1
|
Lu W, Wen J. The relationship among H 2S, neuroinflammation and MMP-9 in BBB injury following ischemic stroke. Int Immunopharmacol 2025; 146:113902. [PMID: 39724730 DOI: 10.1016/j.intimp.2024.113902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 12/10/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024]
Abstract
Blood-brain barrier (BBB) is located at the interface between the central nervous system (CNS) and the circulatory system, which maintains the microenvironmental homeostasis of the CNS. BBB damage is a result of CNS diseases, including ischemic stroke, and is a cause of CNS deterioration. Cerebral ischemia unleashes a profound inflammatory response to remove the damaged tissue in the CNS and prepare the brain for repair. However, the excessive neuroinflammation following stroke onset is associated with BBB breakdown, resulting in neuronal injury and worse neurological outcomes. Additionally, matrix metalloproteinases (MMPs) are likewise responsible for the BBB injury and participate in the pathological processes of neuroinflammation following ischemic stroke. Hydrogen sulfide (H2S) is one of gaseous signaling and freely diffusing molecules. Low concentration of H2S yields the neuroprotection against BBB damage following stroke. This review discussed the current knowledge about the detrimental roles of neuroinflammation and MMPs in BBB injury following ischemic stroke. Specifically, we provided an updated overview of H2S in protecting against BBB injury following ischemic stroke via anti-neuroinflammation and inhibiting MMP-9.
Collapse
Affiliation(s)
- Weizhuo Lu
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China; Medical Branch, Hefei Technology College, Hefei, China.
| | - Jiyue Wen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| |
Collapse
|
2
|
Gu T, Vasilatos SN, Yin J, Qin Y, Zhang L, Davidson NE, Huang Y. Restoration of TFPI2 by LSD1 inhibition suppresses tumor progression and potentiates antitumor immunity in breast cancer. Cancer Lett 2024; 600:217182. [PMID: 39154703 PMCID: PMC11384719 DOI: 10.1016/j.canlet.2024.217182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/16/2024] [Accepted: 08/11/2024] [Indexed: 08/20/2024]
Abstract
Histone lysine-specific demethylase 1 (LSD1) is frequently overexpressed in triple negative breast cancer (TNBC), which is associated with worse clinical outcome in TNBC patients. However, the underlying mechanisms by which LSD1 promotes TNBC progression remain to be identified. We recently established a genetically engineered murine model by crossing mammary gland conditional LSD1 knockout mice with Brca1-deficient mice to explore the role of LSD1 in TNBC pathogenesis. Cre-mediated Brca1 loss led to higher incidence of tumor formation in mouse mammary glands, which was hindered by concurrent depletion of LSD1, indicating a critical role of LSD1 in promoting Brca1-deficient tumors. We also demonstrated that the silencing of a tumor suppressor gene, Tissue Factor Pathway Inhibitor 2 (TFPI2), is functionally associated with LSD1-mediated TNBC progression. Mouse Brca1-deficient tumors exhibited elevated LSD1 expression and decreased TFPI2 level compared to normal mammary tissues. Analysis of TCGA database revealed that TFPI2 expression is significantly lower in aggressive ER-negative or basal-like BC. Restoration of TFPI2 through LSD1 inhibition increased H3K4me2 enrichment at the TFPI2 promoter, suppressed tumor progression, and enhanced antitumor efficacy of chemotherapeutic agent. Induction of TFPI2 by LSD1 ablation downregulates activity of matrix metalloproteinases (MMPs) that in turn increases the level of cytotoxic T lymphocyte attracting chemokines in tumor environment, leading to enhanced tumor infiltration of CD8+ T cells. Moreover, induction of TFPI2 potentiates antitumor effect of LSD1 inhibitor and immune checkpoint blockade in poorly immunogenic TNBC. Together, our study identifies previously unrecognized roles of TFPI2 in LSD1-mediated TNBC progression, therapeutic response, and immunogenic effects.
Collapse
Affiliation(s)
- Tiezheng Gu
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shauna N Vasilatos
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jun Yin
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ye Qin
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lin Zhang
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Medicine, University of Southern California, Keck School of Medicine, Los Angeles, CA, USA
| | - Nancy E Davidson
- Fred Hutchinson Cancer Center and University of Washington, Seattle, WA, USA
| | - Yi Huang
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Internal Medicine, Division of Hematology, Oncology, and Blood & Marrow Transplantation, Carver College of Medicine, University of Iowa, Iowa City, IA, USA; Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
3
|
Zhang Y, Mi X, Zhang Y, Li J, Qin Y, He P, Zhao Y, Su B, He L. Immune checkpoint activity exacerbate renal interstitial fibrosis progression by enhancing PD-L1 expression in renal tubular epithelial cells. Transl Res 2024; 271:52-67. [PMID: 38723861 DOI: 10.1016/j.trsl.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 05/04/2024] [Accepted: 05/06/2024] [Indexed: 05/23/2024]
Abstract
Renal interstitial fibrosis (RIF) is often associated with inflammatory cell infiltration and no effective therapy. Programmed death cell-1 (PD-1) and its ligand PD-L1 were playing critical roles in T cell coinhibition and exhaustion, but the role in RIF is unclear. Here the data analyses of serum from 122 IgA nephrology (IgAN) patients showed that high level of soluble PD-1(sPD-1) was an independent risk factor for RIF and renal function progression. PD-L1 was also overexpressed in renal interstitial tissues from both IgAN patients with high level of sPD-1 and the unilateral ureteral obstruction (UUO) mouse. PD-L1 was significantly overexpressed in HK-2 cells with upregulated collagen and α-SMA when stimulated by inflammation or hypoxia in vitro. Additionally, matrix metalloproteinases (MMP-2) could increase the level of sPD-1 in culture supernatant when added in co-culture system of HK-2 and jurkat cells, which implied serum sPD-1 of IgAN might be cleaved by MMP-2 from T cells infiltrated into the tubulointerstitial inflammatory microenvironment. Crucially, injection of PD-L1 fusion protein, the blocker of sPD-1, could ameliorate kidney fibrosis in UUO mice by increasing T cell coinhibition and exhaustion, suggesting the therapeutic potential of PD-L1 fusion targeting for renal fibrosis. Take together, it reveals a novel causal role of sPD-1 in serum and PD-L1 of renal interstitial tissues in the development of renal fibrosis of IgAN, and targeting sPD-1 in serum by PD-L1 fusion protein is a potential therapeutic approach to prevent renal fibrosis of IgAN.
Collapse
Affiliation(s)
- Yuting Zhang
- Department of Nephrology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Xue Mi
- Department of Nephrology, Xijing Hospital, Air Force Medical University, Xi'an, China; Medical School of Yan'an University, Yan'an, China
| | - Yunchao Zhang
- Department of Nephrology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Jipeng Li
- Department of Nephrology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Yunlong Qin
- Department of Nephrology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Peng He
- Department of Nephrology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Ya Zhao
- Department of Medical Microbiology and Parasitology, Air Force Medical University, Xi'an, China.
| | - Binxiao Su
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Air Force Medical University, Xi'an, China; Department of Intensive Care Unit, Xijing Hospital, Air Force Medical University, Xi'an, China.
| | - Lijie He
- Department of Nephrology, Xijing Hospital, Air Force Medical University, Xi'an, China.
| |
Collapse
|
4
|
Jin P, Duan X, Li L, Zhou P, Zou C, Xie K. Cellular senescence in cancer: molecular mechanisms and therapeutic targets. MedComm (Beijing) 2024; 5:e542. [PMID: 38660685 PMCID: PMC11042538 DOI: 10.1002/mco2.542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 02/28/2024] [Accepted: 03/07/2024] [Indexed: 04/26/2024] Open
Abstract
Aging exhibits several hallmarks in common with cancer, such as cellular senescence, dysbiosis, inflammation, genomic instability, and epigenetic changes. In recent decades, research into the role of cellular senescence on tumor progression has received widespread attention. While how senescence limits the course of cancer is well established, senescence has also been found to promote certain malignant phenotypes. The tumor-promoting effect of senescence is mainly elicited by a senescence-associated secretory phenotype, which facilitates the interaction of senescent tumor cells with their surroundings. Targeting senescent cells therefore offers a promising technique for cancer therapy. Drugs that pharmacologically restore the normal function of senescent cells or eliminate them would assist in reestablishing homeostasis of cell signaling. Here, we describe cell senescence, its occurrence, phenotype, and impact on tumor biology. A "one-two-punch" therapeutic strategy in which cancer cell senescence is first induced, followed by the use of senotherapeutics for eliminating the senescent cells is introduced. The advances in the application of senotherapeutics for targeting senescent cells to assist cancer treatment are outlined, with an emphasis on drug categories, and the strategies for their screening, design, and efficient targeting. This work will foster a thorough comprehension and encourage additional research within this field.
Collapse
Affiliation(s)
- Ping Jin
- State Key Laboratory for Conservation and Utilization of Bio‐Resources in Yunnan, School of Life SciencesYunnan UniversityKunmingYunnanChina
| | - Xirui Duan
- Department of OncologySchool of MedicineSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduSichuanChina
| | - Lei Li
- Department of Anorectal SurgeryHospital of Chengdu University of Traditional Chinese Medicine and Chengdu University of Traditional Chinese MedicineChengduChina
| | - Ping Zhou
- Department of OncologySchool of MedicineSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduSichuanChina
| | - Cheng‐Gang Zou
- State Key Laboratory for Conservation and Utilization of Bio‐Resources in Yunnan, School of Life SciencesYunnan UniversityKunmingYunnanChina
| | - Ke Xie
- Department of OncologySchool of MedicineSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduSichuanChina
| |
Collapse
|
5
|
Yoshida M, Matsuzaki J, Fujita K, Kimura M, Umezu T, Tokuda N, Yamaguchi T, Kuroda M, Ochiya T, Saito Y, Kimura K. Plasma extracellular vesicle microRNAs reflecting the therapeutic effect of the CBP/β-catenin inhibitor PRI-724 in patients with liver cirrhosis. Sci Rep 2024; 14:6266. [PMID: 38491114 PMCID: PMC10943077 DOI: 10.1038/s41598-024-56942-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 03/12/2024] [Indexed: 03/18/2024] Open
Abstract
There is an unmet need for antifibrotic therapies to prevent the progression of liver cirrhosis. Previously, we conducted an exploratory trial to assess the safety and antifibrotic efficacy of PRI-724, a selective CBP/β-catenin inhibitor, in patients with liver cirrhosis. PRI-724 was well tolerated and exerted a potential antifibrotic effect. Here, we investigated whether the profiles of circulating microRNAs packaged in extracellular vesicles (EV-miRNAs) are associated with responses to liver fibrosis treatments. Eighteen patients who received PRI-724 for 12 weeks in a phase 1/2a study were classified as responders (n = 10) or non-responders (n = 8) based on changes in liver stiffness. Plasma samples were obtained before and after PRI-724 administration and the levels of EV-miRNAs were analyzed. Three miRNAs (miR-6510-5p, miR-6772-5p, and miR-4261) were identified as predictors of response or non-response to PRI-724, and the levels of three other miRNAs (miR-939-3p, miR-887-3p, and miR-7112-5p) correlated with the efficacy of treatment. Expression of miR-887-3p was detected in hepatocytes and was decreased significantly in liver tissue following PRI-724 treatment. In addition, transfection of a miR-887-3p mimic activated hepatic stellate cells. Thus, decreases in the miR-887-3p level in blood may reflect recovery from liver fibroses in patients with liver cirrhosis treated with PRI-724, although further validation studies are warranted to confirm this.
Collapse
Affiliation(s)
- Mayu Yoshida
- Division of Pharmacotherapeutics, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| | - Juntaro Matsuzaki
- Division of Pharmacotherapeutics, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan.
| | - Koji Fujita
- Department of Molecular Pathology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan
| | - Masamichi Kimura
- Department of Hepatology, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo, 113-8677, Japan
| | - Tomohiro Umezu
- Department of Molecular Pathology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan
| | - Noi Tokuda
- Division of Pharmacotherapeutics, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| | - Tomoko Yamaguchi
- Division of Pharmacotherapeutics, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| | - Masahiko Kuroda
- Department of Molecular Pathology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan
| | - Takahiro Ochiya
- Department of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Yoshimasa Saito
- Division of Pharmacotherapeutics, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| | - Kiminori Kimura
- Department of Hepatology, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo, 113-8677, Japan
| |
Collapse
|
6
|
Fu S, Li H, Wu Y, Wang J. Nano-/micro-scaled hydroxyapatite ceramic construction and the regulation of immune-associated osteogenic differentiation. J Biomed Mater Res A 2024; 112:193-209. [PMID: 37680167 DOI: 10.1002/jbm.a.37606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/04/2023] [Accepted: 08/24/2023] [Indexed: 09/09/2023]
Abstract
Hydroxyapatite (HA) bioceramic is a promising substitute for bone defects, and the surface properties are major factors that influence bioactivity and osteoinductivity. In this study, two kinds of HA bioceramics with nanoscale (n-HA) and microscale (m-HA) surface topography were designed to mimic the natural bone, thus enhancing the stimulation of osteogenic differentiation and revealing the potential mechanism. Compared to m-HA, n-HA owned a larger surface roughness, a stronger wettability, and reduced hardness and indentation modulus. Based on these properties, n-HA could maintain the conformation of vitronectin better than m-HA, which may contribute to higher cellular activities and a stronger promotion of osteogenic differentiation of mesenchymal stem cells (MSCs). Further RNA sequencing analysis compared the molecular expression between n-HA and m-HA. Six hundred twenty-seven differentially expressed genes were identified in MSCs, and 17 upregulated genes and 610 downregulated genes were included when n-HA compared to m-HA. The GO cluster analysis and enriched Kyoto encyclopedia of genes and genome signaling pathways revealed a close correlation with the immune process in both upregulated (chemokine signaling pathway and cytokine-cytokine receptor interaction) and downregulated pathways (osteoclasts differentiation). It suggested that the nanoscale surface topography of HA enhanced the osteoinductivity of MSCs and could not be separated from its regulation of immune function and the retention of adsorbed protein conformation.
Collapse
Affiliation(s)
- Shijia Fu
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Huishan Li
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Yue Wu
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Jing Wang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| |
Collapse
|
7
|
Quaresma TC, de Aguiar Valentim L, de Sousa JR, de Souza Aarão TL, Fuzii HT, Duarte MIS, de Souza J, Quaresma JAS. Immunohistochemical Characterization of M1, M2, and M4 Macrophages in Leprosy Skin Lesions. Pathogens 2023; 12:1225. [PMID: 37887741 PMCID: PMC10610015 DOI: 10.3390/pathogens12101225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 09/20/2023] [Accepted: 09/26/2023] [Indexed: 10/28/2023] Open
Abstract
Mycobacterium leprae is the etiological agent of leprosy. Macrophages (Mφs) are key players involved in the pathogenesis of leprosy. In this study, immunohistochemical analysis was performed to examine the phenotype of Mφ subpopulations, namely M1, M2, and M4, in the skin lesions of patients diagnosed with leprosy. Based on the database of treatment-naïve patients treated between 2015 and 2019 at the Department of Dermatology of the University of the State of Pará, Belém, routine clinical screening samples were identified. The monolabeling protocol was used for M1 macrophages (iNOS, IL-6, TNF-α) and M2 macrophages (IL-10, IL-13, CD163, Arginase 1, TGF-β, FGFb), and the double-labeling protocol was used for M4 macrophages (IL-6, MMP7, MRP8, TNF-α e CD68). To confirm the M4 macrophage lineage, double labeling of the monoclonal antibodies CD68 and MRP8 was also performed. Our results demonstrated a statistically significant difference for the M1 phenotype among the Virchowian (VV) (4.5 ± 1.3, p < 0.0001), Borderline (1.6 ± 0.4, p < 0.0001), and tuberculoid (TT) (12.5 ± 1.8, p < 0.0001) clinical forms of leprosy. Additionally, the M2 phenotype showed a statistically significant difference among the VV (12.5 ± 2.3, p < 0.0001), Borderline (1.3 ± 0.2, p < 0.0001), and TT (3.2 ± 0.7, p < 0.0001) forms. For the M4 phenotype, a statistically significant difference was observed in the VV (9.8 ± 1.7, p < 0.0001), Borderline (1.2 ± 0.2, p < 0.0001), and TT (2.6 ± 0.7, p < 0.0001) forms. A significant correlation was observed between the VV M1 and M4 (r = 0.8712; p = 0.0000) and between the VV M2 × TT M1 (r = 0.834; p = 0.0002) phenotypes. The M1 Mφs constituted the predominant Mφ subpopulation in the TT and Borderline forms of leprosy, whereas the M2 Mφs showed increased immunoexpression and M4 was the predominant Mφ phenotype in VV leprosy. These results confirm the relationship of the Mφ profile with chronic pathological processes of the inflammatory response in leprosy.
Collapse
Affiliation(s)
- Tatiane Costa Quaresma
- Health Department, Center for Biological and Health Sciences, State University of Para-CCBS, UEPA, Belem 66087-662, Brazil
| | - Lívia de Aguiar Valentim
- Health Department, Center for Biological and Health Sciences, State University of Para-CCBS, UEPA, Belem 66087-662, Brazil
| | - Jorge Rodrigues de Sousa
- Health Department, Center for Biological and Health Sciences, State University of Para-CCBS, UEPA, Belem 66087-662, Brazil
| | - Tinara Leila de Souza Aarão
- Health Department, Center for Biological and Health Sciences, State University of Para-CCBS, UEPA, Belem 66087-662, Brazil
- School of Medicine, Federal University of Para-UFPA, Altamira 68440-000, Brazil
| | - Hellen Thais Fuzii
- Health Department, Tropical Medicine Center, Federal University of Para-NMT-UFPA, Belem 66055-240, Brazil
| | | | - Juarez de Souza
- Health Department, Center for Biological and Health Sciences, State University of Para-CCBS, UEPA, Belem 66087-662, Brazil
| | - Juarez Antônio Simões Quaresma
- Health Department, Center for Biological and Health Sciences, State University of Para-CCBS, UEPA, Belem 66087-662, Brazil
- School of Medicine, Federal University of Para-UFPA, Altamira 68440-000, Brazil
- Health Department, Tropical Medicine Center, Federal University of Para-NMT-UFPA, Belem 66055-240, Brazil
- School of Medicine, Sao Paulo University, Sao Paulo 01246-903, Brazil
| |
Collapse
|
8
|
Ullah A, Wang MJ, Wang YX, Shen B. CXC chemokines influence immune surveillance in immunological disorders: Polycystic ovary syndrome and endometriosis. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166704. [PMID: 37001703 DOI: 10.1016/j.bbadis.2023.166704] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/13/2023] [Accepted: 03/22/2023] [Indexed: 04/05/2023]
Abstract
Reproductive health is a worldwide challenge, but it is of particular significance to women during their reproductive age. Several female reproductive problems, including polycystic ovary syndrome (PCOS) and endometriosis, affect about 10 % of women and have a negative impact on their health, fertility, and quality of life. Small, chemotactic, and secreted cytokines are CXC chemokines. Both PCOS and endometriosis demonstrate dysregulation of CXC chemokines, which are critical to the development and progression of both diseases. Recent research has shown that both in humans and animals, CXC chemokines tend to cause inflammation. It has also been found that CXC chemokines are necessary for promoting angiogenesis and inflammatory responses. CXC chemokine overexpression is frequently associated with poor survival and prognosis. CXC chemokine levels in PCOS and endometriosis patients impact their circumstances significantly. Hence, CXC chemokines have significant potential as diagnostic and prognostic biomarkers and therapeutic targets. The molecular mechanisms through which CXC chemokines promote inflammation and the development of PCOS and endometriosis are currently unknown. This article will discuss the functions of CXC chemokines in the promotion, development, and therapy of PCOS and endometriosis, as well as future research directions. The current state and future prospects of CXC chemokine -based therapeutic strategies in the management of PCOS and endometriosis are also highlighted.
Collapse
|
9
|
Characterization of Active MMP9 in Chronic Inflammatory Diseases Using a Novel Anti-MMP9 Antibody. Antibodies (Basel) 2023; 12:antib12010009. [PMID: 36810514 PMCID: PMC9944116 DOI: 10.3390/antib12010009] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/31/2022] [Accepted: 01/12/2023] [Indexed: 01/21/2023] Open
Abstract
Matrix metalloproteinase 9 (MMP9), a protease implicated in multiple diseases, is secreted as an inactive zymogen and requires proteolytic removal of the pro-domain for activation. The relative levels and functionality of the pro- and active-MMP9 isoforms in tissues are not characterized. We generated a specific antibody that distinguishes an active form of MMP9, F107-MMP9, from the inactive pro-MMP9 isoform. Using multiple in vitro assays and specimen types, we show that F107-MMP9 expression is localized and disease-specific compared with its more abundant parental pro-form. It is detected around sites of active tissue remodeling, including fistulae of inflammatory bowel and dermal fissures in hidradenitis suppurativa, and is expressed by myeloid cells, including macrophages and neutrophils. Together, our findings provide insights into the distribution and potential role of MMP9 in inflammatory diseases.
Collapse
|
10
|
Itoh Y. Proteolytic modulation of tumor microenvironment signals during cancer progression. Front Oncol 2022; 12:935231. [PMID: 36132127 PMCID: PMC9483212 DOI: 10.3389/fonc.2022.935231] [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: 05/03/2022] [Accepted: 08/10/2022] [Indexed: 11/13/2022] Open
Abstract
Under normal conditions, the cellular microenvironment is optimized for the proper functioning of the tissues and organs. Cells recognize and communicate with the surrounding cells and extracellular matrix to maintain homeostasis. When cancer arises, the cellular microenvironment is modified to optimize its malignant growth, evading the host immune system and finding ways to invade and metastasize to other organs. One means is a proteolytic modification of the microenvironment and the signaling molecules. It is now well accepted that cancer progression relies on not only the performance of cancer cells but also the surrounding microenvironment. This mini-review discusses the current understanding of the proteolytic modification of the microenvironment signals during cancer progression.
Collapse
|
11
|
Augoff K, Hryniewicz-Jankowska A, Tabola R, Stach K. MMP9: A Tough Target for Targeted Therapy for Cancer. Cancers (Basel) 2022; 14:cancers14071847. [PMID: 35406619 PMCID: PMC8998077 DOI: 10.3390/cancers14071847] [Citation(s) in RCA: 120] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/27/2022] [Accepted: 03/31/2022] [Indexed: 02/01/2023] Open
Abstract
Having the capability to proteolyze diverse structural and signaling proteins, matrix metalloproteinase 9 (MMP9), one of the best-studied secretory endopeptidases, has been identified as a crucial mediator of processes closely associated with tumorigenesis, such as the extracellular matrix reorganization, epithelial to mesenchymal transition, cell migration, new blood vessel formation, and immune response. In this review, we present the current state of knowledge on MMP9 and its role in cancer growth in the context of cell adhesion/migration, cancer-related inflammation, and tumor microenvironment formation. We also summarize recent achievements in the development of selective MMP9 inhibitors and the limitations of using them as anticancer drugs.
Collapse
Affiliation(s)
- Katarzyna Augoff
- Department of Surgical Education, Wroclaw Medical University, 50-367 Wroclaw, Poland
- Department of Chemistry and Immunochemistry, Wroclaw Medical University, 50-367 Wroclaw, Poland;
- Correspondence:
| | | | - Renata Tabola
- Department of Thoracic Surgery, Wroclaw Medical University, 50-367 Wroclaw, Poland;
| | - Kamilla Stach
- Department of Chemistry and Immunochemistry, Wroclaw Medical University, 50-367 Wroclaw, Poland;
| |
Collapse
|
12
|
Shah MA, Cunningham D, Metges JP, Van Cutsem E, Wainberg Z, Elboudwarej E, Lin KW, Turner S, Zavodovskaya M, Inzunza D, Liu J, Patterson SD, Zhou J, He J, Thai D, Bhargava P, Brachmann CB, Cantenacci DVT. Randomized, open-label, phase 2 study of andecaliximab plus nivolumab versus nivolumab alone in advanced gastric cancer identifies biomarkers associated with survival. J Immunother Cancer 2021; 9:jitc-2021-003580. [PMID: 34893523 PMCID: PMC8666898 DOI: 10.1136/jitc-2021-003580] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/05/2021] [Indexed: 12/13/2022] Open
Abstract
Background Matrix metalloproteinase-9 (MMP9) selectively cleaves extracellular matrix proteins contributing to tumor growth and an immunosuppressive microenvironment. This study evaluated andecaliximab (ADX), an inhibitor of MMP9, in combination with nivolumab (NIVO), for the treatment of advanced gastric cancer. Methods Phase 2, open-label, randomized multicenter study evaluating the efficacy, safety, and pharmacodynamics of ADX+NIVO versus NIVO in patients with pretreated metastatic gastric or gastroesophageal junction (GEJ) adenocarcinoma. The primary endpoint was objective response rate (ORR). Secondary endpoints included progression-free survival (PFS), overall survival (OS), and adverse events (AEs). We explored the correlation of efficacy outcomes with biomarkers. Results 144 patients were randomized; 141 were treated: 81% white, 69% male, median age was 61 years in the ADX+NIVO group and 62 years in the NIVO-alone group. The ORR was 10% (95% CI 4 to 19) in the ADX+NIVO group and 7% (95% CI 2 to 16) in the NIVO-alone group (OR: 1.5 (95% CI 0.4 to 6.1; p=0.8)). There was no response or survival benefit associated with adding ADX. AE rates were comparable in both treatment groups; the most common AEs were fatigue, decreased appetite, nausea, and vomiting. Programmed cell death ligand 1, interferon-γ (IFN), and intratumoral CD8+ cell density were not associated with treatment response or survival. The gene signature most correlated with shorter survival was the epithelial-to-mesenchymal gene signature; high transforming growth factor (TGF)-β fibrosis score was negatively associated with OS (p=0.036). Gene expression analysis of baseline tumors comparing long-(1+ years) and short-term (<1 year) survivors showed that GRB7 was associated with survival beyond 1 year. Human epidermal growth factor receptor 2 (HER2)-positive disease was associated with significantly longer survival (p=0.0077). Median tumor mutation burden (TMB) was 2.01; patients with TMB ≥median had longer survival (p=0.0025) and improved PFS (p=0.016). Based on a model accounting for TMB, TGF-β fibrosis, and HER2, TMB was the main driver of survival in this patient population. Conclusion Combination of ADX+NIVO had a favorable safety profile but did not improve efficacy compared with NIVO alone in patients with pretreated metastatic gastric or GEJ adenocarcinoma. HER2 positivity, higher TMB or GRB7, and lower TGF-β were associated with improved outcomes. Trial registration number NCT02864381 or GS-US-296–-2013.
Collapse
Affiliation(s)
- Manish A Shah
- Medicine, Weill Cornell Medicine, New York, New York, USA .,Medicine, NewYork-Presbyterian Hospital/Weill Cornell Medical Center, New York, New York, USA
| | - David Cunningham
- Gastrointestinal and Lymphoma Unit, The Royal Marsden NHS Foundation Trust, Sutton and London Hospital, Sutton, UK
| | | | - Eric Van Cutsem
- Division Head of Digestive Oncology, University Hospitals Leuven and KU Leuven, Leuven, Belgium
| | - Zev Wainberg
- Gastrointestinal Medical Oncology, University of California Los Angeles School of Medicine, Los Angeles, California, USA
| | | | - Kai-Wen Lin
- Gilead Sciences, Inc, Foster City, California, USA
| | - Scott Turner
- Gilead Sciences, Inc, Foster City, California, USA
| | | | | | - Jinfeng Liu
- Gilead Sciences, Inc, Foster City, California, USA
| | | | - Jingzhu Zhou
- Gilead Sciences, Inc, Foster City, California, USA
| | - Jing He
- Gilead Sciences, Inc, Foster City, California, USA
| | - Dung Thai
- Gilead Sciences, Inc, Foster City, California, USA
| | | | | | - Daniel V T Cantenacci
- Biological Sciences Division, University of Chicago Medical Center, Chicago, Illinois, USA
| |
Collapse
|
13
|
Dual Targeting of Cancer Cells and MMPs with Self-Assembly Hybrid Nanoparticles for Combination Therapy in Combating Cancer. Pharmaceutics 2021; 13:pharmaceutics13121990. [PMID: 34959271 PMCID: PMC8707712 DOI: 10.3390/pharmaceutics13121990] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/05/2021] [Accepted: 11/19/2021] [Indexed: 01/21/2023] Open
Abstract
The co-delivery of chemotherapeutic agents and immune modulators to their targets remains to be a great challenge for nanocarriers. Here, we developed a hybrid thermosensitive nanoparticle (TMNP) which could co-deliver paclitaxel-loaded transferrin (PTX@TF) and marimastat-loaded thermosensitive liposomes (MMST/LTSLs) for the dual targeting of cancer cells and the microenvironment. TMNPs could rapidly release the two payloads triggered by the hyperthermia treatment at the site of tumor. The released PTX@TF entered cancer cells via transferrin-receptor-mediated endocytosis and inhibited the survival of tumor cells. MMST was intelligently employed as an immunomodulator to improve immunotherapy by inhibiting matrix metalloproteinases to reduce chemokine degradation and recruit T cells. The TMNPs promoted the tumor infiltration of CD3+ T cells by 2-fold, including memory/effector CD8+ T cells (4.2-fold) and CD4+ (1.7-fold), but not regulatory T cells. Our in vivo anti-tumor experiment suggested that TMNPs possessed the highest tumor growth inhibitory rate (80.86%) compared with the control group. We demonstrated that the nanoplatform could effectively inhibit the growth of tumors and enhance T cell recruitment through the co-delivery of paclitaxel and marimastat, which could be a promising strategy for the combination of chemotherapy and immunotherapy for cancer treatment.
Collapse
|
14
|
Barillari G. The Impact of Matrix Metalloproteinase-9 on the Sequential Steps of the Metastatic Process. Int J Mol Sci 2020; 21:ijms21124526. [PMID: 32630531 PMCID: PMC7350258 DOI: 10.3390/ijms21124526] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/20/2020] [Accepted: 06/23/2020] [Indexed: 02/07/2023] Open
Abstract
In industrialized countries, cancer is the second leading cause of death after cardiovascular disease. Most cancer patients die because of metastases, which consist of the self-transplantation of malignant cells in anatomical sites other than the one from where the tumor arose. Disseminated cancer cells retain the phenotypic features of the primary tumor, and display very poor differentiation indices and functional regulation. Upon arrival at the target organ, they replace preexisting, normal cells, thereby permanently compromising the patient's health; the metastasis can, in turn, metastasize. The spread of cancer cells implies the degradation of the extracellular matrix by a variety of enzymes, among which the matrix metalloproteinase (MMP)-9 is particularly effective. This article reviews the available published literature concerning the important role that MMP-9 has in the metastatic process. Additionally, information is provided on therapeutic approaches aimed at counteracting, or even preventing, the development of metastasis via the use of MMP-9 antagonists.
Collapse
Affiliation(s)
- Giovanni Barillari
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, 1 via Montpellier, 00133 Rome, Italy
| |
Collapse
|
15
|
Dalby E, Christensen SM, Wang J, Hamidzadeh K, Chandrasekaran P, Hughitt VK, Tafuri WL, Arantes RME, Rodrigues IA, Herbst R, El-Sayed NM, Sims GP, Mosser DM. Immune Complex-Driven Generation of Human Macrophages with Anti-Inflammatory and Growth-Promoting Activity. THE JOURNAL OF IMMUNOLOGY 2020; 205:102-112. [PMID: 32434940 DOI: 10.4049/jimmunol.1901382] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 04/22/2020] [Indexed: 12/23/2022]
Abstract
To maintain homeostasis, macrophages must be capable of assuming either an inflammatory or an anti-inflammatory phenotype. To better understand the latter, we stimulated human macrophages in vitro with TLR ligands in the presence of high-density immune complexes (IC). This combination of stimuli resulted in a broad suppression of inflammatory mediators and an upregulation of molecules involved in tissue remodeling and angiogenesis. Transcriptomic analysis of TLR stimulation in the presence of IC predicted the downstream activation of AKT and the inhibition of GSK3. Consequently, we pretreated LPS-stimulated human macrophages with small molecule inhibitors of GSK3 to partially phenocopy the regulatory effects of stimulation in the presence of IC. The upregulation of DC-STAMP and matrix metalloproteases was observed on these cells and may represent potential biomarkers for this regulatory activation state. To demonstrate the presence of these anti-inflammatory, growth-promoting macrophages in a human infectious disease, biopsies from patients with leprosy (Hanseniasis) were analyzed. The lepromatous form of this disease is characterized by hypergammaglobulinemia and defective cell-mediated immunity. Lesions in lepromatous leprosy contained macrophages with a regulatory phenotype expressing higher levels of DC-STAMP and lower levels of IL-12, relative to macrophages in tuberculoid leprosy lesions. Therefore, we propose that increased signaling by FcγR cross-linking on TLR-stimulated macrophages can paradoxically promote the resolution of inflammation and initiate processes critical to tissue growth and repair. It can also contribute to infectious disease progression.
Collapse
Affiliation(s)
- Elizabeth Dalby
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742
| | - Stephen M Christensen
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742
| | - Jingya Wang
- Department of Respiratory, Inflammation, and Autoimmunity, AstraZeneca, Gaithersburg, MD 20878
| | - Kajal Hamidzadeh
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742
| | - Prabha Chandrasekaran
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742
| | - V Keith Hughitt
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742.,Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD 20742
| | - Wagner Luiz Tafuri
- Department of General Pathology, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil; and
| | - Rosa Maria Esteves Arantes
- Department of General Pathology, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil; and
| | | | - Ronald Herbst
- Department of Respiratory, Inflammation, and Autoimmunity, AstraZeneca, Gaithersburg, MD 20878
| | - Najib M El-Sayed
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742.,Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD 20742
| | - Gary P Sims
- Department of Respiratory, Inflammation, and Autoimmunity, AstraZeneca, Gaithersburg, MD 20878;
| | - David M Mosser
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742;
| |
Collapse
|
16
|
Bronger H, Magdolen V, Goettig P, Dreyer T. Proteolytic chemokine cleavage as a regulator of lymphocytic infiltration in solid tumors. Cancer Metastasis Rev 2020; 38:417-430. [PMID: 31482487 PMCID: PMC6890590 DOI: 10.1007/s10555-019-09807-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In the past decade, immune-based therapies such as monoclonal antibodies against tumor epitopes or immune checkpoint inhibitors have become an integral part of contemporary cancer treatment in many entities. However, a fundamental prerequisite for the success of such therapies is a sufficient trafficking of tumor-infiltrating lymphocytes into the tumor microenvironment. This infiltration is facilitated by chemokines, a group of about 50 small proteins capable of chemotactically guiding leukocytes. Proteolytic inactivation of chemokines leading to an impaired infiltration of immune effector cells appears to be an efficient immune escape mechanism of solid cancers. The CXCR3 and CX3CR1 chemokine receptor ligands CXCL9-11 and CX3CL1, respectively, are mainly responsible for the tumor-suppressive lymphocytic infiltration into the tumor micromilieu. Their structure explains the biochemical basis of their proteolytic cleavage, while in vivo data from mouse models and patient samples shed light on the corresponding processes in cancer. The emerging roles of proteases, e.g., matrix metalloproteinases, cathepsins, and dipeptidyl peptidase 4, in chemokine inactivation define new resistance mechanisms against immunotherapies and identify attractive new targets to enhance immune intervention in cancer.
Collapse
Affiliation(s)
- Holger Bronger
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, Ismaninger Straße 22, D-81675, Munich, Germany.
| | - Viktor Magdolen
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, Ismaninger Straße 22, D-81675, Munich, Germany
| | - Peter Goettig
- Division of Structural Biology, Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Tobias Dreyer
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, Ismaninger Straße 22, D-81675, Munich, Germany
| |
Collapse
|
17
|
Ek M, Roth B, Engström G, Ohlsson B. AXIN1 in Plasma or Serum Is a Potential New Biomarker for Endometriosis. Int J Mol Sci 2019; 20:ijms20010189. [PMID: 30621017 PMCID: PMC6337238 DOI: 10.3390/ijms20010189] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 12/26/2018] [Accepted: 12/30/2018] [Indexed: 12/12/2022] Open
Abstract
Although endometriosis is considered an inflammatory disease, no reliable diagnostic biomarkers exist for use in clinical practice. The aim was to investigate the inflammatory profile in endometriosis using an exploratory approach of inflammation-related proteins. Patients with laparoscopy-verified endometriosis (N = 172), women with microscopic colitis (N = 50), healthy controls (N = 31), and age-matched controls from the general population (N = 100) were enrolled and questionnaires regarding socioeconomic factors, lifestyle habits, and medical history were completed. Sera from patients and healthy controls were analyzed for 92 inflammatory biomarkers using Proximity Extension Assay technology (PEA). Plasma AXIN1 levels were analyzed in patients with endometriosis and controls from the general population by ELISA. General linear model adjusted for age, Mann–Whitney U-test, and principal component analysis (PCA) were used for statistical calculations. Serum levels of AXIN1 and ST1A1 were increased in endometriosis compared with MC (p < 0.001) and healthy controls (p = 0.001), whereas CXCL9 levels were decreased. Plasma levels of AXIN1 were elevated in endometriosis compared with age-matched controls from the general population (30.0 (17.0–38.0) pg/mL vs. 19.5 (15.0–28.0) pg/mL, p < 0.001). PCA analysis identified four clusters of proteins, where one cluster differed between endometriosis and controls, with strong correlations for AXIN1 and ST1A1. Plasma/serum AXIN1 is an interesting biomarker to be further evaluated in endometriosis.
Collapse
Affiliation(s)
- Malin Ek
- Department of Internal Medicine, Skåne University Hospital, Lund University, 221 00 Lund, Sweden.
| | - Bodil Roth
- Department of Internal Medicine, Skåne University Hospital, Lund University, 221 00 Lund, Sweden.
| | - Gunnar Engström
- Department of Clinical Sciences in Malmö, Clinical Research Centre, Lund University, Box 50332, 202 13 Malmö, Sweden.
| | - Bodil Ohlsson
- Department of Internal Medicine, Skåne University Hospital, Lund University, 221 00 Lund, Sweden.
| |
Collapse
|
18
|
The gelatinases, MMP-2 and MMP-9, as fine tuners of neuroinflammatory processes. Matrix Biol 2019; 75-76:102-113. [DOI: 10.1016/j.matbio.2017.11.007] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 11/08/2017] [Accepted: 11/12/2017] [Indexed: 12/11/2022]
|
19
|
MMP-9 inhibition promotes anti-tumor immunity through disruption of biochemical and physical barriers to T-cell trafficking to tumors. PLoS One 2018; 13:e0207255. [PMID: 30500835 PMCID: PMC6267998 DOI: 10.1371/journal.pone.0207255] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 10/26/2018] [Indexed: 01/01/2023] Open
Abstract
Matrix metalloproteinase-9 (MMP-9), whose expression is frequently dysregulated in cancer, promotes tumor growth, invasion, and metastasis by multiple mechanisms, including extracellular matrix remodeling and growth-factor and cytokine activation. We developed a monoclonal antibody against murine MMP-9, which we found decreased growth of established primary tumors in an orthotopic model of HER2-driven breast cancer (HC11-NeuT) in immunocompetent mice. RNA sequencing (RNAseq) profiling of NeuT tumors and additional mouse model tumors revealed that anti-MMP-9 treatment resulted in upregulation of immune signature pathways associated with cytotoxic T-cell response. As there is a need to boost the low response rates observed with anti-PDL1 antibody treatment in the clinical setting, we assessed the potential of anti-MMP-9 to improve T-cell response to immune checkpoint inhibitor anti-PDL1 in NeuT tumors. Anti-MMP-9 and anti-PDL1 cotreatment reduced T-cell receptor (TCR) clonality and increased TCR diversity, as detected by TCR sequencing of NeuT tumors. Flow cytometry analyses of tumors showed that the combination treatment increased the frequency of CD3+ T cells, including memory/effector CD4 and CD8 T cells, but not regulatory T cells, among tumor-infiltrating leukocytes. Moreover, in vitro enzymatic assays corroborated that MMP-9 cleaves key T-cell chemoattractant CXC receptor 3 ligands (CXC ligand [CXCL] 9, CXCL10, and CXCL11) and renders them inactive in T-cell migration assays. Consistent with our in vitro experiments, analysis of NeuT tumor protein lysates showed that anti-MMP-9 treatment increases expression of CXCL10 and other T cell–stimulating factors, such as interleukin (IL)-12p70 and IL-18. We show that inhibition of MMP-9, a key component of the tumor-promoting and immune-suppressive myeloid inflammatory milieu, increases T-helper cell 1 type cytokines, trafficking of effector/memory T cells into tumors, and intratumoral T-cell diversity.
Collapse
|
20
|
How post-translational modifications influence the biological activity of chemokines. Cytokine 2018; 109:29-51. [DOI: 10.1016/j.cyto.2018.02.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 02/27/2018] [Accepted: 02/28/2018] [Indexed: 12/17/2022]
|
21
|
Vandooren J, Goeminne P, Boon L, Ugarte-Berzal E, Rybakin V, Proost P, Abu El-Asrar AM, Opdenakker G. Neutrophils and Activated Macrophages Control Mucosal Immunity by Proteolytic Cleavage of Antileukoproteinase. Front Immunol 2018; 9:1154. [PMID: 29892293 PMCID: PMC5985294 DOI: 10.3389/fimmu.2018.01154] [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: 03/09/2018] [Accepted: 05/08/2018] [Indexed: 12/13/2022] Open
Abstract
Antileukoproteinase or secretory leukocyte peptidase inhibitor is a small protein which protects the mucosal linings against excessive proteolysis, inflammation, and microbial infection. We discovered that gelatinase B or matrix metalloproteinase (MMP)-9, a secreted zinc-dependent endopeptidase typically found at sites of inflammation, destroys antileukoproteinase by cleavages within both of its two functional domains: the anti-microbial N-terminal and the anti-proteolytic C-terminal domains. Cleaved antileukoproteinase possessed a significantly lower ability to bind lipopolysaccharides (LPS) and a reduced capacity to inhibit neutrophil elastase (NE) activity. Whereas intact antileukoproteinase repressed proinflammatory transcript [prostaglandin-endoperoxide synthase 2 (PTGS2) and IL6] synthesis and protein secretion [e.g., of MMP-9] in human CD14+ blood monocytes stimulated with LPS, this effect was reduced or lost for cleaved antileukoproteinase. We demonstrated the in vivo presence of antileukoproteinase cleavage fragments in lower airway secretions of non-cystic fibrosis bronchiectasis patients with considerable levels of neutrophils and, hence, elastase and MMP-9 activity. As a comparison, other MMPs (MMP-2, MMP-7, and MMP-8) and serine proteases (NE, cathepsin G, and proteinase 3) were also able to cleave antileukoproteinase with similar or reduced efficiency. In conclusion, in specific mucosal pathologies, such as bronchiectasis, neutrophils, and macrophage subsets control local immune reactions by proteolytic regulation, here described as the balance between MMPs (in particular MMP-9), serine proteases and local tissue inhibitors.
Collapse
Affiliation(s)
- Jennifer Vandooren
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, KU Leuven, Leuven, Belgium
| | - Pieter Goeminne
- Department of Respiratory Disease, University Hospital of Gasthuisberg, Leuven, Belgium.,Department of Respiratory Disease, AZ Nikolaas, Sint-Niklaas, Belgium
| | - Lise Boon
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, KU Leuven, Leuven, Belgium
| | - Estefania Ugarte-Berzal
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, KU Leuven, Leuven, Belgium
| | - Vasily Rybakin
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, KU Leuven, Leuven, Belgium
| | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, KU Leuven, Leuven, Belgium
| | - Ahmed M Abu El-Asrar
- Department of Ophthalmology and Dr. Nasser Al-Rashid Research Chair in Ophthalmology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Ghislain Opdenakker
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, KU Leuven, Leuven, Belgium
| |
Collapse
|
22
|
Sachdev U, Vodovotz L, Bitner J, Barclay D, Zamora R, Yin J, Simmons RL, Vodovotz Y. Suppressed networks of inflammatory mediators characterize chronic venous insufficiency. J Vasc Surg Venous Lymphat Disord 2018; 6:358-366. [PMID: 29439934 DOI: 10.1016/j.jvsv.2017.11.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 11/05/2017] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Chronic venous insufficiency (CVI) affects 25 million adults in the United States. Little emphasis has been placed on inflammatory changes associated with CVI. We hypothesize that in patients with early to mid-stage benign varicose vein disease, differences in circulating inflammatory mediators will be manifested in blood draining the involved area vs circulating blood in control subjects. METHODS Patients undergoing either endovenous ablation or sclerotherapy for Clinical, Etiology, Anatomy, and Pathophysiology clinical class 3 to 5 disease underwent phlebotomy from regional veins at the time of the procedure. The patient's age, gender, clinical class, duration of symptoms, presence of superficial truncal reflux by duplex ultrasound, and treatment modality were recorded. Plasma from patients and banked blood samples from healthy volunteers (HVs) were subjected to Luminex (EMD Millipore, Billerica, Mass) to evaluate the expression of an established panel of 20 inflammatory mediators. Mediator concentrations were compared between patients and HVs using Mann-Whitney U tests. Importantly, computational analysis allowed us to compare not only the panel of inflammatory mediators but also the inflammatory networks connecting these mediators to one another. Principal components were analyzed to assess network robustness in each group. RESULTS CVI venous blood revealed significantly lower levels of monokine induced by γ interferon, soluble interleukin (IL) 2 receptor α chain, IL-4, IL-6, IL-7, tumor necrosis factor α, eotaxin, and granulocyte-macrophage colony-stimulating factor than blood from controls. Inflammatory networks were significantly less complex and less robust in the CVI patients compared with HVs. Based on principal component analysis, responses among HVs were more varied than those of CVI patients. CONCLUSIONS We demonstrate that patients with CVI have significant differences not only in blood-borne inflammatory mediators but also in the interconnectedness of these mediators with one another and in their principal inflammatory characteristics. Results suggest hypoinflammation in chronic nonhealing changes in CVI. These novel findings, if validated in larger cohorts, may help predict the risk of disease progression or response to therapy in the future and may guide mechanistic studies on tissue responses to CVI.
Collapse
Affiliation(s)
- Ulka Sachdev
- Division of Vascular Surgery, University of Pittsburgh, Pittsburgh, Pa; Department of Surgery, University of Pittsburgh, Pittsburgh, Pa.
| | - Lena Vodovotz
- School of Medicine, University of Pittsburgh, Pittsburgh, Pa
| | - Julie Bitner
- Division of Vascular Surgery, University of Pittsburgh, Pittsburgh, Pa
| | - Derek Barclay
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pa
| | - Ruben Zamora
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pa
| | - Jinling Yin
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pa
| | | | - Yoram Vodovotz
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pa
| |
Collapse
|
23
|
Metzemaekers M, Vanheule V, Janssens R, Struyf S, Proost P. Overview of the Mechanisms that May Contribute to the Non-Redundant Activities of Interferon-Inducible CXC Chemokine Receptor 3 Ligands. Front Immunol 2018; 8:1970. [PMID: 29379506 PMCID: PMC5775283 DOI: 10.3389/fimmu.2017.01970] [Citation(s) in RCA: 196] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 12/20/2017] [Indexed: 12/17/2022] Open
Abstract
The inflammatory chemokines CXCL9, CXCL10, and CXCL11 are predominantly induced by interferon (IFN)-γ and share an exclusive chemokine receptor named CXC chemokine receptor 3 (CXCR3). With a prototype function of directing temporal and spatial migration of activated T cells and natural killer cells, and inhibitory effects on angiogenesis, these CXCR3 ligands have been implicated in infection, acute inflammation, autoinflammation and autoimmunity, as well as in cancer. Intense former research efforts led to recent and ongoing clinical trials using CXCR3 and CXCR3 ligand targeting molecules. Scientific evidence has claimed mutual redundancy, ligand dominance, collaboration or even antagonism, depending on the (patho)physiological context. Most research on their in vivo activity, however, illustrates that CXCL9, CXCL10, and CXCL11 each contribute to the activation and trafficking of CXCR3 expressing cells in a non-redundant manner. When looking into detail, one can unravel a multistep machinery behind final CXCR3 ligand functions. Not only can specific cell types secrete individual CXCR3 interacting chemokines in response to certain stimuli, but also the receptor and glycosaminoglycan interactions, major associated intracellular pathways and susceptibility to processing by particular enzymes, among others, seem ligand-specific. Here, we overview major aspects of the molecular properties and regulatory mechanisms of IFN-induced CXCR3 ligands, and propose that their in vivo non-redundancy is a reflection of the unprecedented degree of versatility that seems inherent to the IFN-related CXCR3 chemokine system.
Collapse
Affiliation(s)
- Mieke Metzemaekers
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute, KU Leuven, Leuven, Belgium
| | - Vincent Vanheule
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute, KU Leuven, Leuven, Belgium
| | - Rik Janssens
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute, KU Leuven, Leuven, Belgium
| | - Sofie Struyf
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute, KU Leuven, Leuven, Belgium
| | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute, KU Leuven, Leuven, Belgium
| |
Collapse
|
24
|
Chaemsaithong P, Romero R, Docheva N, Chaiyasit N, Bhatti G, Pacora P, Hassan SS, Yeo L, Erez O. Comparison of rapid MMP-8 and interleukin-6 point-of-care tests to identify intra-amniotic inflammation/infection and impending preterm delivery in patients with preterm labor and intact membranes . J Matern Fetal Neonatal Med 2018; 31:228-244. [PMID: 28081646 PMCID: PMC5769687 DOI: 10.1080/14767058.2017.1281904] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 01/04/2017] [Accepted: 01/10/2017] [Indexed: 01/30/2023]
Abstract
OBJECTIVE Among patients presenting with preterm labor and intact membranes, those with intra-amniotic inflammation have adverse obstetrical and neonatal outcomes. The diagnosis of intra-amniotic inflammation can easily be made by detecting an elevated concentration of the cytokine interleukin (IL)-6 or the enzyme neutrophil collagenase, also known as matrix metalloproteinase (MMP)-8. The diagnostic performances of MMP-8 and IL-6 enzyme-linked immunosorbent assay tests are similar. Recently, a rapid test has become available for point-of-care determination of either MMP-8 or IL-6. The objectives of this study were to compare the diagnostic indices and predictive values between the rapid MMP-8 and IL-6 tests for the identification of intra-amniotic inflammation in patients with preterm labor and intact membranes. MATERIALS AND METHODS We performed a retrospective cohort study including 124 women with singleton pregnancies who presented with symptoms of preterm labor and underwent transabdominal amniocentesis for the evaluation of microbial invasion of the amniotic cavity (MIAC). MIAC was defined according to amniotic fluid culture results (aerobic and anaerobic bacteria as well as genital Mycoplasmas). Amniotic fluid white blood cell (WBC) counts were determined using a hemocytometer chamber. An elevated amniotic fluid MMP-8 concentration was assessed using Yoon's MMP-8 Check® (cutoff: 10 ng/mL). An elevated amniotic fluid IL-6 concentration was scored when there was a positive result for the lateral flow-based immunoassay (cutoff: ≥745 pg/mL and ≥1000 pg/mL). In order to objectively compare rapid MMP-8 and rapid IL-6 tests to identify intra-amniotic inflammation, an amniotic fluid WBC count of ≥50 cells/mm3 was used to define intra-amniotic inflammation. RESULTS (1) The rapid tests had the same sensitivity for the detection of intra-amniotic inflammation [85.7% (18/21) for all]; (2) the specificity of the rapid MMP-8 test was higher than that of the rapid IL-6 test (cutoff: 745 pg/mL) for the identification of intra-amniotic inflammation [72.8% (75/103) vs. 64.1% (66/103); p < 0.05]; and (3) there were no differences in the sensitivity and specificity between the rapid MMP-8 test and the rapid IL-6 test (cutoff:1000 pg/mL) in the identification of intra-amniotic inflammation. Of 13 patients with discrepant results between the rapid MMP-8 and rapid IL-6 tests, two had a positive MMP-8 but a negative rapid IL-6 test, and both delivered preterm - one within 24 h, and the other within 10 days - and both had acute histologic chorioamnionitis. On the other hand, there were 11 patients with a positive rapid IL-6 but a negative rapid MMP-8 result: 10 delivered preterm, 3 had acute histologic chorioamnionitis and 1 had subacute chorionitis. CONCLUSION We conclude that the rapid MMP-8 test has a better specificity than the rapid IL-6 (cutoff: 745 pg/mL) assay for the detection of intra-amniotic infection. Moreover, we observed that among patients who were not identified as having intra-amniotic infection or inflammation by the standard cultivation technique and amniotic fluid WBC count, those who had a positive MMP-8 rapid test delivered preterm and had acute histologic chorioamnionitis.
Collapse
Affiliation(s)
- Piya Chaemsaithong
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Roberto Romero
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
| | - Nikolina Docheva
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Noppadol Chaiyasit
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Gaurav Bhatti
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Percy Pacora
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Sonia S. Hassan
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Lami Yeo
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Offer Erez
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
25
|
Jansen AFM, Schoffelen T, Textoris J, Mege JL, Nabuurs-Franssen M, Raijmakers RPH, Netea MG, Joosten LAB, Bleeker-Rovers CP, van Deuren M. CXCL9, a promising biomarker in the diagnosis of chronic Q fever. BMC Infect Dis 2017; 17:556. [PMID: 28793883 PMCID: PMC5551022 DOI: 10.1186/s12879-017-2656-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 07/31/2017] [Indexed: 12/29/2022] Open
Abstract
Background In the aftermath of the largest Q fever outbreak in the world, diagnosing the potentially lethal complication chronic Q fever remains challenging. PCR, Coxiella burnetii IgG phase I antibodies, CRP and 18F–FDG-PET/CT scan are used for diagnosis and monitoring in clinical practice. We aimed to identify and test biomarkers in order to improve discriminative power of the diagnostic tests and monitoring of chronic Q fever. Methods We performed a transcriptome analysis on C. burnetii stimulated PBMCs of 4 healthy controls and 6 chronic Q fever patients and identified genes that were most differentially expressed. The gene products were determined using Luminex technology in whole blood samples stimulated with heat-killed C. burnetii and serum samples from chronic Q fever patients and control subjects. Results Gene expression of the chemokines CXCL9, CXCL10, CXCL11 and CCL8 was strongly up-regulated in C. burnetii stimulated PBMCs of chronic Q fever patients, in contrast to healthy controls. In whole blood cultures of chronic Q fever patients, production of all four chemokines was increased upon C. burnetii stimulation, but also healthy controls and past Q fever individuals showed increased production of CXCL9, CXCL10 and CCL8. However, CXCL9 and CXCL11 production was significantly higher for chronic Q fever patients compared to past Q fever individuals. In addition, CXCL9 serum concentrations in chronic Q fever patients were higher than in past Q fever individuals. Conclusion CXCL9 protein, measured in serum or as C. burnetii stimulated production, is a promising biomarker for the diagnosis of chronic Q fever. Electronic supplementary material The online version of this article (doi:10.1186/s12879-017-2656-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anne F M Jansen
- Department of Internal Medicine 463, Radboud center for Infectious Diseases (RCI), Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands. .,Radboud Expert Center for Q fever, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
| | - Teske Schoffelen
- Department of Internal Medicine 463, Radboud center for Infectious Diseases (RCI), Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.,Radboud Expert Center for Q fever, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Julien Textoris
- Université Claude Bernard Lyon 1, Hospices Civils de Lyon, bioMérieux; EA7426 "Pathophysiology of injury induced immunosuppression (PI3)", Hôpital E. Herriot, Lyon, France
| | | | - Marrigje Nabuurs-Franssen
- Department of Medical Microbiology and Infectious Diseases, Canisius Wilhelmina Hospital, Nijmegen, The Netherlands
| | - Ruud P H Raijmakers
- Department of Internal Medicine 463, Radboud center for Infectious Diseases (RCI), Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.,Radboud Expert Center for Q fever, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine 463, Radboud center for Infectious Diseases (RCI), Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.,Radboud Expert Center for Q fever, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine 463, Radboud center for Infectious Diseases (RCI), Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.,Radboud Expert Center for Q fever, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Chantal P Bleeker-Rovers
- Department of Internal Medicine 463, Radboud center for Infectious Diseases (RCI), Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.,Radboud Expert Center for Q fever, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Marcel van Deuren
- Department of Internal Medicine 463, Radboud center for Infectious Diseases (RCI), Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands. .,Radboud Expert Center for Q fever, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
26
|
Multimarker risk stratification approach at multiple sclerosis onset. Clin Immunol 2017; 181:43-50. [PMID: 28578025 DOI: 10.1016/j.clim.2017.05.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/17/2017] [Accepted: 05/26/2017] [Indexed: 12/20/2022]
Abstract
Delay in the diagnosis of multiple sclerosis (MS) stems from the lack of specific clinical and analytical markers to assist in the early diagnosis and prediction of progressive course. We propose a decision-tree model that better defines early at onset MS patients and those with the progressive form by analysing a 12-biomarkers panel in serum and CSF samples of patients with MS, other neurological diseases (OND) and healthy contols. Thus, patients at onset of neurological disease were first classified by serum IL-7 levels <141pg/ml (OR=6.51, p<0.001). Combination of IL-7 and CXCL10 indicated risk for a specific MS clinical form, where IL-7<141 and CXCL10<570pg/ml were associated with the highest risk for PP-MS (OR=22, p=0.01). Unexpectedly, both PP-MS and RR-MS patients shared significantly decreased prototypical biomarkers of inflammation and tissue regeneration in CSF than OND suggesting a defective intrinsic immune response playing a role at the beginning of the disease.
Collapse
|
27
|
Proost P, Struyf S, Van Damme J, Fiten P, Ugarte-Berzal E, Opdenakker G. Chemokine isoforms and processing in inflammation and immunity. J Autoimmun 2017; 85:45-57. [PMID: 28684129 DOI: 10.1016/j.jaut.2017.06.009] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 06/21/2017] [Indexed: 12/16/2022]
Abstract
The first dimension of chemokine heterogeneity is reflected by their discovery and purification as natural proteins. Each of those chemokines attracted a specific inflammatory leukocyte type. With the introduction of genomic technologies, a second wave of chemokine heterogeneity was established by the discovery of putative chemokine-like sequences and by demonstrating chemotactic activity of the gene products in physiological leukocyte homing. In the postgenomic era, the third dimension of chemokine heterogeneity is the description of posttranslational modifications on most chemokines. Proteolysis of chemokines, for instance by dipeptidyl peptidase IV (DPP IV/CD26) and by matrix metalloproteinases (MMPs) is already well established as a biological control mechanism to activate, potentiate, dampen or abrogate chemokine activities. Other posttranslational modifications are less known. Theoretical N-linked and O-linked attachment sites for chemokine glycosylation were searched with bio-informatic tools and it was found that most chemokines are not glycosylated. These findings are corroborated with a low number of experimental studies demonstrating N- or O-glycosylation of natural chemokine ligands. Because attached oligosaccharides protect proteins against proteolytic degradation, their absence may explain the fast turnover of chemokines in the protease-rich environments of infection and inflammation. All chemokines interact with G protein-coupled receptors (GPCRs) and glycosaminoglycans (GAGs). Whether lectin-like GAG-binding induces cellular signaling is not clear, but these interactions are important for leukocyte migration and have already been exploited to reduce inflammation. In addition to selective proteolysis, citrullination and nitration/nitrosylation are being added as biologically relevant modifications contributing to functional chemokine heterogeneity. Resulting chemokine isoforms with reduced affinity for GPCRs reduce leukocyte migration in various models of inflammation. Here, these third dimension modifications are compared, with reflections on the biological and pathological contexts in which these posttranslational modifications take place and contribute to the repertoire of chemokine functions and with an emphasis on autoimmune diseases.
Collapse
Affiliation(s)
- Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, University of Leuven, Herestraat 49, B-3000, Leuven, Belgium.
| | - Sofie Struyf
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, University of Leuven, Herestraat 49, B-3000, Leuven, Belgium.
| | - Jo Van Damme
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, University of Leuven, Herestraat 49, B-3000, Leuven, Belgium.
| | - Pierre Fiten
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, University of Leuven, Herestraat 49, B-3000, Leuven, Belgium.
| | - Estefania Ugarte-Berzal
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, University of Leuven, Herestraat 49, B-3000, Leuven, Belgium.
| | - Ghislain Opdenakker
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, University of Leuven, Herestraat 49, B-3000, Leuven, Belgium.
| |
Collapse
|
28
|
Bronger H, Karge A, Dreyer T, Zech D, Kraeft S, Avril S, Kiechle M, Schmitt M. Induction of cathepsin B by the CXCR3 chemokines CXCL9 and CXCL10 in human breast cancer cells. Oncol Lett 2017; 13:4224-4230. [PMID: 28599423 PMCID: PMC5453043 DOI: 10.3892/ol.2017.5994] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 01/31/2017] [Indexed: 12/25/2022] Open
Abstract
Cathepsin B (CTSB) is a lysosomal cysteine protease that has been linked to the progression of breast cancer, for example by activation of other proteases and tumor-promoting cytokines, thereby supporting tumor invasion and metastasis. Previously, it was shown that CTSB cleaves and inactivates C-X-C motif chemokine receptor 3 (CXCR3) chemokines. As CXCR3 ligands have been demonstrated to induce proteases in cancer cells, the present study hypothesized that they may also affect CTSB in breast cancer cells. The results demonstrated that the human breast cancer tumor cell lines MCF-7 and MDA-MB-231 express the CXCR3 splice variants A and B and CTSB. Upon binding to CXCR3, the two chemokine ligands C-X-C motif chemokine ligand (CXCL) 9 and CXCL10 trigger upregulation of CTSB in these breast cancer cells, whereas the CXCR3-B-specific ligand CXCL4 has no such effect, suggesting the involvement of CXCR3-A in the regulation of CTSB. In early-stage human breast cancer specimens (n=81), overexpression of CXCR3 is associated with statistically significant poorer overall survival, independent of lymph node status, tumor size and nuclear grading (hazard ratio=1.99; 95% confidence interval=1.00–3.97; P=0.050). In conclusion, the data from the current study propose a so far unknown mechanism by which breast cancer cells may exploit tumor-suppressive chemokines to enhance their invasiveness and reduce immune cell infiltration by the degradation of these chemokines. This mechanism may support the established unfavorable prognostic feature of CXCR3 expression in breast cancer.
Collapse
Affiliation(s)
- Holger Bronger
- Department of Gynecology and Obstetrics, Technical University of Munich, D-81675 Munich, Germany
| | - Anne Karge
- Department of Gynecology and Obstetrics, Technical University of Munich, D-81675 Munich, Germany
| | - Tobias Dreyer
- Department of Gynecology and Obstetrics, Technical University of Munich, D-81675 Munich, Germany
| | - Daniela Zech
- Department of Gynecology and Obstetrics, Technical University of Munich, D-81675 Munich, Germany
| | - Sara Kraeft
- Department of Gynecology and Obstetrics, Technical University of Munich, D-81675 Munich, Germany
| | - Stefanie Avril
- Department of Pathology, Technical University of Munich, D-81675 Munich, Germany.,Department of Pathology, Case Western Reserve University School of Medicine, University Hospital Case Medical Center, Cleveland, Ohio 44106-7288, USA
| | - Marion Kiechle
- Department of Gynecology and Obstetrics, Technical University of Munich, D-81675 Munich, Germany
| | - Manfred Schmitt
- Department of Gynecology and Obstetrics, Technical University of Munich, D-81675 Munich, Germany
| |
Collapse
|
29
|
Metzemaekers M, Van Damme J, Mortier A, Proost P. Regulation of Chemokine Activity - A Focus on the Role of Dipeptidyl Peptidase IV/CD26. Front Immunol 2016; 7:483. [PMID: 27891127 PMCID: PMC5104965 DOI: 10.3389/fimmu.2016.00483] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 10/21/2016] [Indexed: 12/15/2022] Open
Abstract
Chemokines are small, chemotactic proteins that play a crucial role in leukocyte migration and are, therefore, essential for proper functioning of the immune system. Chemokines exert their chemotactic effect by activation of chemokine receptors, which are G protein-coupled receptors (GPCRs), and interaction with glycosaminoglycans (GAGs). Furthermore, the exact chemokine function is modulated at the level of posttranslational modifications. Among the different types of posttranslational modifications that were found to occur in vitro and in vivo, i.e., proteolysis, citrullination, glycosylation, and nitration, NH2-terminal proteolysis of chemokines has been described most intensively. Since the NH2-terminal chemokine domain mediates receptor interaction, NH2-terminal modification by limited proteolysis or amino acid side chain modification can drastically affect their biological activity. An enzyme that has been shown to provoke NH2-terminal proteolysis of various chemokines is dipeptidyl peptidase IV or CD26. This multifunctional protein is a serine protease that preferably cleaves dipeptides from the NH2-terminal region of peptides and proteins with a proline or alanine residue in the penultimate position. Various chemokines possess such a proline or alanine residue, and CD26-truncated forms of these chemokines have been identified in cell culture supernatant as well as in body fluids. The effects of CD26-mediated proteolysis in the context of chemokines turned out to be highly complex. Depending on the chemokine ligand, loss of these two NH2-terminal amino acids can result in either an increased or a decreased biological activity, enhanced receptor specificity, inactivation of the chemokine ligand, or generation of receptor antagonists. Since chemokines direct leukocyte migration in homeostatic as well as pathophysiologic conditions, CD26-mediated proteolytic processing of these chemotactic proteins may have significant consequences for appropriate functioning of the immune system. After introducing the chemokine family together with the GPCRs and GAGs, as main interaction partners of chemokines, and discussing the different forms of posttranslational modifications, this review will focus on the intriguing relationship of chemokines with the serine protease CD26.
Collapse
Affiliation(s)
- Mieke Metzemaekers
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute, KU Leuven , Leuven , Belgium
| | - Jo Van Damme
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute, KU Leuven , Leuven , Belgium
| | - Anneleen Mortier
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute, KU Leuven , Leuven , Belgium
| | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute, KU Leuven , Leuven , Belgium
| |
Collapse
|
30
|
Gounko NV, Martens E, Opdenakker G, Rybakin V. Thymocyte development in the absence of matrix metalloproteinase-9/gelatinase B. Sci Rep 2016; 6:29852. [PMID: 27432536 PMCID: PMC4949482 DOI: 10.1038/srep29852] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 06/24/2016] [Indexed: 12/13/2022] Open
Abstract
Matrix metalloproteinases (MMP) play critical roles in a variety of immune reactions by facilitating cell migration, and affect cell communication by processing both cytokines and cell surface receptors. Based on published data indicating that MMP-9 is upregulated upon T cell activation and also in the thymus upon the induction of negative selection, we investigated the contribution of MMP-9 into mouse T cell development and differentiation in the thymus. Our data suggest that MMP-9 deficiency does not result in major abnormalities in the development of any conventionally selected or agonist selected subsets and does not interfere with thymocyte apoptosis and clearance, and that MMP-9 expression is not induced in immature T cells at any stage of their thymic development.
Collapse
Affiliation(s)
- Natalia V Gounko
- Laboratory of Immunobiology, REGA Institute, Department of Microbiology and Immunology, KU Leuven, Minderbroedersstraat 10, Leuven 3000, Belgium.,Electron Microscopy Platform, Center for the Biology of Disease VIB and Center for Human Genetics KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Erik Martens
- Laboratory of Immunobiology, REGA Institute, Department of Microbiology and Immunology, KU Leuven, Minderbroedersstraat 10, Leuven 3000, Belgium
| | - Ghislain Opdenakker
- Laboratory of Immunobiology, REGA Institute, Department of Microbiology and Immunology, KU Leuven, Minderbroedersstraat 10, Leuven 3000, Belgium
| | - Vasily Rybakin
- Laboratory of Immunobiology, REGA Institute, Department of Microbiology and Immunology, KU Leuven, Minderbroedersstraat 10, Leuven 3000, Belgium
| |
Collapse
|
31
|
Decalf J, Tarbell KV, Casrouge A, Price JD, Linder G, Mottez E, Sultanik P, Mallet V, Pol S, Duffy D, Albert ML. Inhibition of DPP4 activity in humans establishes its in vivo role in CXCL10 post-translational modification: prospective placebo-controlled clinical studies. EMBO Mol Med 2016; 8:679-83. [PMID: 27137491 PMCID: PMC4888857 DOI: 10.15252/emmm.201506145] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Biochemical experiments, animal models, and observational studies in humans all support a role of dipeptidyl peptidase 4 (DPP4) in the N‐terminal truncation of CXCL10, which results in the generation of an antagonist form of the chemokine that limits T‐cell and NK cell migration. Motivated by the ability to regulate lymphocyte trafficking in vivo, we conducted two prospective clinical trials to test the effects of DPP4 inhibition on CXCL10 processing in healthy donors and in chronic hepatitis C patients, a disease in which DPP4 levels are found to be elevated. Participants were treated daily with 100 mg sitagliptin, a clinically approved DPP4 inhibitor. Plasma samples were analyzed using an ultrasensitive single‐molecule assay (Simoa) to distinguish the full‐length CXCL101–77 from the NH2‐truncated CXCL103–77, as compared to the total CXCL10 levels. Sitagliptin treatment resulted in a significant decrease in CXCL103–77 concentration, a reciprocal increase in CXCL101–77, with only minimal effects on total levels of the chemokine. These data provide the first direct evidence that in vivo DPP4 inhibition in humans can preserve the bioactive form of CXCL10, offering new therapeutic opportunities for DPP4 inhibitors.
Collapse
Affiliation(s)
- Jérémie Decalf
- The Laboratory of Dendritic Cell Immunobiology, Institut Pasteur, Paris, France INSERM U818, Paris, France
| | - Kristin V Tarbell
- Diabetes Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Armanda Casrouge
- The Laboratory of Dendritic Cell Immunobiology, Institut Pasteur, Paris, France INSERM U818, Paris, France
| | - Jeffrey D Price
- Diabetes Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Grace Linder
- Diabetes Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Estelle Mottez
- Center for Human Immunology, Institut Pasteur, Paris, France
| | - Philippe Sultanik
- Département d'Hépatologie, AP-HP, Hôpital Cochin, Université Paris Descartes, INSERM UMS20, Institut Pasteur, Paris, France
| | - Vincent Mallet
- Département d'Hépatologie, AP-HP, Hôpital Cochin, Université Paris Descartes, INSERM UMS20, Institut Pasteur, Paris, France
| | - Stanislas Pol
- Département d'Hépatologie, AP-HP, Hôpital Cochin, Université Paris Descartes, INSERM UMS20, Institut Pasteur, Paris, France
| | - Darragh Duffy
- The Laboratory of Dendritic Cell Immunobiology, Institut Pasteur, Paris, France INSERM U818, Paris, France Center for Human Immunology, Institut Pasteur, Paris, France
| | - Matthew L Albert
- The Laboratory of Dendritic Cell Immunobiology, Institut Pasteur, Paris, France INSERM U818, Paris, France Center for Human Immunology, Institut Pasteur, Paris, France Department of Cancer Immunotherapy, Genentech, South San Francisco, CA, USA
| |
Collapse
|
32
|
Matrix Metalloproteinases During Axonal Regeneration, a Multifactorial Role from Start to Finish. Mol Neurobiol 2016; 54:2114-2125. [PMID: 26924318 DOI: 10.1007/s12035-016-9801-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 02/16/2016] [Indexed: 12/19/2022]
Abstract
By proteolytic cleavage, matrix metalloproteinases (MMPs) not only remodel the extracellular matrix (ECM) but they also modify the structure and activity of other proteinases, growth factors, signaling molecules, cell surface receptors, etc. Their vast substrate repertoire adds a complex extra dimension of biological control and turns MMPs into important regulatory nodes in the protease web. In the central nervous system (CNS), the detrimental impact of elevated MMP activities has been well-described for traumatic injuries and many neurodegenerative diseases. Nonetheless, there is ample proof corroborating MMPs as fine regulators of CNS physiology, and well-balanced MMP activity is instrumental to development, plasticity, and repair. In this manuscript, we review the emerging evidence for MMPs as beneficial modulators of axonal regeneration in the mammalian CNS. By exploring the multifactorial causes underlying the inability of mature axons to regenerate, and describing how MMPs can help to overcome these hurdles, we emphasize the benign actions of these Janus-faced proteases.
Collapse
|
33
|
Cremers NAJ, Suttorp M, Gerritsen MM, Wong RJ, van Run-van Breda C, van Dam GM, Brouwer KM, Kuijpers-Jagtman AM, Carels CEL, Lundvig DMS, Wagener FADTG. Mechanical Stress Changes the Complex Interplay Between HO-1, Inflammation and Fibrosis, During Excisional Wound Repair. Front Med (Lausanne) 2015; 2:86. [PMID: 26697429 PMCID: PMC4678194 DOI: 10.3389/fmed.2015.00086] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 11/24/2015] [Indexed: 01/11/2023] Open
Abstract
Mechanical stress following surgery or injury can promote pathological wound healing and fibrosis, and lead to functional loss and esthetic problems. Splinted excisional wounds can be used as a model for inducing mechanical stress. The cytoprotective enzyme heme oxygenase-1 (HO-1) is thought to orchestrate the defense against inflammatory and oxidative insults that drive fibrosis. Here, we investigated the activation of the HO-1 system in a splinted and non-splinted full-thickness excisional wound model using HO-1-luc transgenic mice. Effects of splinting on wound closure, HO-1 promoter activity, and markers of inflammation and fibrosis were assessed. After seven days, splinted wounds were more than three times larger than non-splinted wounds, demonstrating a delay in wound closure. HO-1 promoter activity rapidly decreased following removal of the (epi)dermis, but was induced in both splinted and non-splinted wounds during skin repair. Splinting induced more HO-1 gene expression in 7-day wounds; however, HO-1 protein expression remained lower in the epidermis, likely due to lower numbers of keratinocytes in the re-epithelialization tissue. Higher numbers of F4/80-positive macrophages, αSMA-positive myofibroblasts, and increased levels of the inflammatory genes IL-1β, TNF-α, and COX-2 were present in 7-day splinted wounds. Surprisingly, mRNA expression of newly formed collagen (type III) was lower in 7-day wounds after splinting, whereas, VEGF and MMP-9 were increased. In summary, these data demonstrate that splinting delays cutaneous wound closure and HO-1 protein induction. The pro-inflammatory environment following splinting may facilitate higher myofibroblast numbers and increase the risk of fibrosis and scar formation. Therefore, inducing HO-1 activity against mechanical stress-induced inflammation and fibrosis may be an interesting strategy to prevent negative effects of surgery on growth and function in patients with orofacial clefts or in patients with burns.
Collapse
Affiliation(s)
- Niels A J Cremers
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences , Nijmegen , Netherlands ; Experimental Rheumatology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences , Nijmegen , Netherlands
| | - Maarten Suttorp
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences , Nijmegen , Netherlands
| | - Marlous M Gerritsen
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences , Nijmegen , Netherlands
| | - Ronald J Wong
- Department of Pediatrics, Stanford University School of Medicine , Stanford, CA , USA
| | - Coby van Run-van Breda
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences , Nijmegen , Netherlands
| | - Gooitzen M van Dam
- Department of Surgery, University Medical Center Groningen , Groningen , Netherlands
| | - Katrien M Brouwer
- Department of Plastic, Reconstructive and Hand Surgery, VU University Medical Center, MOVE Research Institute Amsterdam , Amsterdam , Netherlands ; Association of Dutch Burn Centers , Beverwijk , Netherlands
| | - Anne Marie Kuijpers-Jagtman
- Department of Orthodontics and Craniofacial Biology, Cleft Palate Craniofacial Center, Radboud University Medical Center , Nijmegen , Netherlands
| | - Carine E L Carels
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences , Nijmegen , Netherlands
| | - Ditte M S Lundvig
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences , Nijmegen , Netherlands
| | - Frank A D T G Wagener
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences , Nijmegen , Netherlands
| |
Collapse
|
34
|
Rees PA, Greaves NS, Baguneid M, Bayat A. Chemokines in Wound Healing and as Potential Therapeutic Targets for Reducing Cutaneous Scarring. Adv Wound Care (New Rochelle) 2015; 4:687-703. [PMID: 26543682 DOI: 10.1089/wound.2014.0568] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Significance: Cutaneous scarring is an almost inevitable end point of adult human wound healing. It is associated with significant morbidity, both physical and psychological. Pathological scarring, including hypertrophic and keloid scars, can be particularly debilitating. Manipulation of the chemokine system may lead to effective therapies for problematic lesions. Recent Advances: Rapid advancement in the understanding of chemokines and their receptors has led to exciting developments in the world of therapeutics. Modulation of their function has led to clinically effective treatments for conditions as diverse as human immunodeficiency virus and inflammatory bowel disease. Potential methods of targeting chemokines include monoclonal antibodies, small-molecule antagonists, interference with glycosaminoglycan binding and the use of synthetic truncated chemokines. Early work has shown promising results on scar development and appearance when the chemokine system is manipulated. Critical Issues: Chemokines are implicated in all stages of wound healing leading to the development of a cutaneous scar. An understanding of entirely regenerative wound healing in the developing fetus and how the expression of chemokines and their receptors change during the transition to the adult phenotype is central to addressing pathological scarring in adults. Future Directions: As our understanding of chemokine/receptor interactions and scar formation evolves it has become apparent that effective therapies will need to mirror the complexities in these diverse biological processes. It is likely that sophisticated treatments that sequentially influence multiple ligand/receptor interactions throughout all stages of wound healing will be required to deliver viable treatment options.
Collapse
Affiliation(s)
- Peter Adam Rees
- Plastic and Reconstructive Surgery Research, Manchester Institute of Biotechnology (MIB), The University of Manchester, Manchester, United Kingdom
- University Hospital of South Manchester NHS Foundation Trust, Wythenshawe Hospital, Manchester, United Kingdom
| | - Nicholas Stuart Greaves
- Plastic and Reconstructive Surgery Research, Manchester Institute of Biotechnology (MIB), The University of Manchester, Manchester, United Kingdom
- University Hospital of South Manchester NHS Foundation Trust, Wythenshawe Hospital, Manchester, United Kingdom
| | - Mohamed Baguneid
- University Hospital of South Manchester NHS Foundation Trust, Wythenshawe Hospital, Manchester, United Kingdom
| | - Ardeshir Bayat
- Plastic and Reconstructive Surgery Research, Manchester Institute of Biotechnology (MIB), The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
35
|
Dezutter-Dambuyant C, Durand I, Alberti L, Bendriss-Vermare N, Valladeau-Guilemond J, Duc A, Magron A, Morel AP, Sisirak V, Rodriguez C, Cox D, Olive D, Caux C. A novel regulation of PD-1 ligands on mesenchymal stromal cells through MMP-mediated proteolytic cleavage. Oncoimmunology 2015; 5:e1091146. [PMID: 27141350 PMCID: PMC4839348 DOI: 10.1080/2162402x.2015.1091146] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 08/28/2015] [Accepted: 08/31/2015] [Indexed: 12/31/2022] Open
Abstract
Whether fibroblasts regulate immune response is a crucial issue in the modulation of inflammatory responses. Herein, we demonstrate that foreskin fibroblasts (FFs) potently inhibit CD3+ T cell proliferation through a mechanism involving early apoptosis of activated T cells. Using blocking antibodies, we demonstrate that the inhibition of T cell proliferation occurs through cell-to-cell interactions implicating PD-1 receptor expressed on T cells and its ligands, PD-L1 and PD-L2, on fibroblasts. Dual PD-1 ligand neutralization is required to abrogate (i) binding of the PD-1-Fc fusion protein, (ii) early apoptosis of T cells, and (iii) inhibition of T cell proliferation. Of utmost importance, we provide the first evidence that PD-1 ligand expression is regulated through proteolytic cleavage by endogenous matrix metalloproteinases (MMPs) without transcriptional alteration during culture-time. Using (i) different purified enzymatic activities, (ii) MMP-specific inhibitors, and (iii) recombinant human MMP-9 and MMP-13, we demonstrated that in contrast to CD80/CD86, PD-L1 was selectively cleaved by MMP-13, while PD-L2 was sensitive to broader MMP activities. Their cleavage by exogenous MMP-9 and MMP-13 with loss of PD-1 binding domain resulted in the reversion of apoptotic signals on mitogen-activated CD3+ T cells. We suggest that MMP-dependent cleavage of PD-1 ligands on fibroblasts may limit their immunosuppressive capacity and thus contribute to the exacerbation of inflammation in tissues. In contrast, carcinoma-associated fibroblasts appear PD-1 ligand-depleted through MMP activity that may impair physical deletion of exhausted defective memory T cells through apoptosis and facilitate their regulatory functions. These observations should be considered when using the powerful PD-1/PD-L1 blocking immunotherapies.
Collapse
Affiliation(s)
- Colette Dezutter-Dambuyant
- Université de Lyon, Lyon, France; Université Lyon 1, ISPB, Lyon, France; INSERM U1052, Center de Recherche en Cancérologie de Lyon, Lyon, France; CNRS UMR5286, Center de Recherche en Cancérologie de Lyon, Lyon, France
| | - Isabelle Durand
- Université de Lyon, Lyon, France; Université Lyon 1, ISPB, Lyon, France; INSERM U1052, Center de Recherche en Cancérologie de Lyon, Lyon, France; CNRS UMR5286, Center de Recherche en Cancérologie de Lyon, Lyon, France
| | - Laurent Alberti
- Université de Lyon, Lyon, France; Université Lyon 1, ISPB, Lyon, France; INSERM U1052, Center de Recherche en Cancérologie de Lyon, Lyon, France; CNRS UMR5286, Center de Recherche en Cancérologie de Lyon, Lyon, France
| | - Nathalie Bendriss-Vermare
- Université de Lyon, Lyon, France; Université Lyon 1, ISPB, Lyon, France; INSERM U1052, Center de Recherche en Cancérologie de Lyon, Lyon, France; CNRS UMR5286, Center de Recherche en Cancérologie de Lyon, Lyon, France
| | - Jenny Valladeau-Guilemond
- Université de Lyon, Lyon, France; Université Lyon 1, ISPB, Lyon, France; INSERM U1052, Center de Recherche en Cancérologie de Lyon, Lyon, France; CNRS UMR5286, Center de Recherche en Cancérologie de Lyon, Lyon, France
| | - Adeline Duc
- Université de Lyon, Lyon, France; Université Lyon 1, ISPB, Lyon, France; INSERM U1052, Center de Recherche en Cancérologie de Lyon, Lyon, France; CNRS UMR5286, Center de Recherche en Cancérologie de Lyon, Lyon, France
| | - Audrey Magron
- Université de Lyon, Lyon, France; Université Lyon 1, ISPB, Lyon, France; INSERM U1052, Center de Recherche en Cancérologie de Lyon, Lyon, France; CNRS UMR5286, Center de Recherche en Cancérologie de Lyon, Lyon, France
| | - Anne-Pierre Morel
- Université de Lyon, Lyon, France; Université Lyon 1, ISPB, Lyon, France; INSERM U1052, Center de Recherche en Cancérologie de Lyon, Lyon, France; CNRS UMR5286, Center de Recherche en Cancérologie de Lyon, Lyon, France
| | - Vanja Sisirak
- Université de Lyon, Lyon, France; Université Lyon 1, ISPB, Lyon, France; INSERM U1052, Center de Recherche en Cancérologie de Lyon, Lyon, France; CNRS UMR5286, Center de Recherche en Cancérologie de Lyon, Lyon, France
| | - Céline Rodriguez
- Université de Lyon, Lyon, France; Université Lyon 1, ISPB, Lyon, France; INSERM U1052, Center de Recherche en Cancérologie de Lyon, Lyon, France; CNRS UMR5286, Center de Recherche en Cancérologie de Lyon, Lyon, France
| | - David Cox
- Université de Lyon, Lyon, France; Université Lyon 1, ISPB, Lyon, France; INSERM U1052, Center de Recherche en Cancérologie de Lyon, Lyon, France; CNRS UMR5286, Center de Recherche en Cancérologie de Lyon, Lyon, France
| | - Daniel Olive
- Aix-Marseille Université, Marseille, France, Inserm U1068, Center de Recherche en Cancérologie de Marseille (CRCM), Immunity & Cancer Institut Paoli-Calmettes; Aix-Marseille Université UM 105, CNRS UMR 7258, IBiSA Cancer Immunomonitoring Platform, Marseilles, France
| | - Christophe Caux
- Université de Lyon, Lyon, France; Université Lyon 1, ISPB, Lyon, France; INSERM U1052, Center de Recherche en Cancérologie de Lyon, Lyon, France; CNRS UMR5286, Center de Recherche en Cancérologie de Lyon, Lyon, France
| |
Collapse
|
36
|
Deleon-Pennell KY, Altara R, Yabluchanskiy A, Modesti A, Lindsey ML. The circular relationship between matrix metalloproteinase-9 and inflammation following myocardial infarction. IUBMB Life 2015; 67:611-8. [PMID: 26269290 DOI: 10.1002/iub.1408] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Accepted: 07/16/2015] [Indexed: 12/26/2022]
Abstract
Matrix metalloproteinase-9 (MMP-9) regulates remodeling of the left ventricle after myocardial infarction (MI) and is tightly linked to the inflammatory response. The inflammatory response serves to recruit leukocytes as part of the wound healing reaction to the MI injury, and infiltrated leukocytes produce cytokines and chemokines that stimulate MMP-9 production and release. In turn, MMP-9 proteolyzes cytokines and chemokines. Although in most cases, MMP-9 cleavage of the cytokine or chemokine substrate serves to increase activity, there are cases where cleavage results in reduced activity. Global MMP-9 deletion in mouse MI models has proven beneficial, suggesting inhibition of some aspects of MMP-9 activity may be valuable for clinical use. At the same time, overexpression of MMP-9 in macrophages has also proven beneficial, indicating that we still do not fully understand the complexity of MMP-9 mechanisms of action. In this review, we summarize the cycle of MMP-9 effects on cytokine production and cleavage to regulate leukocyte functions. Although we use MI as the example process, similar events occur in other inflammatory and wound healing conditions.
Collapse
Affiliation(s)
- Kristine Y Deleon-Pennell
- Department of Physiology and Biophysics, San Antonio Cardiovascular Proteomics Center and Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, MS, USA
| | - Raffaele Altara
- Department of Physiology and Biophysics, San Antonio Cardiovascular Proteomics Center and Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, MS, USA
| | - Andriy Yabluchanskiy
- Department of Physiology and Biophysics, San Antonio Cardiovascular Proteomics Center and Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, MS, USA
| | - Alessandra Modesti
- Dipartimento di Scienze Biomediche, Sperimentali e Cliniche, Università degli Studi di Firenze, Firenze, Italy
| | - Merry L Lindsey
- Department of Physiology and Biophysics, San Antonio Cardiovascular Proteomics Center and Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, MS, USA.,Research Service, G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS, USA
| |
Collapse
|
37
|
Vanheule V, Janssens R, Boff D, Kitic N, Berghmans N, Ronsse I, Kungl AJ, Amaral FA, Teixeira MM, Van Damme J, Proost P, Mortier A. The Positively Charged COOH-terminal Glycosaminoglycan-binding CXCL9(74-103) Peptide Inhibits CXCL8-induced Neutrophil Extravasation and Monosodium Urate Crystal-induced Gout in Mice. J Biol Chem 2015; 290:21292-304. [PMID: 26183778 DOI: 10.1074/jbc.m115.649855] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Indexed: 12/11/2022] Open
Abstract
The ELR(-)CXC chemokine CXCL9 is characterized by a long, highly positively charged COOH-terminal region, absent in most other chemokines. Several natural leukocyte- and fibroblast-derived COOH-terminally truncated CXCL9 forms missing up to 30 amino acids were identified. To investigate the role of the COOH-terminal region of CXCL9, several COOH-terminal peptides were chemically synthesized. These peptides display high affinity for glycosaminoglycans (GAGs) and compete with functional intact chemokines for GAG binding, the longest peptide (CXCL9(74-103)) being the most potent. The COOH-terminal peptide CXCL9(74-103) does not signal through or act as an antagonist for CXCR3, the G protein-coupled CXCL9 receptor, and does not influence neutrophil chemotactic activity of CXCL8 in vitro. Based on the GAG binding data, an anti-inflammatory role for CXCL9(74-103) was further evidenced in vivo. Simultaneous intravenous injection of CXCL9(74-103) with CXCL8 injection in the joint diminished CXCL8-induced neutrophil extravasation. Analogously, monosodium urate crystal-induced neutrophil migration to the tibiofemural articulation, a murine model of gout, is highly reduced by intravenous injection of CXCL9(74-103). These data show that chemokine-derived peptides with high affinity for GAGs may be used as anti-inflammatory peptides; by competing with active chemokines for binding and immobilization on GAGs, these peptides may lower chemokine presentation on the endothelium and disrupt the generation of a chemokine gradient, thereby preventing a chemokine from properly performing its chemotactic function. The CXCL9 peptide may serve as a lead molecule for further development of inhibitors of inflammation based on interference with chemokine-GAG interactions.
Collapse
Affiliation(s)
- Vincent Vanheule
- From the Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute, KU Leuven, 3000 Leuven, Belgium
| | - Rik Janssens
- From the Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute, KU Leuven, 3000 Leuven, Belgium
| | - Daiane Boff
- the Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil, and
| | - Nikola Kitic
- the Department of Pharmaceutical Chemistry, Institute of Pharmaceutical Sciences, Karl-Franzes Universität, 8010 Graz, Austria
| | - Nele Berghmans
- From the Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute, KU Leuven, 3000 Leuven, Belgium
| | - Isabelle Ronsse
- From the Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute, KU Leuven, 3000 Leuven, Belgium
| | - Andreas J Kungl
- the Department of Pharmaceutical Chemistry, Institute of Pharmaceutical Sciences, Karl-Franzes Universität, 8010 Graz, Austria
| | - Flavio Almeida Amaral
- the Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil, and
| | - Mauro Martins Teixeira
- the Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil, and
| | - Jo Van Damme
- From the Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute, KU Leuven, 3000 Leuven, Belgium
| | - Paul Proost
- From the Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute, KU Leuven, 3000 Leuven, Belgium,
| | - Anneleen Mortier
- From the Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
38
|
Fenwick PS, Macedo P, Kilty IC, Barnes PJ, Donnelly LE. Effect of JAK Inhibitors on Release of CXCL9, CXCL10 and CXCL11 from Human Airway Epithelial Cells. PLoS One 2015; 10:e0128757. [PMID: 26090665 PMCID: PMC4474874 DOI: 10.1371/journal.pone.0128757] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 05/01/2015] [Indexed: 12/16/2022] Open
Abstract
Background CD8+ T-cells are located in the small airways of COPD patients and may contribute to pathophysiology. CD8+ cells express the chemokine receptor, CXCR3 that binds CXCL9, CXCL10 and CXCL11, which are elevated in the airways of COPD patients. These chemokines are released from airway epithelial cells via activation of receptor associated Janus kinases (JAK). This study compared the efficacy of two structurally dissimilar pan-JAK inhibitors, PF956980 and PF1367550, and the glucocorticosteroid dexamethasone, in BEAS-2B and human primary airway epithelial cells from COPD patients and control subjects. Methods Cells were stimulated with either IFNγ alone or with TNFα, and release of CXCL9, CXCL10 and CXCL11 measured by ELISA and expression of CXCL9, CXCL10 and CXCL11 by qPCR. Activation of JAK signalling was assessed by STAT1 phosphorylation and DNA binding. Results There were no differences in the levels of release of CXCL9, CXCL10 and CXCL11 from primary airway epithelial cells from any of the subjects or following stimulation with either IFNγ alone or with TNFα. Dexamethasone did not inhibit CXCR3 chemokine release from stimulated BEAS-2B or primary airway epithelial cells. However, both JAK inhibitors suppressed this response with PF1367550 being ~50-65-fold more potent than PF956980. The response of cells from COPD patients did not differ from controls with similar responses regardless of whether inhibitors were added prophylactically or concomitant with stimuli. These effects were mediated by JAK inhibition as both compounds suppressed STAT1 phosphorylation and DNA-binding of STAT1 and gene transcription. Conclusions These data suggest that the novel JAK inhibitor, PF1367550, is more potent than PF956980 and that JAK pathway inhibition in airway epithelium could provide an alternative anti-inflammatory approach for glucocorticosteroid-resistant diseases including COPD.
Collapse
Affiliation(s)
- Peter S Fenwick
- Airway Disease, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Patricia Macedo
- Airway Disease, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Iain C Kilty
- Pfizer Inc, Cambridge, Massachusetts, United States of America
| | - Peter J Barnes
- Airway Disease, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Louise E Donnelly
- Airway Disease, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
39
|
Dipeptidylpeptidase 4 inhibition enhances lymphocyte trafficking, improving both naturally occurring tumor immunity and immunotherapy. Nat Immunol 2015; 16:850-8. [PMID: 26075911 DOI: 10.1038/ni.3201] [Citation(s) in RCA: 233] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 05/17/2015] [Indexed: 12/12/2022]
Abstract
The success of antitumor immune responses depends on the infiltration of solid tumors by effector T cells, a process guided by chemokines. Here we show that in vivo post-translational processing of chemokines by dipeptidylpeptidase 4 (DPP4, also known as CD26) limits lymphocyte migration to sites of inflammation and tumors. Inhibition of DPP4 enzymatic activity enhanced tumor rejection by preserving biologically active CXCL10 and increasing trafficking into the tumor by lymphocytes expressing the counter-receptor CXCR3. Furthermore, DPP4 inhibition improved adjuvant-based immunotherapy, adoptive T cell transfer and checkpoint blockade. These findings provide direct in vivo evidence for control of lymphocyte trafficking via CXCL10 cleavage and support the use of DPP4 inhibitors for stabilizing biologically active forms of chemokines as a strategy to enhance tumor immunotherapy.
Collapse
|
40
|
Yabluchanskiy A, Ma Y, DeLeon-Pennell KY, Altara R, Halade GV, Voorhees AP, Nguyen NT, Jin YF, Winniford MD, Hall ME, Han HC, Lindsey ML. Myocardial Infarction Superimposed on Aging: MMP-9 Deletion Promotes M2 Macrophage Polarization. J Gerontol A Biol Sci Med Sci 2015; 71:475-83. [PMID: 25878031 DOI: 10.1093/gerona/glv034] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
In this study, we examined the combined effect of aging and myocardial infarction on left ventricular remodeling, focusing on matrix metalloproteinase (MMP)-9-dependent mechanisms. We enrolled 55 C57BL/6J wild type (WT) and 85 MMP-9 Null (Null) mice of both sexes at 11-36 months of age and evaluated their response at Day 7 post-myocardial infarction. Plasma MMP-9 levels positively linked to age in WT mice (r = .46, p = .001). MMP-9 deletion improved survival (76% for WT vs 88% for Null, p = .021). Post-myocardial infarction, there was a progressive increase in left ventricular dilation with age in WT but not in Null mice. By inflammatory gene array analysis, WT mice showed linear age-dependent increases in three different proinflammatory genes (C3, CCl4, and CX3CL1; all p < .05), whereas Null mice showed increases in three proinflammatory genes (CCL5, CCL9, and CXCL4; all p < .05) and seven anti-inflammatory genes (CCL1, CCL6, CCR1, IL11, IL1r2, IL8rb, and Mif; all p < .05). Compared with WT, macrophages isolated from Null left ventricle infarct demonstrated enhanced expression of anti-inflammatory M2 markers CD163, MRC1, TGF-β1, and YM1 (all p < .05), without affecting proinflammatory M1 markers. In conclusion, MMP-9 deletion stimulated anti-inflammatory polarization of macrophages to attenuate left ventricle dysfunction in the aging post-myocardial infarction.
Collapse
Affiliation(s)
- Andriy Yabluchanskiy
- Department of Physiology and Biophysics, San Antonio Cardiovascular Proteomics Center, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson
| | - Yonggang Ma
- Department of Physiology and Biophysics, San Antonio Cardiovascular Proteomics Center, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson
| | - Kristine Y DeLeon-Pennell
- Department of Physiology and Biophysics, San Antonio Cardiovascular Proteomics Center, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson
| | - Raffaele Altara
- Department of Physiology and Biophysics, San Antonio Cardiovascular Proteomics Center, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson
| | - Ganesh V Halade
- Department of Physiology and Biophysics, San Antonio Cardiovascular Proteomics Center, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson
| | - Andrew P Voorhees
- Department of Physiology and Biophysics, San Antonio Cardiovascular Proteomics Center, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson. Department of Mechanical Engineering and
| | - Nguyen T Nguyen
- Department of Physiology and Biophysics, San Antonio Cardiovascular Proteomics Center, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson. Department of Electrical and Computer Engineering, University of Texas at San Antonio
| | - Yu-Fang Jin
- Department of Physiology and Biophysics, San Antonio Cardiovascular Proteomics Center, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson. Department of Electrical and Computer Engineering, University of Texas at San Antonio
| | - Michael D Winniford
- Department of Physiology and Biophysics, San Antonio Cardiovascular Proteomics Center, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson. Cardiology Division, University of Mississippi Medical Center, Jackson
| | - Michael E Hall
- Department of Physiology and Biophysics, San Antonio Cardiovascular Proteomics Center, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson. Cardiology Division, University of Mississippi Medical Center, Jackson
| | - Hai-Chao Han
- Department of Physiology and Biophysics, San Antonio Cardiovascular Proteomics Center, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson. Department of Mechanical Engineering and
| | - Merry L Lindsey
- Department of Physiology and Biophysics, San Antonio Cardiovascular Proteomics Center, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson. Research Service, G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS.
| |
Collapse
|
41
|
Gorbachev AV, Fairchild RL. Regulation of chemokine expression in the tumor microenvironment. Crit Rev Immunol 2015; 34:103-20. [PMID: 24940911 DOI: 10.1615/critrevimmunol.2014010062] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Chemokines are chemotactic cytokines critical for homeostatic and inflammation-induced trafficking of leukocytes during immune responses, hematopoesis, wound healing, and tumorigenesis. Despite three decades of intensive study of the chemokine network, the molecular mechanisms regulating chemokine expression during tumor growth are not well understood. In this review, we focus on the role of chemokines in both tumor growth and anti-tumor immune responses and on molecular mechanisms employed by tumor cells to regulate chemokine expression in the tumor microenvironment. Multiple mechanisms used by tumors to regulate chemokine production, including those revealed by very recent studies (such as DNA methylation or post-translational nitrosylation of chemokines) are discussed. Concluding the review, we discuss how understanding of these regulatory mechanisms can be used in cancer therapy to suppress tumor growth and/or to promote immune-mediated eradication of tumors.
Collapse
Affiliation(s)
| | - Robert L Fairchild
- Department of Immunology and Urological Institute, Cleveland Clinic Foundation, Cleveland, OH 44195 and Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| |
Collapse
|
42
|
Song J, Wu C, Korpos E, Zhang X, Agrawal SM, Wang Y, Faber C, Schäfers M, Körner H, Opdenakker G, Hallmann R, Sorokin L. Focal MMP-2 and MMP-9 activity at the blood-brain barrier promotes chemokine-induced leukocyte migration. Cell Rep 2015; 10:1040-54. [PMID: 25704809 DOI: 10.1016/j.celrep.2015.01.037] [Citation(s) in RCA: 158] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Revised: 12/22/2014] [Accepted: 01/14/2015] [Indexed: 01/25/2023] Open
Abstract
Although chemokines are sufficient for chemotaxis of various cells, increasing evidence exists for their fine-tuning by selective proteolytic processing. Using a model of immune cell chemotaxis into the CNS (experimental autoimmune encephalomyelitis [EAE]) that permits precise localization of immigrating leukocytes at the blood-brain barrier, we show that, whereas chemokines are required for leukocyte migration into the CNS, additional MMP-2/9 activities specifically at the border of the CNS parenchyma strongly enhance this transmigration process. Cytokines derived from infiltrating leukocytes regulate MMP-2/9 activity at the parenchymal border, which in turn promotes astrocyte secretion of chemokines and differentially modulates the activity of different chemokines at the CNS border, thereby promoting leukocyte migration out of the cuff. Hence, cytokines, chemokines, and cytokine-induced MMP-2/9 activity specifically at the inflammatory border collectively act to accelerate leukocyte chemotaxis across the parenchymal border.
Collapse
Affiliation(s)
- Jian Song
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, 48149 Muenster, Germany; Cells-in-Motion Cluster of Excellence, University of Muenster, 48149 Muenster, Germany
| | - Chuan Wu
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, 48149 Muenster, Germany
| | - Eva Korpos
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, 48149 Muenster, Germany; Cells-in-Motion Cluster of Excellence, University of Muenster, 48149 Muenster, Germany
| | - Xueli Zhang
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, 48149 Muenster, Germany; Cells-in-Motion Cluster of Excellence, University of Muenster, 48149 Muenster, Germany
| | - Smriti M Agrawal
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, 48149 Muenster, Germany
| | - Ying Wang
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, 48149 Muenster, Germany
| | - Cornelius Faber
- Department of Clinical Radiology, University of Muenster, 48149 Muenster, Germany; Cells-in-Motion Cluster of Excellence, University of Muenster, 48149 Muenster, Germany
| | - Michael Schäfers
- European Institute for Molecular Imaging, University of Muenster, 48149 Muenster, Germany; Cells-in-Motion Cluster of Excellence, University of Muenster, 48149 Muenster, Germany
| | - Heinrich Körner
- Menzies Institute for Medical Research, Hobart, TAS 7000, Australia
| | - Ghislain Opdenakker
- Department of Microbiology and Immunobiology, Rega Institute for Medical Research, University of Leuven, 3000 Leuven, Belgium
| | - Rupert Hallmann
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, 48149 Muenster, Germany; Cells-in-Motion Cluster of Excellence, University of Muenster, 48149 Muenster, Germany
| | - Lydia Sorokin
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, 48149 Muenster, Germany; Cells-in-Motion Cluster of Excellence, University of Muenster, 48149 Muenster, Germany.
| |
Collapse
|
43
|
Salminen A, Åström P, Metso J, Soliymani R, Salo T, Jauhiainen M, Pussinen PJ, Sorsa T. Matrix metalloproteinase 8 degrades apolipoprotein A-I and reduces its cholesterol efflux capacity. FASEB J 2014; 29:1435-45. [PMID: 25550459 DOI: 10.1096/fj.14-262956] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2014] [Accepted: 11/29/2014] [Indexed: 11/11/2022]
Abstract
Various cell types in atherosclerotic lesions express matrix metalloproteinase (MMP)-8. We investigated whether MMP-8 affects the structure and antiatherogenic function of apolipoprotein (apo) A-I, the main protein component of HDL particles. Furthermore, we studied serum lipid profiles and cholesterol efflux capacity in MMP-8-deficient mouse model. Incubation of apoA-I (28 kDa) with activated MMP-8 yielded 22 kDa and 25 kDa apoA-I fragments. Mass spectrometric analyses revealed that apoA-I was cleaved at its carboxyl-terminal part. Treatment of apoA-I and HDL with MMP-8 resulted in significant reduction (up to 84%, P < 0.001) in their ability to facilitate cholesterol efflux from cholesterol-loaded THP-1 macrophages. The cleavage of apoA-I by MMP-8 and the reduction in its cholesterol efflux capacity was inhibited by doxycycline. MMP-8-deficient mice had significantly lower serum triglyceride (TG) levels (P = 0.003) and larger HDL particles compared with wild-type (WT) mice. However, no differences were observed in the apoA-I levels or serum cholesterol efflux capacities between the mouse groups. Proteolytic modification of apoA-I by MMP-8 may impair the first steps of reverse cholesterol transport, leading to increased accumulation of cholesterol in the vessel walls. Eventually, inhibition of MMPs by doxycycline may reduce the risk for atherosclerotic vascular diseases.
Collapse
Affiliation(s)
- Aino Salminen
- *Institute of Dentistry, University of Helsinki, Helsinki, Finland; Department of Oral and Maxillofacial Diseases, Helsinki University Central Hospital, Helsinki, Finland; Department of Diagnostics and Oral Medicine, Institute of Dentistry, and Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland; National Institute for Health and Welfare, Public Health Genomics Unit, Biomedicum, Helsinki, Finland; Meilahti Clinical Proteomics Core Unit, Department of Biochemistry and Developmental Biology, Institute of Biomedicine, Biomedicum Helsinki, University of Helsinki, Finland; and Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Pirjo Åström
- *Institute of Dentistry, University of Helsinki, Helsinki, Finland; Department of Oral and Maxillofacial Diseases, Helsinki University Central Hospital, Helsinki, Finland; Department of Diagnostics and Oral Medicine, Institute of Dentistry, and Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland; National Institute for Health and Welfare, Public Health Genomics Unit, Biomedicum, Helsinki, Finland; Meilahti Clinical Proteomics Core Unit, Department of Biochemistry and Developmental Biology, Institute of Biomedicine, Biomedicum Helsinki, University of Helsinki, Finland; and Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Jari Metso
- *Institute of Dentistry, University of Helsinki, Helsinki, Finland; Department of Oral and Maxillofacial Diseases, Helsinki University Central Hospital, Helsinki, Finland; Department of Diagnostics and Oral Medicine, Institute of Dentistry, and Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland; National Institute for Health and Welfare, Public Health Genomics Unit, Biomedicum, Helsinki, Finland; Meilahti Clinical Proteomics Core Unit, Department of Biochemistry and Developmental Biology, Institute of Biomedicine, Biomedicum Helsinki, University of Helsinki, Finland; and Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Rabah Soliymani
- *Institute of Dentistry, University of Helsinki, Helsinki, Finland; Department of Oral and Maxillofacial Diseases, Helsinki University Central Hospital, Helsinki, Finland; Department of Diagnostics and Oral Medicine, Institute of Dentistry, and Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland; National Institute for Health and Welfare, Public Health Genomics Unit, Biomedicum, Helsinki, Finland; Meilahti Clinical Proteomics Core Unit, Department of Biochemistry and Developmental Biology, Institute of Biomedicine, Biomedicum Helsinki, University of Helsinki, Finland; and Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Tuula Salo
- *Institute of Dentistry, University of Helsinki, Helsinki, Finland; Department of Oral and Maxillofacial Diseases, Helsinki University Central Hospital, Helsinki, Finland; Department of Diagnostics and Oral Medicine, Institute of Dentistry, and Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland; National Institute for Health and Welfare, Public Health Genomics Unit, Biomedicum, Helsinki, Finland; Meilahti Clinical Proteomics Core Unit, Department of Biochemistry and Developmental Biology, Institute of Biomedicine, Biomedicum Helsinki, University of Helsinki, Finland; and Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Matti Jauhiainen
- *Institute of Dentistry, University of Helsinki, Helsinki, Finland; Department of Oral and Maxillofacial Diseases, Helsinki University Central Hospital, Helsinki, Finland; Department of Diagnostics and Oral Medicine, Institute of Dentistry, and Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland; National Institute for Health and Welfare, Public Health Genomics Unit, Biomedicum, Helsinki, Finland; Meilahti Clinical Proteomics Core Unit, Department of Biochemistry and Developmental Biology, Institute of Biomedicine, Biomedicum Helsinki, University of Helsinki, Finland; and Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Pirkko J Pussinen
- *Institute of Dentistry, University of Helsinki, Helsinki, Finland; Department of Oral and Maxillofacial Diseases, Helsinki University Central Hospital, Helsinki, Finland; Department of Diagnostics and Oral Medicine, Institute of Dentistry, and Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland; National Institute for Health and Welfare, Public Health Genomics Unit, Biomedicum, Helsinki, Finland; Meilahti Clinical Proteomics Core Unit, Department of Biochemistry and Developmental Biology, Institute of Biomedicine, Biomedicum Helsinki, University of Helsinki, Finland; and Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Timo Sorsa
- *Institute of Dentistry, University of Helsinki, Helsinki, Finland; Department of Oral and Maxillofacial Diseases, Helsinki University Central Hospital, Helsinki, Finland; Department of Diagnostics and Oral Medicine, Institute of Dentistry, and Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland; National Institute for Health and Welfare, Public Health Genomics Unit, Biomedicum, Helsinki, Finland; Meilahti Clinical Proteomics Core Unit, Department of Biochemistry and Developmental Biology, Institute of Biomedicine, Biomedicum Helsinki, University of Helsinki, Finland; and Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| |
Collapse
|
44
|
Vos R, Verleden SE, Ruttens D, Vandermeulen E, Bellon H, Neyrinck A, Van Raemdonck DE, Yserbyt J, Dupont LJ, Verbeken EK, Moelants E, Mortier A, Proost P, Schols D, Cox B, Verleden GM, Vanaudenaerde BM. Azithromycin and the treatment of lymphocytic airway inflammation after lung transplantation. Am J Transplant 2014; 14:2736-48. [PMID: 25394537 DOI: 10.1111/ajt.12942] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 06/19/2014] [Accepted: 06/22/2014] [Indexed: 01/25/2023]
Abstract
Lymphocytic airway inflammation is a major risk factor for chronic lung allograft dysfunction, for which there is no established treatment. We investigated whether azithromycin could control lymphocytic airway inflammation and improve allograft function. Fifteen lung transplant recipients demonstrating acute allograft dysfunction due to isolated lymphocytic airway inflammation were prospectively treated with azithromycin for at least 6 months (NCT01109160). Spirometry (FVC, FEV1 , FEF25-75 , Tiffeneau index) and FeNO were assessed before and up to 12 months after initiation of azithromycin. Radiologic features, local inflammation assessed on airway biopsy (rejection score, IL-17(+) cells/mm(2) lamina propria) and broncho-alveolar lavage fluid (total and differential cell counts, chemokine and cytokine levels); as well as systemic C-reactive protein levels were compared between baseline and after 3 months of treatment. Airflow improved and FeNO decreased to baseline levels after 1 month of azithromycin and were sustained thereafter. After 3 months of treatment, radiologic abnormalities, submucosal cellular inflammation, lavage protein levels of IL-1β, IL-8/CXCL-8, IP-10/CXCL-10, RANTES/CCL5, MIP1-α/CCL3, MIP-1β/CCL4, Eotaxin, PDGF-BB, total cell count, neutrophils and eosinophils, as well as plasma C-reactive protein levels all significantly decreased compared to baseline (p < 0.05). Administration of azithromycin was associated with suppression of posttransplant lymphocytic airway inflammation and clinical improvement in lung allograft function.
Collapse
Affiliation(s)
- R Vos
- Department of Clinical and Experimental Medicine, Lab of Pneumology, Katholieke Universiteit Leuven and University Hospital Gasthuisberg, Leuven, Belgium; Lung Transplant Unit, Katholieke Universiteit Leuven and University Hospital Gasthuisberg, Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Monin L, Khader SA. Chemokines in tuberculosis: the good, the bad and the ugly. Semin Immunol 2014; 26:552-8. [PMID: 25444549 DOI: 10.1016/j.smim.2014.09.004] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 09/15/2014] [Accepted: 09/21/2014] [Indexed: 10/24/2022]
Abstract
Mycobacterium tuberculosis (Mtb) infects about one-third of the world's population, with a majority of infected individuals exhibiting latent asymptomatic infection, while 5-10% of infected individuals progress to active pulmonary disease. Research in the past two decades has elucidated critical host immune mechanisms that mediate Mtb control. Among these, chemokines have been associated with numerous key processes that lead to Mtb containment, from recruitment of myeloid cells into the lung to activation of adaptive immunity, formation of protective granulomas and vaccine recall responses. However, imbalances in several key chemokine mediators can alter the delicate balance of cytokines and cellular responses that promote mycobacterial containment, instead precipitating terminal tissue destruction and spread of Mtb infection. In this review, we will describe recent insights in the involvement of chemokines in host responses to Mtb infection and Mtb containment (the good), chemokines contributing to inflammation during TB (the bad), and the role of chemokines in driving cavitation and lung pathology (the ugly).
Collapse
Affiliation(s)
- Leticia Monin
- Department of Molecular Microbiology, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA; Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of UPMC, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | - Shabaana A Khader
- Department of Molecular Microbiology, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA.
| |
Collapse
|
46
|
Quillard T, Araújo HA, Franck G, Tesmenitsky Y, Libby P. Matrix metalloproteinase-13 predominates over matrix metalloproteinase-8 as the functional interstitial collagenase in mouse atheromata. Arterioscler Thromb Vasc Biol 2014; 34:1179-86. [PMID: 24723558 DOI: 10.1161/atvbaha.114.303326] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Substantial evidence implicates interstitial collagenases of the matrix metalloproteinase (MMP) family in plaque rupture and fatal thrombosis. Understanding the compensatory mechanisms that may influence the expression of these enzymes and their functions, therefore, has important clinical implications. This study assessed in mice the relative effect of the 2 principal mouse collagenases on collagen content and other plaque characteristics. APPROACH AND RESULTS Apolipoprotein E-deficient (apoE(-/-)) mice, MMP-13(-/-) apoE(-/-), MMP-8(-/-) apoE(-/-) double knockout mice, and MMP-13(-/-) MMP-8(-/-) apoE(-/-) triple knockout mice consumed a high-cholesterol diet for 10 and 24 weeks. Both double knockout and triple knockout mice showed comparable atherosclerotic lesion formation compared with apoE(-/-) controls. Analysis of aortic root sections indicated that lesions of MMP-8/MMP-13-deficient and MMP-13-deficient mice accumulate more fibrillar collagen than apoE(-/-) controls and MMP-8(-/-) apoE(-/-) double knockout. We further tested the relative effect of MMPs on plaque collagenolysis using in situ zymography. MMP-13 deletion alone abrogated collagenolytic activity in lesions, indicating a predominant role for MMP-13 in this process. MMP-13 and MMP-13/MMP-8 deficiency did not alter macrophage content but associated with reduced accumulation of smooth muscle cells. CONCLUSIONS These results show that among MMP interstitial collagenases in mice, MMP-13 prevails over MMP-8 in collagen degradation in atheromata. These findings provide a rationale for the identification and selective targeting a predominant collagenase for modulating key aspects of plaque structure considered critical in clinical complications, although they do not translate directly to human lesions, which also contain MMP-1.
Collapse
Affiliation(s)
- Thibaut Quillard
- From the Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Haniel Alves Araújo
- From the Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Gregory Franck
- From the Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Yevgenia Tesmenitsky
- From the Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Peter Libby
- From the Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.
| |
Collapse
|
47
|
Rainczuk A, Rao JR, Gathercole JL, Fairweather NJ, Chu S, Masadah R, Jobling TW, Deb-Choudhury S, Dyer J, Stephens AN. Evidence for the antagonistic form of CXC-motif chemokine CXCL10 in serous epithelial ovarian tumours. Int J Cancer 2013; 134:530-41. [DOI: 10.1002/ijc.28393] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2013] [Accepted: 07/03/2013] [Indexed: 11/05/2022]
Affiliation(s)
- Adam Rainczuk
- Ovarian Cancer Biomarker Laboratory, Prince Henry's Institute of Medical Research; Clayton VIC Australia
| | - Jyothsna R. Rao
- Ovarian Cancer Biomarker Laboratory, Prince Henry's Institute of Medical Research; Clayton VIC Australia
| | - Jessica L. Gathercole
- Ovarian Cancer Biomarker Laboratory, Prince Henry's Institute of Medical Research; Clayton VIC Australia
| | - Nicole J. Fairweather
- Ovarian Cancer Biomarker Laboratory, Prince Henry's Institute of Medical Research; Clayton VIC Australia
| | - Simon Chu
- Ovarian Cancer Biomarker Laboratory, Prince Henry's Institute of Medical Research; Clayton VIC Australia
| | - Rina Masadah
- Department of Anatomical Pathology; Hasanuddin University; Makassar Indonesia
| | - Thomas W. Jobling
- Department of Obstetrics and Gynaecology; Monash Medical Centre; Clayton VIC Australia
| | | | - Jolon Dyer
- AgResearch Ltd, Lincoln Research Centre; Christchurch New Zealand
| | - Andrew N. Stephens
- Ovarian Cancer Biomarker Laboratory, Prince Henry's Institute of Medical Research; Clayton VIC Australia
| |
Collapse
|
48
|
Tortelli F, Pisano M, Briquez PS, Martino MM, Hubbell JA. Fibronectin binding modulates CXCL11 activity and facilitates wound healing. PLoS One 2013; 8:e79610. [PMID: 24205388 PMCID: PMC3808276 DOI: 10.1371/journal.pone.0079610] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 09/24/2013] [Indexed: 11/18/2022] Open
Abstract
Engineered biomatrices offer the potential to recapitulate the regenerative microenvironment, with important implications in tissue repair. In this context, investigation of the molecular interactions occurring between growth factors, cytokines and extracellular matrix (ECM) has gained increasing interest. Here, we sought to investigate the possible interactions between the ECM proteins fibronectin (FN) and fibrinogen (Fg) with the CXCR3 ligands CXCL9, CXCL10 and CXCL11, which are expressed during wound healing. New binding interactions were observed and characterized. Heparin-binding domains within Fg (residues 15-66 of the β chain, Fg β15-66) and FN (FNI1-5, but not FNIII12-14) were involved in binding to CXCL10 and CXCL11 but not CXCL9. To investigate a possible influence of FN and Fg interactions with CXCL11 in mediating its role during re-epithelialization, we investigated human keratinocyte migration in vitro and wound healing in vivo in diabetic db/db mice. A synergistic effect on CXCL11-induced keratinocyte migration was observed when cells were treated with CXCL11 in combination with FN in a transmigration assay. Moreover, wound healing was enhanced in full thickness excisional wounds treated with fibrin matrices functionalized with FN and containing CXCL11. These findings highlight the importance of the interactions occurring between cytokines and ECM and point to design concepts to develop functional matrices for regenerative medicine.
Collapse
Affiliation(s)
- Federico Tortelli
- Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Marco Pisano
- Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Priscilla S. Briquez
- Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Mikaël M. Martino
- Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Jeffrey A. Hubbell
- Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Institute of Chemical Sciences and Engineering, School of Basic Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- * E-mail:
| |
Collapse
|
49
|
Gómez-Piña V, Martínez E, Fernández-Ruíz I, del Fresno C, Soares-Schanoski A, Jurado T, Siliceo M, Toledano V, Fernández-Palomares R, García-Rio F, Arnalich F, Biswas SK, López-Collazo E. Role of MMPs in orchestrating inflammatory response in human monocytes via a TREM-1-PI3K-NF-κB pathway. J Leukoc Biol 2012; 91:933-945. [DOI: 10.1189/jlb.0711340] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Abstract
ABSTRACTThe MMPs constitute a family of endopeptidases that can cleavage extracellular proteins. They are involved in a number of events; some of these include inflammatory processes. One of its targets is the TREM-1, which has emerged as an important modulator of innate immune responses in mammals. This transmembrane glycoprotein possesses an Ig-like ectodomain readily shed by MMPs to generate sTREM-1. Whereas membrane-anchored TREM-1 amplifies inflammatory responses, sTREM-1 exhibits anti-inflammatory properties. Here we show that sustained cell surface expression of TREM-1 in human monocytes, through metalloproteinase inhibition, counteracts the well-characterized down-regulation of several proinflammatory cytokines during the ET time-frame, also known as M2 or alternative activation. In addition to the cytokines profile, other features of the ET phenotype were underdeveloped when TREM-1 was stabilized at the cell surface. These events were mediated by the signal transducers PI3Ks and Syk. We also show that sTREM-1 counteracts the proinflammatory response obtained by membrane TREM-1 stabilization but failed to induce ET on naïve human monocytes. As the sustained TREM-1 expression at the cell surface suffices to block the progress of a refractory state in human monocytes, our data indicate that TREM-1 and MMPs orchestrate an “adaptive” form of innate immunity by modulating the human monocytes response to endotoxin.
Collapse
Affiliation(s)
| | | | | | | | | | - Teresa Jurado
- Laboratory of Tumor Immunology, IdiPAZ , Madrid, Spain
| | - María Siliceo
- Laboratory of Tumor Immunology, IdiPAZ , Madrid, Spain
| | | | | | | | | | - Subhra K Biswas
- Singapore Immunology Network, Biomedical Sciences Institutes , Singapore
| | | |
Collapse
|
50
|
Casrouge A, Bisiaux A, Stephen L, Schmolz M, Mapes J, Pfister C, Pol S, Mallet V, Albert ML. Discrimination of agonist and antagonist forms of CXCL10 in biological samples. Clin Exp Immunol 2012; 167:137-48. [PMID: 22132893 DOI: 10.1111/j.1365-2249.2011.04488.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The ready access to commercially available multiplex assays and the importance of inflammation in disease pathogenesis has resulted in an abundance of studies aimed at identifying surrogate biomarkers for different clinically important questions. Establishing a link between a biomarker and disease pathogenesis, however, is quite complex, and in some instances this complexity is compounded by post-translational modifications and the use of immunoassays that do not always discriminate between the different forms of the same protein. Herein, we provide a detailed description of an assay system that has been established to discriminate the agonist form of CXCL10 from the NH(2) -terminal truncated form of the molecule generated by dipeptidylpeptidase IV (DPP4) cleavage. We demonstrate the utility of this assay system for monitoring agonist and antagonist forms of CXCL10 in culture supernatant, patient plasma and urine samples. Given the important role of CXCL10 in chronic inflammatory diseases and its suggested role as a predictive marker in managing patients with chronic hepatitis C, asthma, atopic dermatitis, transplantation, tuberculosis, kidney injury, cancer and other diseases, we believe that our method will be of general interest to the research and medical community.
Collapse
Affiliation(s)
- A Casrouge
- Laboratory of Dendritic Cell Biology, Department of Immunology, Institut Pasteur, Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|