1
|
Zhang Y, Gao S, Mao J, Song Y, Wang X, Jiang J, Lv L, Zhou Z, Wang J. The Inhibitory Effect and Mechanism of the Histidine-Rich Peptide rAj-HRP from Apostichopus japonicus on Human Colon Cancer HCT116 Cells. Molecules 2024; 29:5214. [PMID: 39519855 PMCID: PMC11548021 DOI: 10.3390/molecules29215214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/16/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Colon cancer is a common and lethal malignancy, ranking second in global cancer-related mortality, highlighting the urgent need for novel targeted therapies. The sea cucumber (Apostichopus japonicus) is a marine organism known for its medicinal properties. After conducting a bioinformatics analysis of the cDNA library of Apostichopus japonicus, we found and cloned a cDNA sequence encoding histidine-rich peptides, and the recombinant peptide was named rAj-HRP. Human histidine-rich peptides are known for their anti-cancer properties, raising questions as to whether rAj-HRP might exhibit similar effects. To investigate whether rAj-HRP can inhibit colon cancer, we used human colon cancer HCT116 cells as a model and studied the tumor suppressive activity in vitro and in vivo. The results showed that rAj-HRP inhibited HCT116 cell proliferation, migration, and adhesion to extracellular matrix (ECM) proteins in vitro. It also disrupted the cytoskeleton and induced apoptosis in these cells. In vivo, rAj-HRP significantly inhibited the growth of HCT116 tumors in BALB/c mice, reducing tumor volume and weight without affecting the body weight of the tumor-bearing mice. Western blot analysis showed that rAj-HRP inhibited HCT116 cell proliferation and induced apoptosis by upregulating BAX and promoting PARP zymogen degradation. Additionally, rAj-HRP inhibited HCT116 cell adhesion and migration by reducing MMP2 levels. Further research showed that rAj-HRP downregulated EGFR expression in HCT116 cells and inhibited key downstream molecules, including AKT, P-AKT, PLCγ, P38 MAPK, and c-Jun. In conclusion, rAj-HRP exhibits significant inhibitory effects on HCT116 cells in both in vitro and in vivo, primarily through the EGFR and apoptosis pathways. These findings suggest that rAj-HRP has the potential as a novel targeted therapy for colon cancer.
Collapse
Affiliation(s)
- Yuebin Zhang
- School of Life Sciences, Liaoning Normal University, Dalian 116081, China; (Y.Z.); (J.M.); (Y.S.)
| | - Shan Gao
- Liaoning Key Lab of Marine Fishery Molecular Biology, Liaoning Ocean and Fisheries Science Research Institute, Dalian 116023, China; (S.G.); (J.J.)
| | - Jiaming Mao
- School of Life Sciences, Liaoning Normal University, Dalian 116081, China; (Y.Z.); (J.M.); (Y.S.)
| | - Yuyao Song
- School of Life Sciences, Liaoning Normal University, Dalian 116081, China; (Y.Z.); (J.M.); (Y.S.)
| | - Xueting Wang
- Department of Pharmacology, College of Pharmacy, Dalian Medical University, Dalian 116044, China; (X.W.); (L.L.)
| | - Jingwei Jiang
- Liaoning Key Lab of Marine Fishery Molecular Biology, Liaoning Ocean and Fisheries Science Research Institute, Dalian 116023, China; (S.G.); (J.J.)
| | - Li Lv
- Department of Pharmacology, College of Pharmacy, Dalian Medical University, Dalian 116044, China; (X.W.); (L.L.)
| | - Zunchun Zhou
- Liaoning Key Lab of Marine Fishery Molecular Biology, Liaoning Ocean and Fisheries Science Research Institute, Dalian 116023, China; (S.G.); (J.J.)
| | - Jihong Wang
- School of Life Sciences, Liaoning Normal University, Dalian 116081, China; (Y.Z.); (J.M.); (Y.S.)
| |
Collapse
|
2
|
Lorestani P, Dashti M, Nejati N, Habibi MA, Askari M, Robat-Jazi B, Ahmadpour S, Tavakolpour S. The complex role of macrophages in pancreatic cancer tumor microenvironment: a review on cancer progression and potential therapeutic targets. Discov Oncol 2024; 15:369. [PMID: 39186144 PMCID: PMC11347554 DOI: 10.1007/s12672-024-01256-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024] Open
Abstract
Pancreatic cancer (PC) is one of the deadliest cancers worldwide with low survival rates and poor outcomes. The treatment landscape for PC is fraught with obstacles, including drug resistance, lack of effective targeted therapies and the immunosuppressive tumor microenvironment (TME). The resistance of PC to existing immunotherapies highlights the need for innovative approaches, with the TME emerging as a promising therapeutic target. The recent advancements in understanding the role of macrophages, this context highlight their significant impact on tumor development and progression. There are two important types of macrophages: M1 and M2, which play critical roles in the TME. Therapeutics strategies including, depletion of tumor-associated macrophages (TAMs), reprogramming TAMs to promote anti-tumor activity, and targeting macrophage recruitment can lead to promising outcomes. Targeting macrophage-related pathways may offer novel strategies for modulating immune responses, inhibiting angiogenesis, and overcoming resistance to chemotherapy in PC treatment.
Collapse
Affiliation(s)
- Parsa Lorestani
- Students Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohsen Dashti
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Negar Nejati
- Pediatric Cell and Gene Therapy Research Centre, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Amin Habibi
- Department of Neurosurgery, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mandana Askari
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Behruz Robat-Jazi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Sajjad Ahmadpour
- Patient Safety Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| | - Soheil Tavakolpour
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA.
| |
Collapse
|
3
|
Foglia B, Sutti S, Cannito S, Rosso C, Maggiora M, Casalino A, Bocca C, Novo E, Protopapa F, Ramavath NN, Provera A, Gambella A, Bugianesi E, Tacke F, Albano E, Parola M. Histidine-rich glycoprotein in metabolic dysfunction-associated steatohepatitis-related disease progression and liver carcinogenesis. Front Immunol 2024; 15:1342404. [PMID: 38469298 PMCID: PMC10925642 DOI: 10.3389/fimmu.2024.1342404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 01/31/2024] [Indexed: 03/13/2024] Open
Abstract
Background Metabolic dysfunction-associated steatotic liver disease (MASLD), previously non-alcoholic fatty liver disease (NAFLD), is a leading cause of chronic liver disease worldwide. In 20%-30% of MASLD patients, the disease progresses to metabolic dysfunction-associated steatohepatitis (MASH, previously NASH) which can lead to fibrosis/cirrhosis, liver failure as well as hepatocellular carcinoma (HCC). Here we investigated the role of histidine-rich glycoprotein (HRG), a plasma protein produced by hepatocytes, in MASLD/MASH progression and HCC development. Methods The role of HRG was investigated by morphological, cellular, and molecular biology approaches in (a) HRG knock-out mice (HRG-/- mice) fed on a CDAA dietary protocol or a MASH related diethyl-nitrosamine/CDAA protocol of hepatocarcinogenesis, (b) THP1 monocytic cells treated with purified HRG, and (c) well-characterized cohorts of MASLD patients with or without HCC. Results In non-neoplastic settings, murine and clinical data indicate that HRG increases significantly in parallel with disease progression. In particular, in MASLD/MASH patients, higher levels of HRG plasma levels were detected in subjects with extensive fibrosis/cirrhosis. When submitted to the pro-carcinogenic protocol, HRG-/- mice showed a significant decrease in the volume and number of HCC nodules in relation to decreased infiltration of macrophages producing pro-inflammatory mediators, including IL-1β, IL-6, IL-12, IL-10, and VEGF as well as impaired angiogenesis. The histopathological analysis (H-score) of MASH-related HCC indicate that the higher HRG positivity in peritumoral tissue significantly correlates with a lower overall patient survival and an increased recurrence. Moreover, a significant increase in HRG plasma levels was detected in cirrhotic (F4) patients and in patients carrying HCC vs. F0/F1 patients. Conclusion Murine and clinical data indicate that HRG plays a significant role in MASLD/MASH progression to HCC by supporting a specific population of tumor-associated macrophages with pro-inflammatory response and pro-angiogenetic capabilities which critically support cancer cell survival. Furthermore, our data suggest HRG as a possible prognostic predictor in HCC patients with MASLD/MASH-related HCCs.
Collapse
Affiliation(s)
- Beatrice Foglia
- Department Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Torino, Torino, Italy
| | - Salvatore Sutti
- Department Health Sciences and Interdisciplinary Research Centre for Autoimmune Diseases, University Amedeo Avogadro of Eastern Piedmont, Novara, Italy
| | - Stefania Cannito
- Department Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Torino, Torino, Italy
| | - Chiara Rosso
- Department Medical Sciences, University of Torino, and Division of Gastroenterology, San Giovanni Hospital, Torino, Italy
| | - Marina Maggiora
- Department Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Torino, Torino, Italy
| | - Alice Casalino
- Department Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Torino, Torino, Italy
| | - Claudia Bocca
- Department Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Torino, Torino, Italy
| | - Erica Novo
- Department Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Torino, Torino, Italy
| | - Francesca Protopapa
- Department Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Torino, Torino, Italy
| | - Naresh Naik Ramavath
- Department of Pediatrics, School of Medicine, Washington University, St Louis, MO, United States
| | - Alessia Provera
- Department Health Sciences and Interdisciplinary Research Centre for Autoimmune Diseases, University Amedeo Avogadro of Eastern Piedmont, Novara, Italy
| | - Alessandro Gambella
- Department Medical Sciences, University of Torino, and Division of Gastroenterology, San Giovanni Hospital, Torino, Italy
| | - Elisabetta Bugianesi
- Department Medical Sciences, University of Torino, and Division of Gastroenterology, San Giovanni Hospital, Torino, Italy
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité-Universitatsmedizin Berlin, Berlin, Germany
| | - Emanuele Albano
- Department Health Sciences and Interdisciplinary Research Centre for Autoimmune Diseases, University Amedeo Avogadro of Eastern Piedmont, Novara, Italy
| | - Maurizio Parola
- Department Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Torino, Torino, Italy
| |
Collapse
|
4
|
Nishibori M. Novel aspects of sepsis pathophysiology: NETs, plasma glycoproteins, endotheliopathy and COVID-19. J Pharmacol Sci 2022; 150:9-20. [PMID: 35926948 PMCID: PMC9197787 DOI: 10.1016/j.jphs.2022.06.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/02/2022] [Accepted: 06/07/2022] [Indexed: 12/13/2022] Open
Abstract
In 2016, sepsis was newly defined as life-threatening organ dysfunction caused by a dysregulated host response to infection. Sepsis remains one of the crucial medical problems to be solved worldwide. Although the world health organization has made sepsis a global health priority, there remain no specific and effective therapy for sepsis so far. Indeed, over the previous decades almost all attempts to develop novel drugs have failed. This may be partly ascribable to the multifactorial complexity of the septic cascade and the resultant difficulties of identifying drug targets. In addition, there might still be missing links among dysregulated host responses in vital organs. In this review article, recent advances in understanding of the complex pathophysiology of sepsis are summarized, with a focus on neutrophil extracellular traps (NETs), the significant role of NETs in thrombosis/embolism, and the functional roles of plasma proteins, histidine-rich glycoprotein (HRG) and inter-alpha-inhibitor proteins (IAIPs). The specific plasma proteins that are markedly decreased in the acute phase of sepsis may play important roles in the regulation of blood cells, vascular endothelial cells and coagulation. The accumulating evidence may provide us with insights into a novel aspect of the pathophysiology of sepsis and septic ARDS, including that in COVID-19.
Collapse
Affiliation(s)
- M Nishibori
- Department of Translational Research and Drug Development, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan.
| |
Collapse
|
5
|
Quartz Crystal Microbalance Measurement of Histidine-Rich Glycoprotein and Stanniocalcin-2 Binding to Each Other and to Inflammatory Cells. Cells 2022; 11:cells11172684. [PMID: 36078092 PMCID: PMC9454698 DOI: 10.3390/cells11172684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 12/02/2022] Open
Abstract
The plasma protein histidine-rich glycoprotein (HRG) is implicated in the polarization of macrophages to an M1 antitumoral phenotype. The broadly expressed secreted protein stanniocalcin 2 (STC2), also implicated in tumor inflammation, is an HRG interaction partner. With the aim to biochemically characterize the HRG and STC2 complex, binding of recombinant HRG and STC2 preparations to each other and to cells was explored using the quartz crystal microbalance (QCM) methodology. The functionality of recombinant proteins was tested in a phagocytosis assay, where HRG increased phagocytosis by monocytic U937 cells while STC2 suppressed HRG-induced phagocytosis. The binding of HRG to STC2, measured using QCM, showed an affinity between the proteins in the nanomolar range, and both HRG and STC2 bound individually and in combination to vitamin D3-treated, differentiated U937 monocytes. HRG, but not STC2, also bound to formaldehyde-fixed U937 cells irrespective of their differentiation stage in part through the interaction with heparan sulfate. These data show that HRG and STC2 bind to each other as well as to U937 monocytes with high affinity, supporting the relevance of these interactions in monocyte/macrophage polarity.
Collapse
|
6
|
Rose M, Huth S, Wiesehöfer M, Ehling J, Henkel C, Steitz J, Lammers T, Kistermann J, Klaas O, Koch M, Rushrush S, Knüchel R, Dahl E. ITIH5-Derived Polypeptides Covering the VIT Domain Suppress the Growth of Human Cancer Cells In Vitro. Cancers (Basel) 2022; 14:cancers14030488. [PMID: 35158755 PMCID: PMC8833355 DOI: 10.3390/cancers14030488] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/07/2022] [Accepted: 01/11/2022] [Indexed: 01/27/2023] Open
Abstract
Oncogenic drivers such as mutated EGFR are the preferred targets in modern drug development. However, restoring the lost function of tumor suppressor proteins could also be a valid approach to combatting cancer. ITIH5 has been revealed as a potent metastasis suppressor in both breast and pancreatic cancer. Here, we show that ITIH5 overexpression in MDA-MB-231 breast cancer cells can also locally suppress tumor growth by 85%, when transplanted into the mammary fat pad of nude mice. For a potential drug development approach, we further aimed to define downsized ITIH5 polypeptides that still are capable of mediating growth inhibitory effects. By cloning truncated and His-tagged ITIH5 fragments, we synthesized two recombinant N-terminal polypeptides (ITIH5681aa and ITIH5161aa), both covering the ITI heavy chain specific “vault protein inter-alpha-trypsin” (VIT) domain. Truncated ITIH5 variants caused dose-dependent cell growth inhibition by up to 50% when applied to various cancer cell lines (e.g., MDA-MB-231, SCaBER, A549) reflecting breast, bladder and lung cancer in vitro. Thus, our data suggest the substantial role of the ITIH5-specific VIT domain in ITIH5-mediated suppression of tumor cell proliferation. As extracellularly administered ITIH5 peptides mimic the growth-inhibitory effects of the full-length ITIH5 tumor suppressor protein, they may constitute the basis for developing anticancer drugs in the future.
Collapse
Affiliation(s)
- Michael Rose
- Institute of Pathology, RWTH Aachen University, 52074 Aachen, Germany; (S.H.); (M.W.); (C.H.); (J.K.); (O.K.); (M.K.); (S.R.); (R.K.)
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), 52074 Aachen, Germany
- Correspondence: (M.R.); (E.D.); Tel.: +49-241-80-89715 (M.R.); +49-241-80-88431 (E.D.); Fax: +49-241-8082439 (M.R. & E.D.)
| | - Sebastian Huth
- Institute of Pathology, RWTH Aachen University, 52074 Aachen, Germany; (S.H.); (M.W.); (C.H.); (J.K.); (O.K.); (M.K.); (S.R.); (R.K.)
- Department of Dermatology and Allergology, RWTH Aachen University, 52074 Aachen, Germany
| | - Marc Wiesehöfer
- Institute of Pathology, RWTH Aachen University, 52074 Aachen, Germany; (S.H.); (M.W.); (C.H.); (J.K.); (O.K.); (M.K.); (S.R.); (R.K.)
| | - Josef Ehling
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074 Aachen, Germany; (J.E.); (T.L.)
| | - Corinna Henkel
- Institute of Pathology, RWTH Aachen University, 52074 Aachen, Germany; (S.H.); (M.W.); (C.H.); (J.K.); (O.K.); (M.K.); (S.R.); (R.K.)
- Bruker Daltonik GmbH, 28359 Bremen, Germany
| | - Julia Steitz
- Institute for Laboratory Animal Science, University Hospital, RWTH Aachen University, 52074 Aachen, Germany;
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074 Aachen, Germany; (J.E.); (T.L.)
| | - Jennifer Kistermann
- Institute of Pathology, RWTH Aachen University, 52074 Aachen, Germany; (S.H.); (M.W.); (C.H.); (J.K.); (O.K.); (M.K.); (S.R.); (R.K.)
| | - Oliver Klaas
- Institute of Pathology, RWTH Aachen University, 52074 Aachen, Germany; (S.H.); (M.W.); (C.H.); (J.K.); (O.K.); (M.K.); (S.R.); (R.K.)
| | - Maximilian Koch
- Institute of Pathology, RWTH Aachen University, 52074 Aachen, Germany; (S.H.); (M.W.); (C.H.); (J.K.); (O.K.); (M.K.); (S.R.); (R.K.)
| | - Sandra Rushrush
- Institute of Pathology, RWTH Aachen University, 52074 Aachen, Germany; (S.H.); (M.W.); (C.H.); (J.K.); (O.K.); (M.K.); (S.R.); (R.K.)
| | - Ruth Knüchel
- Institute of Pathology, RWTH Aachen University, 52074 Aachen, Germany; (S.H.); (M.W.); (C.H.); (J.K.); (O.K.); (M.K.); (S.R.); (R.K.)
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), 52074 Aachen, Germany
| | - Edgar Dahl
- Institute of Pathology, RWTH Aachen University, 52074 Aachen, Germany; (S.H.); (M.W.); (C.H.); (J.K.); (O.K.); (M.K.); (S.R.); (R.K.)
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), 52074 Aachen, Germany
- Correspondence: (M.R.); (E.D.); Tel.: +49-241-80-89715 (M.R.); +49-241-80-88431 (E.D.); Fax: +49-241-8082439 (M.R. & E.D.)
| |
Collapse
|
7
|
Men WQ, Xu SG, Mou R. Hepatic transcriptome study of Taenia asiatica infection in suckling pigs. Microb Pathog 2020; 152:104598. [PMID: 33157217 DOI: 10.1016/j.micpath.2020.104598] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/18/2020] [Accepted: 10/19/2020] [Indexed: 01/05/2023]
Abstract
Taenia asiatica is a crucial Taenia that is prevalent in East and Southeast Asia. Domestic pigs and wild boars are essential intermediate hosts for Taenia. Cysticercus larvae are mainly parasitic in the liver of domestic pigs. The Taenia asiatica was collected from Liangmu Township, Duyun City, Guizhou Province. Twelve Yorkshire Suckling pigs of 20 days of age were randomly divided into an experimental and control group of 6 pigs each. RNA sequencing (RNA-seq) technology was used to detect the expression differences of the mRNA transcriptomes in the liver of the experimental and control group at different infection times. Differential genes were analyzed by bioinformatics and verified by Real Time-PCR(RT-PCR). On the 15th and 75th days after infection, 152 and 558 differentially expressed genes were detected in the liver of the experimental group, respectively, accounting for 0.85% and 3.12% of all identified transcribed RNA genes, respectively. Through GO and KEGG related bioinformatics analysis, it was found that these differentially expressed genes are involved in the immune response, material metabolism, fibrosis, and tissue proliferation and repair of suckling pig liver, and related to MHC antigen processing and presentation, cytochrome P450, transforming growth factor-beta (TGF-β) signaling pathway and so on. Cysticercus asiatica parasites cause significant differential gene expression in the liver of suckling pigs. Specific differentially expressed genes are involved in biological processes such as liver metabolism, immune response, and tissue repair or regeneration in suckling pigs. The immune evasion is related to the immuno-suppressive response of the intermediate host.
Collapse
Affiliation(s)
- Wan-Qi Men
- Department of Parasitology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550025, China; Characteristic and Key Laboratory of Modern Pathogenic Biology, Guizhou Medical University, Guiyang, 550025, China
| | - Shi-Gang Xu
- Department of Parasitology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550025, China; Characteristic and Key Laboratory of Modern Pathogenic Biology, Guizhou Medical University, Guiyang, 550025, China
| | - Rong Mou
- Department of Parasitology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550025, China; Characteristic and Key Laboratory of Modern Pathogenic Biology, Guizhou Medical University, Guiyang, 550025, China.
| |
Collapse
|
8
|
Razzaque MS, Atfi A. TGIF1-Twist1 axis in pancreatic ductal adenocarcinoma. Comput Struct Biotechnol J 2020; 18:2568-2572. [PMID: 33005315 PMCID: PMC7520386 DOI: 10.1016/j.csbj.2020.09.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/11/2020] [Accepted: 09/11/2020] [Indexed: 12/18/2022] Open
Abstract
TG-interacting factor 1 (TGIF1) exerts inhibitory effects on transforming growth factor-beta (TGF-β) signaling by suppressing Smad signaling pathway at multiple levels. TGIF1 activity is important for normal embryogenesis and organogenesis, yet its dysregulation can culminate in tumorigenesis. For instance, increased expression of TGIF1 correlates with poor prognosis in triple-negative breast cancer patients, and enforced expression of TGIF1 facilitates Wnt-driven mammary tumorigenesis, suggesting that TGIF1 might function as an oncoprotein. Quite surprisingly, TGIF1 has recently been shown to function as a tumor suppressor in pancreatic ductal adenocarcinoma (PDAC), possibly owing to its ability to antagonize the pro-malignant transcription factor Twist1. In this article, we will briefly elaborate on the biological and clinical significance of the unique tumor-suppressive function of TGIF1 and its functional interaction with Twist1 in the context of PDAC pathogenesis and progression.
Collapse
Affiliation(s)
- Mohammed S Razzaque
- Department of Pathology, Lake Erie College of Osteopathic Medicine, Erie, PA, USA
| | - Azeddine Atfi
- Department of Pathology and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
9
|
Albonici L, Giganti MG, Modesti A, Manzari V, Bei R. Multifaceted Role of the Placental Growth Factor (PlGF) in the Antitumor Immune Response and Cancer Progression. Int J Mol Sci 2019; 20:ijms20122970. [PMID: 31216652 PMCID: PMC6627047 DOI: 10.3390/ijms20122970] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 06/10/2019] [Accepted: 06/14/2019] [Indexed: 12/22/2022] Open
Abstract
The sharing of molecules function that affects both tumor growth and neoangiogenesis with cells of the immune system creates a mutual interplay that impairs the host’s immune response against tumor progression. Increasing evidence shows that tumors are able to create an immunosuppressive microenvironment by recruiting specific immune cells. Moreover, molecules produced by tumor and inflammatory cells in the tumor microenvironment create an immunosuppressive milieu able to inhibit the development of an efficient immune response against cancer cells and thus fostering tumor growth and progression. In addition, the immunoediting could select cancer cells that are less immunogenic or more resistant to lysis. In this review, we summarize recent findings regarding the immunomodulatory effects and cancer progression of the angiogenic growth factor namely placental growth factor (PlGF) and address the biological complex effects of this cytokine. Different pathways of the innate and adaptive immune response in which, directly or indirectly, PlGF is involved in promoting tumor immune escape and metastasis will be described. PlGF is important for building up vascular structures and functions. Although PlGF effects on vascular and tumor growth have been widely summarized, its functions in modulating the immune intra-tumoral microenvironment have been less highlighted. In agreement with PlGF functions, different antitumor strategies can be envisioned.
Collapse
Affiliation(s)
- Loredana Albonici
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy.
| | - Maria Gabriella Giganti
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy.
| | - Andrea Modesti
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy.
| | - Vittorio Manzari
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy.
| | - Roberto Bei
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy.
| |
Collapse
|
10
|
Guo L, Ren H, Zeng H, Gong Y, Ma X. Proteomic analysis of cerebrospinal fluid in pediatric acute lymphoblastic leukemia patients: a pilot study. Onco Targets Ther 2019; 12:3859-3868. [PMID: 31190885 PMCID: PMC6527054 DOI: 10.2147/ott.s193616] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Purpose Involvement of central nervous system in acute lymphoblastic leukemia (CNSL) remains one of the major causes of pediatric acute lymphoblastic leukemia (ALL) treatment failure. However, the current understanding of the pathological process of CNSL is still limited. This study aimed to better understand the protein expression in cerebrospinal fluid (CSF) of ALL and discover valuable prognostic biomarkers. Materials and methods CSF samples were obtained from ALL patients and healthy controls. Comparative proteomic profiling using label-free liquid chromatography-tandem mass spectrometry was performed to detect differentially expressed proteins. Results In the present study, 51 differentially expressed proteins were found. Among them, two core clusters including ten proteins (TIMP1, LGALS3BP, A2M, FN1, AHSG, HRG, ITIH4, CF I, C2, and C4a) might be crucial for tumorigenesis and progression of ALL and can be potentially valuable indicators of CNSL. Conclusion These differentially expressed proteins of ALL children with central nervous system involvement and normal children may work as diagnostic and prognostic factors of ALL patients.
Collapse
Affiliation(s)
- Linghong Guo
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, People's Republic of China, .,West China School of Medicine, Sichuan University, Chengdu, People's Republic of China
| | - Honghong Ren
- West China School of Medicine, Sichuan University, Chengdu, People's Republic of China
| | - Hao Zeng
- West China School of Medicine, Sichuan University, Chengdu, People's Republic of China
| | - Yanqiu Gong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xuelei Ma
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, People's Republic of China,
| |
Collapse
|
11
|
Decreased Levels of Histidine-Rich Glycoprotein in Advanced Lung Cancer: Association with Prothrombotic Alterations. DISEASE MARKERS 2019; 2019:8170759. [PMID: 30944671 PMCID: PMC6421726 DOI: 10.1155/2019/8170759] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 01/10/2019] [Indexed: 11/18/2022]
Abstract
Background Histidine-rich glycoprotein (HRG) displays anticoagulant and antifibrinolytic properties in animal models, but its effects in humans are unclear. We investigated serum HRG levels and their associations with the disease stage and prothrombotic alterations in lung cancer (LC) patients. Methods In 148 patients with advanced LC prior to anticancer therapy (87 non-small-cell LC and 61 small-cell LC) versus 100 well-matched controls, we measured HRG levels in association with clot permeability (Ks), clot turbidimetry (lag phase and maximum absorbance), and clot lysis time (CLT). Results Compared to controls, LC patients had 45.9% lower HRG levels with no associations with demographics and comorbidities. Decreased HRG, defined as the 90th percentile of control values (<52.7 μg/ml), was 16 times more common in subjects with than without LC (OR = 16.4, 95% CI 9.2-23.5, p < 0.01). HRG < 38 μg/ml discriminated stage IIIAB/limited disease from IV/extensive disease (ED) LC. In LC patients, HRG correlated inversely with CLT (r = −0.41, p < 0.001), but not with other fibrin variables. Among stage IV/ED LC, HRG correlated significantly with Ks and lag phase (r = 0.28 and r = 0.33, respectively, both p < 0.001). LC patients with low Ks (10th percentile of control values) combined with prolonged CLT (90th percentile of control values) had reduced HRG levels compared to the remainder (p = 0.003). No such observations were noted in controls. Conclusions Our study is the first to show that decreased HRG levels occur in advanced LC and are associated with the disease stage and hypofibrinolysis.
Collapse
|
12
|
Theek B, Baues M, Gremse F, Pola R, Pechar M, Negwer I, Koynov K, Weber B, Barz M, Jahnen-Dechent W, Storm G, Kiessling F, Lammers T. Histidine-rich glycoprotein-induced vascular normalization improves EPR-mediated drug targeting to and into tumors. J Control Release 2018; 282:25-34. [PMID: 29730154 PMCID: PMC6130770 DOI: 10.1016/j.jconrel.2018.05.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 04/26/2018] [Accepted: 05/02/2018] [Indexed: 11/18/2022]
Abstract
Tumors are characterized by leaky blood vessels, and by an abnormal and heterogeneous vascular network. These pathophysiological characteristics contribute to the enhanced permeability and retention (EPR) effect, which is one of the key rationales for developing tumor-targeted drug delivery systems. Vessel abnormality and heterogeneity, however, which typically result from excessive pro-angiogenic signaling, can also hinder efficient drug delivery to and into tumors. Using histidine-rich glycoprotein (HRG) knockout and wild type mice, and HRG-overexpressing and normal t241 fibrosarcoma cells, we evaluated the effect of genetically induced and macrophage-mediated vascular normalization on the tumor accumulation and penetration of 10-20 nm-sized polymeric drug carriers based on poly(N-(2-hydroxypropyl)methacrylamide). Multimodal and multiscale optical imaging was employed to show that normalizing the tumor vasculature improves the accumulation of fluorophore-labeled polymers in tumors, and promotes their penetration out of tumor blood vessels deep into the interstitium.
Collapse
Affiliation(s)
- Benjamin Theek
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic and Helmholtz Institute for Biomedical Engineering, Aachen, Germany; Department of Targeted Therapeutics, Biomaterial Science and Technology, University of Twente, Enschede, The Netherlands
| | - Maike Baues
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic and Helmholtz Institute for Biomedical Engineering, Aachen, Germany
| | - Felix Gremse
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic and Helmholtz Institute for Biomedical Engineering, Aachen, Germany
| | - Robert Pola
- Institute of Macromolecular Chemistry, Czech Academy of Science, Prague, Czech Republic
| | - Michal Pechar
- Institute of Macromolecular Chemistry, Czech Academy of Science, Prague, Czech Republic
| | - Inka Negwer
- Max Planck Institute for Polymer Research, Mainz, Germany
| | - Kaloian Koynov
- Max Planck Institute for Polymer Research, Mainz, Germany
| | - Benjamin Weber
- Institute of Organic Chemistry, Johannes Gutenberg University, Mainz, Germany
| | - Matthias Barz
- Institute of Organic Chemistry, Johannes Gutenberg University, Mainz, Germany
| | - Willi Jahnen-Dechent
- Biointerface Laboratory, RWTH Aachen University Clinic and Helmholtz Institute for Biomedical Engineering, Aachen, Germany
| | - Gert Storm
- Department of Targeted Therapeutics, Biomaterial Science and Technology, University of Twente, Enschede, The Netherlands; Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Fabian Kiessling
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic and Helmholtz Institute for Biomedical Engineering, Aachen, Germany
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic and Helmholtz Institute for Biomedical Engineering, Aachen, Germany; Department of Targeted Therapeutics, Biomaterial Science and Technology, University of Twente, Enschede, The Netherlands; Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
13
|
Roche FP, Pietilä I, Kaito H, Sjöström EO, Sobotzki N, Noguer O, Skare TP, Essand M, Wollscheid B, Welsh M, Claesson-Welsh L. Leukocyte Differentiation by Histidine-Rich Glycoprotein/Stanniocalcin-2 Complex Regulates Murine Glioma Growth through Modulation of Antitumor Immunity. Mol Cancer Ther 2018; 17:1961-1972. [PMID: 29945872 DOI: 10.1158/1535-7163.mct-18-0097] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 04/21/2018] [Accepted: 06/19/2018] [Indexed: 11/16/2022]
Abstract
The plasma-protein histidine-rich glycoprotein (HRG) is implicated in phenotypic switching of tumor-associated macrophages, regulating cytokine production and phagocytotic activity, thereby promoting vessel normalization and antitumor immune responses. To assess the therapeutic effect of HRG gene delivery on CNS tumors, we used adenovirus-encoded HRG to treat mouse intracranial GL261 glioma. Delivery of Ad5-HRG to the tumor site resulted in a significant reduction in glioma growth, associated with increased vessel perfusion and increased CD45+ leukocyte and CD8+ T-cell accumulation in the tumor. Antibody-mediated neutralization of colony-stimulating factor-1 suppressed the effects of HRG on CD45+ and CD8+ infiltration. Using a novel protein interaction-decoding technology, TRICEPS-based ligand receptor capture (LRC), we identified Stanniocalcin-2 (STC2) as an interacting partner of HRG on the surface of inflammatory cells in vitro and colocalization of HRG and STC2 in gliomas. HRG reduced the suppressive effects of STC2 on monocyte CD14+ differentiation and STC2-regulated immune response pathways. In consequence, Ad5-HRG-treated gliomas displayed decreased numbers of IL35+ Treg cells, providing a mechanistic rationale for the reduction in GL261 growth in response to Ad5-HRG delivery. We conclude that HRG suppresses glioma growth by modulating tumor inflammation through monocyte infiltration and differentiation. Moreover, HRG acts to balance the regulatory effects of its partner, STC2, on inflammation and innate and/or acquired immunity. HRG gene delivery therefore offers a potential therapeutic strategy to control antitumor immunity. Mol Cancer Ther; 17(9); 1961-72. ©2018 AACR.
Collapse
Affiliation(s)
- Francis P Roche
- Uppsala University, Department of Immunology, Genetics and Pathology, Science for Life Laboratory, The Rudbeck Laboratory, Uppsala, Sweden
| | - Ilkka Pietilä
- Uppsala University, Department of Medical Cell Biology, Biomedical Center, Uppsala, Sweden
| | - Hiroshi Kaito
- Uppsala University, Department of Immunology, Genetics and Pathology, Science for Life Laboratory, The Rudbeck Laboratory, Uppsala, Sweden
| | - Elisabet O Sjöström
- Uppsala University, Department of Immunology, Genetics and Pathology, Science for Life Laboratory, The Rudbeck Laboratory, Uppsala, Sweden
| | - Nadine Sobotzki
- ETH Zürich, Department of Health Sciences and Technology & Institute of Molecular Systems Biology, Zürich, Switzerland
| | - Oriol Noguer
- Uppsala University, Department of Immunology, Genetics and Pathology, Science for Life Laboratory, The Rudbeck Laboratory, Uppsala, Sweden
| | - Tor Persson Skare
- Uppsala University, Department of Immunology, Genetics and Pathology, Science for Life Laboratory, The Rudbeck Laboratory, Uppsala, Sweden
| | - Magnus Essand
- Uppsala University, Department of Immunology, Genetics and Pathology, Science for Life Laboratory, The Rudbeck Laboratory, Uppsala, Sweden
| | - Bernd Wollscheid
- ETH Zürich, Department of Health Sciences and Technology & Institute of Molecular Systems Biology, Zürich, Switzerland
| | - Michael Welsh
- Uppsala University, Department of Medical Cell Biology, Biomedical Center, Uppsala, Sweden
| | - Lena Claesson-Welsh
- Uppsala University, Department of Immunology, Genetics and Pathology, Science for Life Laboratory, The Rudbeck Laboratory, Uppsala, Sweden.
| |
Collapse
|
14
|
Cui R, Yue W, Lattime EC, Stein MN, Xu Q, Tan XL. Targeting tumor-associated macrophages to combat pancreatic cancer. Oncotarget 2018; 7:50735-50754. [PMID: 27191744 PMCID: PMC5226617 DOI: 10.18632/oncotarget.9383] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 05/05/2016] [Indexed: 12/18/2022] Open
Abstract
The tumor microenvironment is replete with cells that evolve with and provide support to tumor cells during the transition to malignancy. The hijacking of the immune system in the pancreatic tumor microenvironment is suggested to contribute to the failure to date to produce significant improvements in pancreatic cancer survival by various chemotherapeutics. Regulatory T cells, myeloid derived suppressor cells, and fibroblasts, all of which constitute a complex ecology microenvironment, can suppress CD8+ T cells and NK cells, thus inhibiting effector immune responses. Tumor-associated macrophages (TAM) are versatile immune cells that can express different functional programs in response to stimuli in tumor microenvironment at different stages of pancreatic cancer development. TAM have been implicated in suppression of anti-tumorigenic immune responses, promotion of cancer cell proliferation, stimulation of tumor angiogenesis and extracellular matrix breakdown, and subsequent enhancement of tumor invasion and metastasis. Many emerging agents that have demonstrated efficacy in combating other types of tumors via modulation of macrophages in tumor microenvironments are, however, only marginally studied for pancreatic cancer prevention and treatment. A better understanding of the paradoxical roles of TAM in pancreatic cancer may pave the way to novel preventive and therapeutic approaches. Here we give an overview of the recruitment and differentiation of macrophages, TAM and pancreatic cancer progression and prognosis, as well as the potential preventive and therapeutic targets that interact with TAM for pancreatic cancer prevention and treatment.
Collapse
Affiliation(s)
- Ran Cui
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, P. R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Wen Yue
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Edmund C Lattime
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Mark N Stein
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Qing Xu
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, P. R. China
| | - Xiang-Lin Tan
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.,Department of Epidemiology, School of Public Health, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| |
Collapse
|
15
|
Chang JH, Jiang Y, Pillarisetty VG. Role of immune cells in pancreatic cancer from bench to clinical application: An updated review. Medicine (Baltimore) 2016; 95:e5541. [PMID: 27930550 PMCID: PMC5266022 DOI: 10.1097/md.0000000000005541] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Pancreatic cancer (PC) remains difficult to treat, despite the recent advances in various anticancer therapies. Immuno-inflammatory response is considered to be a major risk factor for the development of PC in addition to a combination of genetic background and environmental factors. Although patients with PC exhibit evidence of systemic immune dysfunction, the PC microenvironment is replete with immune cells. METHODS We searched PubMed for all relevant English language articles published up to March 2016. They included clinical trials, experimental studies, observational studies, and reviews. Trials enrolled at Clinical trial.gov were also searched. RESULTS PC induces an immunosuppressive microenvironment, and intratumoral activation of immunity in PC is attenuated by inhibitory signals that limit immune effector function. Multiple types of immune responses can promote an immunosuppressive microenvironment; key regulators of the host tumor immune response are dendritic cells, natural killer cells, macrophages, myeloid derived suppressor cells, and T cells. The function of these immune cells in PC is also influenced by chemotherapeutic agents and the components in tumor microenvironment such as pancreatic stellate cells. Immunotherapy of PC employs monoclonal antibodies/effector cells generated in vitro or vaccination to stimulate antitumor response. Immune therapy in PC has failed to improve overall survival; however, combination therapies comprising immune checkpoint inhibitors and vaccines have been attempted to increase the response. CONCLUSION A number of studies have begun to elucidate the roles of immune cell subtypes and their capacity to function or dysfunction in the tumor microenvironment of PC. It will not be long before immune therapy for PC becomes a clinical reality.
Collapse
Affiliation(s)
- Jae Hyuck Chang
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yongjian Jiang
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Venu G. Pillarisetty
- Department of Surgery, University of Washington Medical Center, Seattle, University of Washington, Seattle, WA
| |
Collapse
|
16
|
Zhang Q, Jiang K, Li Y, Gao D, Sun L, Zhang S, Liu T, Guo K, Liu Y. Histidine-rich glycoprotein function in hepatocellular carcinoma depends on its N-glycosylation status, and it regulates cell proliferation by inhibiting Erk1/2 phosphorylation. Oncotarget 2016; 6:30222-31. [PMID: 26336134 PMCID: PMC4745792 DOI: 10.18632/oncotarget.4997] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 07/31/2015] [Indexed: 11/30/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the third most common cause of cancer mortality. Significantly downregulated histidine-rich glycoprotein (HRG) during the dynamic stages (WB, WB7, and WB11) of neoplastic transformation of WB F344 hepatic oval-like cells was screened out by iTRAQ labeling followed by 2DLC-ESI-MS/MS analysis. HRG expression was significantly lower in HCC tissues. HRG overexpression in Huh7 and MHCC-97H hepatoma cell lines led to decreased cell proliferation, colony-forming ability, and tumor growth, and increased cell apoptosis. HRG could inhibit cell proliferation via the FGF-Erk1/2 signaling pathway by reducing Erk1/2 phosphorylation. On the other hand, the functional expression of HRG was also dependent on the glycosylation status at its N-terminal, especially at the glycosylation site Asn 125. The glycosylation of HRG may play a key competitive role in the interaction between HRG and heparin sulfate for binding bFGF and activating the FGF receptor. These findings provide novel insights into the molecular mechanism of HRG in HCC.
Collapse
Affiliation(s)
- Qinle Zhang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Cancer Research Center, Institute of Biomedical Science, Fudan University, Shanghai 200032, China
| | - Kai Jiang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Cancer Research Center, Institute of Biomedical Science, Fudan University, Shanghai 200032, China
| | - Yan Li
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Dongmei Gao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Lu Sun
- Cancer Research Center, Institute of Biomedical Science, Fudan University, Shanghai 200032, China
| | - Shu Zhang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Tianhua Liu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Cancer Research Center, Institute of Biomedical Science, Fudan University, Shanghai 200032, China
| | - Kun Guo
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yinkun Liu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Cancer Research Center, Institute of Biomedical Science, Fudan University, Shanghai 200032, China
| |
Collapse
|
17
|
Bartneck M, Fech V, Ehling J, Govaere O, Warzecha KT, Hittatiya K, Vucur M, Gautheron J, Luedde T, Trautwein C, Lammers T, Roskams T, Jahnen-Dechent W, Tacke F. Histidine-rich glycoprotein promotes macrophage activation and inflammation in chronic liver disease. Hepatology 2016; 63:1310-24. [PMID: 26699087 DOI: 10.1002/hep.28418] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 12/20/2015] [Indexed: 01/07/2023]
Abstract
UNLABELLED Pathogen- and injury-related danger signals as well as cytokines released by immune cells influence the functional differentiation of macrophages in chronic inflammation. Recently, the liver-derived plasma protein, histidine-rich glycoprotein (HRG), was demonstrated, in mouse tumor models, to mediate the transition of alternatively activated (M2) to proinflammatory (M1) macrophages, which limit tumor growth and metastasis. We hypothesized that liver-derived HRG is a critical endogenous modulator of hepatic macrophage functionality and investigated its implications for liver inflammation and fibrosis by comparing C57BL/6N wild-type (WT) and Hrg(-/-) mice. In homeostatic conditions, hepatic macrophages were overall reduced and preferentially polarized toward the anti-inflammatory M2 subtype in Hrg(-/-) mice. Upon chronic liver damage induced by CCl4 or methionine-choline-deficient (MCD) diet, liver injury and fibrosis were attenuated in Hrg(-/-) , compared to WT, mice. Macrophage populations were reduced and skewed toward M2 polarization in injured livers of Hrg(-/-) mice. Moreover, HRG-deficient mice showed significantly enhanced hepatic vascularization by micro-computed tomography and histology, corroborating proangiogenic activities of M2-polarized liver macrophages. Purified HRG protein induced, but HRG-deficient serum prevented, M1 macrophage differentiation in vitro. Accordingly, Hrg(-/-) mice transplanted with Hrg(+/+) bone marrow, but not Hrg(-/-) -transplanted Hrg(+/+) mice, remained protected from experimental steatohepatitis. Consistent with these findings, patients with chronic hepatitis C and nonalcoholic steatohepatitis significantly up-regulated hepatocytic HRG expression, which was associated with M1 polarization of adjacent macrophages. CONCLUSIONS Liver-derived HRG, similar to alarmins, appears to be an endogenous molecular factor promoting polarization of hepatic macrophages toward the M1 phenotype, thereby promoting chronic liver injury and fibrosis progression, but limiting angiogenesis. Therefore, controlling tissue levels of HRG or PGF might be a promising strategy in chronic inflammatory liver diseases.
Collapse
Affiliation(s)
- Matthias Bartneck
- Department of Medicine III, Helmholtz Institute for Biomedical Engineering, RWTH University-Hospital Aachen, Aachen, Germany
| | - Viktor Fech
- Department of Medicine III, Helmholtz Institute for Biomedical Engineering, RWTH University-Hospital Aachen, Aachen, Germany
| | - Josef Ehling
- Department of Experimental Molecular Imaging, Helmholtz Institute for Biomedical Engineering, RWTH University-Hospital Aachen, Aachen, Germany
| | - Olivier Govaere
- Translational Cell & Tissue Research Unit, Department of Imaging & Pathology, KU Leuven, Belgium
| | - Klaudia Theresa Warzecha
- Department of Medicine III, Helmholtz Institute for Biomedical Engineering, RWTH University-Hospital Aachen, Aachen, Germany
| | | | - Mihael Vucur
- Department of Medicine III, Helmholtz Institute for Biomedical Engineering, RWTH University-Hospital Aachen, Aachen, Germany
| | - Jérémie Gautheron
- Department of Medicine III, Helmholtz Institute for Biomedical Engineering, RWTH University-Hospital Aachen, Aachen, Germany
| | - Tom Luedde
- Department of Medicine III, Helmholtz Institute for Biomedical Engineering, RWTH University-Hospital Aachen, Aachen, Germany
| | - Christian Trautwein
- Department of Medicine III, Helmholtz Institute for Biomedical Engineering, RWTH University-Hospital Aachen, Aachen, Germany
| | - Twan Lammers
- Department of Experimental Molecular Imaging, Helmholtz Institute for Biomedical Engineering, RWTH University-Hospital Aachen, Aachen, Germany.,Department of Targeted Therapeutics, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands.,Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Tania Roskams
- Translational Cell & Tissue Research Unit, Department of Imaging & Pathology, KU Leuven, Belgium
| | - Willi Jahnen-Dechent
- Helmholtz-Institute for Biomedical Engineering, Biointerface Laboratory, RWTH Aachen, Germany
| | - Frank Tacke
- Department of Medicine III, Helmholtz Institute for Biomedical Engineering, RWTH University-Hospital Aachen, Aachen, Germany
| |
Collapse
|
18
|
Fedeli C, Segat D, Tavano R, Bubacco L, De Franceschi G, de Laureto PP, Lubian E, Selvestrel F, Mancin F, Papini E. The functional dissection of the plasma corona of SiO₂-NPs spots histidine rich glycoprotein as a major player able to hamper nanoparticle capture by macrophages. NANOSCALE 2015; 7:17710-17728. [PMID: 26451907 DOI: 10.1039/c5nr05290d] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
A coat of strongly-bound host proteins, or hard corona, may influence the biological and pharmacological features of nanotheranostics by altering their cell-interaction selectivity and macrophage clearance. With the goal of identifying specific corona-effectors, we investigated how the capture of amorphous silica nanoparticles (SiO2-NPs; Ø = 26 nm; zeta potential = -18.3 mV) by human lymphocytes, monocytes and macrophages is modulated by the prominent proteins of their plasma corona. LC MS/MS analysis, western blotting and quantitative SDS-PAGE densitometry show that Histidine Rich Glycoprotein (HRG) is the most abundant component of the SiO2-NP hard corona in excess plasma from humans (HP) and mice (MP), together with minor amounts of the homologous Kininogen-1 (Kin-1), while it is remarkably absent in their Foetal Calf Serum (FCS)-derived corona. HRG binds with high affinity to SiO2-NPs (HRG Kd ∼2 nM) and competes with other plasma proteins for the NP surface, so forming a stable and quite homogeneous corona inhibiting nanoparticles binding to the macrophage membrane and their subsequent uptake. Conversely, in the case of lymphocytes and monocytes not only HRG but also several common plasma proteins can interchange in this inhibitory activity. The depletion of HRG and Kin-1 from HP or their plasma exhaustion by increasing NP concentration (>40 μg ml(-1) in 10% HP) lead to a heterogeneous hard corona, mostly formed by fibrinogen (Fibr), HDLs, LDLs, IgGs, Kallikrein and several minor components, allowing nanoparticle binding to macrophages. Consistently, the FCS-derived SiO2-NP hard corona, mainly formed by hemoglobin, α2 macroglobulin and HDLs but lacking HRG, permits nanoparticle uptake by macrophages. Moreover, purified HRG competes with FCS proteins for the NP surface, inhibiting their recruitment in the corona and blocking NP macrophage capture. HRG, the main component of the plasma-derived SiO2-NPs' hard corona, has antiopsonin characteristics and uniquely confers to these particles the ability to evade macrophage capture.
Collapse
Affiliation(s)
- Chiara Fedeli
- Centro di Ricerca Interdipartimentale per le Biotecnologie Innovative, Università di Padova, via U. Bassi 58/B, I-35131, Padova, Italy. and Dipartimento di Scienze Biomediche, Università di Padova, via U. Bassi 58/B, I-35131, Padova, Italy
| | - Daniela Segat
- Dipartimento di Biologia, Università di Padova, via U. Bassi 58/B, I-35131, Padova, Italy
| | - Regina Tavano
- Centro di Ricerca Interdipartimentale per le Biotecnologie Innovative, Università di Padova, via U. Bassi 58/B, I-35131, Padova, Italy. and Dipartimento di Scienze Biomediche, Università di Padova, via U. Bassi 58/B, I-35131, Padova, Italy
| | - Luigi Bubacco
- Dipartimento di Biologia, Università di Padova, via U. Bassi 58/B, I-35131, Padova, Italy
| | - Giorgia De Franceschi
- Centro di Ricerca Interdipartimentale per le Biotecnologie Innovative, Università di Padova, via U. Bassi 58/B, I-35131, Padova, Italy.
| | - Patrizia Polverino de Laureto
- Centro di Ricerca Interdipartimentale per le Biotecnologie Innovative, Università di Padova, via U. Bassi 58/B, I-35131, Padova, Italy.
| | - Elisa Lubian
- Dipartimento di Scienze Chimiche, Università di Padova, via Marzolo 1, I -35131, Padova, Italy.
| | - Francesco Selvestrel
- Dipartimento di Scienze Chimiche, Università di Padova, via Marzolo 1, I -35131, Padova, Italy.
| | - Fabrizio Mancin
- Dipartimento di Scienze Chimiche, Università di Padova, via Marzolo 1, I -35131, Padova, Italy.
| | - Emanuele Papini
- Centro di Ricerca Interdipartimentale per le Biotecnologie Innovative, Università di Padova, via U. Bassi 58/B, I-35131, Padova, Italy. and Dipartimento di Scienze Biomediche, Università di Padova, via U. Bassi 58/B, I-35131, Padova, Italy
| |
Collapse
|
19
|
Hu H, Jiao F, Han T, Wang LW. Functional significance of macrophages in pancreatic cancer biology. Tumour Biol 2015; 36:9119-26. [PMID: 26411672 PMCID: PMC4689759 DOI: 10.1007/s13277-015-4127-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 09/20/2015] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) is a lethal disease that is usually diagnosed at late stage with few effective therapies. Despite the rapid progress on the genomics and proteomics of the neoplastic cells, therapies that targeted the pancreatic cancer cells proved to be inefficient, which promoted the researchers to turn their attentions to the microenvironment. Currently, various studies had proposed the microenvironment to be a contributing factor for PDA and pervasive researches showed that macrophages within the malignancy correlate with the malignant phenotype of the disease and were reported to a new therapeutic target. Generally, the pro-tumoral effects of macrophages can be summarized as angiogenesis promotion, immunosuppression, matrix remodeling and so on. Hence, a comprehensive understanding of the biologic behaviors of macrophages and their critical role in PDA development may provide new directions for the managements of the lethal disease. In this review, we will summarize the recent advancements on macrophages as pivotal players in PDA biology and the current knowledge about anti-macrophages as a novel strategy against cancer, with the expectation that more efficient therapies will be developed in the near future.
Collapse
Affiliation(s)
- Hai Hu
- Department of Medical Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 650 New Songjiang Road, Shanghai, 201620, China
| | - Feng Jiao
- Department of Medical Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 650 New Songjiang Road, Shanghai, 201620, China
| | - Ting Han
- Department of Medical Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 650 New Songjiang Road, Shanghai, 201620, China
| | - Li-Wei Wang
- Department of Medical Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 650 New Songjiang Road, Shanghai, 201620, China.
| |
Collapse
|
20
|
Roche F, Sipilä K, Honjo S, Johansson S, Tugues S, Heino J, Claesson-Welsh L. Histidine-rich glycoprotein blocks collagen-binding integrins and adhesion of endothelial cells through low-affinity interaction with α2 integrin. Matrix Biol 2015; 48:89-99. [PMID: 26051322 DOI: 10.1016/j.matbio.2015.06.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 06/02/2015] [Accepted: 06/02/2015] [Indexed: 11/19/2022]
Abstract
The plasma protein histidine-rich glycoprotein (HRG) affects the morphology and function of both endothelial cells (ECs) and monocytes/macrophages in cancer. Here, we examined the mechanism of action of HRG's effect on ECs. HRG suppressed adhesion, spreading and migration of ECs specifically on collagen I (COL I) whereas ECs seeded on other extracellular matrix proteins were insensitive to HRG. HRG did not bind specifically to COL I or to the α-integrin binding site on collagen, GFOGER. Furthermore, HRG's inhibition of EC adhesion was not dependent upon heparan sulfate (HS) moieties as heparitinase-treated ECs remained sensitive to HRG. C2C12 cells expressing α2 integrin, the major collagen-binding α-integrin subunit in ECs, showed increased binding of HRG compared with wild type C2C12 cells lacking the α2 subunit. Recombinant α2 I-domain protein bound HRG and to a higher extent when in active conformation. However, the α2 I-domain bound weakly to HRG compared with COL I and the purified α2β1 ectodomain complex failed to retain HRG. We conclude that HRG binds to α2 integrin through low-affinity interactions in a HS-independent manner, thereby blocking EC-adhesion to COL I.
Collapse
Affiliation(s)
- Francis Roche
- Uppsala University, Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Dag Hammarskjöldv. 20, 751 85 Uppsala, Sweden.
| | - Kalle Sipilä
- University of Turku, Department of Biochemistry and MediCity Research Laboratory, FI-20014 Turku, Finland
| | - Satoshi Honjo
- Uppsala University, Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Dag Hammarskjöldv. 20, 751 85 Uppsala, Sweden
| | - Staffan Johansson
- Uppsala University, Department of Medical Biochemistry and Microbiology, Biomedical Center, Box 582, 751 23 Uppsala, Sweden
| | - Sònia Tugues
- Uppsala University, Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Dag Hammarskjöldv. 20, 751 85 Uppsala, Sweden
| | - Jyrki Heino
- University of Turku, Department of Biochemistry and MediCity Research Laboratory, FI-20014 Turku, Finland
| | - Lena Claesson-Welsh
- Uppsala University, Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Dag Hammarskjöldv. 20, 751 85 Uppsala, Sweden
| |
Collapse
|
21
|
Nikpour M, Gustafsson K, Vågesjö E, Seignez C, Giraud A, Phillipson M, Welsh M. Shb deficiency in endothelium but not in leucocytes is responsible for impaired vascular performance during hindlimb ischaemia. Acta Physiol (Oxf) 2015; 214:200-9. [PMID: 25561022 DOI: 10.1111/apha.12448] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 12/31/2014] [Accepted: 01/01/2015] [Indexed: 12/17/2022]
Abstract
AIM Myeloid cells have been suggested to participate in angiogenesis and regulation of vascular function. Shb-deficient mice display both vascular and myeloid cell abnormalities with possible consequences for recovery after hindlimb ischaemia. This study was conducted in order to assess the contribution of Shb deficiency in myeloid cells to impaired vascular function in ischaemia. METHODS Wild type and Shb-deficient mice were subjected to peritoneal vascular endothelial growth factor A (VEGFA) followed by intraperitoneal lavage, after which blood and peritoneal cells were stained for myeloid markers. VEGFA-induced leucocyte recruitment to cremaster muscle was investigated using intravital microscopy of both mouse strains. Blood flow after femoral artery ligation was determined on chimeric mice after bone marrow transplantation. RESULTS No differences in neutrophil numbers or cell surface phenotypes were detected. Moreover, neutrophil extravasation in VEGFA-activated cremaster muscle was unaffected by Shb deficiency. However, blood and peritoneal CXCR4+ monocytes/macrophages were reduced in response to intraperitoneal VEGFA but not lipopolysaccharide (LPS) in the absence of Shb. Furthermore, the macrophage population in ischaemic muscle was unaffected by Shb deficiency after 2 days but reduced 7 days after injury. The bone marrow transplantation experiments revealed that mice with wild type vasculature showed better blood flow than those with Shb-deficient vasculature irrespective of leucocyte genotype. CONCLUSION The observed aberrations in myeloid cell properties in Shb-deficient mice are likely consequences of an abnormal vascular compartment and are not responsible for reduced muscle blood flow. Structural vascular abnormalities seem to be the primary cause of poor vascular performance under provoked vascular stress in this genetic model.
Collapse
Affiliation(s)
- M. Nikpour
- Department of Medical Cell Biology; Uppsala University; Uppsala Sweden
| | - K. Gustafsson
- Department of Medical Cell Biology; Uppsala University; Uppsala Sweden
| | - E. Vågesjö
- Department of Medical Cell Biology; Uppsala University; Uppsala Sweden
| | - C. Seignez
- Department of Medical Cell Biology; Uppsala University; Uppsala Sweden
| | - A. Giraud
- Department of Medical Cell Biology; Uppsala University; Uppsala Sweden
| | - M. Phillipson
- Department of Medical Cell Biology; Uppsala University; Uppsala Sweden
| | - M. Welsh
- Department of Medical Cell Biology; Uppsala University; Uppsala Sweden
| |
Collapse
|
22
|
Deuschle U, Birkel M, Hambruch E, Hornberger M, Kinzel O, Perović-Ottstadt S, Schulz A, Hahn U, Burnet M, Kremoser C. The nuclear bile acid receptor FXR controls the liver derived tumor suppressor histidine-rich glycoprotein. Int J Cancer 2014; 136:2693-704. [PMID: 25363753 DOI: 10.1002/ijc.29312] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 10/16/2014] [Indexed: 12/19/2022]
Abstract
The nuclear bile acid receptor Farnesoid X receptor (FXR) is strongly expressed in liver and intestine, controls bile acid and lipid homeostasis and exerts tumor-protective functions in liver and intestine. Histidine-rich glycoprotein (HRG) is an abundant plasma protein produced by the liver with the proposed function as a pattern recognition molecule involved in the clearance of immune complexes, necrotic cells and pathogens, the modulation of angiogenesis, the normalization of deranged endothelial vessel structure in tumors and tumor suppression. FXR recognition sequences were identified within a human HRG promoter fragment that mediated FXR/FXR-agonist dependent reporter gene activity in vitro. We show that HRG is a novel transcriptional target gene of FXR in human hepatoma cells, human upcyte® primary hepatocytes and 3D human liver microtissues in vitro and in mouse liver in vivo. Prolonged administration of the potent nonsteroidal FXR agonist PX20606 increases HRG levels in mouse plasma. Finally, daily oral administration of this FXR agonist for seven days resulted in a significant increase of HRG levels in the plasma of healthy human male volunteers during a clinical Phase I safety study. HRG might serve as a surrogate marker indicative of liver-specific FXR activation in future human clinical studies. Furthermore, potent FXR agonists might be beneficial in serious health conditions where HRG is reduced, for example, in hepatocellular carcinoma but also other solid cancers, liver failure, sepsis and pre-eclampsia.
Collapse
Affiliation(s)
- Ulrich Deuschle
- Phenex Pharmaceuticals AG, Waldhofer Str. 104, 69123, Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Tugues S, Roche F, Noguer O, Orlova A, Bhoi S, Padhan N, Åkerud P, Honjo S, Selvaraju RK, Mazzone M, Tolmachev V, Claesson-Welsh L. Histidine-rich glycoprotein uptake and turnover is mediated by mononuclear phagocytes. PLoS One 2014; 9:e107483. [PMID: 25243896 PMCID: PMC4171488 DOI: 10.1371/journal.pone.0107483] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 08/13/2014] [Indexed: 01/14/2023] Open
Abstract
Histidine-rich glycoprotein (HRG) is implicated in tumor growth and metastasis by regulation of angiogenesis and inflammation. HRG is produced by hepatocytes and carried to tissues via the circulation. We hypothesized that HRG's tissue distribution and turnover may be mediated by inflammatory cells. Biodistribution parameters were analyzed by injection of radiolabeled, bioactive HRG in the circulation of healthy and tumor-bearing mice. 125I-HRG was cleared rapidly from the blood and taken up in tissues of healthy and tumor-bearing mice, followed by degradation, to an increased extent in the tumor-bearing mice. Steady state levels of HRG in the circulation were unaffected by the tumor disease both in murine tumor models and in colorectal cancer (CRC) patients. Importantly, stromal pools of HRG, detected in human CRC microarrays, were associated with inflammatory cells. In agreement, microautoradiography identified 125I-HRG in blood vessels and on CD45-positive leukocytes in mouse tissues. Moreover, radiolabeled HRG bound in a specific, heparan sulfate-independent manner, to differentiated human monocytic U937 cells in vitro. Suppression of monocyte differentiation by systemic treatment of mice with anti-colony stimulating factor-1 neutralizing antibodies led to reduced blood clearance of radiolabeled HRG and to accumulation of endogenous HRG in the blood. Combined, our data show that mononuclear phagocytes have specific binding sites for HRG and that these cells are essential for uptake of HRG from blood and distribution of HRG in tissues. Thereby, we confirm and extend our previous report that inflammatory cells mediate the effect of HRG on tumor growth and metastatic spread.
Collapse
Affiliation(s)
- Sònia Tugues
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Francis Roche
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Oriol Noguer
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Anna Orlova
- Department of Medicinal Chemistry, Preclinical PET Platform, Uppsala University, Uppsala, Sweden
| | - Sujata Bhoi
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Narendra Padhan
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Peter Åkerud
- Department of Surgical Sciences, Uppsala University, University Hospital, Uppsala, Sweden
| | - Satoshi Honjo
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Ram Kumar Selvaraju
- Department of Medicinal Chemistry, Preclinical PET Platform, Uppsala University, Uppsala, Sweden
| | - Massimiliano Mazzone
- Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, Dept of Oncology, Katholieke Universiteit Leuven, Leuven, Belgium
- Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, Vlaams Instituut voor Biotechnologie, Leuven, Belgium
| | - Vladimir Tolmachev
- Department of Radiology, Oncology and Radiation sciences, The Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Lena Claesson-Welsh
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
- * E-mail:
| |
Collapse
|
24
|
Abstract
During cancer progression, bone marrow derived myeloid cells, including immature myeloid cells and macrophages, progressively accumulate at the primary tumour site where they contribute to the establishment of a tumour promoting microenvironment. A marked infiltration of macrophages into the stromal compartment and the generation of a desmoplastic stromal reaction is a particular characteristic of pancreatic ductal adenocarcinoma (PDA) and is thought to play a key role in disease progression and its response to therapy. Tumour associated macrophages (TAMs) foster PDA tumour progression by promoting angiogenesis, metastasis, and by suppressing an anti-tumourigenic immune response. Recent work also suggests that TAMs contribute to resistance to chemotherapy and to the emergence of cancer stem-like cells. Here we will review the current understanding of the biology and the pro-tumourigenic functions of TAMs in cancer and specifically in PDA, and highlight potential therapeutic strategies to target TAMs and to improve current therapies for pancreatic cancer.
Collapse
Affiliation(s)
- Ainhoa Mielgo
- Liverpool Cancer Research UK Centre, Liverpool L7 8XP, UK
| | | |
Collapse
|
25
|
Cedervall J, Zhang Y, Ringvall M, Thulin A, Moustakas A, Jahnen-Dechent W, Siegbahn A, Olsson AK. HRG regulates tumor progression, epithelial to mesenchymal transition and metastasis via platelet-induced signaling in the pre-tumorigenic microenvironment. Angiogenesis 2013; 16:889-902. [PMID: 23793459 DOI: 10.1007/s10456-013-9363-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2013] [Accepted: 06/12/2013] [Indexed: 01/26/2023]
Abstract
Mice lacking histidine-rich glycoprotein (HRG) display an accelerated angiogenic switch and larger tumors-a phenotype caused by enhanced platelet activation in the HRG-deficient mice. Here we show that platelets induce molecular changes in the pre-tumorigenic environment in HRG-deficient mice, promoting cell survival, angiogenesis and epithelial-to-mesenchymal transition (EMT) and that these effects involved signaling via TBK1, Akt2 and PDGFRβ. These early events subsequently translate into an enhanced rate of spontaneous metastasis to distant organs in mice lacking HRG. Later in tumor development characteristic features of pathological angiogenesis, such as decreased perfusion and pericyte coverage, are more pronounced in HRG-deficient mice. At this stage, platelets are essential to support the larger tumor volumes formed in mice lacking HRG by keeping their tumor vasculature sufficiently functional. We conclude that HRG-deficiency promotes tumor progression via enhanced platelet activity and that platelets play a dual role in this process. During early stages of transformation, activated platelets promote tumor cell survival, the angiogenic switch and invasiveness. In the more progressed tumor, platelets support the enhanced pathological angiogenesis and hence increased tumor growth seen in the absence of HRG. Altogether, our findings strengthen the notion of HRG as a potent tumor suppressor, with capacity to attenuate the angiogenic switch, tumor growth, EMT and subsequent metastatic spread, by regulating platelet activity.
Collapse
Affiliation(s)
- Jessica Cedervall
- Department of Medical Biochemistry and Microbiology, Uppsala Biomedical Center, Uppsala University, Husargatan 3, 751 23, Uppsala, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Patel KK, Poon IKH, Talbo GH, Perugini MA, Taylor NL, Ralph TJ, Hoogenraad NJ, Hulett MD. New method for purifying histidine-rich glycoprotein from human plasma redefines its functional properties. IUBMB Life 2013; 65:550-63. [DOI: 10.1002/iub.1168] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 03/05/2013] [Indexed: 12/12/2022]
|
27
|
Tang X, Mo C, Wang Y, Wei D, Xiao H. Anti-tumour strategies aiming to target tumour-associated macrophages. Immunology 2013; 138:93-104. [PMID: 23113570 DOI: 10.1111/imm.12023] [Citation(s) in RCA: 200] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Revised: 10/19/2012] [Accepted: 10/22/2012] [Indexed: 02/05/2023] Open
Abstract
Tumour-associated macrophages (TAMs) represent a predominant population of inflammatory cells that present in solid tumours. TAMs are mostly characterized as alternatively activated M2-like macrophages and are known to orchestrate nearly all stages of tumour progression. Experimental investigations indicate that TAMs contribute to drug-resistance and radio-protective effects, and clinical evidence shows that an elevated number of TAMs and their M2 profile are correlated with therapy failure and poor prognosis in cancer patients. Recently, many studies on TAM-targeted strategies have made significant progress and some pilot works have achieved encouraging results. Among these, connections between some anti-tumour drugs and their influence on TAMs have been suggested. In this review, we will summarize recent advances in TAM-targeted strategies for tumour therapy. Based on the proposed mechanisms, those strategies are grouped into four categories: (i) inhibiting macrophage recruitment; (ii) suppressing TAM survival; (iii) enhancing M1-like tumoricidal activity of TAMs; (iv) blocking M2-like tumour-promoting activity of TAMs. It is desired that further attention be drawn to this research field and more effort be made to promote TAM-targeted tumour therapy.
Collapse
Affiliation(s)
- Xiaoqiang Tang
- Department of Geriatrics, State Key Laboratory of Biotherapy and Cancer Centre, West China Hospital, Sichuan University, Chengdu, China
| | | | | | | | | |
Collapse
|
28
|
New Insights into the Functions of Histidine-Rich Glycoprotein. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 304:467-93. [DOI: 10.1016/b978-0-12-407696-9.00009-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
29
|
Chen P, Bonaldo P. Role of macrophage polarization in tumor angiogenesis and vessel normalization: implications for new anticancer therapies. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 301:1-35. [PMID: 23317816 DOI: 10.1016/b978-0-12-407704-1.00001-4] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Angiogenesis, the formation of new capillary blood vessels from preexisting vasculature, is one of the hallmarks of cancer that is pivotal for tumor growth and metastasis. Tumor vessels are known to be abnormal, with typically aberrant, leaky and disordered vessels. Thus, the combination of angiogenesis inhibition and vessel normalization is a potential strategy for anticancer therapy. The solid tumor is composed of not only cancer cells, but also the nonmalignant resident stromal cells, such as bone-marrow-derived cells (BMDCs) and cancer-associated fibroblasts (CAFs). Tumor-associated macrophages (TAMs) are the most abundant cell components of BMDCs, which play a significant role in promoting tumor progression. Accumulating evidences from both patient biopsies and experimental animal models have shown that TAMs function in tumor angiogenesis and vessel abnormalization in a density- and phenotype-dependent manner. This chapter will discuss the evidence for the factors and signaling pathways that are involved in macrophage recruitment and polarization in the tumor microenvironment, and it summarizes the role and underlying molecular mechanisms of macrophage polarization in tumor angiogenesis and vessel normalization. In addition, an overview of the potential of targeting TAM polarization for anticancer therapy will be provided.
Collapse
Affiliation(s)
- Peiwen Chen
- Department of Biomedical Sciences, University of Padova, Padova, Italy.
| | | |
Collapse
|
30
|
Lee HW, Choi HJ, Ha SJ, Lee KT, Kwon YG. Recruitment of monocytes/macrophages in different tumor microenvironments. Biochim Biophys Acta Rev Cancer 2012; 1835:170-9. [PMID: 23287570 DOI: 10.1016/j.bbcan.2012.12.007] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Revised: 12/16/2012] [Accepted: 12/20/2012] [Indexed: 01/21/2023]
Abstract
After emigration from the bone marrow into the peripheral blood, monocytes enter tissues and differentiate into macrophages. Monocytes/macrophages have many roles in immune regulation, angiogenesis, and tumor metastasis and invasion. In addition, studies have revealed that these cells are essential to tumor progression. Recently, an accumulation of evidence has indicated that macrophages in distinct regions of tumor masses have distinct origins. For instance, classical monocytes appear to be a major source of macrophages in tumor epithelial, perivascular, and hypoxic regions. In contrast, non-classical monocytes are an important source of macrophages in the tumor perivascular region. During the past century, it has been demonstrated that several chemoattractants can regulate the recruitment of monocytes/macrophages to tumor sites. Despite the importance of monocytes/macrophages in tumor progression, there had been, until recently, no efforts to summarize receptor-ligand pairs between tumor-derived chemokines and corresponding receptors in monocytes in different microenvironments. In this review, we present a cohesive view of the distinct expression patterns of chemokine receptors in two different monocyte subsets (classical and non-classical monocytes) and describe their roles in monocyte/macrophage recruitment into distinct tumor microenvironments. This review provides insight into the behavior of monocytes/macrophages in different tumor microenvironments.
Collapse
Affiliation(s)
- Heon-Woo Lee
- Department of Biochemistry, Yonsei University, Seoul, Republic of Korea
| | | | | | | | | |
Collapse
|
31
|
Hale JS, Li M, Sinyuk M, Jahnen-Dechent W, Lathia JD, Silverstein RL. Context dependent role of the CD36--thrombospondin--histidine-rich glycoprotein axis in tumor angiogenesis and growth. PLoS One 2012; 7:e40033. [PMID: 22808089 PMCID: PMC3393734 DOI: 10.1371/journal.pone.0040033] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Accepted: 06/03/2012] [Indexed: 11/19/2022] Open
Abstract
The angiogenic switch is a promising therapeutic target in cancer. Work by our laboratory and others has described an important endogenous anti-angiogenic pathway mediated by interactions of CD36, a receptor on microvascular endothelial cells, with proteins containing thrombospondin (TSP) type I repeat domains (TSR). Recent studies revealed that circulating Histidine Rich Glycoprotein (HRG) inhibits the anti-angiogenic potential of the CD36-TSR pathway by functioning as a decoy receptor that binds and sequesters TSR proteins. As tumors of different origin display variable expression profiles of numerous targets, we hypothesized that the TSP-CD36-HRG axis regulates vascularization and growth in the tumor microenvironment in a context, or tumor type, dependent manner. Growth of Lewis Lung Carcinoma (LL2) and B16F1 Melanoma tumor cell implants in syngeneic wild type (WT), hrg, or cd36 null mice were used as a model to interrogate this signaling axis. LL2 tumor volumes were greater in cd36 null mice and smaller in hrg null mice compared to WT. Immunofluorescent staining showed increased vascularity in cd36 null vs. WT and WT vs. hrg null mice. No differences in tumor growth or vascularity were observed with B16F1 implants, consistent with lack of expression of TSP-1 in B16F1 cells. When TSR expression was induced in B16F1 cells by cDNA transfection, tumor growth and vascularity were similar to that seen with LL2 cells. These data show a role for CD36-mediated anti-angiogenic activity in the tumor microenvironment when TSR proteins are available and demonstrate that HRG modulates this activity. Further, they suggest a mechanism by which tumor microenvironments may regulate sensitivity to TSR containing proteins.
Collapse
MESH Headings
- Animals
- CD36 Antigens/deficiency
- CD36 Antigens/genetics
- Carcinoma, Lewis Lung/blood supply
- Carcinoma, Lewis Lung/genetics
- Carcinoma, Lewis Lung/metabolism
- Carcinoma, Lewis Lung/pathology
- Cell Line, Tumor
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Gene Expression Regulation, Neoplastic
- Male
- Melanoma, Experimental/blood supply
- Melanoma, Experimental/genetics
- Melanoma, Experimental/metabolism
- Melanoma, Experimental/pathology
- Mice
- Mice, Knockout
- Neovascularization, Pathologic
- Organ Specificity
- Protein Binding
- Proteins/genetics
- Proteins/metabolism
- RNA, Small Interfering/genetics
- Signal Transduction
- Skin Neoplasms/blood supply
- Skin Neoplasms/genetics
- Skin Neoplasms/metabolism
- Skin Neoplasms/pathology
- Thrombospondin 1/genetics
- Thrombospondin 1/metabolism
- Tumor Burden/genetics
- Tumor Microenvironment/genetics
Collapse
Affiliation(s)
- James Scott Hale
- Department of Biological, Geological and Environmental Sciences, Cleveland State University, Cleveland, Ohio, United States of America
- Department of Cell Biology, Lerner Research Institute, Cleveland Clinic Foundation and Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Cleveland, Ohio, United States of America
| | - Meizhang Li
- Department of Cell Biology, Lerner Research Institute, Cleveland Clinic Foundation and Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Cleveland, Ohio, United States of America
| | - Maksim Sinyuk
- Department of Cell Biology, Lerner Research Institute, Cleveland Clinic Foundation and Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Cleveland, Ohio, United States of America
| | - Willi Jahnen-Dechent
- Helmholtz Institute for Biomedical Engineering, Rheinisch-Westfälische Technische Hochschule Aachen University Hospital, Aachen, Germany
| | - Justin Durla Lathia
- Department of Cell Biology, Lerner Research Institute, Cleveland Clinic Foundation and Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Cleveland, Ohio, United States of America
| | - Roy Lee Silverstein
- Department of Biological, Geological and Environmental Sciences, Cleveland State University, Cleveland, Ohio, United States of America
- Department of Cell Biology, Lerner Research Institute, Cleveland Clinic Foundation and Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Cleveland, Ohio, United States of America
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|