, Mantovani A, Lambris JD. Complement in cancer: untangling an intricate relationship. Nat Rev Immunol 2018; 18:5-18. [PMID: 28920587 PMCID: PMC5816344 DOI: 10.1038/nri.2017.97] [Citation(s) in RCA: 291] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In tumour immunology, complement has traditionally been considered as an adjunctive component that enhances the cytolytic effects of antibody-based immunotherapies, such as rituximab. Remarkably, research in the past decade has uncovered novel molecular mechanisms linking imbalanced complement activation in the tumour microenvironment with inflammation and suppression of antitumour immune responses. These findings have prompted new interest in manipulating the complement system for cancer therapy. This Review summarizes our current understanding of complement-mediated effector functions in the tumour microenvironment, focusing on how complement activation can act as a negative or positive regulator of tumorigenesis. It also offers insight into clinical aspects, including the feasibility of using complement biomarkers for cancer diagnosis and the use of complement inhibitors during cancer treatment.
Collapse
Affiliation(s)
- Edimara S Reis
- Department of Pathology and Laboratory Medicine, School of Medicine, University of Pennsylvania 19104, Philadelphia, Pennsylvania, USA
| | | | - Daniel Ricklin
- Department of Pharmaceutical Sciences, University of Basel, Basel 4056, Switzerland
| | - Alberto Mantovani
- Humanitas Clinical and Research Center and Humanitas University, Rozzano-Milan 20089, Italy
| | - John D Lambris
- Department of Pathology and Laboratory Medicine, School of Medicine, University of Pennsylvania 19104, Philadelphia, Pennsylvania, USA
| |
Collapse
|
63
|
Ajona D, Ortiz-Espinosa S, Pio R. Complement anaphylatoxins C3a and C5a: Emerging roles in cancer progression and treatment. Semin Cell Dev Biol 2017; 85:153-163. [PMID: 29155219 DOI: 10.1016/j.semcdb.2017.11.023] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 11/07/2017] [Accepted: 11/15/2017] [Indexed: 02/06/2023]
Abstract
Recent insights into the role of complement anaphylatoxins C3a and C5a in cancer provide new opportunities for the development of innovative biomarkers and therapeutic strategies. These two complement activation products can maintain chronic inflammation, promote an immunosuppressive microenvironment, induce angiogenesis, and increase the motility and metastatic potential of cancer cells. Still, the diverse heterogeneity of responses mediated by these peptides poses a challenge both to our understanding of the role played by these molecules in cancer progression and to the development of effective treatments. This review attempts to summarize the evidence surrounding the involvement of anaphylatoxins in the biological contexts associated with tumor progression. We also describe the recent developments that support the inhibition of anaphylatoxins, or their cognate receptors C3aR and C5aR1, as a treatment option for maximizing the clinical efficacy of current immunotherapies that target the PD-1/PD-L1 immune checkpoint.
Collapse
Affiliation(s)
- Daniel Ajona
- University of Navarra, Center for Applied Medical Research (CIMA), Program in Solid Tumors and Biomarkers, Pamplona, Spain; Navarra's Health Research Institute (IdiSNA), Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Spain; University of Navarra, School of Sciences, Department of Biochemistry and Genetics, Pamplona, Spain
| | - Sergio Ortiz-Espinosa
- University of Navarra, Center for Applied Medical Research (CIMA), Program in Solid Tumors and Biomarkers, Pamplona, Spain; University of Navarra, School of Sciences, Department of Biochemistry and Genetics, Pamplona, Spain
| | - Ruben Pio
- University of Navarra, Center for Applied Medical Research (CIMA), Program in Solid Tumors and Biomarkers, Pamplona, Spain; Navarra's Health Research Institute (IdiSNA), Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Spain; University of Navarra, School of Sciences, Department of Biochemistry and Genetics, Pamplona, Spain.
| |
Collapse
|
64
|
Kwak JW, Laskowski J, Li HY, McSharry MV, Sippel TR, Bullock BL, Johnson AM, Poczobutt JM, Neuwelt AJ, Malkoski SP, Weiser-Evans MC, Lambris JD, Clambey ET, Thurman JM, Nemenoff RA. Complement Activation via a C3a Receptor Pathway Alters CD4 + T Lymphocytes and Mediates Lung Cancer Progression. Cancer Res 2017; 78:143-156. [PMID: 29118090 DOI: 10.1158/0008-5472.can-17-0240] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 06/12/2017] [Accepted: 11/01/2017] [Indexed: 11/16/2022]
Abstract
The complement cascade is a part of the innate immune system that acts primarily to remove pathogens and injured cells. However, complement activation is also peculiarly associated with tumor progression. Here we report mechanistic insights into this association in multiple immunocompetent orthotopic models of lung cancer. After tumor engraftment, we observed systemic activation of the complement cascade as reflected by elevated levels of the key regulator C3a. Notably, growth of primary tumors and metastases was both strongly inhibited in C3-deficient mice (C3-/- mice), with tumors undetectable in many subjects. Growth inhibition was associated with increased numbers of IFNγ+/TNFα+/IL10+ CD4+ and CD8+ T cells. Immunodepletion of CD4+ but not CD8+ T cells in tumor-bearing subjects reversed the inhibitory effects of C3 deletion. Similarly, antagonists of the C3a or C5a receptors inhibited tumor growth. Investigations using multiple tumor cell lines in the orthotopic model suggested the involvement of a C3/C3 receptor autocrine signaling loop in regulating tumor growth. Overall, our findings offer functional evidence that complement activation serves as a critical immunomodulator in lung cancer progression, acting to drive immune escape via a C3/C5-dependent pathway.Significance: This provocative study suggests that inhibiting complement activation may heighten immunotherapeutic responses in lung cancer, offering findings with immediate implications, given the existing clinical availability of complement antagonists. Cancer Res; 78(1); 143-56. ©2017 AACR.
Collapse
Affiliation(s)
- Jeff W Kwak
- Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | | | - Howard Y Li
- Department of Medicine, University of Colorado Denver, Aurora, Colorado.,Veterans Affairs Medical Center, Denver, Colorado
| | - Maria V McSharry
- Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Trisha R Sippel
- Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Bonnie L Bullock
- Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Amber M Johnson
- Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | | | | | | | - Mary C Weiser-Evans
- Department of Medicine, University of Colorado Denver, Aurora, Colorado.,Department of Pharmacology, University of Colorado Denver, Aurora, Colorado
| | - John D Lambris
- Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado
| | - Eric T Clambey
- Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado
| | - Joshua M Thurman
- Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Raphael A Nemenoff
- Department of Medicine, University of Colorado Denver, Aurora, Colorado. .,Department of Pharmacology, University of Colorado Denver, Aurora, Colorado
| |
Collapse
|
65
|
Ratajczak MZ. HO-1 inhibits migration of leukemic cells. Oncotarget 2017; 8:89429-89430. [PMID: 29163759 PMCID: PMC5685680 DOI: 10.18632/oncotarget.21044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 09/15/2017] [Indexed: 01/25/2023] Open
Affiliation(s)
- Mariusz Z Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, KY, USA and Department of Regenerative Medicine Warsaw Medical University, Warsaw, Poland
| |
Collapse
|
66
|
Jacamo R, Davis RE, Ling X, Sonnylal S, Wang Z, Ma W, Zhang M, Ruvolo P, Ruvolo V, Wang RY, McQueen T, Lowe S, Zuber J, Kornblau SM, Konopleva M, Andreeff M. Tumor Trp53 status and genotype affect the bone marrow microenvironment in acute myeloid leukemia. Oncotarget 2017; 8:83354-83369. [PMID: 29137349 PMCID: PMC5663521 DOI: 10.18632/oncotarget.19042] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 06/03/2017] [Indexed: 02/06/2023] Open
Abstract
The genetic heterogeneity of acute myeloid leukemia (AML) and the variable responses of individual patients to therapy suggest that different AML genotypes may influence the bone marrow (BM) microenvironment in different ways. We performed gene expression profiling of bone marrow mesenchymal stromal cells (BM-MSC) isolated from normal C57BL/6 mice or mice inoculated with syngeneic murine leukemia cells carrying different human AML genotypes, developed in mice with Trp53 wild-type or nullgenetic backgrounds. We identified a set of genes whose expression in BM-MSC was modulated by all four AML genotypes tested. In addition, there were sets of differentially-expressed genes in AML-exposed BM-MSC that were unique to the particular AML genotype or Trp53 status. Our findings support the hypothesis that leukemia cells alter the transcriptome of surrounding BM stromal cells, in both common and genotype-specific ways. These changes are likely to be advantageous to AML cells, affecting disease progression and response to chemotherapy, and suggest opportunities for stroma-targeting therapy, including those based on AML genotype.
Collapse
Affiliation(s)
- Rodrigo Jacamo
- Department of Leukemia, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - R. Eric Davis
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiaoyang Ling
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sonali Sonnylal
- Department of Leukemia, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zhiqiang Wang
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wencai Ma
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Min Zhang
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Peter Ruvolo
- Department of Leukemia, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vivian Ruvolo
- Department of Leukemia, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rui-Yu Wang
- Department of Leukemia, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Teresa McQueen
- Department of Leukemia, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Scott Lowe
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Johannes Zuber
- Research Institute of Molecular Pathology, Vienna, Austria
| | - Steven M. Kornblau
- Department of Leukemia, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marina Konopleva
- Department of Leukemia, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael Andreeff
- Department of Leukemia, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
67
|
Ji Q, Wang W, Luo Y, Cai F, Lu Y, Deng W, Li Q, Su S. Characteristic proteins in the plasma of postoperative colorectal and liver cancer patients with Yin deficiency of liver-kidney syndrome. Oncotarget 2017; 8:103223-103235. [PMID: 29262557 PMCID: PMC5732723 DOI: 10.18632/oncotarget.21735] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 09/22/2017] [Indexed: 12/24/2022] Open
Abstract
Systems biology and bioinformatics provide the feasibility for the basic research associated with “same traditional Chinese medicine (TCM) syndrome in different diseases”. In this study, the plasma proteins in postoperative colorectal (PCC) and postoperative liver cancer (PLC) patients with YDLKS (Yin deficiency of liver-kidney syndrome) were screened out using iTRAQ combined with LC-MS/MS technology. The results demonstrated that, KNG1, AMBP, SERPING1, etc, were all differentially expressed in both PCC and PLC patients with YDLKS, and associated closely with complement and coagulation cascades pathway. C7 and C2 were another two representative factors involving in former pathway. Further validation showed that, the C7 levels were increased significantly in PLC (P < 0.05) and PCC (P < 0.05) with YDLKS group compared to those of NS (no obvious TCM syndromes) group. The AMBP levels were down-regulated significantly in PLC with YDLKS group compared to those of PCC with YDLKS group (P < 0.05). The significant differences of SERPING1 levels (and C2 levels) were shown between YDLKS and NS in PCC (P < 0.01). There were also significant differences of C2 levels between PCC and PLC patients with YDLKS (P < 0.05). Moreover, significant differences of C2 levels were also found between PLC and PCC patients with YDLKS (P < 0.01). ROC curves indicated that, C7 and SERPING1 independently had a potential diagnostic value in distinguishing YDLKS from NS in PLC and PCC, providing the evidences for the material basis of “same TCM syndrome in different diseases” in PCC and PLC patients with YDLKS.
Collapse
Affiliation(s)
- Qing Ji
- Research Center for Traditional Chinese Medicine Complexity System, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.,Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wenhai Wang
- Department of Medical Oncology, Shuguang Hospital Affiliated Baoshan Branch, Shanghai University of Traditional Chinese Medicine, Shanghai 201901, China
| | - Yunquan Luo
- Department of Liver and Gallbladder surgery, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Feifei Cai
- Research Center for Traditional Chinese Medicine Complexity System, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yiyu Lu
- Research Center for Traditional Chinese Medicine Complexity System, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wanli Deng
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qi Li
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Shibing Su
- Research Center for Traditional Chinese Medicine Complexity System, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
68
|
Abstract
BACKGROUND Glioblastoma is the most common and aggressive type of primary brain tumor in adults. A key problem is the capacity of glioma cells to inactivate the body's immune response. The complement system acts as a functional bridge between the innate and adaptive immune response. Still, the role of the complement system has almost been forgotten in glioma research. In our present study, we hypothesize that C1 inactivator (C1-IA) is upregulated in astrocytoma grade IV, and that its inhibition of the complement system has beneficial effects upon survival. METHODS AND RESULTS We have explored this hypothesis both on gene and protein levels and found an upregulation of C1-IA in human glioblastoma cells using data from a publicly available database and our own mRNA material from glioblastoma patients. Furthermore, we demonstrated the presence of C1-IA by using immunohistochemistry on glioma cells from both humans and rats in vitro. Finally, we could demonstrate a significantly increased survival in vivo in animals inoculated intracerebrally with glioma cells pre-coated with C1-IA antibodies as compared to control animals. CONCLUSIONS Our findings indicate that overexpression of C1-IA is present in glioblastomas. This could be demonstrated both at the gene level from patients with glioblastoma, on mRNA level and with immunohistochemistry. Treatment with antibodies against C1-IA had beneficial effects on survival when tested in vivo.
Collapse
|
69
|
Berraondo P, Minute L, Ajona D, Corrales L, Melero I, Pio R. Innate immune mediators in cancer: between defense and resistance. Immunol Rev 2017; 274:290-306. [PMID: 27782320 DOI: 10.1111/imr.12464] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Chronic inflammation in the tumor microenvironment and evasion of the antitumor effector immune response are two of the emerging hallmarks required for oncogenesis and cancer progression. The innate immune system not only plays a critical role in perpetuating these tumor-promoting hallmarks but also in developing antitumor adaptive immune responses. Thus, understanding the dual role of the innate system in cancer immunology is required for the design of combined immunotherapy strategies able to tackle established tumors. Here, we review recent advances in the understanding of the role of cell populations and soluble components of the innate immune system in cancer, with a focus on complement, the adapter molecule Stimulator of Interferon Genes, natural killer cells, myeloid cells, and B cells.
Collapse
Affiliation(s)
- Pedro Berraondo
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Luna Minute
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Daniel Ajona
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Program of Solid Tumors and Biomarkers, CIMA, Pamplona, Spain.,Deparment of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | | | - Ignacio Melero
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Ruben Pio
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain. .,Program of Solid Tumors and Biomarkers, CIMA, Pamplona, Spain. .,Deparment of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain.
| |
Collapse
|
70
|
MALDI-TOF-MS analysis in discovery and identification of serum proteomic patterns of ovarian cancer. BMC Cancer 2017; 17:472. [PMID: 28683725 PMCID: PMC5501370 DOI: 10.1186/s12885-017-3467-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 06/30/2017] [Indexed: 12/11/2022] Open
Abstract
Background Due to high mortality and lack of efficient screening, new tools for ovarian cancer (OC) diagnosis are urgently needed. To broaden the knowledge on the pathological processes that occur during ovarian cancer tumorigenesis, protein-peptide profiling was proposed. Methods Serum proteomic patterns in samples from OC patients were obtained using matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF). Eighty nine serum samples (44 ovarian cancer and 45 healthy controls) were pretreated using solid-phase extraction method. Next, a classification model with the most discriminative factors was identified using chemometric algorithms. Finally, the results were verified by external validation on an independent test set of samples. Results Main outcome of this study was an identification of potential OC biomarkers by applying liquid chromatography coupled with tandem mass spectrometry. Application of this novel strategy enabled the identification of four potential OC serum biomarkers (complement C3, kininogen-1, inter-alpha-trypsin inhibitor heavy chain H4, and transthyretin). The role of these proteins was discussed in relation to OC pathomechanism. Conclusions The study results may contribute to the development of clinically useful multi-component diagnostic tools in OC. In addition, identifying a novel panel of discriminative proteins could provide a new insight into complex signaling and functional networks associated with this multifactorial disease. Electronic supplementary material The online version of this article (doi:10.1186/s12885-017-3467-2) contains supplementary material, which is available to authorized users.
Collapse
|
71
|
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
|
72
|
Saraswat M, Mäkitie A, Agarwal R, Joenväärä S, Renkonen S. Oral squamous cell carcinoma patients can be differentiated from healthy individuals with label-free serum proteomics. Br J Cancer 2017. [PMID: 28632724 PMCID: PMC5537490 DOI: 10.1038/bjc.2017.172] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background: No blood biomarkers to detect early oral cavity squamous cell carcinoma (OSCC) without clinical signs exist – diagnosis is solely based on histology of a visible tumour. Most OSCC patients are diagnosed at advanced stage, which leads to significant morbidity and poor survival. Our aim was to find the serum screening or detection biomarkers in OSCC. Methods: Serum samples from patients with OSCC treated at the Department of Otorhinolaryngology – Head and Neck Surgery, Helsinki University Hospital (Finland) were collected. Age- and gender-matched healthy individuals served as controls. Quantitative label-free proteomics in high definition MSE mode(HDMSE) was performed on 13 patients and 12 healthy samples. Various statistical analyses were performed on quantitative proteomics data to obtain the most influential proteins, which classify the patients vs healthy samples. Results: In quantitative proteomic analysis (HDMSE), 388 proteins were quantified in our pilot study. A complete separation between cases and controls was seen in supervised and unsupervised classification techniques such as orthogonal projections on latent structure-discriminant analysis (OPLS-DA) and self-organising maps. Using OPLS-DA S-plot, we identified a set of eight proteins that completely separated OSCC patients from healthy individuals. Conclusions: Although the tumour stages varied from I to IVa, these potential biomarkers were able to identify all OSCCs demonstrating their sensitivity to detect tumours of all stages. We are the first to suggest a set of serum biomarkers in our pilot study to be evaluated further as a diagnostic panel to detect preclinical OSCC in risk patients.
Collapse
Affiliation(s)
- Mayank Saraswat
- Transplantation Laboratory, University of Helsinki, Haartmaninkatu 3, PO Box 21, Helsinki FI-00014, Finland.,HUSLAB, Helsinki University Hospital, Helsinki 00290, Finland
| | - Antti Mäkitie
- Department of Otorhinolaryngology - Head and Neck Surgery, University of Helsinki and Helsinki University Hospital, Helsinki 00290, Finland
| | - Rahul Agarwal
- GenXPro GmbH, Altenhöferallee 3, Frankfurt am Main 60438, Germany
| | - Sakari Joenväärä
- Transplantation Laboratory, University of Helsinki, Haartmaninkatu 3, PO Box 21, Helsinki FI-00014, Finland.,HUSLAB, Helsinki University Hospital, Helsinki 00290, Finland
| | - Suvi Renkonen
- Department of Otorhinolaryngology - Head and Neck Surgery, University of Helsinki and Helsinki University Hospital, Helsinki 00290, Finland.,Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm 11382, Sweden
| |
Collapse
|
73
|
Okumura F, Joo-Okumura A, Nakatsukasa K, Kamura T. Hypoxia-inducible factor-2α stabilizes the von Hippel-Lindau (VHL) disease suppressor, Myb-related protein 2. PLoS One 2017; 12:e0175593. [PMID: 28394947 PMCID: PMC5386292 DOI: 10.1371/journal.pone.0175593] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 03/28/2017] [Indexed: 11/18/2022] Open
Abstract
Ubiquitin ligase von Hippel–Lindau tumor suppressor (pVHL) negatively regulates protein levels of hypoxia-inducible factor-α (HIF-α). Loss of pVHL causes HIF-α accumulation, which contributes to the pathogenesis of von Hippel-Lindau (VHL) disease. In contrast, v-Myb avian myeloblastosis viral oncogene homolog–like 2 (MYBL2; B-Myb), a transcription factor, prevents VHL pathogenesis by regulating gene expression of HIF-independent pathways. Both HIF-α and B-Myb are targets of pVHL-mediated polyubiquitination and proteasomal degradation. Here, we show that knockdown of HIF-2α induces downregulation of B-Myb in 786-O cells, which are deficient in pVHL, and this downregulation is prevented by proteasome inhibition. In the presence of pVHL and under hypoxia-like conditions, B-Myb and HIF-2α are both upregulated, and the upregulation of B-Myb requires expression of HIF-2α. We also show that HIF-2α and B-Myb interact in the nucleus, and this interaction is mediated by the central region of HIF-2α and the C-terminal region of B-Myb. These data indicate that oncogenic HIF-2α stabilizes B-Myb to suppress VHL pathogenesis.
Collapse
Affiliation(s)
- Fumihiko Okumura
- Division of Biological Science, Graduate School of Science, Nagoya University, Aichi, Japan
- * E-mail: (FO); (TK)
| | - Akiko Joo-Okumura
- Division of Biological Science, Graduate School of Science, Nagoya University, Aichi, Japan
| | - Kunio Nakatsukasa
- Division of Biological Science, Graduate School of Science, Nagoya University, Aichi, Japan
| | - Takumi Kamura
- Division of Biological Science, Graduate School of Science, Nagoya University, Aichi, Japan
- * E-mail: (FO); (TK)
| |
Collapse
|
74
|
A review of human diseases caused or exacerbated by aberrant complement activation. Neurobiol Aging 2017; 52:12-22. [DOI: 10.1016/j.neurobiolaging.2016.12.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 11/15/2016] [Accepted: 12/18/2016] [Indexed: 12/14/2022]
|
75
|
Ajona D, Ortiz-Espinosa S, Moreno H, Lozano T, Pajares MJ, Agorreta J, Bértolo C, Lasarte JJ, Vicent S, Hoehlig K, Vater A, Lecanda F, Montuenga LM, Pio R. A Combined PD-1/C5a Blockade Synergistically Protects against Lung Cancer Growth and Metastasis. Cancer Discov 2017; 7:694-703. [PMID: 28288993 DOI: 10.1158/2159-8290.cd-16-1184] [Citation(s) in RCA: 153] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 01/27/2017] [Accepted: 03/10/2017] [Indexed: 11/16/2022]
Abstract
Disruption of the programmed cell death protein 1 (PD-1) pathway with immune checkpoint inhibitors represents a major breakthrough in the treatment of non-small cell lung cancer. We hypothesized that combined inhibition of C5a/C5aR1 and PD-1 signaling may have a synergistic antitumor effect. The RMP1-14 antibody was used to block PD-1, and an L-aptamer was used to inhibit signaling of complement C5a with its receptors. Using syngeneic models of lung cancer, we demonstrate that the combination of C5a and PD-1 blockade markedly reduces tumor growth and metastasis and leads to prolonged survival. This effect is accompanied by a negative association between the frequency of CD8 T cells and myeloid-derived suppressor cells within tumors, which may result in a more complete reversal of CD8 T-cell exhaustion. Our study provides support for the clinical evaluation of anti-PD-1 and anti-C5a drugs as a novel combination therapeutic strategy for lung cancer.Significance: Using a variety of preclinical models of lung cancer, we demonstrate that the blockade of C5a results in a substantial improvement in the efficacy of anti-PD-1 antibodies against lung cancer growth and metastasis. This study provides the preclinical rationale for the combined blockade of PD-1/PD-L1 and C5a to restore antitumor immune responses, inhibit tumor cell growth, and improve outcomes of patients with lung cancer. Cancer Discov; 7(7); 694-703. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 653.
Collapse
Affiliation(s)
- Daniel Ajona
- University of Navarra, Center for Applied Medical Research (CIMA), Program in Solid Tumors and Biomarkers, Pamplona, Spain.,Navarra's Health Research Institute (IdiSNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,University of Navarra, School of Sciences, Department of Biochemistry and Genetics, Pamplona, Spain
| | - Sergio Ortiz-Espinosa
- University of Navarra, Center for Applied Medical Research (CIMA), Program in Solid Tumors and Biomarkers, Pamplona, Spain.,University of Navarra, School of Sciences, Department of Biochemistry and Genetics, Pamplona, Spain
| | - Haritz Moreno
- University of Navarra, Center for Applied Medical Research (CIMA), Program in Solid Tumors and Biomarkers, Pamplona, Spain.,Navarra's Health Research Institute (IdiSNA), Pamplona, Spain
| | - Teresa Lozano
- Navarra's Health Research Institute (IdiSNA), Pamplona, Spain.,University of Navarra, CIMA, Program in Immunology and Immunotherapy, Pamplona, Spain
| | - María J Pajares
- University of Navarra, Center for Applied Medical Research (CIMA), Program in Solid Tumors and Biomarkers, Pamplona, Spain.,Navarra's Health Research Institute (IdiSNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,University of Navarra, School of Medicine, Department of Histology and Pathology, Pamplona, Spain
| | - Jackeline Agorreta
- University of Navarra, Center for Applied Medical Research (CIMA), Program in Solid Tumors and Biomarkers, Pamplona, Spain.,Navarra's Health Research Institute (IdiSNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,University of Navarra, School of Medicine, Department of Histology and Pathology, Pamplona, Spain
| | - Cristina Bértolo
- University of Navarra, Center for Applied Medical Research (CIMA), Program in Solid Tumors and Biomarkers, Pamplona, Spain
| | - Juan J Lasarte
- Navarra's Health Research Institute (IdiSNA), Pamplona, Spain.,University of Navarra, CIMA, Program in Immunology and Immunotherapy, Pamplona, Spain
| | - Silvestre Vicent
- University of Navarra, Center for Applied Medical Research (CIMA), Program in Solid Tumors and Biomarkers, Pamplona, Spain.,Navarra's Health Research Institute (IdiSNA), Pamplona, Spain.,University of Navarra, School of Medicine, Department of Histology and Pathology, Pamplona, Spain
| | | | | | - Fernando Lecanda
- University of Navarra, Center for Applied Medical Research (CIMA), Program in Solid Tumors and Biomarkers, Pamplona, Spain.,Navarra's Health Research Institute (IdiSNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,University of Navarra, School of Medicine, Department of Histology and Pathology, Pamplona, Spain
| | - Luis M Montuenga
- University of Navarra, Center for Applied Medical Research (CIMA), Program in Solid Tumors and Biomarkers, Pamplona, Spain.,Navarra's Health Research Institute (IdiSNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,University of Navarra, School of Medicine, Department of Histology and Pathology, Pamplona, Spain
| | - Ruben Pio
- University of Navarra, Center for Applied Medical Research (CIMA), Program in Solid Tumors and Biomarkers, Pamplona, Spain. .,Navarra's Health Research Institute (IdiSNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,University of Navarra, School of Sciences, Department of Biochemistry and Genetics, Pamplona, Spain
| |
Collapse
|
76
|
Abstract
In addition to being a component of innate immunity and an ancient defense mechanism against invading pathogens, complement activation also participates in the adaptive immune response, inflammation, hemostasis, embryogenesis, and organ repair and development. Activation of the complement system via classical, lectin, or alternative pathways generates anaphylatoxins (C3a and C5a) and membrane attack complex (C5b-9) and opsonizes targeted cells. Complement activation end products and their receptors mediate cell-cell interactions that regulate several biological functions in the extravascular tissue. Signaling of anaphylatoxin receptors or assembly of membrane attack complex promotes cell dedifferentiation, proliferation, and migration in addition to reducing apoptosis. As a result, complement activation in the tumor microenvironment enhances tumor growth and increases metastasis. In this Review, I discuss immune and nonimmune functions of complement proteins and the tumor-promoting effect of complement activation.
Collapse
|
77
|
Markiewski MM, Vadrevu SK, Sharma SK, Chintala NK, Ghouse S, Cho JH, Fairlie DP, Paterson Y, Astrinidis A, Karbowniczek M. The Ribosomal Protein S19 Suppresses Antitumor Immune Responses via the Complement C5a Receptor 1. THE JOURNAL OF IMMUNOLOGY 2017; 198:2989-2999. [PMID: 28228558 DOI: 10.4049/jimmunol.1602057] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 01/27/2017] [Indexed: 01/01/2023]
Abstract
Relatively little is known about factors that initiate immunosuppression in tumors and act at the interface between tumor cells and host cells. In this article, we report novel immunosuppressive properties of the ribosomal protein S19 (RPS19), which is upregulated in human breast and ovarian cancer cells and released from apoptotic tumor cells, whereupon it interacts with the complement C5a receptor 1 expressed on tumor infiltrating myeloid-derived suppressor cells. This interaction promotes tumor growth by facilitating recruitment of these cells to tumors. RPS19 also induces the production of immunosuppressive cytokines, including TGF-β, by myeloid-derived suppressor cells in tumor-draining lymph nodes, leading to T cell responses skewed toward Th2 phenotypes. RPS19 promotes generation of regulatory T cells while reducing infiltration of CD8+ T cells into tumors. Reducing RPS19 in tumor cells or blocking the C5a receptor 1-RPS19 interaction decreases RPS19-mediated immunosuppression, impairs tumor growth, and delays the development of tumors in a transgenic model of breast cancer. This work provides initial preclinical evidence for targeting RPS19 for anticancer therapy enhancing antitumor T cell responses.
Collapse
Affiliation(s)
- Maciej M Markiewski
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Science Center, Abilene, TX 79601;
| | - Surya Kumari Vadrevu
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Science Center, Abilene, TX 79601
| | - Sharad K Sharma
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Science Center, Abilene, TX 79601
| | - Navin Kumar Chintala
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Science Center, Abilene, TX 79601
| | - Shanawaz Ghouse
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Science Center, Abilene, TX 79601
| | - Jun-Hung Cho
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Science Center, Abilene, TX 79601
| | - David P Fairlie
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, The University of Queensland, Brisbane, Queensland 4072, Australia; and
| | - Yvonne Paterson
- Department of Microbiology, School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Aristotelis Astrinidis
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Science Center, Abilene, TX 79601
| | - Magdalena Karbowniczek
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Science Center, Abilene, TX 79601;
| |
Collapse
|
78
|
Abdelbaset-Ismail A, Borkowska-Rzeszotek S, Kubis E, Bujko K, Brzeźniakiewicz-Janus K, Bolkun L, Kloczko J, Moniuszko M, Basak GW, Wiktor-Jedrzejczak W, Ratajczak MZ. Activation of the complement cascade enhances motility of leukemic cells by downregulating expression of HO-1. Leukemia 2017; 31:446-458. [PMID: 27451975 PMCID: PMC5288274 DOI: 10.1038/leu.2016.198] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 07/06/2016] [Accepted: 07/08/2016] [Indexed: 02/07/2023]
Abstract
As a crucial arm of innate immunity, the complement cascade (ComC) is involved both in mobilization of normal hematopoietic stem/progenitor cells (HSPCs) from bone marrow (BM) into peripheral blood and in their homing to BM. Despite the fact that ComC cleavage fragments alone do not chemoattract normal HSPCs, we found that leukemia cell lines as well as clonogenic blasts from chronic myeloid leukemia and acute myeloid leukemia patients respond robustly to C3 and C5 cleavage fragments by chemotaxis and increased adhesion. This finding was supported by the detection of C3a and C5a receptors in cells from human malignant hematopoietic cell lines and patient blasts at the mRNA (reverse transcriptase-polymerase chain reaction) and protein level (fluorescence-activated cell sorting), and by the demonstration that these receptors respond to stimulation by C3a and C5a by phosphorylation of p42/44 and p38 mitogen-activated protein kinases (MAPK), and protein kinase B (PKB/AKT). We also found that inducible heme oxygenase 1 (HO-1) is a negative regulator of ComC-mediated trafficking of leukemic cells, and that stimulation of leukemic cells by C3 or C5 cleavage fragments activates p38 MAPK, which downregulates HO-1 expression, rendering cells more mobile. We conclude that activation of the ComC in leukemia/lymphoma patients (for example, as a result of accompanying infections) enhances the motility of malignant cells and contributes to their spread in a p38 MAPK-HO-1-dependent manner. Therefore, inhibition of p38 MAPK or upregulation of HO-1 by small-molecule modulators would have a beneficial effect on ameliorating cell migration-mediated expansion of leukemia/lymphoma cells when the ComC becomes activated.
Collapse
Affiliation(s)
- A Abdelbaset-Ismail
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | | | - E Kubis
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - K Bujko
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | | | - L Bolkun
- Department of Regenerative Medicine, Medical University of Bialystok, Bialystok, Poland
- Department of Hematology, Medical University of Bialystok, Bialystok, Poland
| | - J Kloczko
- Department of Regenerative Medicine, Medical University of Bialystok, Bialystok, Poland
- Department of Hematology, Medical University of Bialystok, Bialystok, Poland
| | - M Moniuszko
- Department of Regenerative Medicine, Medical University of Bialystok, Bialystok, Poland
| | - G W Basak
- Department of Hematology, Warsaw Medical University, Warsaw, Poland
| | | | - M Z Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
- Department of Regenerative Medicine, Warsaw Medical University, Warsaw, Poland
| |
Collapse
|
79
|
Al-Rayahi IAM, Browning MJ, Stover C. Tumour cell conditioned medium reveals greater M2 skewing of macrophages in the absence of properdin. IMMUNITY INFLAMMATION AND DISEASE 2017; 5:68-77. [PMID: 28250926 PMCID: PMC5322164 DOI: 10.1002/iid3.142] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 09/14/2016] [Accepted: 10/28/2016] [Indexed: 11/29/2022]
Abstract
Introduction The tumour microenvironment is shaped by the interaction of immune, non immune, and tumour cells present in close proximity. Tumour cells direct the development of a locally immune suppressed state, affecting the activity of anti tumour T cells and preparing the escape phase of tumour development. Macrophages in the tumour typically develop into so‐called tumour associated macrophages with a distinct profile of activities which lead to a reduction in inflammation and antigen presentation. The direct impact of tumour cell conditioned medium on the activity profile of macrophages in dependence of their complement component expression has not yet been investigated. Methods In our in vitro study, macrophages differentiated from bone marrows of properdin deficient and wildtype mice were stimulated with conditioned medium of a syngeneic tumour cell line, B16F10, a mouse melanoma subline. Results In comparison with macrophages from wildtype mice, those from congenic properdin deficient mice showed skewing towards M2 profile, encompassing mRNA expression for genes involved in arginine metabolism, production of type 2 cytokines, and relatively lower surface expression of molecules needed for antigen presentation. Conclusions These data suggest that properdin insufficiency promotes a tumour environment that helps the tumour evade the immune response.
Collapse
Affiliation(s)
- Izzat A M Al-Rayahi
- Department of Infection, Immunity and InflammationUniversity of LeicesterLeicesterUK; Department of Medical Laboratory TechnologyCollege of Health and Medical TechnologyBaghdadIraq
| | - Michael J Browning
- Department of Infection, Immunity and InflammationUniversity of LeicesterLeicesterUK; Department of ImmunologyLeicester Royal InfirmaryLeicesterUK
| | - Cordula Stover
- Department of Infection, Immunity and Inflammation University of Leicester Leicester UK
| |
Collapse
|
80
|
Successful chemoimmunotherapy against hepatocellular cancer in a novel murine model. J Hepatol 2017; 66:75-85. [PMID: 27520877 PMCID: PMC5167655 DOI: 10.1016/j.jhep.2016.07.044] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 07/11/2016] [Accepted: 07/26/2016] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS We have established a clinically relevant animal model of hepatocellular cancer (HCC) in immune competent mice to elucidate the complex dialog between host immunity and tumors during HCC initiation and progression. Mechanistic findings have been leveraged to develop a clinically feasible anti-tumor chemoimmunotherapeutic strategy. METHODS Intraperitoneal injection of carbon tetrachloride and intrasplenic inoculation of oncogenic hepatocytes were combined to induce progressive HCCs in fibrotic livers of immunocompetent mice. Immunization and adoptive cell transfer (ACT) were used to dissect the tumor antigen-specific immune response. The ability of the tyrosine kinase inhibitor sunitinib to enhance immunotherapy in the setting of HCC was evaluated. RESULTS This new mouse model mimics human HCC and reflects its typical features. Tumor-antigen-specific CD8+ T cells maintained a naïve phenotype and remained responsive during early-stage tumor progression. Late tumor progression produced circulating tumor cells, tumor migration into draining lymph nodes, and profound exhaustion of tumor-antigen-specific CD8+ T cells associated with accumulation of programmed cell death protein 1 (PD-1)hi CD8+ T cells and regulatory T cells (Tregs). Sunitinib-mediated tumoricidal effect and Treg suppression synergized with antibody-mediated blockade of PD-1 to powerfully suppress tumor growth and activate anti-tumor immunity. CONCLUSION Treg accumulation and upregulation of PD-1 provide two independent mechanisms to induce profound immune tolerance in HCC. Chemoimmunotherapy using Food and Drug Administration-approved sunitinib with anti-PD-1 antibodies achieved significant tumor control, supporting translation of this approach for the treatment of HCC patients. LAY SUMMARY In the current study, we have established a clinically relevant mouse model which mimics human liver cancer. Using this unique model, we studied the response of the immune system to this aggressive cancer. Findings from this trial have led to the development of an innovative and clinically feasible chemoimmunotherapeutic strategy.
Collapse
|
81
|
Bandini S, Macagno M, Hysi A, Lanzardo S, Conti L, Bello A, Riccardo F, Ruiu R, Merighi IF, Forni G, Iezzi M, Quaglino E, Cavallo F. The non-inflammatory role of C1q during Her2/neu-driven mammary carcinogenesis. Oncoimmunology 2016; 5:e1253653. [PMID: 28123895 DOI: 10.1080/2162402x.2016.1253653] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 10/12/2016] [Accepted: 10/23/2016] [Indexed: 12/13/2022] Open
Abstract
There is an ever increasing amount of evidence to support the hypothesis that complement C1q, the first component of the classical complement pathway, is involved in the regulation of cancer growth, in addition to its role in fighting infections. It has been demonstrated that C1q is expressed in the microenvironment of various types of human tumors, including breast adenocarcinomas. This study compares carcinogenesis progression in C1q deficient (neuT-C1KO) and C1q competent neuT mice in order to investigate the role of C1q in mammary carcinogenesis. Significantly accelerated autochthonous neu+ carcinoma progression was paralleled by accelerated spontaneous lung metastases occurrence in C1q deficient mice. Surprisingly, this effect was not caused by differences in the tumor-infiltrating cells or in the activation of the complement classical pathway, since neuT-C1KO mice did not display a reduction in C3 fragment deposition at the tumor site. By contrast, a significant higher number of intratumor blood vessels and a decrease in the activation of the tumor suppressor WW domain containing oxidoreductase (WWOX) were observed in tumors from neuT-C1KO as compare with neuT mice. In parallel, an increase in Her2/neu expression was observed on the membrane of tumor cells. Taken together, our findings suggest that C1q plays a direct role both on halting tumor angiogenesis and on inducing apoptosis in mammary cancer cells by coordinating the signal transduction pathways linked to WWOX and, furthermore, highlight the role of C1q in mammary tumor immune surveillance regardless of complement system activation.
Collapse
Affiliation(s)
- Silvio Bandini
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino , Torino, Italy
| | - Marco Macagno
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino , Torino, Italy
| | - Albana Hysi
- Department of Medicine Science, Center of Excellence on Aging and Translational Medicine (CeSI-Met), G. d'Annunzio University of Chieti Pescara , Italy
| | - Stefania Lanzardo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino , Torino, Italy
| | - Laura Conti
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino , Torino, Italy
| | - Amanda Bello
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino , Torino, Italy
| | - Federica Riccardo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino , Torino, Italy
| | - Roberto Ruiu
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino , Torino, Italy
| | - Irene Fiore Merighi
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino , Torino, Italy
| | - Guido Forni
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino , Torino, Italy
| | - Manuela Iezzi
- Department of Medicine Science, Center of Excellence on Aging and Translational Medicine (CeSI-Met), G. d'Annunzio University of Chieti Pescara , Italy
| | - Elena Quaglino
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino , Torino, Italy
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino , Torino, Italy
| |
Collapse
|
82
|
Agatea L, Crotti S, Ragazzi E, Bedin C, Urso E, Mammi I, Traldi P, Pucciarelli S, Nitti D, Agostini M. Peptide Patterns as Discriminating Biomarkers in Plasma of Patients With Familial Adenomatous Polyposis. Clin Colorectal Cancer 2016; 15:e75-92. [DOI: 10.1016/j.clcc.2015.12.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 12/04/2015] [Accepted: 12/09/2015] [Indexed: 11/28/2022]
|
83
|
Chen J, Yang WJ, Sun HJ, Yang X, Wu YZ. C5b-9 Staining Correlates With Clinical and Tumor Stage in Gastric Adenocarcinoma. Appl Immunohistochem Mol Morphol 2016; 24:470-5. [PMID: 26186252 PMCID: PMC4979623 DOI: 10.1097/pai.0000000000000218] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 04/10/2015] [Indexed: 01/26/2023]
Abstract
The complement system is a critical part of the immune response, acting in defense against viral infections, clearance of immune complexes, and maintenance of tissue homeostasis. Upregulated expression of the terminal complement complex, C5b-9, has been observed on various tumor cells, such as stomach carcinoma cells, and on cells in the necrotic regions of these tumors as well; however, whether and how C5b-9 is related to gastric cancer progression and severity remains unknown. In this study, human gastric adenocarcinoma (HGAC) tissues (n=47 cases) and patient-matched adjacent nontumoral parenchyma (n=20 cases) were evaluated by tissue microarray and immunohistochemistry. The HGAC tissues showed upregulated C5b-9 expression. Multinomial logistic regression and likelihood ratio testing showed that overexpression of C5b-9 in HGAC tissue was significantly correlated with clinical stage (P=0.007) and tumor stage (P=0.005), but not with tumor distant organ metastasis, lymphoid nodal status, sex, or age. Patients with late-stage gastric adenocarcinoma had a higher amount of tumor cells showing positive staining for C5b-9 than patients with early-stage disease. These results may help in diagnosis and assessment of disease severity of human gastric carcinoma.
Collapse
Affiliation(s)
- Jian Chen
- Institute of Immunology, Third Military Medical University, Chongqing
| | - Wei-jun Yang
- Department of General Surgery, First People’s Hospital of Guiyang, Guiyang, P. R. China
| | - Hai-jian Sun
- Institute of Immunology, Third Military Medical University, Chongqing
| | - Xia Yang
- Institute of Immunology, Third Military Medical University, Chongqing
| | - Yu-zhang Wu
- Institute of Immunology, Third Military Medical University, Chongqing
| |
Collapse
|
84
|
Ricklin D, Lambris JD. New milestones ahead in complement-targeted therapy. Semin Immunol 2016; 28:208-22. [PMID: 27321574 PMCID: PMC5404743 DOI: 10.1016/j.smim.2016.06.001] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 05/26/2016] [Accepted: 06/01/2016] [Indexed: 02/08/2023]
Abstract
The complement system is a powerful effector arm of innate immunity that typically confers protection from microbial intruders and accumulating debris. In many clinical situations, however, the defensive functions of complement can turn against host cells and induce or exacerbate immune, inflammatory, and degenerative conditions. Although the value of inhibiting complement in a therapeutic context has long been recognized, bringing complement-targeted drugs into clinical use has proved challenging. This important milestone was finally reached a decade ago, yet the clinical availability of complement inhibitors has remained limited. Still, the positive long-term experience with complement drugs and their proven effectiveness in various diseases has reinvigorated interest and confidence in this approach. Indeed, a broad variety of clinical candidates that act at almost any level of the complement activation cascade are currently in clinical development, with several of them being evaluated in phase 2 and phase 3 trials. With antibody-related drugs dominating the panel of clinical candidates, the emergence of novel small-molecule, peptide, protein, and oligonucleotide-based inhibitors offers new options for drug targeting and administration. Whereas all the currently approved and many of the proposed indications for complement-targeted inhibitors belong to the rare disease spectrum, these drugs are increasingly being evaluated for more prevalent conditions. Fortunately, the growing experience from preclinical and clinical use of therapeutic complement inhibitors has enabled a more evidence-based assessment of suitable targets and rewarding indications as well as related technical and safety considerations. This review highlights recent concepts and developments in complement-targeted drug discovery, provides an overview of current and emerging treatment options, and discusses the new milestones ahead on the way to the next generation of clinically available complement therapeutics.
Collapse
Affiliation(s)
- Daniel Ricklin
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, USA.
| | - John D Lambris
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, USA.
| |
Collapse
|
85
|
Parallel Regulation of von Hippel-Lindau Disease by pVHL-Mediated Degradation of B-Myb and Hypoxia-Inducible Factor α. Mol Cell Biol 2016; 36:1803-17. [PMID: 27090638 DOI: 10.1128/mcb.00067-16] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 04/11/2016] [Indexed: 11/20/2022] Open
Abstract
pVHL, the protein product of the von Hippel-Lindau (VHL) tumor suppressor gene, is a ubiquitin ligase that targets hypoxia-inducible factor α (HIF-α) for proteasomal degradation. Although HIF-α activation is necessary for VHL disease pathogenesis, constitutive activation of HIF-α alone did not induce renal clear cell carcinomas and pheochromocytomas in mice, suggesting the involvement of an HIF-α-independent pathway in VHL pathogenesis. Here, we show that the transcription factor B-Myb is a pVHL substrate that is degraded via the ubiquitin-proteasome pathway and that vascular endothelial growth factor (VEGF)- and/or platelet-derived growth factor (PDGF)-dependent tyrosine 15 phosphorylation of B-Myb prevents its degradation. Mice injected with B-Myb knockdown 786-O cells developed dramatically larger tumors than those bearing control cell tumors. Microarray screening of B-Myb-regulated genes showed that the expression of HIF-α-dependent genes was not affected by B-Myb knockdown, indicating that B-Myb prevents HIF-α-dependent tumorigenesis through an HIF-α-independent pathway. These data indicate that the regulation of B-Myb by pVHL plays a critical role in VHL disease.
Collapse
|
86
|
Lynam-Lennon N, Bibby BA, Mongan AM, Marignol L, Paxton CN, Geiersbach K, Bronner MP, O'Sullivan J, Reynolds J, Maher SG. Low miR-187 expression promotes resistance to chemoradiation therapy in vitro and correlates with treatment failure in patients with esophageal adenocarcinoma. Mol Med 2016; 22:molmed.2016.00020. [PMID: 27254108 DOI: 10.2119/molmed.2016.00020] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 05/11/2016] [Indexed: 12/13/2022] Open
Abstract
Esophageal adenocarcinoma (EAC) has a poor prognosis and is increasing in incidence in many western populations. Neoadjuvant chemoradiation therapy (CRT) followed by surgery is increasingly the standard of care for locally advanced EAC; however, resistance to treatment is a significant clinical problem. The identification of both novel biomarkers predicting response to treatment and novel therapeutic targets to enhance the efficacy of CRT are key to improving survival rates in EAC. In this study we performed global microRNA (miRNA) profiling of pre-treatment EAC biopsies and identified 67 miRNA significantly altered in patients who are resistant to CRT. One of these miRNA, miR-187, was significantly decreased in pre-treatment EAC tumors from patients having a poor response to neoadjuvant CRT, highlighting downregulation of miR-187 as a potential mechanism of treatment resistance in EAC. In vitro, miR-187 was demonstrated to play a functional role in modulating sensitivity to X-ray radiation and cisplatin in EAC and its dysregulation was demonstrated to be due to chromosomal alterations. In vitro, miR-187 altered expression of a diverse array of pathways, including the immune regulator complement component 3 (C3), serum levels of which we have previously demonstrated to predict patient response to CRT. In vivo, expression of C3 was significantly increased in tumors from patients having a poor response to CRT. This study highlights for the first time a role for miR-187 as a novel biomarker of response to CRT and a potential therapeutic target for enhancing the efficacy of CRT in EAC.
Collapse
Affiliation(s)
| | - Becky A Bibby
- University of Hull, Hull, Kingston upon Hull, United Kingdom of Great Britain and Northern Ireland
| | | | | | | | - Katherine Geiersbach
- University of Utah and ARUP Laboratories, United States.,Arup Institute for Clinical and Experimental Pathology, United States
| | - Mary P Bronner
- University of Utah and ARUP Laboratories, United States.,Arup Institute for Clinical and Experimental Pathology, United States
| | | | - John Reynolds
- University of Dublin Trinity College, Dublin, Ireland
| | - Stephen G Maher
- University of Dublin Trinity College, Dublin, Ireland.,University of Hull, Hull, Kingston upon Hull, United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
87
|
Poersch A, Grassi ML, Carvalho VPD, Lanfredi GP, Palma CDS, Greene LJ, de Sousa CB, Carrara HHA, Candido Dos Reis FJ, Faça VM. A proteomic signature of ovarian cancer tumor fluid identified by highthroughput and verified by targeted proteomics. J Proteomics 2016; 145:226-236. [PMID: 27222041 DOI: 10.1016/j.jprot.2016.05.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 04/27/2016] [Accepted: 05/04/2016] [Indexed: 10/21/2022]
Abstract
UNLABELLED Tumor fluid samples have emerged as a rich source for the identification of ovarian cancer in the context of proteomics studies. To uncover differences among benign and malignant ovarian samples, we performed a quantitative proteomic study consisting of albumin immunodepletion, isotope labeling with acrylamide and in-depth proteomic profiling by LC-MS/MS in a pool of 10 samples of each histological type. 1135 proteins were identified, corresponding to 505 gene products. 223 proteins presented associated quantification and the comparative analysis of histological types revealed 75 differentially abundant proteins. Based on this, we developed a panel for targeted proteomic analysis using the multiple reaction monitoring (MRM) method for validation of 51 proteins in individual samples of high-grade serous ovarian tumor fluids (malignant) and benign serous cystadenoma tumor fluids. This analysis showed concordant results in terms of average amounts of proteins, and APOE, SERPINF2, SERPING1, ADAM17, CD44 and OVGP1 were statistically significant between benign and malignant group. The results observed in the MRM for APOE were confirmed by western blotting, where APOE was more abundant in malignant samples. This molecular signature can contribute to improve tumor stratification and shall be investigated in combination with current biomarkers in larger cohorts to improve ovarian cancer diagnosis. BIOLOGICAL SIGNIFICANCE Despite advances in cancer research, ovarian cancer has a high mortality and remains a major challenge due to a number of particularities of the disease, especially late diagnosis caused by vague clinical symptoms, the cellular and molecular heterogeneity of tumors, and the lack of effective treatment. Thus, efforts are directed to better understand this neoplasia, its origin, development and, particularly the identification and validation of biomarkers for early detection of the disease in asymptomatic stage. In the present work, we confirmed by MRM method in individual ovarian tumor fluid samples the regulation of 27 proteins out of 33 identified in a highthroughput study. We speculate that the presence and/or differential abundance observed in tumor fluid is a cooperation primarily of high rates of secretion of such tumor proteins to extra tumor environment that will at the end accumulate in plasma, and also the accumulation of acute-phase proteins throughout the entire body. On top of that, consideration of physiological influences in the interpretation of expression observed, including age, menopause status, route-of-elimination kinetics and metabolism of the tumor marker, coexisting disease, hormonal imbalances, life-style influences (smoking, alcoholism, obesity), among others, are mandatory to enable the selection of good protein tumor marker candidates for extensive validation.
Collapse
Affiliation(s)
- Aline Poersch
- Department of Biochemistry and Immunology, FMRP,University of São Paulo, Av. Bandeirantes, 3900, 14040-900 Ribeirão Preto, SP, Brazil; Department of Gynecology and Obstetrics, FMRP, University of São Paulo, Av. Bandeirantes, 3900, 14040-900 Ribeirão Preto, SP, Brazil; Center for Cell Based Therapy, Hemotherapy Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, 14051-140, Ribeirão Preto, SP, Brazil.
| | - Mariana Lopes Grassi
- Department of Biochemistry and Immunology, FMRP,University of São Paulo, Av. Bandeirantes, 3900, 14040-900 Ribeirão Preto, SP, Brazil; Center for Cell Based Therapy, Hemotherapy Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, 14051-140, Ribeirão Preto, SP, Brazil
| | - Vinícius Pereira de Carvalho
- Department of Biochemistry and Immunology, FMRP,University of São Paulo, Av. Bandeirantes, 3900, 14040-900 Ribeirão Preto, SP, Brazil; Medical School of Barão de Mauá University, Rua Ramos de Azevedo, 423, 14090-180 Ribeirão Preto, SP, Brazil
| | - Guilherme Pauperio Lanfredi
- Department of Biochemistry and Immunology, FMRP,University of São Paulo, Av. Bandeirantes, 3900, 14040-900 Ribeirão Preto, SP, Brazil
| | - Camila de Souza Palma
- Department of Biochemistry and Immunology, FMRP,University of São Paulo, Av. Bandeirantes, 3900, 14040-900 Ribeirão Preto, SP, Brazil; Center for Cell Based Therapy, Hemotherapy Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, 14051-140, Ribeirão Preto, SP, Brazil
| | - Lewis Joel Greene
- Center for Cell Based Therapy, Hemotherapy Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, 14051-140, Ribeirão Preto, SP, Brazil; Department of Cell and Molecular Biology and Pathogenic Bioagents, FMRP, University of São Paulo, Av. Bandeirantes, 3900, 14040-900 Ribeirão Preto, SP, Brazil
| | - Christiani Bisinoto de Sousa
- Department of Gynecology and Obstetrics, FMRP, University of São Paulo, Av. Bandeirantes, 3900, 14040-900 Ribeirão Preto, SP, Brazil
| | - Hélio Humberto Angotti Carrara
- Department of Gynecology and Obstetrics, FMRP, University of São Paulo, Av. Bandeirantes, 3900, 14040-900 Ribeirão Preto, SP, Brazil
| | - Francisco José Candido Dos Reis
- Department of Gynecology and Obstetrics, FMRP, University of São Paulo, Av. Bandeirantes, 3900, 14040-900 Ribeirão Preto, SP, Brazil
| | - Vitor Marcel Faça
- Department of Biochemistry and Immunology, FMRP,University of São Paulo, Av. Bandeirantes, 3900, 14040-900 Ribeirão Preto, SP, Brazil; Center for Cell Based Therapy, Hemotherapy Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, 14051-140, Ribeirão Preto, SP, Brazil
| |
Collapse
|
88
|
Towner LD, Wheat RA, Hughes TR, Morgan BP. Complement Membrane Attack and Tumorigenesis: A SYSTEMS BIOLOGY APPROACH. J Biol Chem 2016; 291:14927-38. [PMID: 27226542 DOI: 10.1074/jbc.m115.708446] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Indexed: 12/21/2022] Open
Abstract
Tumor development driven by inflammation is now an established phenomenon, but the role that complement plays remains uncertain. Recent evidence has suggested that various components of the complement (C) cascade may influence tumor development in disparate ways; however, little attention has been paid to that of the membrane attack complex (MAC). This is despite abundant evidence documenting the effects of this complex on cell behavior, including cell activation, protection from/induction of apoptosis, release of inflammatory cytokines, growth factors, and ECM components and regulators, and the triggering of the NLRP3 inflammasome. Here we present a novel approach to this issue by using global gene expression studies in conjunction with a systems biology analysis. Using network analysis of MAC-responsive expression changes, we demonstrate a cluster of co-regulated genes known to have impact in the extracellular space and on the supporting stroma and with well characterized tumor-promoting roles. Network analysis highlighted the central role for EGF receptor activation in mediating the observed responses to MAC exposure. Overall, the study sheds light on the mechanisms by which sublytic MAC causes tumor cell responses and exposes a gene expression signature that implicates MAC as a driver of tumor progression. These findings have implications for understanding of the roles of complement and the MAC in tumor development and progression, which in turn will inform future therapeutic strategies in cancer.
Collapse
Affiliation(s)
- Laurence D Towner
- From the Complement Biology Group, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff CF14 4XN, Wales, United Kingdom
| | - Richard A Wheat
- From the Complement Biology Group, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff CF14 4XN, Wales, United Kingdom
| | - Timothy R Hughes
- From the Complement Biology Group, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff CF14 4XN, Wales, United Kingdom
| | - B Paul Morgan
- From the Complement Biology Group, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff CF14 4XN, Wales, United Kingdom
| |
Collapse
|
89
|
Menietti E, Xu X, Ostano P, Joseph JM, Lefort K, Dotto GP. Negative control of CSL gene transcription by stress/DNA damage response and p53. Cell Cycle 2016; 15:1767-78. [PMID: 27163456 DOI: 10.1080/15384101.2016.1186317] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
CSL is a key transcriptional repressor and mediator of Notch signaling. Despite wide interest in CSL, mechanisms responsible for its own regulation are little studied. CSL down-modulation in human dermal fibroblasts (HDFs) leads to conversion into cancer associated fibroblasts (CAF), promoting keratinocyte tumors. We show here that CSL transcript levels differ among HDF strains from different individuals, with negative correlation with genes involved in DNA damage/repair. CSL expression is negatively regulated by stress/DNA damage caused by UVA, Reactive Oxygen Species (ROS), smoke extract, and doxorubicin treatment. P53, a key effector of the DNA damage response, negatively controls CSL gene transcription, through suppression of CSL promoter activity and, indirectly, by increased p21 expression. CSL was previously shown to bind p53 suppressing its activity. The present findings indicate that p53, in turn, decreases CSL expression, which can serve to enhance p53 activity in acute DNA damage response of cells.
Collapse
Affiliation(s)
- Elena Menietti
- a Department of Biochemistry , University of Lausanne , Epalinges , Switzerland
| | - Xiaoying Xu
- a Department of Biochemistry , University of Lausanne , Epalinges , Switzerland
| | - Paola Ostano
- b Cancer Genomics Laboratory, Edo and Elvo Tempia Valenta Foundation , Biella , Italy
| | - Jean-Marc Joseph
- c Pediatric surgery Department , University Hospital CHUV , Lausanne , Switzerland
| | - Karine Lefort
- a Department of Biochemistry , University of Lausanne , Epalinges , Switzerland.,d Department of Dermatology , University Hospital CHUV , Lausanne , Switzerland
| | - G Paolo Dotto
- a Department of Biochemistry , University of Lausanne , Epalinges , Switzerland.,e Cutaneous Biology Research Center, Massachusetts General Hospital , Charlestown , MA , USA
| |
Collapse
|
90
|
Cimmino F, Avitabile M, Pezone L, Scalia G, Montanaro D, Andreozzi M, Terracciano L, Iolascon A, Capasso M. CD55 is a HIF-2α marker with anti-adhesive and pro-invading properties in neuroblastoma. Oncogenesis 2016; 5:e212. [PMID: 27043658 PMCID: PMC4848835 DOI: 10.1038/oncsis.2016.20] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 12/31/2015] [Accepted: 01/14/2016] [Indexed: 02/08/2023] Open
Abstract
CD55 has been revealed to have an important role in tumor genesis, and presence of small populations of cells with strong CD55 expression would be sufficient to predict poor prognosis of several tumors. In our study we revealed that CD55 is a novel target of hypoxia-inducible factor HIF-2α in neuroblastoma (NB) cells. We show that HIF-2α expression is sufficient to sustain stem-like features of NB cells, whereas CD55 protein upon HIF-2α expression contributes to growth of colonies and to invasion of cells, but not to stemness features. Interestingly, in NB tissues, CD55 expression is limited to quite a small population of cells that are HIF-2α positive, and the gene expression of CD55 in the NB data set reveals that the presence of CD55(high) affects prognosis of NB patients. The functional characterization of CD55-positive populations within heterogeneous NB monoclonal cell lines shows that CD55 has pro-invading and anti-adhesive properties that might provide the basis for the ability of solid tumors to survive as microscopic residual disease. The easy accessibility to CD55 membrane antigen will offer the possibility of a novel antibody approach in the treatment of recurrent tumors and will provide a ready target for antibody-based visualization in NB diagnosis and prognosis.
Collapse
Affiliation(s)
- F Cimmino
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli ‘Federico II', Naples, Italy
- CEINGE Biotecnologie Avanzate, Naples, Italy
| | - M Avitabile
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli ‘Federico II', Naples, Italy
- CEINGE Biotecnologie Avanzate, Naples, Italy
| | - L Pezone
- CEINGE Biotecnologie Avanzate, Naples, Italy
- Dipartimento di Medicina, Scuola di Medicina e Chirurgia, Università degli Studi di Verona, Verona, Italy
| | - G Scalia
- CEINGE Biotecnologie Avanzate, Naples, Italy
| | - D Montanaro
- CEINGE Biotecnologie Avanzate, Naples, Italy
| | - M Andreozzi
- Institute of Pathology, University of Basel, Basel, Switzerland
| | - L Terracciano
- Institute of Pathology, University of Basel, Basel, Switzerland
| | - A Iolascon
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli ‘Federico II', Naples, Italy
- CEINGE Biotecnologie Avanzate, Naples, Italy
| | - M Capasso
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli ‘Federico II', Naples, Italy
- CEINGE Biotecnologie Avanzate, Naples, Italy
| |
Collapse
|
91
|
Identification of Serum Peptidome Signatures of Non-Small Cell Lung Cancer. Int J Mol Sci 2016; 17:410. [PMID: 27043541 PMCID: PMC4848884 DOI: 10.3390/ijms17040410] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 03/07/2016] [Accepted: 03/14/2016] [Indexed: 12/26/2022] Open
Abstract
Due to high mortality rates of lung cancer, there is a need for identification of new, clinically useful markers, which improve detection of this tumor in early stage of disease. In the current study, serum peptide profiling was evaluated as a diagnostic tool for non-small cell lung cancer patients. The combination of the ZipTip technology with matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF-MS) for the analysis of peptide pattern of cancer patients (n = 153) and control subjects (n = 63) was presented for the first time. Based on the observed significant differences between cancer patients and control subjects, the classification model was created, which allowed for accurate group discrimination. The model turned out to be robust enough to discriminate a new validation set of samples with satisfactory sensitivity and specificity. Two peptides from the diagnostic pattern for non-small cell lung cancer (NSCLC) were identified as fragments of C3 and fibrinogen α chain. Since ELISA test did not confirm significant differences in the expression of complement component C3, further study will involve a quantitative approach to prove clinical utility of the other proteins from the proposed multi-peptide cancer signature.
Collapse
|
92
|
Scherzer MT, Waigel S, Donninger H, Arumugam V, Zacharias W, Clark G, Siskind LJ, Soucy P, Beverly L. Fibroblast-Derived Extracellular Matrices: An Alternative Cell Culture System That Increases Metastatic Cellular Properties. PLoS One 2015; 10:e0138065. [PMID: 26371754 PMCID: PMC4570771 DOI: 10.1371/journal.pone.0138065] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 08/26/2015] [Indexed: 12/31/2022] Open
Abstract
Poor survival rates from lung cancer can largely be attributed to metastatic cells that invade and spread throughout the body. The tumor microenvironment (TME) is composed of multiple cell types, as well as non-cellular components. The TME plays a critical role in the development of metastatic cancers by providing migratory cues and changing the properties of the tumor cells. The Extracellular Matrix (ECM), a main component of the TME, has been shown to change composition during tumor progression, contributing to cancer cell invasion and survival away from the primary cancer site. Although the ECM is well-known to influence the fate of tumor progression, little is known about the molecular mechanisms that are affected by the cancer cell-ECM interactions. It is imperative that these mechanisms are elucidated in order to properly understand and prevent lung cancer dissemination. However, common in vitro studies do not incorporate these interactions into everyday cell culture assays. We have adopted a model that examines decellularized human fibroblast-derived ECM as a 3-dimensional substrate for growth of lung adenocarcinoma cell lines. Here, we have characterized the effect of fibroblast-derived matrices on the properties of various lung-derived epithelial cell lines, including cancerous and non-transformed cells. This work highlights the significance of the cell-ECM interaction and its requirement for incorporation into in vitro experiments. Implementation of a fibroblast-derived ECM as an in vitro technique will provide researchers with an important factor to manipulate to better recreate and study the TME.
Collapse
Affiliation(s)
- Michael T. Scherzer
- J. G. Brown Cancer Center, University of Louisville, Louisville, Kentucky, 40202, United States of America
- Department of Bioengineering, University of Louisville, Louisville, Kentucky, 40202, United States of America
| | - Sabine Waigel
- J. G. Brown Cancer Center, University of Louisville, Louisville, Kentucky, 40202, United States of America
| | - Howard Donninger
- J. G. Brown Cancer Center, University of Louisville, Louisville, Kentucky, 40202, United States of America
- Department of Medicine, University of Louisville, Louisville, Kentucky, 40202, United States of America
| | - Vennila Arumugam
- J. G. Brown Cancer Center, University of Louisville, Louisville, Kentucky, 40202, United States of America
| | - Wolfgang Zacharias
- J. G. Brown Cancer Center, University of Louisville, Louisville, Kentucky, 40202, United States of America
- Department of Medicine, University of Louisville, Louisville, Kentucky, 40202, United States of America
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, 40202, United States of America
| | - Geoffrey Clark
- J. G. Brown Cancer Center, University of Louisville, Louisville, Kentucky, 40202, United States of America
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, 40202, United States of America
| | - Leah J. Siskind
- J. G. Brown Cancer Center, University of Louisville, Louisville, Kentucky, 40202, United States of America
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, 40202, United States of America
| | - Patricia Soucy
- Department of Bioengineering, University of Louisville, Louisville, Kentucky, 40202, United States of America
| | - Levi Beverly
- J. G. Brown Cancer Center, University of Louisville, Louisville, Kentucky, 40202, United States of America
- Department of Bioengineering, University of Louisville, Louisville, Kentucky, 40202, United States of America
- Department of Medicine, University of Louisville, Louisville, Kentucky, 40202, United States of America
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, 40202, United States of America
- * E-mail:
| |
Collapse
|
93
|
Khan MA, Assiri AM, Broering DC. Complement and macrophage crosstalk during process of angiogenesis in tumor progression. J Biomed Sci 2015; 22:58. [PMID: 26198107 PMCID: PMC4511526 DOI: 10.1186/s12929-015-0151-1] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 05/25/2015] [Indexed: 12/27/2022] Open
Abstract
The complement system, which contains some of the most potent pro-inflammatory mediators in the tissue including the anaphylatoxins C3a and C5a are the vital parts of innate immunity. Complement activation seems to play a more critical role in tumor development, but little attention has been given to the angiogenic balance of the activated complement mediators and macrophage polarization during tumor progression. The tumor growth mainly supported by the infiltration of M2- tumor-associated macrophages, and high levels of C3a and C5a, whereas M1-macrophages contribute to immune-mediated tumor suppression. Macrophages express a cognate receptors for both C3a and C5a on their cell surface, and specific binding of C3a and C5a affects the functional modulation and angiogenic properties. Activation of complement mediators induce angiogenesis, favors an immunosuppressive microenvironment, and activate cancer-associated signaling pathways to assist chronic inflammation. In this review manuscript, we highlighted the specific roles of complement activation and macrophage polarization during uncontrolled angiogenesis in tumor progression, and therefore blocking of complement mediators would be an alternative therapeutic option for treating cancer.
Collapse
Affiliation(s)
- M Afzal Khan
- Department Comparative Medicine, King Faisal Specialist Hospital and Research Centre, MBC 03, P.O. Box 3354, Riyadh, 11211, Kingdom of Saudi Arabia.
| | - A M Assiri
- Department Comparative Medicine, King Faisal Specialist Hospital and Research Centre, MBC 03, P.O. Box 3354, Riyadh, 11211, Kingdom of Saudi Arabia
| | - D C Broering
- Organ Transplant Centre, King Faisal Specialist Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
94
|
Hwang HJ, Sohn KY, Han YH, Chong S, Yoon SY, Kim YJ, Jeong J, Kim SH, Kim JW. Effect of 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol on Immune Functions in Healthy Adults in a Randomized Controlled Trial. Immune Netw 2015; 15:150-60. [PMID: 26140047 PMCID: PMC4486778 DOI: 10.4110/in.2015.15.3.150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 06/01/2015] [Accepted: 06/08/2015] [Indexed: 12/01/2022] Open
Abstract
We previously reported that 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) accelerates hematopoiesis and has an improving effect on animal disease models such as sepsis and asthma. The effects of PLAG supplementation on immune modulation were assessed in healthy men and women. The objective was to evaluate the effects of PLAG supplementation on immune regulatory functions such as activities of immune cells and cytokine production. A randomized double blind placebo-controlled trial was conducted. Seventy-five participants were assigned to one of two groups; all participants had an appropriate number of white blood cells on the testing day. The PLAG group (n=27) received oral PLAG supplements and the control group (n=22) received oral soybean oil supplements. IL-4 and IL-6 production by peripheral blood mononuclear cells (PBMC) were lower (p<0.001 and p<0.001, respectively) with PLAG than with soybean oil. However, the production of IL-2 and IFN-γ by PBMC was unaltered with PLAG supplementation. The B cell proliferation decreased significantly in the PLAG group compared to the soybean oil control (p<0.05). The intake of PLAG in healthy adults for 4 weeks was deemed safe. These data suggest that PLAG has an immunomodulatory function that inhibits the excessive immune activity of immunological disorders such as atopic and autoimmune diseases. PLAG could improve the condition of these diseases safely as a health food supplement.
Collapse
Affiliation(s)
- Hee-Jin Hwang
- Department of Family Medicine, International St. Mary's Hospital, Catholic Kwandong University College of Medicine, Incheon 404-834, Korea
| | | | | | - Saeho Chong
- ENZYCHEM Lifesciences, Daejeon 305-732, Korea
| | | | - Young-Jun Kim
- Biomedical Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Korea
| | - Jinseoun Jeong
- Biomedical Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Korea
| | - Sang-Hwan Kim
- Institute for Geriatric Medicine, Yonsei Woori Geriatric Hospital, Goyang 412-802, Korea
| | - Jae Wha Kim
- Biomedical Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Korea
| |
Collapse
|
95
|
Evgin L, Acuna SA, Tanese de Souza C, Marguerie M, Lemay CG, Ilkow CS, Findlay CS, Falls T, Parato KA, Hanwell D, Goldstein A, Lopez R, Lafrance S, Breitbach CJ, Kirn D, Atkins H, Auer RC, Thurman JM, Stahl GL, Lambris JD, Bell JC, McCart JA. Complement inhibition prevents oncolytic vaccinia virus neutralization in immune humans and cynomolgus macaques. Mol Ther 2015; 23:1066-1076. [PMID: 25807289 DOI: 10.1038/mt.2015.49] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 03/16/2015] [Indexed: 02/07/2023] Open
Abstract
Oncolytic viruses (OVs) have shown promising clinical activity when administered by direct intratumoral injection. However, natural barriers in the blood, including antibodies and complement, are likely to limit the ability to repeatedly administer OVs by the intravenous route. We demonstrate here that for a prototype of the clinical vaccinia virus based product Pexa-Vec, the neutralizing activity of antibodies elicited by smallpox vaccination, as well as the anamnestic response in hyperimmune virus treated cancer patients, is strictly dependent on the activation of complement. In immunized rats, complement depletion stabilized vaccinia virus in the blood and led to improved delivery to tumors. Complement depletion also enhanced tumor infection when virus was directly injected into tumors in immunized animals. The feasibility and safety of using a complement inhibitor, CP40, in combination with vaccinia virus was tested in cynomolgus macaques. CP40 pretreatment elicited an average 10-fold increase in infectious titer in the blood early after the infusion and prolonged the time during which infectious virus was detectable in the blood of animals with preexisting immunity. Capitalizing on the complement dependence of antivaccinia antibody with adjunct complement inhibitors may increase the infectious dose of oncolytic vaccinia virus delivered to tumors in virus in immune hosts.
Collapse
Affiliation(s)
- Laura Evgin
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Sergio A Acuna
- Division of Experimental Therapeutics, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | | | - Monique Marguerie
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Chantal G Lemay
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Carolina S Ilkow
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - C Scott Findlay
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Theresa Falls
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Kelley A Parato
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - David Hanwell
- Animal Resources Centre, University Health Network, Toronto, Ontario, Canada
| | - Alyssa Goldstein
- Animal Resources Centre, University Health Network, Toronto, Ontario, Canada
| | - Roberto Lopez
- Animal Resources Centre, University Health Network, Toronto, Ontario, Canada
| | - Sandra Lafrance
- Animal Resources Centre, University Health Network, Toronto, Ontario, Canada
| | | | | | - Harold Atkins
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Rebecca C Auer
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Joshua M Thurman
- Department of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | - Gregory L Stahl
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesia, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Institutes of Medicine, Boston, Massachusetts, USA
| | - John D Lambris
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - John C Bell
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada.
| | - J Andrea McCart
- Division of Experimental Therapeutics, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada; Department of Surgery, Mount Sinai Hospital and University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
96
|
Edwards RP, Huang X, Vlad AM. Chronic inflammation in endometriosis and endometriosis-associated ovarian cancer: New roles for the "old" complement pathway. Oncoimmunology 2015; 4:e1002732. [PMID: 26155393 DOI: 10.1080/2162402x.2014.1002732] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 12/23/2014] [Indexed: 02/03/2023] Open
Abstract
Immune escape is consequential for cancer development. Identifying abnormalities of the immune microenvironment during early carcinogenesis can provide insight into disease pathogenesis and unravel new preventive or therapeutic targets. We recently conducted a comprehensive immune gene expression analysis in endometriosis and endometriosis-associated ovarian cancer and explored new mechanistic roles for the complement pathway.
Collapse
Affiliation(s)
- Robert P Edwards
- Department of Obstetrics, Gynecology and Reproductive Sciences; University of Pittsburgh School of Medicine and Magee Women's Research Institute (MWRI) ; Pittsburgh, PA USA ; Magee-Womens Hospital of University of Pittsburgh Medical Center ; Pittsburgh, PA USA
| | - Xin Huang
- Department of Obstetrics, Gynecology and Reproductive Sciences; University of Pittsburgh School of Medicine and Magee Women's Research Institute (MWRI) ; Pittsburgh, PA USA
| | - Anda M Vlad
- Department of Obstetrics, Gynecology and Reproductive Sciences; University of Pittsburgh School of Medicine and Magee Women's Research Institute (MWRI) ; Pittsburgh, PA USA
| |
Collapse
|
97
|
Al-Rayahi IAM, Sanyi RHH. The overlapping roles of antimicrobial peptides and complement in recruitment and activation of tumor-associated inflammatory cells. Front Immunol 2015; 6:2. [PMID: 25657649 PMCID: PMC4302985 DOI: 10.3389/fimmu.2015.00002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 01/04/2015] [Indexed: 12/16/2022] Open
Abstract
Antimicrobial peptides (AMPs) represent a group of small (6-100 amino acids), biologically active molecules, which are produced by plants, mammals, and microorganisms (1). An important element of the innate immune response, AMP, possesses potent antibiotic, antifungal, and antiviral activities. Furthermore, AMP may be involved in a number of other processes such as angiogenesis and modulation of the immune response such as stimulation of chemokines and chemotaxis of leukocytes. AMPs have been proposed as alternative therapies for infectious diseases. AMP may also exert cytotoxic activity against tumor cells. Further understanding of the biological function of these peptides during tumor development and progression may aid in the development of novel anti-tumor therapies with refined application of innate molecules. AMP and complement have distinct roles to play in shaping the microenvironment (Table 1). Components of the complement system are integral contributors in responding to infection and sterile inflammation. Moreover, complement plays a role in the trafficking of cells in the tumor microenvironment, and thereby possibly in the immune response to cancer. This article will try to outline characteristics of AMP and complement in mobilization and recruitment of cells in tumor microenvironment.
Collapse
Affiliation(s)
- Izzat A M Al-Rayahi
- Department of Infection, Immunity and Inflammation, College of Medicine, Biological Sciences and Psychology, University of Leicester , Leicester , UK ; The Ministry of Higher Education , Baghdad , Iraq
| | - Raghad H H Sanyi
- Department of Infection, Immunity and Inflammation, College of Medicine, Biological Sciences and Psychology, University of Leicester , Leicester , UK ; The Ministry of Higher Education , Baghdad , Iraq
| |
Collapse
|
98
|
Storm L, Christensen IJ, Jensenius JC, Nielsen HJ, Thiel S. Evaluation of complement proteins as screening markers for colorectal cancer. Cancer Immunol Immunother 2015; 64:41-50. [PMID: 25261356 PMCID: PMC11028411 DOI: 10.1007/s00262-014-1615-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 09/17/2014] [Indexed: 01/28/2023]
Abstract
BACKGROUND Colorectal cancer (CRC) is a leading cause of cancer death worldwide. Lack of symptoms results in late detection and increased mortality. Inflammation, including complement activation, plays an important role in tumorigenesis. EXPERIMENTAL DESIGN The concentrations of nine proteins of the lectin pathway of the complement system were determined using time-resolved immunofluorometric assays. The first cohort investigated comprised a matched case-control study of 95 patients with CRC, 48 patients with adenomas and 48 individuals without neoplastic findings. Based on the results, Collectin-liver 1 (CL-L1), M-ficolin and MAp44 were determined as the most promising biomarkers and were subsequently evaluated in a case-control study of 99 CRC patients, 196 patients with adenomas and 696 individuals without neoplastic bowel lesions. RESULTS Using logistic regression, we found that CL-L1, M-ficolin and MAp44 levels could significantly distinguish between patients with CRC, patients with adenomas and individuals without neoplastic bowel lesions. Higher levels of CL-L1 or MAp44 were associated with lower odds of CRC (OR 0.42 (0.25-0.70) p = 0.0003 and OR 0.39 (0.23-0.65) p = 0.0003, respectively), whereas higher levels of M-ficolin were associated with higher odds of CRC compared to individuals without CRC (OR 1.94 (1.46-2.59) p < 0.0001). The combination of CL-L1, M-ficolin and MAp44 in a test of CRC versus individuals without CRC resulted in 36 % sensitivity at 83 % specificity. CONCLUSION CL-L1, M-ficolin and MAp44 in combination discriminate between CRC and patients without cancer. The markers did not have sufficient discriminatory value for CRC detection, but may prove useful for screening when combined with other markers.
Collapse
Affiliation(s)
- Line Storm
- Department of Biomedicine, Faculty of Health Sciences, Aarhus University, Wilhelm Meyers Allé 4, Aarhus C, Denmark
| | - Ib J. Christensen
- Finsen Laboratory, Rigshospitalet and Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Jens C. Jensenius
- Department of Biomedicine, Faculty of Health Sciences, Aarhus University, Wilhelm Meyers Allé 4, Aarhus C, Denmark
| | - Hans J. Nielsen
- Department of Surgical Gastroenterology, Hvidovre Hospital, University of Copenhagen, Hvidovre, Denmark
| | - Steffen Thiel
- Department of Biomedicine, Faculty of Health Sciences, Aarhus University, Wilhelm Meyers Allé 4, Aarhus C, Denmark
| | - the Danish Study Group on Early Detection of Colorectal Cancer
- Department of Biomedicine, Faculty of Health Sciences, Aarhus University, Wilhelm Meyers Allé 4, Aarhus C, Denmark
- Finsen Laboratory, Rigshospitalet and Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark
- Department of Surgical Gastroenterology, Hvidovre Hospital, University of Copenhagen, Hvidovre, Denmark
| |
Collapse
|
99
|
Zhang J, Zhang C. Research Progress on the Antineoplastic Pharmacological Effects and Mechanisms of Litchi Seeds. Chin Med 2015. [DOI: 10.4236/cm.2015.61003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
100
|
Khorasani AA, Weaver JL, Salvador-Morales C. Closing the gap: accelerating the translational process in nanomedicine by proposing standardized characterization techniques. Int J Nanomedicine 2014; 9:5729-51. [PMID: 25525356 PMCID: PMC4268909 DOI: 10.2147/ijn.s72479] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
On the cusp of widespread permeation of nanomedicine, academia, industry, and government have invested substantial financial resources in developing new ways to better treat diseases. Materials have unique physical and chemical properties at the nanoscale compared with their bulk or small-molecule analogs. These unique properties have been greatly advantageous in providing innovative solutions for medical treatments at the bench level. However, nanomedicine research has not yet fully permeated the clinical setting because of several limitations. Among these limitations are the lack of universal standards for characterizing nanomaterials and the limited knowledge that we possess regarding the interactions between nanomaterials and biological entities such as proteins. In this review, we report on recent developments in the characterization of nanomaterials as well as the newest information about the interactions between nanomaterials and proteins in the human body. We propose a standard set of techniques for universal characterization of nanomaterials. We also address relevant regulatory issues involved in the translational process for the development of drug molecules and drug delivery systems. Adherence and refinement of a universal standard in nanomaterial characterization as well as the acquisition of a deeper understanding of nanomaterials and proteins will likely accelerate the use of nanomedicine in common practice to a great extent.
Collapse
Affiliation(s)
- Ali A Khorasani
- Department of Chemistry and Biochemistry, George Mason University, Fairfax, VA, USA
- Bioengineering Department, George Mason University, Fairfax, VA, USA
- Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA, USA
| | - James L Weaver
- Division of Applied Regulatory Science, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Carolina Salvador-Morales
- Bioengineering Department, George Mason University, Fairfax, VA, USA
- Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA, USA
| |
Collapse
|