1
|
Gupta S, Sharma A, Petrovski G, Verma RS. Vascular reconstruction of the decellularized biomatrix for whole-organ engineering-a critical perspective and future strategies. Front Bioeng Biotechnol 2023; 11:1221159. [PMID: 38026872 PMCID: PMC10680456 DOI: 10.3389/fbioe.2023.1221159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 10/09/2023] [Indexed: 12/01/2023] Open
Abstract
Whole-organ re-engineering is the most challenging goal yet to be achieved in tissue engineering and regenerative medicine. One essential factor in any transplantable and functional tissue engineering is fabricating a perfusable vascular network with macro- and micro-sized blood vessels. Whole-organ development has become more practical with the use of the decellularized organ biomatrix (DOB) as it provides a native biochemical and structural framework for a particular organ. However, reconstructing vasculature and re-endothelialization in the DOB is a highly challenging task and has not been achieved for constructing a clinically transplantable vascularized organ with an efficient perfusable capability. Here, we critically and articulately emphasized factors that have been studied for the vascular reconstruction in the DOB. Furthermore, we highlighted the factors used for vasculature development studies in general and their application in whole-organ vascular reconstruction. We also analyzed in detail the strategies explored so far for vascular reconstruction and angiogenesis in the DOB for functional and perfusable vasculature development. Finally, we discussed some of the crucial factors that have been largely ignored in the vascular reconstruction of the DOB and the future directions that should be addressed systematically.
Collapse
Affiliation(s)
- Santosh Gupta
- Stem Cell and Molecular Biology, Laboratory, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences. Indian Institute of Technology Madras, Chennai, India
- Center for Eye Research and Innovative Diagnostics, Department of Ophthalmology, Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Akriti Sharma
- Stem Cell and Molecular Biology, Laboratory, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences. Indian Institute of Technology Madras, Chennai, India
| | - Goran Petrovski
- Center for Eye Research and Innovative Diagnostics, Department of Ophthalmology, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Ophthalmology, Oslo University Hospital, Oslo, Norway
- Department of Ophthalmology, University of Split School of Medicine and University Hospital Centre, Split, Croatia
| | - Rama Shanker Verma
- Stem Cell and Molecular Biology, Laboratory, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences. Indian Institute of Technology Madras, Chennai, India
| |
Collapse
|
2
|
Korbecki J, Kupnicka P, Barczak K, Bosiacki M, Ziętek P, Chlubek D, Baranowska-Bosiacka I. The Role of CXCR1, CXCR2, CXCR3, CXCR5, and CXCR6 Ligands in Molecular Cancer Processes and Clinical Aspects of Acute Myeloid Leukemia (AML). Cancers (Basel) 2023; 15:4555. [PMID: 37760523 PMCID: PMC10526350 DOI: 10.3390/cancers15184555] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/09/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Acute myeloid leukemia (AML) is a type of leukemia known for its unfavorable prognoses, prompting research efforts to discover new therapeutic targets. One area of investigation involves examining extracellular factors, particularly CXC chemokines. While CXCL12 (SDF-1) and its receptor CXCR4 have been extensively studied, research on other CXC chemokine axes in AML is less developed. This study aims to bridge that gap by providing an overview of the significance of CXC chemokines other than CXCL12 (CXCR1, CXCR2, CXCR3, CXCR5, and CXCR6 ligands and CXCL14 and CXCL17) in AML's oncogenic processes. We explore the roles of all CXC chemokines other than CXCL12, in particular CXCL1 (Gro-α), CXCL8 (IL-8), CXCL10 (IP-10), and CXCL11 (I-TAC) in AML tumor processes, including their impact on AML cell proliferation, bone marrow angiogenesis, interaction with non-leukemic cells like MSCs and osteoblasts, and their clinical relevance. We delve into how they influence prognosis, association with extramedullary AML, induction of chemoresistance, effects on bone marrow microvessel density, and their connection to French-American-British (FAB) classification and FLT3 gene mutations.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (J.K.); (P.K.); (M.B.); (D.C.)
- Department of Anatomy and Histology, Collegium Medicum, University of Zielona Góra, Zyty 28, 65-046 Zielona Góra, Poland
| | - Patrycja Kupnicka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (J.K.); (P.K.); (M.B.); (D.C.)
| | - Katarzyna Barczak
- Department of Conservative Dentistry and Endodontics, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland;
| | - Mateusz Bosiacki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (J.K.); (P.K.); (M.B.); (D.C.)
| | - Paweł Ziętek
- Department of Orthopaedics, Traumatology and Orthopaedic Oncology, Pomeranian Medical University, Unii Lubelskiej 1, 71-252 Szczecin, Poland;
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (J.K.); (P.K.); (M.B.); (D.C.)
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (J.K.); (P.K.); (M.B.); (D.C.)
| |
Collapse
|
3
|
Li C, Kuang K, Du J, Eymin B, Jia T. Far beyond anti-angiogenesis: Benefits for anti-basicFGF therapy in cancer. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119253. [PMID: 35259425 DOI: 10.1016/j.bbamcr.2022.119253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/02/2022] [Accepted: 03/03/2022] [Indexed: 12/28/2022]
Abstract
Basic FGF (bFGF) was discovered as a typical inducer of angiogenesis and has already been studied for 3 decades. Recent evidence indicates that bFGF plays different roles and controls signaling pathways that participate in the hallmarks of cancer, underscoring bFGF an appealing target for anti-cancer therapy. However, the early clinical trials designed to block bFGF signaling showed safety without satisfiable benefits for cancer patients. In this review, we firstly discuss bFGF's canonical signaling pathways and later review newly identified bFGF's functions that contribute to the cancer hallmarks besides its typical role in angiogenesis. After, we summarize the role of bFGF as a therapeutic target in response to different cancer therapies including radiotherapy, chemotherapy, targeted therapy, immunotherapy, and highlight the difficulties we must solve regarding the design of drugs targeting specifically bFGF. We also emphasize the need, especially for natural bFGF traps, to deepen their molecular mechanisms of action considering the specific context of cancer with different FGFR status, as well as the urgence of stratifying patients for both anti-bFGF first line and second line anti-cancer therapy. Finally, a perspective on potential feed-forward oncogenic signaling pathways mediated by bFGF is made. We discuss the importance of developing additional robust biomarkers to select patients who will benefit from bFGF-targeted therapy, as well as the rationale of developing combinatory therapies targeting either bFGF and/or its intracellular (co)effectors. This would ultimately provide novel therapeutic strategies to fight cancer.
Collapse
Affiliation(s)
- ChunYan Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - KeLi Kuang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - JunRong Du
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Beatrice Eymin
- INSERM U1209, CNRS UMR5309, Institute For Advanced Biosciences, 38700 La Tronche, France; University Grenoble Alpes, 38000 Grenoble, France
| | - Tao Jia
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
4
|
Al-Shammari AM, Al-Mudhafr MA, Chalap Al- Grawi ED, Al-Hili ZA, Yaseen N. Newcastle disease virus suppresses angiogenesis in mammary adenocarcinoma models. BULGARIAN JOURNAL OF VETERINARY MEDICINE 2022. [DOI: 10.15547/bjvm.2020-0019] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Cancer cells heavily utilise angiogenesis process to increase vascularisation for tumour mass growth and spread, so targeting this process is important to create an effective therapy. The AMHA1 strain of Newcastle disease virus (NDV) is an RNA virus with natural oncotropism. NDV induces direct tumour cytolysis, apoptosis, and immune stimulation. This work aimed to test NDV anti-angiogenic activity in a breast cancer model. To evaluate NDV’s antitumour effect in vivo, NDV was tested against mammary adenocarcinoma AN3 transplanted in syngeneic immunocompetent mice. In vivo antiangiogenic activity was evaluated by quantifying the blood vessels in treated and control tumour sections. In vitro experiments that exposed AMN3 mammary adenocarcinoma cells and Hep-2 laryngeal carcinoma cells to NDV at different time intervals were performed to identify the exact mechanism of anti-angiogenesis by using angiogenesis microarray slides. In vivo results showed significant tumour regression and significant decrease in blood vessel formation in treated tumour sections. The in vitro microarray analysis of 14 different angiogenesis factors revealed that NDV downregulated angiopoietin-1, angiopoietin-2, and epidermal growth factor in mammary adenocarcinoma cells. However, NDV elicited a different effect on Hep-2 as represented by the downregulation of inducible protein 10, intracellular adhesion molecule-1, and basic fibroblast growth factor beta in NDV-infected tumour cells. It was found out that microarray analysis results helped interpret the in vivo data. The results suggested that the NDV oncolytic strain reduced angiogenesis by interfering with angiogenesis factors that might reduce tumour cell proliferation, infiltration, and invasion.
Collapse
Affiliation(s)
- A. M. Al-Shammari
- University of Mustansiriyah, Iraqi Center for Cancer and Medical Genetic Research, Experimental Therapy Department, Baghdad, Iraq
| | - M. A. Al-Mudhafr
- University of Kufa, Faculty of Veterinary Medicine, Department of Microbiology
| | | | - Z. A. Al-Hili
- University of Mustansiriyah, Iraqi Center for Cancer and Medical Genetic Research, Experimental Therapy Department, Baghdad, Iraq
| | - N. Yaseen
- University of Mustansiriyah, Iraqi Center for Cancer and Medical Genetic Research, Experimental Therapy Department, Baghdad, Iraq
| |
Collapse
|
5
|
CXCL13 in Cancer and Other Diseases: Biological Functions, Clinical Significance, and Therapeutic Opportunities. Life (Basel) 2021; 11:life11121282. [PMID: 34947813 PMCID: PMC8708574 DOI: 10.3390/life11121282] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/31/2021] [Accepted: 11/12/2021] [Indexed: 12/11/2022] Open
Abstract
The development of cancer is a multistep and complex process involving interactions between tumor cells and the tumor microenvironment (TME). C-X-C chemokine ligand 13 (CXCL13) and its receptor, CXCR5, make crucial contributions to this process by triggering intracellular signaling cascades in malignant cells and modulating the sophisticated TME in an autocrine or paracrine fashion. The CXCL13/CXCR5 axis has a dominant role in B cell recruitment and tertiary lymphoid structure formation, which activate immune responses against some tumors. In most cancer types, the CXCL13/CXCR5 axis mediates pro-neoplastic immune reactions by recruiting suppressive immune cells into tumor tissues. Tobacco smoke and haze (smohaze) and the carcinogen benzo(a)pyrene induce the secretion of CXCL13 by lung epithelial cells, which contributes to environmental lung carcinogenesis. Interestingly, the knockout of CXCL13 inhibits benzo(a)pyrene-induced lung cancer and azoxymethane/dextran sodium sulfate-induced colorectal cancer in mice. Thus, a better understanding of the context-dependent functions of the CXCL13/CXCR5 axis in tumor tissue and the TME is required to design an efficient immune-based therapy. In this review, we summarize the molecular events and TME alterations caused by CXCL13/CXCR5 and briefly discuss the potentials of agents targeting this axis in different malignant tumors.
Collapse
|
6
|
Ma Q, Chen Y, Qin Q, Guo F, Wang YS, Li D. CXCL13 expression in mouse 4T1 breast cancer microenvironment elicits antitumor immune response by regulating immune cell infiltration. PRECISION CLINICAL MEDICINE 2021; 4:155-167. [PMID: 35693216 PMCID: PMC8982548 DOI: 10.1093/pcmedi/pbab020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/14/2021] [Accepted: 07/29/2021] [Indexed: 02/05/2023] Open
Abstract
Breast cancer is the most commonly diagnosed cancer type and the leading cause of cancer-related deaths among women worldwide. Previous studies have reported contradictory performance of chemokine CXC motif ligand 13 (CXCL13) in breast cancer. In this study, The Cancer Genome Atlas database analysis revealed that CXCL13 was overexpressed in various human cancers including breast carcinoma, and associated with good clinical prognosis in breast cancer. Flow cytometry detection also found upregulated intracellular CXCL13 expression in human breast cancer cell lines. To explore the possible role of CXCL13 in the breast cancer microenvironment, mouse triple negative breast cancer (TNBC) was lentivirally transfected to stably overexpress mouse CXCL13 (4T1-CXCL13). Both parental 4T1 and 4T1-CXCL13 strains showed no in vitro or in vivo endogenous cell surface CXCR5 expression. In immune-competent BALB/c mice, the in vivo tumor growth of 4T1-CXCL13 was significantly inhibited and even completely eradicated, accompanied with increased infiltrations of CD4+, CD8+ T lymphocytes and CD11b+CD11c+ DCs. Further investigations showed that CXCL13 expression in the 4T1 tumor microenvironment elicited long-term antitumor immune memory, and rejection of distal parental tumor. The antitumor activity of CXCL13 was remarkedly impaired in BALB/cA-nu nude mice, or in BALB/c mice with CD8+ T lymphocyte or NK cell depletion. Our investigation indicated that CXCL13 expression in TNBC triggered effective antitumor immunity by chemoattracting immune cell infiltrations and could be considered as a novel prognostic marker for TNBC.
Collapse
Affiliation(s)
- Qizhi Ma
- Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yue Chen
- Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qing Qin
- Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Fuchun Guo
- Institute of Drug Clinical Trial, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yong-sheng Wang
- Institute of Drug Clinical Trial, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Dan Li
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, and Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
7
|
Chen W, Wang Y, Zhou T, Xu Y, Zhan J, Wu J. CXCL13 Is Involved in the Lipopolysaccharide-Induced Hyperpermeability of Umbilical Vein Endothelial Cells. Inflammation 2021; 43:1789-1796. [PMID: 32500306 PMCID: PMC7476967 DOI: 10.1007/s10753-020-01253-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Sepsis is a disease that is characterized by a severe systemic inflammatory response to microbial infection and lipopolysaccharide (LPS) and is a well-known inducer of sepsis, as well as endothelial cell hyperpermeability. In the present study, we confirm the elevation of CXC chemokine ligand 13 (CXCL13) in sepsis patients. We also show that LPS exposure increases the release of CXCL13, as well as the mRNA and protein expression of CXCL13 and its receptor, CXC chemokine receptor 5 (CXCR5) in human umbilical vein endothelial cells (HUVECs) in a dose- and time-dependent manner. We also examined the effects of CXCL13 knockdown on LPS-mediated endothelial hyperpermeability and tight junction (TJ) protein expression in HUVECs. Our results show that HUVECs exposed to LPS result in a significant decrease in transendothelial electrical resistance (TER) and TJ protein (Zonula occluden-1, occludin, and claudin-4) expression, and a notable increase in fluorescein isothiocyanate (FITC)-dextran flux and p38 phosphorylation, which was partially reversed by CXCL13 knockdown. Recombinant CXCL13 treatment had a similar effect as LPS exposure, which was attenuated by a p38 inhibitor, SB203580. Moreover, the CXCL13-neutralizing antibody significantly increased the survival rate of LPS-induced sepsis mice. Collectively, our results show that CXCL13 plays a key role in LPS-induced endothelium hyperpermeability via regulating p38 signaling and suggests that therapeutically targeting CXCL13 may be beneficial for the treatment of sepsis.
Collapse
Affiliation(s)
- Wen Chen
- Department of General Practice, Hangzhou First People's Hospital affiliated to Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yi Wang
- Department of Emergency, Hangzhou First People's Hospital affiliated to Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ting Zhou
- Department of Emergency, Hangzhou First People's Hospital affiliated to Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yuansheng Xu
- Department of Emergency, Hangzhou First People's Hospital affiliated to Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jianwei Zhan
- Department of Emergency, Hangzhou First People's Hospital affiliated to Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jinhong Wu
- Department of Emergency, Hangzhou First People's Hospital affiliated to Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
8
|
Korbecki J, Kojder K, Kapczuk P, Kupnicka P, Gawrońska-Szklarz B, Gutowska I, Chlubek D, Baranowska-Bosiacka I. The Effect of Hypoxia on the Expression of CXC Chemokines and CXC Chemokine Receptors-A Review of Literature. Int J Mol Sci 2021; 22:ijms22020843. [PMID: 33467722 PMCID: PMC7830156 DOI: 10.3390/ijms22020843] [Citation(s) in RCA: 125] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/06/2021] [Accepted: 01/12/2021] [Indexed: 12/26/2022] Open
Abstract
Hypoxia is an integral component of the tumor microenvironment. Either as chronic or cycling hypoxia, it exerts a similar effect on cancer processes by activating hypoxia-inducible factor-1 (HIF-1) and nuclear factor (NF-κB), with cycling hypoxia showing a stronger proinflammatory influence. One of the systems affected by hypoxia is the CXC chemokine system. This paper reviews all available information on hypoxia-induced changes in the expression of all CXC chemokines (CXCL1, CXCL2, CXCL3, CXCL4, CXCL5, CXCL6, CXCL7, CXCL8 (IL-8), CXCL9, CXCL10, CXCL11, CXCL12 (SDF-1), CXCL13, CXCL14, CXCL15, CXCL16, CXCL17) as well as CXC chemokine receptors—CXCR1, CXCR2, CXCR3, CXCR4, CXCR5, CXCR6, CXCR7 and CXCR8. First, we present basic information on the effect of these chemoattractant cytokines on cancer processes. We then discuss the effect of hypoxia-induced changes on CXC chemokine expression on the angiogenesis, lymphangiogenesis and recruitment of various cells to the tumor niche, including myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), regulatory T cells (Tregs) and tumor-infiltrating lymphocytes (TILs). Finally, the review summarizes data on the use of drugs targeting the CXC chemokine system in cancer therapies.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (P.K.); (P.K.); (D.C.)
| | - Klaudyna Kojder
- Department of Anaesthesiology and Intensive Care, Pomeranian Medical University in Szczecin, Unii Lubelskiej 1, 71-281 Szczecin, Poland;
| | - Patrycja Kapczuk
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (P.K.); (P.K.); (D.C.)
| | - Patrycja Kupnicka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (P.K.); (P.K.); (D.C.)
| | - Barbara Gawrońska-Szklarz
- Department of Pharmacokinetics and Therapeutic Drug Monitoring, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland;
| | - Izabela Gutowska
- Department of Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72 Av., 70-111 Szczecin, Poland;
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (P.K.); (P.K.); (D.C.)
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (P.K.); (P.K.); (D.C.)
- Correspondence: ; Tel.: +48-914661515
| |
Collapse
|
9
|
Tan Y, Qiao Y, Chen Z, Liu J, Guo Y, Tran T, Tan KS, Wang DY, Yan Y. FGF2, an Immunomodulatory Factor in Asthma and Chronic Obstructive Pulmonary Disease (COPD). Front Cell Dev Biol 2020; 8:223. [PMID: 32300593 PMCID: PMC7142218 DOI: 10.3389/fcell.2020.00223] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 03/16/2020] [Indexed: 12/14/2022] Open
Abstract
The fibroblast growth factor 2 (FGF2) is a potent mitogenic factor belonging to the FGF family. It plays a role in airway remodeling associated with chronic inflammatory airway diseases, including asthma and chronic obstructive pulmonary disease (COPD). Recently, research interest has been raised in the immunomodulatory function of FGF2 in asthma and COPD, through its involvement in not only the regulation of inflammatory cells but also its participation as a mediator between immune cells and airway structural cells. Herein, this review provides the current knowledge on the biology of FGF2, its expression pattern in asthma and COPD patients, and its role as an immunomodulatory factor. The potential that FGF2 is involved in regulating inflammation indicates that FGF2 could be a therapeutic target for chronic inflammatory diseases.
Collapse
Affiliation(s)
- Yuanyang Tan
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | | | - Zhuanggui Chen
- Department of Pediatrics, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jing Liu
- Department of Respiratory Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Yanrong Guo
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Thai Tran
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kai Sen Tan
- Department of Otolaryngology, Yong Loo Lin School of Medicine, University Health System, National University of Singapore, Singapore, Singapore
| | - De-Yun Wang
- Department of Otolaryngology, Yong Loo Lin School of Medicine, University Health System, National University of Singapore, Singapore, Singapore
| | - Yan Yan
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China.,Center for Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| |
Collapse
|
10
|
Taniguchi T, Miyagawa T, Toyama S, Yamashita T, Nakamura K, Saigusa R, Ichimura Y, Takahashi T, Toyama T, Yoshizaki A, Sato S, Asano Y. CXCL13 produced by macrophages due to Fli1 deficiency may contribute to the development of tissue fibrosis, vasculopathy and immune activation in systemic sclerosis. Exp Dermatol 2018; 27:1030-1037. [PMID: 29947047 DOI: 10.1111/exd.13724] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 06/13/2018] [Accepted: 06/22/2018] [Indexed: 02/06/2023]
Abstract
CXCL13, a chemokine for B cells, follicular T cells, T helper 17 cells, and regulatory T cells, is reported to contribute to the development of systemic sclerosis (SSc), reflecting aberrant activation of immune system. To better understand the role of CXCL13 in SSc, we investigated the influence of Fli1 deficiency, a potential predisposing factor of this disease, on CXCL13 expression and assessed the clinical correlation of serum CXCL13 levels by multivariate regression analysis. Haploinsufficient loss of Fli1 remarkably induced CXCL13 expression in murine peritoneal macrophages, while gene silencing of FLI1 did not affect the expression of CXCL13 in human dermal fibroblasts and human dermal microvascular endothelial cells. Serum CXCL13 levels were elevated in SSc patients compared with healthy controls and correlated positively with skin score and negatively with pulmonary function test results. SSc patients with elevated serum CXCL13 levels had longer disease duration, diffuse cutaneous involvement, interstitial lung disease (ILD), heart involvement, pulmonary arterial hypertension, Raynaud's phenomenon, pitting scars, digital ulcers, telangiectasia, and high serum IgG levels more frequently than the other patients. In particular, serum CXCL13 levels were associated with ILD and digital ulcers by multivariate regression analysis. Taken together, these results indicate that CXCL13 expression is upregulated by Fli1 deficiency in macrophages, potentially contributing to the development of tissue fibrosis, vasculopathy and immune activation in SSc, especially ILD and digital ulcers.
Collapse
Affiliation(s)
- Takashi Taniguchi
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Takuya Miyagawa
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Satoshi Toyama
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Takashi Yamashita
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Kouki Nakamura
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Ryosuke Saigusa
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Yohei Ichimura
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Takehiro Takahashi
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Tetsuo Toyama
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Ayumi Yoshizaki
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Shinichi Sato
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Yoshihide Asano
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
11
|
Sakamoto S, Takahashi H, Tan X, Inoue Y, Nomura Y, Arai Y, Fujino Y, Kawashima H, Yanagi Y. Changes in multiple cytokine concentrations in the aqueous humour of neovascular age-related macular degeneration after 2 months of ranibizumab therapy. Br J Ophthalmol 2017; 102:448-454. [PMID: 28765149 PMCID: PMC5890644 DOI: 10.1136/bjophthalmol-2017-310284] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 07/06/2017] [Accepted: 07/11/2017] [Indexed: 12/12/2022]
Abstract
Purpose To determine changes in multiple cytokine concentrations in the anterior chamber during the induction phase of ranibizumab treatment in patients with neovascular age-related macular degeneration (AMD). Methods This prospective study included 48 treatment-naïve neovascular AMD eyes of 48 patients who received three consecutive monthly injections of ranibizumab at the Japan Community Health Care Organization Tokyo Shinjuku Medical Center between November 2010 and August 2012. We collected ~0.2 mL aqueous humour before the first and third (2 months later) injections. Controls were 80 eyes with cataracts without retinal disease. The cytokines C-X-C motif chemokine ligand 1 (CXCL1), interferon-γ-induced protein 10 (IP-10), C-X-C motif chemokine ligand 12 (CXCL12), C-X-C motif chemokine ligand 13 (CXCL13), monocyte chemoattractant protein 1 (MCP-1), CCL11, C-C motif chemokine ligand 11 (CCL11), interleukin-6 (IL-6), interleukin-10 (IL-10) and matrix metalloproteinase 9 (MMP-9) were analysed using multiplex cytokine assays. Results Mean ages of the patients with AMD and controls were 73 and 75 years, respectively, and 31 (65%) and 37 (46%) subjects were men, respectively. Polypoidal choroidal vasculopathy was found in 27 eyes (56%). Mean concentrations of cytokines in aqueous humour in patients with neovascular AMD before the first and third ranibizumab injections were as follows (in pg/mL): CXCL1, 8.4 and 3.3; IP-10, 110 and 55; CXCL12, 480 and 240; CXCL13, 9.2 and 2.6; MCP-1, 620 and 220; CCL11, 7.1 and 2.8; IL-6, 5.9 and 1.6; IL-10, 0.15 and 0.015 (all p<0.0001), and MMP-9, 0.92 and 1.5 (p=0.0216), respectively. Concentrations of all cytokines decreased significantly after two consecutive ranibizumab injections, except for MMP-9, which increased significantly. Conclusions After two monthly consecutive antivascular endothelial growth factor injections, inflammatory cytokine levels in the aqueous humour of the eyes with AMD were strongly suppressed, while MMP-9 levels increased.
Collapse
Affiliation(s)
- Shinichi Sakamoto
- Department of Ophthalmology, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Hidenori Takahashi
- Department of Ophthalmology, Jichi Medical University, Shimotsuke, Tochigi, Japan.,Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.,Department of Ophthalmology, Japan Community Health Care Organization Tokyo Shinjuku Medical Center, Tokyo, Japan
| | - Xue Tan
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yuji Inoue
- Department of Ophthalmology, Jichi Medical University, Shimotsuke, Tochigi, Japan.,Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yoko Nomura
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yusuke Arai
- Department of Ophthalmology, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Yujiro Fujino
- Department of Ophthalmology, Japan Community Health Care Organization Tokyo Shinjuku Medical Center, Tokyo, Japan
| | - Hidetoshi Kawashima
- Department of Ophthalmology, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Yasuo Yanagi
- Department of Medical Retina, Singapore National Eye Centre, Singapore, Singapore.,Department of Medical Retina, Singapore Eye Research Institute, Singapore, Singapore.,Department of Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS, Medical School, National University of Singapore, Singapore, Singapore
| |
Collapse
|
12
|
Akl MR, Nagpal P, Ayoub NM, Tai B, Prabhu SA, Capac CM, Gliksman M, Goy A, Suh KS. Molecular and clinical significance of fibroblast growth factor 2 (FGF2 /bFGF) in malignancies of solid and hematological cancers for personalized therapies. Oncotarget 2016; 7:44735-44762. [PMID: 27007053 PMCID: PMC5190132 DOI: 10.18632/oncotarget.8203] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 03/10/2016] [Indexed: 12/30/2022] Open
Abstract
Fibroblast growth factor (FGF) signaling is essential for normal and cancer biology. Mammalian FGF family members participate in multiple signaling pathways by binding to heparan sulfate and FGF receptors (FGFR) with varying affinities. FGF2 is the prototype member of the FGF family and interacts with its receptor to mediate receptor dimerization, phosphorylation, and activation of signaling pathways, such as Ras-MAPK and PI3K pathways. Excessive mitogenic signaling through the FGF/FGFR axis may induce carcinogenic effects by promoting cancer progression and increasing the angiogenic potential, which can lead to metastatic tumor phenotypes. Dysregulated FGF/FGFR signaling is associated with aggressive cancer phenotypes, enhanced chemotherapy resistance and poor clinical outcomes. In vitro experimental settings have indicated that extracellular FGF2 affects proliferation, drug sensitivity, and apoptosis of cancer cells. Therapeutically targeting FGF2 and FGFR has been extensively assessed in multiple preclinical studies and numerous drugs and treatment options have been tested in clinical trials. Diagnostic assays are used to quantify FGF2, FGFRs, and downstream signaling molecules to better select a target patient population for higher efficacy of cancer therapies. This review focuses on the prognostic significance of FGF2 in cancer with emphasis on therapeutic intervention strategies for solid and hematological malignancies.
Collapse
Affiliation(s)
- Mohamed R. Akl
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Poonam Nagpal
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Nehad M. Ayoub
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Betty Tai
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Sathyen A. Prabhu
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Catherine M. Capac
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Matthew Gliksman
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Andre Goy
- Lymphoma Division, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - K. Stephen Suh
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| |
Collapse
|
13
|
Lovely CB, Swartz ME, McCarthy N, Norrie JL, Eberhart JK. Bmp signaling mediates endoderm pouch morphogenesis by regulating Fgf signaling in zebrafish. Development 2016; 143:2000-11. [PMID: 27122171 DOI: 10.1242/dev.129379] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 04/12/2016] [Indexed: 02/03/2023]
Abstract
The endodermal pouches are a series of reiterated structures that segment the pharyngeal arches and help pattern the vertebrate face. Multiple pathways regulate the complex process of endodermal development, including the Bone morphogenetic protein (Bmp) pathway. However, the role of Bmp signaling in pouch morphogenesis is poorly understood. Using genetic and chemical inhibitor approaches, we show that pouch morphogenesis requires Bmp signaling from 10-18 h post-fertilization, immediately following gastrulation. Blocking Bmp signaling during this window results in morphological defects to the pouches and craniofacial skeleton. Using genetic chimeras we show that Bmp signals directly to the endoderm for proper morphogenesis. Time-lapse imaging and analysis of reporter transgenics show that Bmp signaling is necessary for pouch outpocketing via the Fibroblast growth factor (Fgf) pathway. Double loss-of-function analyses demonstrate that Bmp and Fgf signaling interact synergistically in craniofacial development. Collectively, our analyses shed light on the tissue and signaling interactions that regulate development of the vertebrate face.
Collapse
Affiliation(s)
- C Ben Lovely
- Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Mary E Swartz
- Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Neil McCarthy
- Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | | | - Johann K Eberhart
- Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
14
|
Takahashi H, Nomura Y, Tan X, Fujino Y, Kawashima H, Yanagi Y. Effects of posterior vitreous detachment on aqueous humour levels of VEGF and inflammatory cytokines. Br J Ophthalmol 2015; 99:1065-9. [DOI: 10.1136/bjophthalmol-2014-306051] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 01/19/2015] [Indexed: 01/31/2023]
|
15
|
Rainczuk A, Rao J, Gathercole J, Stephens AN. The emerging role of CXC chemokines in epithelial ovarian cancer. Reproduction 2012; 144:303-17. [PMID: 22771929 DOI: 10.1530/rep-12-0153] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
In recent years, chemokines have generated intense investigations due to their involvement in both physiological and pathological processes of inflammation, particularly in ovarian biology. The physiological process of ovulation in the normal ovary involves various chemokines that mediate the healing of the ruptured endometrium. It is now being reported that many of these chemokines are also associated with the cancer of the ovary. Chronic inflammation underlies the progression of ovarian cancer; therefore, it raises the possibility that chemokines are involved in the inflammatory process and mediate immune responses that may favour or inhibit tumour progression. Ovarian cancer is a gynaecological cancer responsible for highest rate of mortality in women. Although there have been several investigations and advances in surgery and chemotherapy, the survival rate for this disease remains low. This is mainly because of a lack of specific symptoms and biomarkers for detection. In this review, we have discussed the emerging role of the CXC chemokines in epithelial ovarian cancer (EOC). The CXC group of chemokines is gaining importance in the field of ovarian cancer for being angiostatic and angiogenic in function. While there have been several studies on the angiogenesis function, emerging research shows that ELR(-) CXC chemokines, CXCL9 and CXCL10, are angiostatic. Importantly, the angiostatic chemokines can inhibit the progression of EOC. Given that there are currently no biomarkers or specific therapeutic targets for the disease, these chemokines are emerging as promising targets for therapy.
Collapse
Affiliation(s)
- Adam Rainczuk
- Prince Henry's Institute, Monash Medical Centre, Clayton, Victoria 3168, Australia
| | | | | | | |
Collapse
|
16
|
Verbeke H, Struyf S, Laureys G, Van Damme J. The expression and role of CXC chemokines in colorectal cancer. Cytokine Growth Factor Rev 2011; 22:345-58. [PMID: 22000992 DOI: 10.1016/j.cytogfr.2011.09.002] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Accepted: 09/06/2011] [Indexed: 12/14/2022]
Abstract
Cancer is a life-threatening disease world-wide and colorectal cancer is the second common cause of cancer mortality. The interaction between tumor cells and stromal cells plays a crucial role in tumor initiation and progression and is partially mediated by chemokines. Chemokines predominantly participate in the chemoattraction of leukocytes to inflammatory sites. Nowadays, it is clear that CXC chemokines and their receptors (CXCR) may also modulate tumor behavior by several important mechanisms: regulation of angiogenesis, activation of a tumor-specific immune response by attracting leukocytes, stimulation of tumor cell proliferation and metastasis. Here, we review the expression and complex roles of CXC chemokines (CXCL1 to CXCL16) and their receptors (CXCR1 to CXCR6) in colorectal cancer. Overall, increased expression levels of CXC chemokines correlate with poor prognosis.
Collapse
Affiliation(s)
- Hannelien Verbeke
- Laboratory of Molecular Immunology, Rega Institute for Medical Research, University of Leuven (K.U. Leuven), Minderbroedersstraat 10, B-3000 Leuven, Belgium
| | | | | | | |
Collapse
|
17
|
Thrombospondin-1 as a Paradigm for the Development of Antiangiogenic Agents Endowed with Multiple Mechanisms of Action. Pharmaceuticals (Basel) 2010; 3:1241-1278. [PMID: 27713299 PMCID: PMC4034032 DOI: 10.3390/ph3041241] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2010] [Revised: 04/20/2010] [Accepted: 04/22/2010] [Indexed: 12/12/2022] Open
Abstract
Uncontrolled neovascularization occurs in several angiogenesis-dependent diseases, including cancer. Neovascularization is tightly controlled by the balance between angiogenic growth factors and antiangiogenic agents. The various natural angiogenesis inhibitors identified so far affect neovascularization by different mechanisms of action. Thrombospondin-1 (TSP-1) is a matricellular modular glycoprotein that acts as a powerful endogenous inhibitor of angiogenesis. It acts both indirectly, by sequestering angiogenic growth factors and effectors in the extracellular environment, and directly, by inducing an antiangiogenic program in endothelial cells following engagement of specific receptors including CD36, CD47, integrins and proteoglycans (all involved in angiogenesis ). In view of its central, multifaceted role in angiogenesis, TSP-1 has served as a source of antiangiogenic tools, including TSP-1 fragments, synthetic peptides and peptidomimetics, gene therapy strategies, and agents that up-regulate TSP-1 expression. This review discusses TSP-1-based inhibitors of angiogenesis, their mechanisms of action and therapeutic potential, drawing our experience with angiogenic growth factor-interacting TSP-1 peptides, and the possibility of exploiting them to design novel antiangiogenic agents.
Collapse
|
18
|
Rusnati M, Bugatti A, Mitola S, Leali D, Bergese P, Depero LE, Presta M. Exploiting Surface Plasmon Resonance (SPR) Technology for the Identification of Fibroblast Growth Factor-2 (FGF2) Antagonists Endowed with Antiangiogenic Activity. SENSORS (BASEL, SWITZERLAND) 2009; 9:6471-503. [PMID: 22454596 PMCID: PMC3312455 DOI: 10.3390/s90806471] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Revised: 08/17/2009] [Accepted: 08/19/2009] [Indexed: 12/31/2022]
Abstract
Angiogenesis, the process of new blood vessel formation, is implicated in various physiological/pathological conditions, including embryonic development, inflammation and tumor growth. Fibroblast growth factor-2 (FGF2) is a heparin-binding angiogenic growth factor involved in various physiopathological processes, including tumor neovascularization. Accordingly, FGF2 is considered a target for antiangiogenic therapies. Thus, numerous natural/synthetic compounds have been tested for their capacity to bind and sequester FGF2 in the extracellular environment preventing its interaction with cellular receptors. We have exploited surface plasmon resonance (SPR) technique in search for antiangiogenic FGF2 binders/antagonists. In this review we will summarize our experience in SPR-based angiogenesis research, with the aim to validate SPR as a first line screening for the identification of antiangiogenic compounds.
Collapse
Affiliation(s)
- Marco Rusnati
- Unit of General Pathology and Immunology, Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Brescia, 25123, Italy; E-Mails: (M.R.); (A.B.); (S.M.); (D.L.)
| | - Antonella Bugatti
- Unit of General Pathology and Immunology, Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Brescia, 25123, Italy; E-Mails: (M.R.); (A.B.); (S.M.); (D.L.)
| | - Stefania Mitola
- Unit of General Pathology and Immunology, Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Brescia, 25123, Italy; E-Mails: (M.R.); (A.B.); (S.M.); (D.L.)
| | - Daria Leali
- Unit of General Pathology and Immunology, Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Brescia, 25123, Italy; E-Mails: (M.R.); (A.B.); (S.M.); (D.L.)
| | - Paolo Bergese
- Chemistry for Technologies Laboratory and Department of Mechanical and Industrial Engineering, School of Engineering, University of Brescia, Brescia, 25123, Italy; E-Mails: (P.B.); (L.E.D.)
| | - Laura E. Depero
- Chemistry for Technologies Laboratory and Department of Mechanical and Industrial Engineering, School of Engineering, University of Brescia, Brescia, 25123, Italy; E-Mails: (P.B.); (L.E.D.)
| | - Marco Presta
- Unit of General Pathology and Immunology, Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Brescia, 25123, Italy; E-Mails: (M.R.); (A.B.); (S.M.); (D.L.)
| |
Collapse
|
19
|
Kim MJ, Romero R, Kim CJ, Tarca AL, Chhauy S, LaJeunesse C, Lee DC, Draghici S, Gotsch F, Kusanovic JP, Hassan SS, Kim JS. Villitis of unknown etiology is associated with a distinct pattern of chemokine up-regulation in the feto-maternal and placental compartments: implications for conjoint maternal allograft rejection and maternal anti-fetal graft-versus-host disease. THE JOURNAL OF IMMUNOLOGY 2009; 182:3919-27. [PMID: 19265171 DOI: 10.4049/jimmunol.0803834] [Citation(s) in RCA: 144] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The co-presence of histoincompatible fetal and maternal cells is a characteristic of human placental inflammation. Villitis of unknown etiology (VUE), a destructive inflammatory lesion of villous placenta, is characterized by participation of Hofbauer cells (placental macrophages) and maternal T cells. In contrast to acute chorioamnionitis of infection-related origin, the fundamental immunopathology of VUE is unknown. This study was performed to investigate the placental transcriptome of VUE and to determine whether VUE is associated with systemic maternal and/or fetal inflammatory response(s). Comparison of the transcriptome between term placentas without and with VUE revealed differential expression of 206 genes associated with pathways related to immune response. The mRNA expression of a subset of chemokines and their receptors (CXCL9, CXCL10, CXCL11, CXCL13, CCL4, CCL5, CXCR3, CCR5) was higher in VUE placentas than in normal placentas (p < 0.05). Analysis of blood cell mRNA showed a higher expression of CXCL9 and CXCL13 in the mother, and CXCL11 and CXCL13 in the fetus of VUE cases (p < 0.05). The median concentrations of CXCL9, CXCL10, and CXCL11 in maternal and fetal plasma were higher in VUE (p < 0.05). Comparison of preterm cases without and with acute chorioamnionitis revealed elevated CXCL9, CXCL10, CXCL11, and CXCL13 concentrations in fetal plasma (p < 0.05), but not in maternal plasma with chorioamnionitis. We report for the first time the placental transcriptome of VUE. A systemic derangement of CXC chemokines in maternal and fetal circulation distinguishes VUE from acute chorioamnionitis. We propose that VUE be a unique state combining maternal allograft rejection and maternal antifetal graft-vs-host disease mechanisms.
Collapse
Affiliation(s)
- Mi Jeong Kim
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development/National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892 and Detroit, MI 48201, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Nhan-Chang CL, Romero R, Kusanovic JP, Gotsch F, Edwin SS, Erez O, Mittal P, Kim CJ, Kim MJ, Espinoza J, Friel LA, Vaisbuch E, Than NG, Mazaki-Tovi S, Hassan SS. A role for CXCL13 (BCA-1) in pregnancy and intra-amniotic infection/inflammation. J Matern Fetal Neonatal Med 2008; 21:763-75. [PMID: 19031272 PMCID: PMC3169890 DOI: 10.1080/14767050802244946] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVES CXCL13 is a potent chemokine, produced by mature and recently recruited macrophages to sites of inflammation, which has antimicrobial and anti-angiogenic properties. The purpose of this study was to: (1) determine whether CXCL13 is present in maternal serum, umbilical cord blood, and amniotic fluid (AF); (2) to determine if AF concentration changes with intra-amniotic infection/inflammation (IAI); and (3) to localize the production of CXCL13 in chorioamniotic membranes and umbilical cord. STUDY DESIGN A cross-sectional study on maternal serum was performed including patients in the following groups: (1) non-pregnant women (n = 20), (2) normal pregnant women (n = 49), (3) patients at term not in labor (n = 30), and (4) patients in spontaneous labor at term (n = 29). Umbilical cord blood was collected from term neonates with (n = 30) and without labor (n = 28). Amniotic fluid was obtained from patients in the following groups: (1) midtrimester (n = 65); (2) term not in labor (n = 22); (3) term in labor (n = 47); (4) preterm labor (PTL) with intact membranes leading to term delivery (n = 70); and (5) PTL leading to preterm delivery with IAI (n = 79) and without IAI (n = 60). CXCL13 concentrations were determined by enzyme-linked immunosorbent assay. Chorioamniotic membranes and umbilical cords were examined with immunohistochemistry. Non-parametric statistics were used for analysis. RESULTS (1) CXCL13 was present in 100% of serum and cord blood samples, and 99% of AF samples (339/343). (2) Serum CXCL13 concentration was significantly higher in pregnant women when compared to non-pregnant women (median 313.3 pg/mL (interquartile range (IQR) 197.2-646.9) vs. 40.5 pg/mL (IQR 29.5-93.5), respectively; p < 0.001). (3) Serum CXCL13 concentration decreased with advancing gestational age (Spearman's Rho = -0.424; p < 0.001). (4) There were no significant differences in the median serum CXCL13 concentration between women at term with and without labor (371.6 pg/mL (IQR 194.3-614.3) vs. 235.1 pg/mL (IQR 182.8-354.7), respectively; p = 0.6). (5) The concentration of CXCL13 in AF did not change with gestational age (p = 0.1). (6) Patients with PTL and delivery with IAI had a significantly higher median concentration of CXCL13 than those without IAI (median 513.2 pg/mL (IQR 199.7-2505.5) vs. 137.3 pg/mL (IQR 96.7-209.6), respectively; p < 0.001) and those who delivered at term (133.7 pg/mL (IQR 97.8-174.8); p < 0.001). (7) Spontaneous labor did not result in a change in the median AF concentration of CXCL13 (labor: 86.9 pg/mL (IQR 55.6-152.0) vs. no labor: 77.8 pg/mL (IQR 68.0-98.0); p = 0.8). (8) CXCL13 was immunolocalized to macrophages in fetal membranes and umbilical vein. CONCLUSIONS (1) We report for the first time the presence of CXCL13 in AF. (2) AF CXCL13 concentrations are dramatically increased in IAI. (3) Unlike other chemokines, AF and serum CXCL13 concentrations did not change with spontaneous parturition.
Collapse
Affiliation(s)
- Chia-Ling Nhan-Chang
- Perinatology Research Branch, Intramural Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Hutzel Women’s Hospital, Bethesda, MD, and Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University/Hutzel Women’s Hospital, Detroit, MI
| | - Roberto Romero
- Perinatology Research Branch, Intramural Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Hutzel Women’s Hospital, Bethesda, MD, and Detroit, MI
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI
| | - Juan Pedro Kusanovic
- Perinatology Research Branch, Intramural Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Hutzel Women’s Hospital, Bethesda, MD, and Detroit, MI
| | - Francesca Gotsch
- Perinatology Research Branch, Intramural Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Hutzel Women’s Hospital, Bethesda, MD, and Detroit, MI
| | - Samuel S. Edwin
- Perinatology Research Branch, Intramural Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Hutzel Women’s Hospital, Bethesda, MD, and Detroit, MI
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI
| | - Offer Erez
- Department of Obstetrics and Gynecology, Wayne State University/Hutzel Women’s Hospital, Detroit, MI
| | - Pooja Mittal
- Perinatology Research Branch, Intramural Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Hutzel Women’s Hospital, Bethesda, MD, and Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University/Hutzel Women’s Hospital, Detroit, MI
| | - Chong Jai Kim
- Perinatology Research Branch, Intramural Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Hutzel Women’s Hospital, Bethesda, MD, and Detroit, MI
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI
| | - Mi Jeong Kim
- Perinatology Research Branch, Intramural Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Hutzel Women’s Hospital, Bethesda, MD, and Detroit, MI
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI
| | - Jimmy Espinoza
- Perinatology Research Branch, Intramural Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Hutzel Women’s Hospital, Bethesda, MD, and Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University/Hutzel Women’s Hospital, Detroit, MI
| | - Lara A. Friel
- Perinatology Research Branch, Intramural Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Hutzel Women’s Hospital, Bethesda, MD, and Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University/Hutzel Women’s Hospital, Detroit, MI
| | - Edi Vaisbuch
- Perinatology Research Branch, Intramural Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Hutzel Women’s Hospital, Bethesda, MD, and Detroit, MI
| | - Nandor Gabor Than
- Perinatology Research Branch, Intramural Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Hutzel Women’s Hospital, Bethesda, MD, and Detroit, MI
| | - Shali Mazaki-Tovi
- Perinatology Research Branch, Intramural Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Hutzel Women’s Hospital, Bethesda, MD, and Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University/Hutzel Women’s Hospital, Detroit, MI
| | - Sonia S. Hassan
- Perinatology Research Branch, Intramural Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Hutzel Women’s Hospital, Bethesda, MD, and Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University/Hutzel Women’s Hospital, Detroit, MI
| |
Collapse
|
21
|
A systematic methodology for proteome-wide identification of peptides inhibiting the proliferation and migration of endothelial cells. Proc Natl Acad Sci U S A 2008; 105:13775-80. [PMID: 18780781 DOI: 10.1073/pnas.0803241105] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
We introduce a systematic computational methodology based on bioinformatics that has enabled us to identify and classify >120 endogenous peptide inhibitors of endothelial cell proliferation and migration. These peptides are derived from members of the type IV collagen, thrombospondin, and CXC chemokine protein families, as well as somatotropin hormones, serpins, and various kringle-containing proteins. Their activity in suppressing the proliferation and migration of endothelial cells in vitro provides proof of principle for the validity of this computational method. Interestingly, some of the peptides are derived from proteins known to be proangiogenic. By performing receptor neutralization studies, we have identified receptors to which these peptides bind. On the basis of this receptor-binding information, we evaluated several examples of peptide-based combinatorial screening strategies. In some cases, this combinatorial screening identified strong synergism between peptides. The current work provides a guideline for a computational-based peptidomics approach for the discovery of endogenous bioactive peptides.
Collapse
|
22
|
Vandercappellen J, Van Damme J, Struyf S. The role of CXC chemokines and their receptors in cancer. Cancer Lett 2008; 267:226-44. [PMID: 18579287 DOI: 10.1016/j.canlet.2008.04.050] [Citation(s) in RCA: 487] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2008] [Revised: 04/28/2008] [Accepted: 04/29/2008] [Indexed: 12/16/2022]
Abstract
Chemokines, or chemotactic cytokines, and their receptors have been discovered as essential and selective mediators in leukocyte migration to inflammatory sites and to secondary lymphoid organs. Besides their functions in the immune system, they also play a critical role in tumor initiation, promotion and progression. There are four subgroups of chemokines: CXC, CC, CX(3)C, and C chemokine ligands. The CXC or alpha subgroup is further subdivided in the ELR(+) and ELR(-) chemokines. Members that contain the ELR motif bind to CXC chemokine receptor 2 (CXCR2) and are angiogenic. In contrast, most of the CXC chemokines without ELR motif bind to CXCR3 and are angiostatic. An exception is the angiogenic ELR(-)CXC chemokine stromal cell-derived factor-1 (CXCL12/SDF-1), which binds to CXCR4 and CXCR7 and is implicated in tumor metastasis. This review is focusing on the role of CXC chemokines and their receptors in tumorigenesis, including angiogenesis, attraction of leukocytes to tumor sites and induction of tumor cell migration and homing in metastatic sites. Finally, their therapeutic use in cancer treatment is discussed.
Collapse
Affiliation(s)
- Jo Vandercappellen
- Laboratory of Molecular Immunology, Rega Institute for Medical Research, University of Leuven, Minderbroedersstraat 10, B-3000 Leuven, Belgium
| | | | | |
Collapse
|
23
|
Beyer T, Meyer-Hermann M. Mechanisms of organogenesis of primary lymphoid follicles. Int Immunol 2008; 20:615-23. [DOI: 10.1093/intimm/dxn020] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
24
|
Schutyser E, Su Y, Yu Y, Gouwy M, Zaja-Milatovic S, Van Damme J, Richmond A. Hypoxia enhances CXCR4 expression in human microvascular endothelial cells and human melanoma cells. Eur Cytokine Netw 2007; 18:59-70. [PMID: 17594938 PMCID: PMC2665278 DOI: 10.1684/ecn.2007.0087] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2007] [Indexed: 11/17/2022]
Abstract
The influence of environmental factors (cytokines, matrix components, serum factors and O(2) level) on expression of receptors for angiogenic versus angiostatic CXC chemokines in human microvascular endothelial cells has not been extensively investigated. Our semi-quantitative RT-PCR analysis demonstrated that TNF-alpha and IFN-gamma repressed CXCR4 mRNA levels in immortalized human microvascular endothelial HMEC-1 cells after 4 h, whereas only TNF-alpha displayed inhibitory activity in primary human microvascular endothelial cells (HMVEC). CXCR4 mRNA expression was not affected by VEGF, GM-CSF, IL-1beta or various basal membrane matrix components, but was significantly up-regulated after serum starvation and/or hypoxic treatment of the microvascular endothelial cells. The alternative CXCL12 receptor, CXCR7/RDC1, was also up-regulated by hypoxia in HMEC-1 cells, although less consistently than CXCR4. Furthermore, hypoxia and serum starvation were required for cell surface display of CXCR4 and CXCL12 induction of ERK activation in HMEC-1 cells. In contrast, CXCR2 and CXCR3 mRNA levels remained, respectively, low and undetectable under all the conditions tested, and surface expression of CXCR2, CXCR3 and CXCR7 on the HMEC- 1 cells could not be demonstrated by FACS. In the human SK-MEL-5 melanoma cell line, CXCR4 mRNA expression was also increased under hypoxic conditions, whereas CXCR2 mRNA levels remained low and levels of CXCR3 and CXCR7 were undetectable. However, immunohistochemical staining of human metastatic melanoma sections demonstrated that CXCR2, CXCR3, CXCR4 and CXCR7 are expressed on tumor cells and, to a lesser extent, on endothelial cells. These results demonstrate that the tumor microenvironment regulates chemokine receptor expression through both cytokine and oxygen levels.
Collapse
Affiliation(s)
- Evemie Schutyser
- Laboratory of Molecular Immunology, Rega Institute for Medical Research, University of Leuven, Belgium
| | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
The chemokine system controls leukocyte trafficking during homeostasis as well as during inflammation and is necessary for the linkage between innate and adaptive immunity. Tissue regulation outside the hematopoietic compartment, for instance, angiogenesis, organogenesis and tumor development, growth and metastasis, is another important function of the chemokine system. The chemokine-mediated regulation of angiogenesis is highly sophisticated and fine tuned, and involves pro-angiogenic chemokines, for instance, CXCL8/IL8 interacting with the CXCR2 receptor, and anti-angiogenic (i.e. angiostatic) chemokines, for instance, CXCL10/IP10 interacting with the CXCR3 receptor. Chemokines also regulate angiogenesis in a receptor-independent manner by means of a perturbation of bFGF and VEGF function. The current review focuses on the influence of the chemokine system in angiogenesis. Examples of the delicate angiogenesis regulation by the chemokine system in, for instance, wound healing and of the dysregulation in, for instance, tumor development are provided along with the interesting phenomenon of molecular piracy of host-encoded genes within the chemokine system. This phenomenon is a general strategy to circumvent and exploit the immune system -- and thereby improve survival -- for many viruses. Yet, a certain group of herpesviruses -- the gamma2-herpesviruses -- encode a functional CXCR2 receptor homolog that is activated by angiogenic chemokines and antagonized by angiostatic chemokines, and this particular gene seems to cause the development of a vascular tumor -- Kaposi's sarcoma -- in the host.
Collapse
Affiliation(s)
- Mette M Rosenkilde
- Laboratory for Molecular Pharmacology, Department of Pharmacology, The Panum Institute, Copenhagen, Denmark.
| | | |
Collapse
|
26
|
Schmutz C, Hulme A, Burman A, Salmon M, Ashton B, Buckley C, Middleton J. Chemokine receptors in the rheumatoid synovium: upregulation of CXCR5. Arthritis Res Ther 2004; 7:R217-29. [PMID: 15743468 PMCID: PMC1065316 DOI: 10.1186/ar1475] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2004] [Revised: 10/07/2004] [Accepted: 11/12/2004] [Indexed: 02/08/2023] Open
Abstract
In patients with rheumatoid arthritis (RA), chemokine and chemokine receptor interactions play a central role in the recruitment of leukocytes into inflamed joints. This study was undertaken to characterize the expression of chemokine receptors in the synovial tissue of RA and non-RA patients. RA synovia (n = 8) were obtained from knee joint replacement operations and control non-RA synovia (n = 9) were obtained from arthroscopic knee biopsies sampled from patients with recent meniscal or articular cartilage damage or degeneration. The mRNA expression of chemokine receptors and their ligands was determined using gene microarrays and PCR. The protein expression of these genes was demonstrated by single-label and double-label immunohistochemistry. Microarray analysis showed the mRNA for CXCR5 to be more abundant in RA than non-RA synovial tissue, and of the chemokine receptors studied CXCR5 showed the greatest upregulation. PCR experiments confirmed the differential expression of CXCR5. By immunohistochemistry we were able to detect CXCR5 in all RA and non-RA samples. In the RA samples the presence of CXCR5 was observed on B cells and T cells in the infiltrates but also on macrophages and endothelial cells. In the non-RA samples the presence of CXCR5 was limited to macrophages and endothelial cells. CXCR5 expression in synovial fluid macrophages and peripheral blood monocytes from RA patients was confirmed by PCR. The present study shows that CXCR5 is upregulated in RA synovial tissue and is expressed in a variety of cell types. This receptor may be involved in the recruitment and positioning of B cells, T cells and monocytes/macrophages in the RA synovium. More importantly, the increased level of CXCR5, a homeostatic chemokine receptor, in the RA synovium suggests that non-inflammatory receptor–ligand pairs might play an important role in the pathogenesis of RA.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Animals
- Antigens, CD20/analysis
- Arthritis, Rheumatoid/genetics
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/pathology
- Autoimmune Diseases/genetics
- Autoimmune Diseases/immunology
- Autoimmune Diseases/metabolism
- Autoimmune Diseases/pathology
- CD3 Complex/analysis
- Endothelium, Vascular/metabolism
- Female
- Gene Expression Profiling
- Humans
- Knee
- Lymphocyte Subsets/metabolism
- Macrophages/metabolism
- Male
- Mice
- Middle Aged
- Oligonucleotide Array Sequence Analysis
- RNA, Messenger/biosynthesis
- Receptors, CXCR5
- Receptors, Chemokine/biosynthesis
- Receptors, Chemokine/genetics
- Receptors, Cytokine/biosynthesis
- Receptors, Cytokine/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- Synovial Membrane/immunology
- Synovial Membrane/metabolism
- Synovial Membrane/pathology
- Up-Regulation
Collapse
Affiliation(s)
- Caroline Schmutz
- Leopold Muller Arthritis Research Centre, Robert Jones and Agnes Hunt Orthopaedic Hospital, Oswestry, UK
| | - Alison Hulme
- Leopold Muller Arthritis Research Centre, Robert Jones and Agnes Hunt Orthopaedic Hospital, Oswestry, UK
| | - Angela Burman
- Division of Immunity and Infection, Medical Research Council Centre for Immune Regulation, University of Birmingham, Edgbaston, UK
| | - Mike Salmon
- Division of Immunity and Infection, Medical Research Council Centre for Immune Regulation, University of Birmingham, Edgbaston, UK
| | - Brian Ashton
- Leopold Muller Arthritis Research Centre, Robert Jones and Agnes Hunt Orthopaedic Hospital, Oswestry, UK
- Institute for Science and Technology in Medicine, Medical School, Keele University, Stoke-on-Trent, UK
| | - Christopher Buckley
- Division of Immunity and Infection, Medical Research Council Centre for Immune Regulation, University of Birmingham, Edgbaston, UK
| | - Jim Middleton
- Leopold Muller Arthritis Research Centre, Robert Jones and Agnes Hunt Orthopaedic Hospital, Oswestry, UK
- Institute for Science and Technology in Medicine, Medical School, Keele University, Stoke-on-Trent, UK
| |
Collapse
|
27
|
Shi X, Cao S, Mitsuhashi M, Xiang Z, Ma X. Genome-wide analysis of molecular changes in IL-12-induced control of mammary carcinoma via IFN-gamma-independent mechanisms. THE JOURNAL OF IMMUNOLOGY 2004; 172:4111-22. [PMID: 15034023 PMCID: PMC2956987 DOI: 10.4049/jimmunol.172.7.4111] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IL-12 is a major activator of tumor-killing NK cells and CTL. IFN-gamma mediates most of the well-known immunological activities of IL-12. In this study, we report IFN-gamma-independent activities induced by therapeutic application of rIL-12 in restricting tumor growth and metastasis in the 4T1 murine mammary carcinoma model. IFN-gamma-deficient mice carrying 4T1 tumor exhibit no gross defect in the number of tumor-infiltrating lymphocytes but have exaggerated angiogenesis in the tumor. Administration of IL-12 is able to constrict blood vessels in the tumor in the absence of IFN-gamma, and retains certain therapeutic efficacy even when applied late during tumor progression. IL-12 exposure in vivo does not irreversibly alter the immunogenicity of the tumor. Finally, global gene expression analysis of primary tumors reveals IL-12-induced molecular patterns and changes, implicating a number of novel genes potentially important for IFN-gamma-independent immune responses against the tumor, for IL-12-mediated antiproliferation, antimetastasis, and antiangiogenesis activities.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/administration & dosage
- Antineoplastic Agents/therapeutic use
- Cell Line, Tumor
- Chemokines/biosynthesis
- Chemokines/genetics
- Female
- Gene Expression Profiling/methods
- Gene Expression Regulation, Neoplastic/immunology
- Growth Inhibitors/administration & dosage
- Growth Inhibitors/therapeutic use
- Interferon-gamma/deficiency
- Interferon-gamma/genetics
- Interferon-gamma/physiology
- Interleukin-12/administration & dosage
- Interleukin-12/therapeutic use
- Lung Neoplasms/genetics
- Lung Neoplasms/immunology
- Lung Neoplasms/prevention & control
- Lung Neoplasms/secondary
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/pathology
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/prevention & control
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Neoplasm Transplantation
- Neovascularization, Pathologic/immunology
- Neovascularization, Pathologic/prevention & control
- Oligonucleotide Array Sequence Analysis/methods
- Recombinant Proteins/administration & dosage
- Recombinant Proteins/therapeutic use
Collapse
Affiliation(s)
| | | | | | | | - Xiaojing Ma
- Address correspondence and reprint requests to Dr. Xiaojing Ma, Department of, Microbiology and Immunology, Weill Medical College of Cornell University, 1300, York Avenue, New York, NY 10021.
| |
Collapse
|
28
|
Romagnani P, Lasagni L, Annunziato F, Serio M, Romagnani S. CXC chemokines: the regulatory link between inflammation and angiogenesis. Trends Immunol 2004; 25:201-9. [PMID: 15039047 DOI: 10.1016/j.it.2004.02.006] [Citation(s) in RCA: 297] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
MESH Headings
- Angiogenesis Modulating Agents/immunology
- Angiogenesis Modulating Agents/metabolism
- Animals
- Chemokines, CXC/physiology
- Endothelial Cells/physiology
- Endothelium, Vascular/physiology
- Endothelium, Vascular/physiopathology
- Humans
- Inflammation/immunology
- Inflammation/physiopathology
- Inflammation Mediators/immunology
- Inflammation Mediators/physiology
- Models, Biological
- Neovascularization, Pathologic/immunology
- Neovascularization, Pathologic/physiopathology
- Neovascularization, Physiologic/immunology
- Neovascularization, Physiologic/physiology
- Receptors, CXCR3
- Receptors, Chemokine/physiology
- Wound Healing/immunology
- Wound Healing/physiology
Collapse
Affiliation(s)
- Paola Romagnani
- Department of Clinical Pathophysiology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy.
| | | | | | | | | |
Collapse
|
29
|
Mazzetti I, Magagnoli G, Paoletti S, Uguccioni M, Olivotto E, Vitellozzi R, Cattini L, Facchini A, Borzì RM. A role for chemokines in the induction of chondrocyte phenotype modulation. ACTA ACUST UNITED AC 2004; 50:112-22. [PMID: 14730607 DOI: 10.1002/art.11474] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
OBJECTIVE To extend the study of the chemokine receptor repertoire on human chondrocytes to receptors with reported housekeeping functions (CXCR3, CXCR4, CXCR5, and CCR6) and to evaluate whether ligands of these receptors play a role in chondrocyte phenotype modulation and proliferation. METHODS Chemokine receptor expression was determined by flow cytometry. Subcultures of chondrocytes were collected and fixed at confluence or during the exponential phase of growth and analyzed for chemokine receptor modulation. The effects of chemokines on isolated cells as well as chondrocytes cultured within an intact extracellular matrix were investigated. Isolated human chondrocytes were stimulated with 100 nM chemokines (monokine induced by interferon-gamma, stromal cell-derived factor 1alpha [SDF-1alpha], B cell-attracting chemokine 1 [BCA-1], or macrophage inflammatory protein 3alpha), and conditioned media were assessed for matrix-degrading enzyme contents (matrix metalloproteinases [MMPs] 1, 3, and 13, and N-acetyl-beta-D-glucosaminidase [NAG]). Cell proliferation and phenotype modulation were evaluated by bromodeoxyuridine incorporation and cathepsin B production. Induction of cell proliferation was assessed in cartilage explants by immunodetection of the proliferation-associated antigen S100A4. RESULTS CXCR3, CXCR4, CXCR5, and CCR6 were detected on human chondrocytes. CXCR3 and CXCR4 expression was increased in exponentially growing chondrocyte subcultures. Ligands of all receptors enhanced the release of MMPs 1, 3, and 13. Release of NAG and cathepsin B was significantly higher in chemokine-stimulated cultures than in unstimulated cultures. SDF-1alpha and BCA-1 also induced DNA synthesis and chondrocyte proliferation, as was shown by the up-regulation of S100A4 in cartilage explants as well. CONCLUSION Our findings extend the repertoire of functional responses elicited by the activity of chemokines on chondrocytes and open new avenues in our understanding of the control of chondrocyte differentiation status by chemokines and their receptors.
Collapse
|
30
|
Salcedo R, Oppenheim JJ. Role of chemokines in angiogenesis: CXCL12/SDF-1 and CXCR4 interaction, a key regulator of endothelial cell responses. Microcirculation 2003; 10:359-70. [PMID: 12851652 DOI: 10.1038/sj.mn.7800200] [Citation(s) in RCA: 121] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2002] [Accepted: 02/24/2003] [Indexed: 11/09/2022]
Abstract
Chemokines are small proteins that act as cell attractants via the activation of G protein-coupled receptors. Chemokines play an important role in several pathophysiological processes such as inflammation and immunity. Many proinflammatory chemokines also support the development of vascular blood supply at the site of inflammation. Similarly, tumor-generated chemokines can contribute to tumor growth by promoting angiogenesis. Recently, significant advances have been made in understanding the contribution of chemokines to the angiogenesis process. This review will discuss first the evidence supporting the direct contribution of different chemokine subfamily members, including CC, CXC, and CX3C chemokines, as positive or negative regulators of the angiogenesis process based on the expression of their cognate receptors on endothelial cells. Additionally, the relationship between classic angiogenic factors and chemokine receptor expression on endothelial cells, and the implications of chemokine production by cancer cells will be analyzed with particular emphasis on the CXCL12/stromal-cell derived factor-1 interaction with CXCR4.
Collapse
Affiliation(s)
- Rosalba Salcedo
- Laboratory of Molecular Immunoregulation, Division of Basic Sciences, National Cancer Institute at Frederick, Frederick, MD 21702-1201, USA
| | | |
Collapse
|
31
|
Buschmann I, Heil M, Jost M, Schaper W. Influence of inflammatory cytokines on arteriogenesis. Microcirculation 2003; 10:371-9. [PMID: 12851653 DOI: 10.1038/sj.mn.7800199] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2002] [Accepted: 01/10/2003] [Indexed: 01/10/2023]
Abstract
Blood vessel growth after birth is limited to two major processes. Angiogenesis is the growth of new capillaries by sprouting or intussusception. The major stimulus for angiogenesis is ischemia. In contrast, arteriogenesis describes the remodeling and growth of collateral arteries from a preexisting arteriolar network. Arteriogenesis is induced after the occlusion of a major artery which induces hemodynamic and mechanical effects on the collateral vessel wall which occur with increasing blood flow velocity due to the low pressure at the reentrant site of the collateral vessel. A variety of different cytokines that act by stimulating endothelial and smooth muscle cell proliferation and migration or recruitment and activation of monocytes have been identified to stimulate angiogenesis and/or arteriogenesis (i.e., MCP-1, FGF-2, TGF-beta, VEGF, and GM-CSF). Several clinical trials have been published in that field to suggest the feasibility and safety of treatment with such cytokines or their genes. However, the results indicate that further studies are needed before proangiogenic and proarteriogenic therapies are ready for clinical application.
Collapse
Affiliation(s)
- Ivo Buschmann
- Research Group for Experimental and Clinical Arteriogenesis, Department for Cardiology and Angiology, Albert Ludwigs University, Freiburg, Germany
| | | | | | | |
Collapse
|
32
|
Bernardini G, Ribatti D, Spinetti G, Morbidelli L, Ziche M, Santoni A, Capogrossi MC, Napolitano M. Analysis of the role of chemokines in angiogenesis. J Immunol Methods 2003; 273:83-101. [PMID: 12535800 DOI: 10.1016/s0022-1759(02)00420-9] [Citation(s) in RCA: 143] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Chemokines, a large family of inflammatory cytokines, have been shown to play a critical role in the regulation of angiogenesis during several pathophysiologic processes, such as tumor growth, wound healing and ischemia. Semiquantitative or quantitative angiogenesis assays are commonly utilized to screen the angiogenic or angiostatic activity of chemokines. These include in vitro endothelial cell activation assays and ex vivo or in vivo models of neovascularization. Chemokines may exert their regulatory activity on angiogenesis directly or as a consequence of leukocyte infiltration and/or the induction of growth factor expression. The effect of chemokines on endothelium can be assessed by performing in vitro assays on purified endothelial cell populations or by in vivo assays. Nevertheless, each model used to evaluate the angiogenic or angiostatic activity of a discrete factor has advantages and limitations. Thus, in order to avoid under- or overestimating the regulatory effect of chemokines on angiogenesis and to evaluate all aspects of the angiogenic process, multiple assays are usually performed. This review summarizes past and recent studies on chemokines as modulators of angiogenesis with particular emphasis on the methods currently used for the assessment of chemokine-mediated angiogenic or angiostatic responses.
Collapse
Affiliation(s)
- Giovanni Bernardini
- Department of Experimental Medicine and Pathology, University of Rome, Viale Regina Elena 324, La Sapienza 00161, Italy.
| | | | | | | | | | | | | | | |
Collapse
|