1
|
Hatipoglu OF, Nishinaka T, Yaykasli KO, Mori S, Watanabe M, Toyomura T, Nishibori M, Hirohata S, Wake H, Takahashi H. Histidine-rich glycoprotein inhibits TNF-α-induced tube formation in human vascular endothelial cells. Front Pharmacol 2025; 16:1561628. [PMID: 40191430 PMCID: PMC11969118 DOI: 10.3389/fphar.2025.1561628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 03/06/2025] [Indexed: 04/09/2025] Open
Abstract
Introduction Tumor necrosis factor-α (TNF-α)-induced angiogenesis plays a critical role in tumor progression and metastasis, making it an important therapeutic target in cancer treatment. Suppressing angiogenesis can effectively limit tumor growth and metastasis. However, despite advancements in understanding angiogenic pathways, effective strategies to inhibit TNF-α-mediated angiogenesis remain limited. Methods This study investigates the antiangiogenic effects of histidine-rich glycoprotein (HRG), a multifunctional plasma protein with potent antiangiogenic properties, on TNF-α-stimulated human endothelial cells (EA.hy926). Tube formation assays were performed to assess angiogenesis, and gene/protein expression analyses were conducted to evaluate HRG's effects on integrins αV and β8. The role of nuclear factor erythroid 2-related factor 2 (NRF2) in HRG-mediated antiangiogenic activity was also examined through nuclear translocation assays and NRF2 activation studies. Results At physiological concentrations, HRG effectively suppressed TNF-α-induced tube formation in vitro and downregulated TNF-α-induced expression of integrins αV and β8 at both the mRNA and protein levels. HRG treatment promoted NRF2 nuclear translocation in a time-dependent manner. Furthermore, activation of NRF2 significantly reduced TNF-α-induced tube formation and integrin expression, suggesting that NRF2 plays a key role in HRG-mediated antiangiogenic effects. Discussion and Conclusion Our findings indicate that HRG suppresses TNF-α-induced angiogenesis by promoting NRF2 nuclear translocation and transcriptional activation, which in turn inhibits integrin αV and β8 expression. Given the essential role of angiogenesis in tumor progression, HRG's ability to regulate this process presents a promising therapeutic strategy for cancer treatment.
Collapse
Affiliation(s)
- Omer Faruk Hatipoglu
- Department of Pharmacology, Kindai University Faculty of Medicine, Osakasayama, Japan
| | - Takashi Nishinaka
- Department of Pharmacology, Kindai University Faculty of Medicine, Osakasayama, Japan
| | - Kursat Oguz Yaykasli
- Department of Internal Medicine 3—Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Shuji Mori
- Department of Pharmacology, School of Pharmacy, Shujitsu University, Okayama, Japan
| | - Masahiro Watanabe
- Department of Pharmacology, School of Pharmacy, Shujitsu University, Okayama, Japan
| | - Takao Toyomura
- Department of Pharmacology, School of Pharmacy, Shujitsu University, Okayama, Japan
| | - Masahiro Nishibori
- Department of Translational Research and Dug Development, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Satoshi Hirohata
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, Okayama, Japan
| | - Hidenori Wake
- Department of Pharmacology, Kindai University Faculty of Medicine, Osakasayama, Japan
| | - Hideo Takahashi
- Department of Pharmacology, Kindai University Faculty of Medicine, Osakasayama, Japan
| |
Collapse
|
2
|
Salemi M, Schillaci FA, Lanza G, Marchese G, Salluzzo MG, Cordella A, Caniglia S, Bruccheri MG, Truda A, Greco D, Ferri R, Romano C. Transcriptome Study in Sicilian Patients with Autism Spectrum Disorder. Biomedicines 2024; 12:1402. [PMID: 39061976 PMCID: PMC11274004 DOI: 10.3390/biomedicines12071402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/12/2024] [Accepted: 06/14/2024] [Indexed: 07/28/2024] Open
Abstract
ASD is a complex condition primarily rooted in genetics, although influenced by environmental, prenatal, and perinatal risk factors, ultimately leading to genetic and epigenetic alterations. These mechanisms may manifest as inflammatory, oxidative stress, hypoxic, or ischemic damage. To elucidate potential variances in gene expression in ASD, a transcriptome analysis of peripheral blood mononuclear cells was conducted via RNA-seq on 12 ASD patients and 13 healthy controls, all of Sicilian ancestry to minimize environmental confounds. A total of 733 different statistically significant genes were identified between the two cohorts. Gene Set Enrichment Analysis (GSEA) and Gene Ontology (GO) terms were employed to explore the pathways influenced by differentially expressed mRNAs. GSEA revealed GO pathways strongly associated with ASD, namely the GO Biological Process term "Response to Oxygen-Containing Compound". Additionally, the GO Cellular Component pathway "Mitochondrion" stood out among other pathways, with differentially expressed genes predominantly affiliated with this specific pathway, implicating the involvement of different mitochondrial functions in ASD. Among the differentially expressed genes, FPR2 was particularly highlighted, belonging to three GO pathways. FPR2 can modulate pro-inflammatory responses, with its intracellular cascades triggering the activation of several kinases, thus suggesting its potential utility as a biomarker of pro-inflammatory processes in ASD.
Collapse
Affiliation(s)
- Michele Salemi
- Oasi Research Institute—IRCCS, 94018 Troina, Italy; (F.A.S.); (G.L.); (M.G.S.); (S.C.); (M.G.B.); (D.G.); (R.F.); (C.R.)
| | - Francesca A. Schillaci
- Oasi Research Institute—IRCCS, 94018 Troina, Italy; (F.A.S.); (G.L.); (M.G.S.); (S.C.); (M.G.B.); (D.G.); (R.F.); (C.R.)
| | - Giuseppe Lanza
- Oasi Research Institute—IRCCS, 94018 Troina, Italy; (F.A.S.); (G.L.); (M.G.S.); (S.C.); (M.G.B.); (D.G.); (R.F.); (C.R.)
- Department of Surgery and Medical—Surgical Specialties, University of Catania, 95124 Catania, Italy
| | - Giovanna Marchese
- Genomix4Life S.r.l., 84081 Baronissi, Italy; (G.M.); (A.C.); (A.T.)
- Genome Research Center for Health—CRGS, 84081 Baronissi, Italy
| | - Maria Grazia Salluzzo
- Oasi Research Institute—IRCCS, 94018 Troina, Italy; (F.A.S.); (G.L.); (M.G.S.); (S.C.); (M.G.B.); (D.G.); (R.F.); (C.R.)
| | - Angela Cordella
- Genomix4Life S.r.l., 84081 Baronissi, Italy; (G.M.); (A.C.); (A.T.)
- Genome Research Center for Health—CRGS, 84081 Baronissi, Italy
| | - Salvatore Caniglia
- Oasi Research Institute—IRCCS, 94018 Troina, Italy; (F.A.S.); (G.L.); (M.G.S.); (S.C.); (M.G.B.); (D.G.); (R.F.); (C.R.)
| | - Maria Grazia Bruccheri
- Oasi Research Institute—IRCCS, 94018 Troina, Italy; (F.A.S.); (G.L.); (M.G.S.); (S.C.); (M.G.B.); (D.G.); (R.F.); (C.R.)
| | - Anna Truda
- Genomix4Life S.r.l., 84081 Baronissi, Italy; (G.M.); (A.C.); (A.T.)
- Genome Research Center for Health—CRGS, 84081 Baronissi, Italy
| | - Donatella Greco
- Oasi Research Institute—IRCCS, 94018 Troina, Italy; (F.A.S.); (G.L.); (M.G.S.); (S.C.); (M.G.B.); (D.G.); (R.F.); (C.R.)
| | - Raffaele Ferri
- Oasi Research Institute—IRCCS, 94018 Troina, Italy; (F.A.S.); (G.L.); (M.G.S.); (S.C.); (M.G.B.); (D.G.); (R.F.); (C.R.)
| | - Corrado Romano
- Oasi Research Institute—IRCCS, 94018 Troina, Italy; (F.A.S.); (G.L.); (M.G.S.); (S.C.); (M.G.B.); (D.G.); (R.F.); (C.R.)
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95124 Catania, Italy
| |
Collapse
|
3
|
Soontara C, Uchuwittayakul A, Kayansamruaj P, Amparyup P, Wongpanya R, Srisapoome P. Adjuvant Effects of a CC Chemokine for Enhancing the Efficacy of an Inactivated Streptococcus agalactiae Vaccine in Nile Tilapia ( Oreochromis niloticus). Vaccines (Basel) 2024; 12:641. [PMID: 38932370 PMCID: PMC11209360 DOI: 10.3390/vaccines12060641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
In this study, the ability of a CC chemokine (On-CC1) adjuvant to enhance the efficacy of a formalin-killed Streptococcus agalactiae vaccine (WC) in inducing immune responses against S. agalactiae in Nile tilapia was investigated through immune-related gene expression analysis, enzyme-linked immunosorbent assay (ELISA), transcriptome sequencing, and challenge tests. Significantly higher S. agalactiae-specific IgM levels were detected in fish in the WC+CC group than in the WC alone or control groups at 8 days postvaccination (dpv). The WC vaccine group exhibited increased specific IgM levels at 15 dpv, comparable to those of the WC+CC group, with sustained higher levels observed in the latter group at 29 dpv and after challenge with S. agalactiae for 14 days. Immune-related gene expression analysis revealed upregulation of all target genes in the control group compared to those in the vaccinated groups, with notable differences between the WC and WC+CC groups at various time intervals. Additionally, transcriptome analysis revealed differential gene expression profiles between the vaccinated (24 and 96 hpv) and control groups, with notable upregulation of immune-related genes in the vaccinated fish. Differential gene expression (DGE) analysis revealed significant upregulation of immunoglobulin and other immune-related genes in the control group compared to those in the vaccinated groups (24 and 96 hpv), with distinct patterns observed between the WC and WC+CC vaccine groups. Finally, challenge with a virulent strain of S. agalactiae resulted in significantly higher survival rates for fish in the WC and WC+CC groups compared to fish in the control group, with a notable increase in survival observed in fish in the WC+CC group.
Collapse
Affiliation(s)
- Chayanit Soontara
- Laboratory of Aquatic Animal Health Management, Department of Aquaculture, Faculty of Fisheries, Kasetsart University, 50 Paholayothin Rd., Ladyao, Chatuchak, Bangkok 10900, Thailand; (C.S.); (A.U.); (P.K.)
- Center of Excellence in Aquatic Animal Health Management, Faculty of Fisheries, Kasetsart University, 50 Paholayothin Rd., Ladyao, Chatuchak, Bangkok 10900, Thailand
| | - Anurak Uchuwittayakul
- Laboratory of Aquatic Animal Health Management, Department of Aquaculture, Faculty of Fisheries, Kasetsart University, 50 Paholayothin Rd., Ladyao, Chatuchak, Bangkok 10900, Thailand; (C.S.); (A.U.); (P.K.)
- Center of Excellence in Aquatic Animal Health Management, Faculty of Fisheries, Kasetsart University, 50 Paholayothin Rd., Ladyao, Chatuchak, Bangkok 10900, Thailand
| | - Pattanapon Kayansamruaj
- Laboratory of Aquatic Animal Health Management, Department of Aquaculture, Faculty of Fisheries, Kasetsart University, 50 Paholayothin Rd., Ladyao, Chatuchak, Bangkok 10900, Thailand; (C.S.); (A.U.); (P.K.)
- Center of Excellence in Aquatic Animal Health Management, Faculty of Fisheries, Kasetsart University, 50 Paholayothin Rd., Ladyao, Chatuchak, Bangkok 10900, Thailand
| | - Piti Amparyup
- Marine Biotechnology Research Team, Integrative Aquaculture Biotechnology Research Group, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), 113 Paholyothin Road, Klong 1, Khlong Luang 12120, Thailand;
| | - Ratree Wongpanya
- Department of Biochemistry, Faculty of Science, Kasetsart University, 50 Ngamwongwan Road, Bangkok 10900, Thailand;
| | - Prapansak Srisapoome
- Laboratory of Aquatic Animal Health Management, Department of Aquaculture, Faculty of Fisheries, Kasetsart University, 50 Paholayothin Rd., Ladyao, Chatuchak, Bangkok 10900, Thailand; (C.S.); (A.U.); (P.K.)
- Center of Excellence in Aquatic Animal Health Management, Faculty of Fisheries, Kasetsart University, 50 Paholayothin Rd., Ladyao, Chatuchak, Bangkok 10900, Thailand
| |
Collapse
|
4
|
Boles J, Uriarte Huarte O, Tansey MG. Peripheral endotoxin exposure in mice activates crosstalk between phagocytes in the brain and periphery. RESEARCH SQUARE 2024:rs.3.rs-4478250. [PMID: 38883776 PMCID: PMC11177977 DOI: 10.21203/rs.3.rs-4478250/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Background Inflammation is a central process of many neurological diseases, and a growing number of studies suggest that non-brain-resident immune cells may contribute to this neuroinflammation. However, the unique contributions of specific immune cell subsets to neuroinflammation are presently unknown, and it is unclear how communication between brain-resident and non-resident immune cells underlies peripheral immune cell involvement in neuroinflammation. Methods In this study, we employed the well-established model of lipopolysaccharide (LPS)-induced neuroinflammation and captured brain-resident and non-resident immune cells from the brain and its vasculature by magnetically enriching cell suspensions from the non-perfused brain for CD45 + cells. Then, we identified immune subtype-specific neuroinflammatory processes using single-cell genomics and predicted the crosstalk between immune cell subtypes by analyzing the simultaneous expression of ligands and receptors. Results We observed a greater abundance of peripheral phagocytes associated with the brain in this model of neuroinflammation, and report that these professional phagocytes activated similar transcriptional profiles to microglia during LPS-induced neuroinflammation. And, we observed that the probable crosstalk between microglia and peripheral phagocytes was activated in this model while homotypic microglial communication was likely to be decreased. Conclusions Our novel findings reveal that microglia signaling to non-brain-resident peripheral phagocytes is preferentially triggered by peripheral inflammation, which is associated with brain infiltration of peripheral cells. Overall, our study supports the involvement of peripheral immune cells in neuroinflammation and suggests several possible molecular signaling pathways between microglia and peripheral cells that may facilitate central-peripheral crosstalk during inflammation. Examining these molecular mediators in human disease and other rodent models may reveal novel targets that modify brain health, especially in comorbidities characterized by peripheral inflammation.
Collapse
|
5
|
Suárez Vázquez TA, López López N, Salinas Carmona MC. MASTer cell: chief immune modulator and inductor of antimicrobial immune response. Front Immunol 2024; 15:1360296. [PMID: 38638437 PMCID: PMC11024470 DOI: 10.3389/fimmu.2024.1360296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/26/2024] [Indexed: 04/20/2024] Open
Abstract
Mast cells have long been recognized for their involvement in allergic pathology through the immunoglobulin E (IgE)-mediated degranulation mechanism. However, there is growing evidence of other "non-canonical" degranulation mechanisms activated by certain pathogen recognition receptors. Mast cells release several mediators, including histamine, cytokines, chemokines, prostaglandins, and leukotrienes, to initiate and enhance inflammation. The chemical nature of activating stimuli influences receptors, triggering mechanisms for the secretion of formed and new synthesized mediators. Mast cells have more than 30 known surface receptors that activate different pathways for direct and indirect activation by microbes. Different bacterial strains stimulate mast cells through various ligands, initiating the innate immune response, which aids in clearing the bacterial burden. Mast cell interactions with adaptative immune cells also play a crucial role in infections. Recent publications revealed another "non-canonical" degranulation mechanism present in tryptase and chymase mast cells in humans and connective tissue mast cells in mice, occurring through the activation of the Mas-related G protein-coupled receptor (MRGPRX2/b2). This receptor represents a new therapeutic target alongside antibiotic therapy. There is an urgent need to reconsider and redefine the biological role of these MASTer cells of innate immunity, extending beyond their involvement in allergic pathology.
Collapse
Affiliation(s)
| | | | - Mario César Salinas Carmona
- Department of Immunology, School of Medicine and Dr. Jose Eleuterio Gonzalez University Hospital, Universidad Autónoma de Nuevo León, Monterrey, Mexico
| |
Collapse
|
6
|
Tansey M, Boles J, Uriarte Huarte O. Microfluidics-free single-cell genomics reveals complex central-peripheral immune crosstalk in the mouse brain during peripheral inflammation. RESEARCH SQUARE 2023:rs.3.rs-3428910. [PMID: 37886510 PMCID: PMC10602178 DOI: 10.21203/rs.3.rs-3428910/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Inflammation is a realized detriment to brain health in a growing number of neurological diseases, but querying neuroinflammation in its cellular complexity remains a challenge. This manuscript aims to provide a reliable and accessible strategy for examining the brain's immune system. We compare the efficacy of cell isolation methods in producing ample and pure immune samples from mouse brains. Then, with the high-input single-cell genomics platform PIPseq, we generate a rich neuroimmune dataset containing microglia and many peripheral immune populations. To demonstrate this strategy's utility, we interrogate the well-established model of LPS-induced neuroinflammation with single-cell resolution. We demonstrate the activation of crosstalk between microglia and peripheral phagocytes and highlight the unique contributions of microglia and peripheral immune cells to neuroinflammation. Our approach enables the high-depth evaluation of inflammation in longstanding rodent models of neurological disease to reveal novel insight into the contributions of the immune system to brain health.
Collapse
|
7
|
McMinn PH, Ahmed A, Huttenlocher A, Beebe DJ, Kerr SC. The lymphatic endothelium-derived follistatin: activin A axis regulates neutrophil motility in response to Pseudomonas aeruginosa. Integr Biol (Camb) 2023; 15:zyad003. [PMID: 36781971 PMCID: PMC10101905 DOI: 10.1093/intbio/zyad003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 12/02/2022] [Accepted: 01/23/2023] [Indexed: 02/15/2023]
Abstract
The lymphatic system plays an active role during infection, however the role of lymphatic-neutrophil interactions in host-defense responses is not well understood. During infection with pathogens such as Pseudomonas aeruginosa, Staphylococcus aureus and Yersinia pestis, neutrophils traffic from sites of infection through the lymphatic vasculature, to draining lymph nodes to interact with resident lymphocytes. This process is poorly understood, in part, due to the lack of in vitro models of the lymphatic system. Here we use a 3D microscale lymphatic vessel model to examine neutrophil-lymphatic cell interactions during host defense responses to pathogens. In previous work, we have shown that follistatin is secreted at high concentrations by lymphatic endothelial cells during inflammation. Follistatin inhibits activin A, a member of the TGF-β superfamily, and, together, these molecules form a signaling pathway that plays a role in regulating both innate and adaptive immune responses. Although follistatin and activin A are constitutively produced in the pituitary, gonads and skin, their major source in the serum and their effects on neutrophils are poorly understood. Here we report a microfluidic model that includes both blood and lymphatic endothelial vessels, and neutrophils to investigate neutrophil-lymphatic trafficking during infection with P. aeruginosa. We found that lymphatic endothelial cells produce secreted factors that increase neutrophil migration toward P. aeruginosa, and are a significant source of both follistatin and activin A during Pseudomonas infection. We determined that follistatin produced by lymphatic endothelial cells inhibits activin A, resulting in increased neutrophil migration. These data suggest that the follistatin:activin A ratio influences neutrophil trafficking during infection with higher ratios increasing neutrophil migration.
Collapse
Affiliation(s)
- Patrick H McMinn
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Adeel Ahmed
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Anna Huttenlocher
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI, USA
| | - David J Beebe
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Sheena C Kerr
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
8
|
Vugler A, O’Connell J, Nguyen MA, Weitz D, Leeuw T, Hickford E, Verbitsky A, Ying X, Rehberg M, Carrington B, Merriman M, Moss A, Nicholas JM, Stanley P, Wright S, Bourne T, Foricher Y, Zhu Z, Brookings D, Horsley H, Heer J, Schio L, Herrmann M, Rao S, Kohlmann M, Florian P. An orally available small molecule that targets soluble TNF to deliver anti-TNF biologic-like efficacy in rheumatoid arthritis. Front Pharmacol 2022; 13:1037983. [PMID: 36467083 PMCID: PMC9709720 DOI: 10.3389/fphar.2022.1037983] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/21/2022] [Indexed: 07/30/2023] Open
Abstract
Tumor necrosis factor (TNF) is a pleiotropic cytokine belonging to a family of trimeric proteins with both proinflammatory and immunoregulatory functions. TNF is a key mediator in autoimmune diseases and during the last couple of decades several biologic drugs have delivered new therapeutic options for patients suffering from chronic autoimmune diseases such as rheumatoid arthritis and chronic inflammatory bowel disease. Attempts to design small molecule therapies directed to this cytokine have not led to approved products yet. Here we report the discovery and development of a potent small molecule inhibitor of TNF that was recently moved into phase 1 clinical trials. The molecule, SAR441566, stabilizes an asymmetrical form of the soluble TNF trimer, compromises downstream signaling and inhibits the functions of TNF in vitro and in vivo. With SAR441566 being studied in healthy volunteers we hope to deliver a more convenient orally bioavailable and effective treatment option for patients suffering with chronic autoimmune diseases compared to established biologic drugs targeting TNF.
Collapse
Affiliation(s)
- Alexander Vugler
- Immunology Therapeutic Area, PV Early Solutions, UCB Pharma, Slough, United Kingdom
| | - James O’Connell
- Discovery Sciences, PV Early Solutions, UCB Pharma, Slough, United Kingdom
| | - Mai Anh Nguyen
- Sanofi R&D, TMED Pharmacokinetics Dynamics and Metabolism, Frankfurt am Main, Germany
| | - Dietmar Weitz
- Sanofi R&D, Drug Metabolism and Pharmacokinetics, Frankfurt am Main, Germany
| | - Thomas Leeuw
- Sanofi R&D, Type 1/17 Immunology, Immunology & Inflammation Research TA, Frankfurt, Germany
| | - Elizabeth Hickford
- Development Science, PV Early Solutions, UCB Pharma, Slough, United Kingdom
| | | | - Xiaoyou Ying
- Sanofi R&D, Translation In vivo Models, Cambridge, MA, United States
| | - Markus Rehberg
- Sanofi R&D, Translational Disease Modelling, Frankfurt am Main, Germany
| | - Bruce Carrington
- Discovery Sciences, PV Early Solutions, UCB Pharma, Slough, United Kingdom
| | - Mark Merriman
- Immunology Therapeutic Area, PV Early Solutions, UCB Pharma, Slough, United Kingdom
| | - Andrew Moss
- Translational Medicine Immunology, PV Early Solutions, UCB Pharma, Slough, United Kingdom
| | - Jean-Marie Nicholas
- Development Science, Drug Metabolism and Pharmacokinetics, UCB Pharma, Braine-I’Alleud, Belgium
| | - Phil Stanley
- Immunology Therapeutic Area, PV Early Solutions, UCB Pharma, Slough, United Kingdom
| | - Sara Wright
- Early PV Missions, PV Early Solutions, UCB Pharma, Slough, United Kingdom
| | - Tim Bourne
- Milvuswood Consultancy, Penn, United Kingdom
| | - Yann Foricher
- Sanofi R&D, Integrated Drug Discovery, Vitry-sur-Seine, France
| | - Zhaoning Zhu
- Global Chemistry, Discovery Sciences, PV Early Solutions, UCB Pharma, Slough, United Kingdom
| | - Daniel Brookings
- Global Chemistry, Discovery Sciences, PV Early Solutions, UCB Pharma, Slough, United Kingdom
| | - Helen Horsley
- Global Chemistry, Discovery Sciences, PV Early Solutions, UCB Pharma, Slough, United Kingdom
| | - Jag Heer
- Global Chemistry, Discovery Sciences, PV Early Solutions, UCB Pharma, Slough, United Kingdom
| | - Laurent Schio
- Sanofi R&D, Integrated Drug Discovery, Vitry-sur-Seine, France
| | - Matthias Herrmann
- Sanofi R&D, Type 1/17 Immunology, Immunology & Inflammation Research TA, Frankfurt, Germany
| | - Srinivas Rao
- Sanofi R&D, Translation In vivo Models, Cambridge, MA, United States
| | - Markus Kohlmann
- Sanofi R&D, Early Clinical Development, Therapeutic Area Immunology and Inflammation, Frankfurt am Main, Germany
| | - Peter Florian
- Sanofi R&D, Type 1/17 Immunology, Immunology & Inflammation Research TA, Frankfurt, Germany
| |
Collapse
|
9
|
Dib K, El Banna A, Radulescu C, Lopez Campos G, Sheehan G, Kavanagh K. Histamine Produced by Gram-Negative Bacteria Impairs Neutrophil's Antimicrobial Response by Engaging the Histamine 2 Receptor. J Innate Immun 2022; 15:153-173. [PMID: 35858582 PMCID: PMC10643892 DOI: 10.1159/000525536] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 06/12/2022] [Indexed: 11/19/2022] Open
Abstract
We found that histamine (10-9 M) did not have any effect on the in vitro capture of Escherichia coli by neutrophils but accelerated its intracellular killing. In contrast, histamine (10-6 M) delayed the capture of Escherichia coli by neutrophils and reduced the amounts of pHrodo zymosan particles inside acidic mature phagosomes. Histamine acted through the H4R and the H2R, which are coupled to the Src family tyrosine kinases or the cAMP/protein kinase A pathway, respectively. The protein kinase A inhibitor H-89 abrogated the delay in bacterial capture induced by histamine (10-6 M) and the Src family tyrosine kinase inhibitor PP2 blocked histamine (10-9 M) induced acceleration of bacterial intracellular killing and tyrosine phosphorylation of proteins. To investigate the role of histamine in pathogenicity, we designed an Acinetobacter baumannii strain deficient in histamine production (hdc::TOPO). Galleria mellonella larvae inoculated with the wild-type Acinetobacter baumannii ATCC 17978 strain (1.1 × 105 CFU) died rapidly (100% death within 40 h) but not when inoculated with the Acinetobacter baumannii hdc::TOPO mutant (10% mortality). The concentration of histamine rose in the larval haemolymph upon inoculation of the wild type but not the Acinetobacter baumannii hdc::TOPO mutant, such concentration of histamine blocks the ability of hemocytes from Galleria mellonella to capture Candida albicans in vitro. Thus, bacteria-producing histamine, by maintaining high levels of histamine, may impair neutrophil phagocytosis by hijacking the H2R.
Collapse
Affiliation(s)
- Karim Dib
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Amal El Banna
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Clara Radulescu
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Guillermo Lopez Campos
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Gerard Sheehan
- Department of Biology, Maynooth University, Maynooth, Ireland
| | - Kevin Kavanagh
- Department of Biology, Maynooth University, Maynooth, Ireland
| |
Collapse
|
10
|
Yamada E, Martin CG, Moreno-Huizar N, Fouquier J, Neff CP, Coleman SL, Schneider JM, Huber J, Nusbacher NM, McCarter M, Campbell TB, Lozupone CA, Palmer BE. Intestinal microbial communities and Holdemanella isolated from HIV+/- men who have sex with men increase frequencies of lamina propria CCR5 + CD4 + T cells. Gut Microbes 2022; 13:1997292. [PMID: 34818131 PMCID: PMC8632320 DOI: 10.1080/19490976.2021.1997292] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Men who have sex with men (MSM), regardless of HIV infection status, have an intestinal microbiome that is compositionally distinct from men who have sex with women (MSW) and women. We recently showed HIV-negative MSM have elevated levels of intestinal CD4+ T cells expressing CCR5, a critical co-receptor for HIV. Whether elevated expression of CCR5 is driven by the altered gut microbiome composition in MSM has not been explored. Here we used in vitro stimulation of gut Lamina Propria Mononuclear Cells (LPMCs) with whole intact microbial cells isolated from stool to demonstrate that fecal bacterial communities (FBCs) from HIV-positive/negative MSM induced higher frequencies of CCR5+ CD4+ T cells compared to FBCs from HIV-negative MSW and women. To identify potential microbial drivers, we related the frequency of CCR5+ CD4+ T cells to the abundance of individual microbial taxa in rectal biopsy of HIV-positive/negative MSM and controls, and Holdemanella biformis was strongly associated with increased frequency of CCR5+ CD4+ T cells. We used in vitro stimulation of gut LPMCs with the type strain of H. biformis, a second strain of H.biformis and an isolate of the closely related Holdemanella porci , cultured from either a HIV-positive or a HIV-negative MSM stool. H. porci elevated the frequency of both CCR5+ CD4+ T cells and the ratio of TNF-α/IL-10 Genomic comparisons of the 3 Holdemanella isolates revealed unique cell wall and capsular components, which may be responsible for their differences in immunogenicity. These findings describe a novel mechanism potentially linking intestinal dysbiosis in MSM to HIV transmission and mucosal pathogenesis.
Collapse
Affiliation(s)
- Eiko Yamada
- Division of Allergy and Clinical Immunology, Department of Medicine, University of Colorado Anschutz, Aurora, Colorado, USA
| | - Casey G. Martin
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Anschutz, Aurora, Colorado, USA
| | - Nancy Moreno-Huizar
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Anschutz, Aurora, Colorado, USA
| | - Jennifer Fouquier
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Anschutz, Aurora, Colorado, USA
| | - C. Preston Neff
- Division of Allergy and Clinical Immunology, Department of Medicine, University of Colorado Anschutz, Aurora, Colorado, USA
| | | | - Jennifer M. Schneider
- Division of Allergy and Clinical Immunology, Department of Medicine, University of Colorado Anschutz, Aurora, Colorado, USA
| | - Jonathan Huber
- Division of Allergy and Clinical Immunology, Department of Medicine, University of Colorado Anschutz, Aurora, Colorado, USA
| | - Nichole M. Nusbacher
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Anschutz, Aurora, Colorado, USA
| | - Martin McCarter
- Department of Surgery, University of Colorado Anschutz, Aurora, Colorado, USA
| | - Thomas B. Campbell
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz, Aurora, Colorado, USA
| | - Catherine A. Lozupone
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Anschutz, Aurora, Colorado, USA
| | - Brent E. Palmer
- Division of Allergy and Clinical Immunology, Department of Medicine, University of Colorado Anschutz, Aurora, Colorado, USA,CONTACT Brent E. Palmer Division of Allergy and Clinical Immunology, Department of Medicine, University of Colorado Anschutz, Aurora, Colorado, USA
| |
Collapse
|
11
|
Li J, Kumari T, Barazia A, Jha V, Jeong SY, Olson A, Kim M, Lee BK, Manickam V, Song Z, Clemens R, Razani B, Kim J, Dinauer MC, Cho J. Neutrophil DREAM promotes neutrophil recruitment in vascular inflammation. J Exp Med 2022; 219:e20211083. [PMID: 34751735 PMCID: PMC8719643 DOI: 10.1084/jem.20211083] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 09/21/2021] [Accepted: 10/19/2021] [Indexed: 01/02/2023] Open
Abstract
The interaction between neutrophils and endothelial cells is critical for the pathogenesis of vascular inflammation. However, the regulation of neutrophil adhesive function remains not fully understood. Intravital microscopy demonstrates that neutrophil DREAM promotes neutrophil recruitment to sites of inflammation induced by TNF-α but not MIP-2 or fMLP. We observe that neutrophil DREAM represses expression of A20, a negative regulator of NF-κB activity, and enhances expression of pro-inflammatory molecules and phosphorylation of IκB kinase (IKK) after TNF-α stimulation. Studies using genetic and pharmacologic approaches reveal that DREAM deficiency and IKKβ inhibition significantly diminish the ligand-binding activity of β2 integrins in TNF-α-stimulated neutrophils or neutrophil-like HL-60 cells. Neutrophil DREAM promotes degranulation through IKKβ-mediated SNAP-23 phosphorylation. Using sickle cell disease mice lacking DREAM, we show that hematopoietic DREAM promotes vaso-occlusive events in microvessels following TNF-α challenge. Our study provides evidence that targeting DREAM might be a novel therapeutic strategy to reduce excessive neutrophil recruitment in inflammatory diseases.
Collapse
Affiliation(s)
- Jing Li
- Department of Pharmacology, University of Illinois at Chicago College of Medicine, Chicago, IL
| | - Tripti Kumari
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Andrew Barazia
- Department of Pharmacology, University of Illinois at Chicago College of Medicine, Chicago, IL
| | - Vishwanath Jha
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Si-Yeon Jeong
- Department of Pharmacology, University of Illinois at Chicago College of Medicine, Chicago, IL
| | - Amber Olson
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Mijeong Kim
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX
| | - Bum-Kyu Lee
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX
| | - Vijayprakash Manickam
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Zhimin Song
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Regina Clemens
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Babak Razani
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO
- John Cochran VA Medical Center, St. Louis, MO
| | - Jonghwan Kim
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX
| | - Mary C. Dinauer
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Jaehyung Cho
- Department of Pharmacology, University of Illinois at Chicago College of Medicine, Chicago, IL
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
12
|
Nichols BE, Hook JS, Weng K, Ahn C, Moreland JG. Novel neutrophil phenotypic signature in pediatric patients with type 1 diabetes and diabetic ketoacidosis. J Leukoc Biol 2021; 111:849-856. [PMID: 34342036 DOI: 10.1002/jlb.3a1220-826r] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Type 1 diabetes (T1D) is a chronic inflammatory condition sometimes complicated by acute diabetic ketoacidosis (DKA). A subset of patients with T1D develop DKA independent of known risk factors. This study tested the hypothesis that circulating polymorphonuclear leukocytes (PMN) from children with T1D and DKA would exhibit a primed phenotype and that the signature would be unique in patients predisposed to have DKA. Using a prospective cohort study design, neutrophil phenotype was assessed in 30 patients with T1D seen in endocrinology clinic for routine care, 30 patients with acute DKA, and 36 healthy donors. Circulating PMN from patients with DKA display a primed phenotype with increased basal cell-surface CD11b, L-selectin shedding, and enhanced fMLF-elicited reactive oxygen species (ROS) production. Moreover, PMN from T1D patients both with and without DKA lack the capacity to be further primed by incubation with TNF-α, a classic priming stimulus. Primed PMN phenotypic signatures demonstrated are independent of hemoglobin A1c, the premier biological marker for DKA risk, and are consistent with a hyperinflammatory state. A single nucleotide polymorphism in TLR-1 (1805G>T), known to be associated with a hyperinflammatory PMN phenotype, correlated with DKA. This study elucidated a novel phenotypic signature in circulating PMN from children with T1D with and without DKA, and suggests the possibility of a previously unrecognized PMN phenotype with potential clinical implications. Immunophenotype and genotype may be applicable as biomarkers for DKA risk stratification in patients with T1D.
Collapse
Affiliation(s)
- Blake E Nichols
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jessica S Hook
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Kayson Weng
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Chul Ahn
- Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jessica G Moreland
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
13
|
Belchamber KBR, Hughes MJ, Spittle DA, Walker EM, Sapey E. New Pharmacological Tools to Target Leukocyte Trafficking in Lung Disease. Front Immunol 2021; 12:704173. [PMID: 34367163 PMCID: PMC8334730 DOI: 10.3389/fimmu.2021.704173] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 06/24/2021] [Indexed: 01/13/2023] Open
Abstract
Infection and inflammation of the lung results in the recruitment of non-resident immune cells, including neutrophils, eosinophils and monocytes. This swift response should ensure clearance of the threat and resolution of stimuli which drive inflammation. However, once the threat is subdued this influx of immune cells should be followed by clearance of recruited cells through apoptosis and subsequent efferocytosis, expectoration or retrograde migration back into the circulation. This cycle of cell recruitment, containment of threat and then clearance of immune cells and repair is held in exquisite balance to limit host damage. Advanced age is often associated with detrimental changes to the balance described above. Cellular functions are altered including a reduced ability to traffic accurately towards inflammation, a reduced ability to clear pathogens and sustained inflammation. These changes, seen with age, are heightened in lung disease, and most chronic and acute lung diseases are associated with an exaggerated influx of immune cells, such as neutrophils, to the airways as well as considerable inflammation. Indeed, across many lung diseases, pathogenesis and progression has been associated with the sustained presence of trafficking cells, with examples including chronic diseases such as Chronic Obstructive Pulmonary Disease and Idiopathic Pulmonary Fibrosis and acute infections such as Pneumonia and Pneumonitis. In these instances, there is evidence that dysfunctional and sustained recruitment of cells to the airways not only increases host damage but impairs the hosts ability to effectively respond to microbial invasion. Targeting leukocyte migration in these instances, to normalise cellular responses, has therapeutic promise. In this review we discuss the current evidence to support the trafficking cell as an immunotherapeutic target in lung disease, and which potential mechanisms or pathways have shown promise in early drug trials, with a focus on the neutrophil, as the quintessential trafficking immune cell.
Collapse
Affiliation(s)
- Kylie B. R. Belchamber
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Michael J. Hughes
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Daniella A. Spittle
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Eloise M. Walker
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Elizabeth Sapey
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
- NIHR Clinical Research Facility Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| |
Collapse
|
14
|
The Antimicrobial Activity of Peripheral Blood Neutrophils Is Altered in Patients with Primary Ciliary Dyskinesia. Int J Mol Sci 2021; 22:ijms22126172. [PMID: 34201048 PMCID: PMC8230338 DOI: 10.3390/ijms22126172] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/28/2021] [Accepted: 06/02/2021] [Indexed: 01/13/2023] Open
Abstract
The airways of patients with primary ciliary dyskinesia (PCD) contain persistently elevated neutrophil numbers and CXCL8 levels. Despite their abundance, neutrophils fail to clear the airways from bacterial infections. We investigated whether neutrophil functions are altered in patients with PCD. Neutrophils from patients and healthy controls (HC) were isolated from peripheral blood and exposed to various bacterial stimuli or cytokines. Neutrophils from patients with PCD were less responsive to low levels of fMLF in three different chemotaxis assays (p < 0.05), but expression of the fMLF receptors was unaltered. PCD neutrophils showed normal phagocytic function and expression of adhesion molecules. However, PCD neutrophils produced less reactive oxygen species upon stimulation with bacterial products or cytokines compared to HC neutrophils (p < 0.05). Finally, the capacity to release DNA, as observed during neutrophil extracellular trap formation, seemed to be reduced in patients with PCD compared to HC (p = 0.066). These results suggest that peripheral blood neutrophils from patients with PCD, in contrast to those of patients with cystic fibrosis or COPD, do not show features of over-activation, neither on baseline nor after stimulation. If these findings extend to lung-resident neutrophils, the reduced neutrophil activity could possibly contribute to the recurrent respiratory infections in patients with PCD.
Collapse
|
15
|
Lung macrophages drive mucus production and steroid-resistant inflammation in chronic bronchitis. Respir Res 2021; 22:172. [PMID: 34098956 PMCID: PMC8186034 DOI: 10.1186/s12931-021-01762-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 05/27/2021] [Indexed: 11/10/2022] Open
Abstract
Background Patients with chronic obstructive pulmonary disease (COPD) frequently suffer from chronic bronchitis (CB) and display steroid-resistant inflammation with increased sputum neutrophils and macrophages. Recently, a causal link between mucus hyper-concentration and disease progression of CB has been suggested. Methods In this study, we have evaluated the steroid sensitivity of purified, patient-derived sputum and alveolar macrophages and used a novel mechanistic cross-talk assay to examine how macrophages and bronchial epithelial cells cross-talk to regulate MUC5B production. Results We demonstrate that sputum plug macrophages isolated from COPD patients with chronic bronchitis (COPD/CB) are chronically activated and only partially respond to ex vivo corticosteroid treatment compared to alveolar macrophages isolated from lung resections. Further, we show that pseudo-stratified bronchial epithelial cells grown in air–liquid-interface are inert to direct bacterial lipopolysaccharide stimulation and that macrophages are able to relay this signal and activate the CREB/AP-1 transcription factor complex and subsequent MUC5B expression in epithelial cells through a soluble mediator. Using recombinant protein and neutralizing antibodies, we identified a key role for TNFα in this cross-talk. Conclusions For the first time, we describe ex vivo pharmacology in purified human sputum macrophages isolated from chronic bronchitis COPD patients and identify a possible basis for the steroid resistance frequently seen in this population. Our data pinpoint a critical role for chronically activated sputum macrophages in perpetuating TNFα-dependent signals driving mucus hyper-production. Targeting the chronically activated mucus plug macrophage phenotype and interfering with aberrant macrophage-epithelial cross-talk may provide a novel strategy to resolve chronic inflammatory lung disease. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-021-01762-4.
Collapse
|
16
|
Gasparoto TH, Dalboni TM, Amôr NG, Abe AE, Perri G, Lara VS, Vieira NA, Gasparoto CT, Campanelli AP. Fcγ receptors on aging neutrophils. J Appl Oral Sci 2021; 29:e20200770. [PMID: 33825754 PMCID: PMC8011831 DOI: 10.1590/1678-7757-2020-0770] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 12/15/2020] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE Neutrophils are key effector cells of the innate immune system. They recognize antigens through membrane receptors, which are expressed during their maturation and activation. Neutrophils express FcγRII (CD32), FcγRIII (CD16), and FcγRI (CD64) after being activated by different factors such as cytokines and bacterial products. These receptors are involved with phagocytosis of IgG-opsonized microbes and enhance defense mechanisms. Based on that, our study seeks to compare the expression of FcγRII, FcγRIII, FcγRI, and CD11b on neutrophils from elderly and young subjects and their expression after in vitro activation with cytokines and LPS. METHODOLOGY Neutrophils were isolated from human peripheral blood and from mice bone marrow by density gradient. After isolation, FCγRs expression was immediately analyzed by flow cytometry or after in vitro stimulation. RESULTS In freshly isolated cells, the percentage of FcγRIIIb+ and CD11b+ neutrophils were higher in samples from young individuals; FcγRIIIa expression was more prominent on aged neutrophils; FcγRIA expression was similar in all samples analyzed. Exposure to CXCL8 and LPS resulted in a higher percentage of FcγRIa+ neutrophils on elderly individuals' samples but lower when compared with neutrophils from young donors. We observed that LPS caused an increase in FcγRIIa expression on aging human neutrophils. In contrast, FcγRIIIb expression in response to CXCL8 and LPS stimulation was not altered in the four groups. CD11b expression was lower in neutrophils from elderly individuals even in response to LPS and CXCL8. In mice, we observed differences only regarding CD11b expression, which was increased on aged neutrophils. LPS exposure caused an increase in all FcγRs. CONCLUSIONS Our results suggest that, in humans, the overall pattern of FcγR expression and integrin CD11b are altered during aging and immunosenescence might contribute to age-related infection.
Collapse
Affiliation(s)
- Thaís Helena Gasparoto
- Universidade de São Paulo, Faculdade de Odontologia de Bauru, Departamento de Ciências Biológicas, Bauru, SP, Brasil
| | - Thalita Marcato Dalboni
- Universidade de São Paulo, Faculdade de Odontologia de Bauru, Departamento de Ciências Biológicas, Bauru, SP, Brasil
| | - Nádia Ghinelli Amôr
- Universidade de São Paulo, Faculdade de Odontologia de Bauru, Departamento de Ciências Biológicas, Bauru, SP, Brasil
| | - Aneli Eiko Abe
- Universidade de São Paulo, Faculdade de Odontologia de Bauru, Departamento de Ciências Biológicas, Bauru, SP, Brasil
| | - Graziela Perri
- Universidade de São Paulo, Faculdade de Odontologia de Bauru, Departamento de Ciências Biológicas, Bauru, SP, Brasil
| | - Vanessa Soares Lara
- Universidade de São Paulo, Faculdade de Odontologia de Bauru, Departamento de Estomatologia (Patologia Oral), Bauru, SP, Brasil
| | | | - Carlos Teodoro Gasparoto
- Universidade de São Paulo, Faculdade de Medicina de São Paulo, Departamento de Saúde Pública, São Paulo, Brasil
| | - Ana Paula Campanelli
- Universidade de São Paulo, Faculdade de Odontologia de Bauru, Departamento de Ciências Biológicas, Bauru, SP, Brasil
| |
Collapse
|
17
|
Directional mast cell degranulation of tumor necrosis factor into blood vessels primes neutrophil extravasation. Immunity 2021; 54:468-483.e5. [PMID: 33484643 DOI: 10.1016/j.immuni.2020.12.017] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 11/10/2020] [Accepted: 12/23/2020] [Indexed: 12/13/2022]
Abstract
Tissue resident mast cells (MCs) rapidly initiate neutrophil infiltration upon inflammatory insult, yet the molecular mechanism is still unknown. Here, we demonstrated that MC-derived tumor necrosis factor (TNF) was crucial for neutrophil extravasation to sites of contact hypersensitivity-induced skin inflammation by promoting intraluminal crawling. MC-derived TNF directly primed circulating neutrophils via TNF receptor-1 (TNFR1) while being dispensable for endothelial cell activation. The MC-derived TNF was infused into the bloodstream by directional degranulation of perivascular MCs that were part of the vascular unit with access to the vessel lumen. Consistently, intravenous administration of MC granules boosted neutrophil extravasation. Pronounced and rapid intravascular MC degranulation was also observed upon IgE crosslinking or LPs challenge indicating a universal MC potential. Consequently, the directional MC degranulation of pro-inflammatory mediators into the bloodstream may represent an important target for therapeutic approaches aimed at dampening cytokine storm syndromes or shock symptoms, or intentionally pushing immune defense.
Collapse
|
18
|
Liberale L, Bertolotto M, Minetti S, Contini P, Verzola D, Ameri P, Ghigliotti G, Pende A, Camici GG, Carbone F, Montecucco F. Recombinant Tissue Plasminogen Activator (r-tPA) Induces In-Vitro Human Neutrophil Migration via Low Density Lipoprotein Receptor-Related Protein 1 (LRP-1). Int J Mol Sci 2020; 21:7014. [PMID: 32977685 PMCID: PMC7582901 DOI: 10.3390/ijms21197014] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 01/04/2023] Open
Abstract
Thrombolysis is the gold standard treatment for acute ischemic stroke. Besides its fibrinolytic role, recombinant tissue plasminogen activator (r-tPA) holds several non-fibrinolytic functions. Here, we investigated the potential role of r-tPA on human primary neutrophil migration in vitro. By means of modified Boyden chamber migration assay and checkerboard analysis we showed a dose-dependent chemotactic effect of r-TPA with a maximum effect reached by 0.03 mg/mL (0.003-1 mg/mL). Pre-incubation with MAP kinases inhibitors allowed the identification of PI3K/Akt, but not ERK1/2 as the intracellular pathway mediating the observed effects. Furthermore, by means of real-time PCR, immunocytochemistry and cytofluorimetry we demonstrated that the r-tPA receptor low density lipoprotein receptor-related protein 1 (LRP-1) is synthetized and expressed by neutrophils in response to r-tPA and TNF-α. Inhibition of LRP-1 by receptor-associated protein (RAP), prevented r-tPA-mediated F-actin polymerization, migration and signal through Akt but not ERK1/2. Lastly, also neutrophil degranulation in response to r-tPA seems to be mediated by LRP-1 under adhesion conditions. In conclusion, we show that r-tPA induces neutrophil chemotaxis through LRP-1/Akt pathway. Blunting r-tPA-mediated neutrophil activation might be beneficial as an adjuvant therapy to thrombolysis in this setting.
Collapse
Affiliation(s)
- Luca Liberale
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, 8952 Schlieren, Switzerland; (L.L.); (G.G.C.)
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy; (M.B.); (S.M.); (F.C.)
| | - Maria Bertolotto
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy; (M.B.); (S.M.); (F.C.)
| | - Silvia Minetti
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy; (M.B.); (S.M.); (F.C.)
| | - Paola Contini
- Clinical Immunology, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy;
| | - Daniela Verzola
- Division of Nephrology, Dialysis and Transplantation, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy;
| | - Pietro Ameri
- IRCCS Ospedale Policlinico San Martino Genoa—Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa, Italy; (P.A.); (G.G.); (A.P.)
- Laboratory of Cardiovascular Biology, IRCCS Ospedale Policlinico San Martino & Department of Internal Medicine, University of Genoa, 16126 Genoa, Italy
| | - Giorgio Ghigliotti
- IRCCS Ospedale Policlinico San Martino Genoa—Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa, Italy; (P.A.); (G.G.); (A.P.)
- Laboratory of Cardiovascular Biology, IRCCS Ospedale Policlinico San Martino & Department of Internal Medicine, University of Genoa, 16126 Genoa, Italy
| | - Aldo Pende
- IRCCS Ospedale Policlinico San Martino Genoa—Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa, Italy; (P.A.); (G.G.); (A.P.)
- Clinic of Emergency Medicine, Department of Emergency Medicine, University of Genoa, 16126 Genoa, Italy
| | - Giovanni G. Camici
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, 8952 Schlieren, Switzerland; (L.L.); (G.G.C.)
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy; (M.B.); (S.M.); (F.C.)
- IRCCS Ospedale Policlinico San Martino Genoa—Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa, Italy; (P.A.); (G.G.); (A.P.)
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy; (M.B.); (S.M.); (F.C.)
- IRCCS Ospedale Policlinico San Martino Genoa—Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa, Italy; (P.A.); (G.G.); (A.P.)
| |
Collapse
|
19
|
Presicce P, Cappelletti M, Senthamaraikannan P, Ma F, Morselli M, Jackson CM, Mukherjee S, Miller LA, Pellegrini M, Jobe AH, Chougnet CA, Kallapur SG. TNF-Signaling Modulates Neutrophil-Mediated Immunity at the Feto-Maternal Interface During LPS-Induced Intrauterine Inflammation. Front Immunol 2020; 11:558. [PMID: 32308656 PMCID: PMC7145904 DOI: 10.3389/fimmu.2020.00558] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 03/11/2020] [Indexed: 01/10/2023] Open
Abstract
Accumulation of activated neutrophils at the feto-maternal interface is a defining hallmark of intrauterine inflammation (IUI) that might trigger an excessive immune response during pregnancy. Mechanisms responsible of this massive neutrophil recruitment are poorly investigated. We have previously showed that intraamniotic injection of LPS in rhesus macaques induced a neutrophil predominant inflammatory response similar to that seen in human IUI. Here, we demonstrate that anti-TNF antibody (Adalimumab) inhibited ~80% of genes induced by LPS involved in inflammatory signaling and innate immunity in chorio-decidua neutrophils. Consistent with the gene expression data, TNF-blockade decreased LPS-induced neutrophil accumulation and activation at the feto-maternal interface. We also observed a reduction in IL-6 and other pro-inflammatory cytokines but not prostaglandins concentrations in the amniotic fluid. Moreover, TNF-blockade decreased mRNA expression of inflammatory cytokines in the chorio-decidua but not in the uterus, suggesting that inhibition of TNF-signaling decreased the inflammation in a tissue-specific manner within the uterine compartment. Taken together, our results demonstrate a predominant role for TNF-signaling in modulating the neutrophilic infiltration at the feto-maternal interface during IUI and suggest that blockade of TNF-signaling could be considered as a therapeutic approach for IUI, the major leading cause of preterm birth.
Collapse
Affiliation(s)
- Pietro Presicce
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Monica Cappelletti
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Paranthaman Senthamaraikannan
- Division of Neonatology/Pulmonary Biology, Cincinnati Children's Hospital Research Foundation, The University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Feiyang Ma
- Department of Molecular, Cell and Developmental Biology Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Marco Morselli
- Department of Molecular, Cell and Developmental Biology Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Institute for Quantitative and Computational Biosciences-Collaboratory, University of California, Los Angeles, Los Angeles, CA, United States
| | - Courtney M Jackson
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, The University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Shibabrata Mukherjee
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, The University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Lisa A Miller
- California National Primate Research Center, University of California, Davis, Davis, CA, United States.,Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Institute for Quantitative and Computational Biosciences-Collaboratory, University of California, Los Angeles, Los Angeles, CA, United States
| | - Alan H Jobe
- Division of Neonatology/Pulmonary Biology, Cincinnati Children's Hospital Research Foundation, The University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Claire A Chougnet
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, The University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Suhas G Kallapur
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
20
|
Schloss MJ, Horckmans M, Guillamat-Prats R, Hering D, Lauer E, Lenglet S, Weber C, Thomas A, Steffens S. 2-Arachidonoylglycerol mobilizes myeloid cells and worsens heart function after acute myocardial infarction. Cardiovasc Res 2020; 115:602-613. [PMID: 30295758 DOI: 10.1093/cvr/cvy242] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 06/06/2018] [Accepted: 10/04/2018] [Indexed: 12/19/2022] Open
Abstract
AIMS Myocardial infarction (MI) leads to an enhanced release of endocannabinoids and a massive accumulation of neutrophils and monocytes within the ischaemic myocardium. These myeloid cells originate from haematopoietic precursors in the bone marrow and are rapidly mobilized in response to MI. We aimed to determine whether endocannabinoid signalling is involved in myeloid cell mobilization and cardiac recruitment after ischaemia onset. METHODS AND RESULTS Intravenous administration of endocannabinoid 2-arachidonoylglycerol (2-AG) into wild type (WT) C57BL6 mice induced a rapid increase of blood neutrophil and monocyte counts as measured by flow cytometry. This effect was blunted when using cannabinoid receptor 2 knockout mice. In response to MI induced in WT mice, the lipidomic analysis revealed significantly elevated plasma and cardiac levels of the endocannabinoid 2-AG 24 h after infarction, but no changes in anandamide, palmitoylethanolamide, and oleoylethanolamide. This was a consequence of an increased expression of 2-AG synthesizing enzyme diacylglycerol lipase and a decrease of metabolizing enzyme monoacylglycerol lipase (MAGL) in infarcted hearts, as determined by quantitative RT-PCR analysis. The opposite mRNA expression pattern was observed in bone marrow. Pharmacological blockade of MAGL with JZL184 and thus increased systemic 2-AG levels in WT mice subjected to MI resulted in elevated cardiac CXCL1, CXCL2, and MMP9 protein levels as well as higher cardiac neutrophil and monocyte counts 24 h after infarction compared with vehicle-treated mice. Increased post-MI inflammation in these mice led to an increased infarct size, an impaired ventricular scar formation assessed by histology and a worsened cardiac function in echocardiography evaluations up to 21 days. Likewise, JZL184-administration in a myocardial ischaemia-reperfusion model increased cardiac myeloid cell recruitment and resulted in a larger fibrotic scar size. CONCLUSION These findings suggest that changes in endocannabinoid gradients due to altered tissue levels contribute to myeloid cell recruitment from the bone marrow to the infarcted heart, with crucial consequences on cardiac healing and function.
Collapse
Affiliation(s)
- Maximilian J Schloss
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University (LMU) Munich, Pettenkoferstr. 9, Munich, Germany
| | - Michael Horckmans
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University (LMU) Munich, Pettenkoferstr. 9, Munich, Germany.,Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles (U.L.B.), Brussels, Belgium
| | - Raquel Guillamat-Prats
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University (LMU) Munich, Pettenkoferstr. 9, Munich, Germany
| | - Daniel Hering
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University (LMU) Munich, Pettenkoferstr. 9, Munich, Germany
| | - Estelle Lauer
- Unit of Toxicology, CURML, Lausanne University Hospital, Geneva University Hospitals, rue Michel-Servet 1, Geneva CH-1211, Switzerland
| | - Sebastien Lenglet
- Unit of Toxicology, CURML, Lausanne University Hospital, Geneva University Hospitals, rue Michel-Servet 1, Geneva CH-1211, Switzerland
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University (LMU) Munich, Pettenkoferstr. 9, Munich, Germany.,Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands.,German Centre for Cardiovascular Research (DZHK), Partner Site, Munich Heart Alliance, Munich, Germany
| | - Aurelien Thomas
- Unit of Toxicology, CURML, Lausanne University Hospital, Geneva University Hospitals, rue Michel-Servet 1, Geneva CH-1211, Switzerland.,Faculty of Biology and Medicine, University of Lausanne, Vulliette 04, Lausanne 1000, Switzerland
| | - Sabine Steffens
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University (LMU) Munich, Pettenkoferstr. 9, Munich, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site, Munich Heart Alliance, Munich, Germany
| |
Collapse
|
21
|
van Gemmeren T, Schuppner R, Grosse GM, Fering J, Gabriel MM, Huber R, Worthmann H, Lichtinghagen R, Weissenborn K. Early Post-Stroke Infections Are Associated with an Impaired Function of Neutrophil Granulocytes. J Clin Med 2020; 9:jcm9030872. [PMID: 32209993 PMCID: PMC7141520 DOI: 10.3390/jcm9030872] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/11/2020] [Accepted: 03/18/2020] [Indexed: 01/02/2023] Open
Abstract
To investigate whether neutrophil granulocytes’ function relates to post-stroke infections and clinical outcome after stroke, we prospectively recruited 95 patients after ischemic stroke and tested them for their microbiocidal neutrophil functions in this exploratory study. Additionally, 24 age-adjusted controls were examined regarding neutrophil function. Phagocytic capacity and the ability of the neutrophil granulocytes to produce reactive oxygen species (ROS) as well as CD11b and CD16 receptor expression profile were measured by flow cytometry at days 1, 3, 7, and 90 after symptom onset. Primary outcome was the development of an infection within the first week after stroke. Results of neutrophil functional measurements were compared between patients with and without infection as well as between all stroke patients and controls. Further risk factors for the development of infections were summarized in an infection-risk score for the purpose of multivariate statistical analysis. The ROS production in neutrophils after stimulation with formyl-methionyl-leucyl-phenylalanine (fMLP) was reduced at baseline in patients with post-stroke infections compared to those without (p = 0.013). This difference proved to be independent from the infection-risk score in the binary logistic regression (p = 0.011). Phagocytosis and oxidative bursts were not significantly reduced in the whole stroke patient group compared to controls. Dysfunction of neutrophil granulocytes seems to play a significant role in the development of post-stroke infections. Further studies are warranted to investigate neutrophil granulocytes´ function as a potential biomarker of post-stroke infections.
Collapse
Affiliation(s)
- Till van Gemmeren
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany; (T.v.G.); (G.M.G.); (J.F.); (M.M.G.); (H.W.); (K.W.)
| | - Ramona Schuppner
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany; (T.v.G.); (G.M.G.); (J.F.); (M.M.G.); (H.W.); (K.W.)
- Correspondence: ; Tel.: +49-511-532-3580; Fax: +49-511-532-18625
| | - Gerrit M. Grosse
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany; (T.v.G.); (G.M.G.); (J.F.); (M.M.G.); (H.W.); (K.W.)
| | - Jessica Fering
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany; (T.v.G.); (G.M.G.); (J.F.); (M.M.G.); (H.W.); (K.W.)
| | - Maria M. Gabriel
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany; (T.v.G.); (G.M.G.); (J.F.); (M.M.G.); (H.W.); (K.W.)
| | - René Huber
- Institute of Clinical Chemistry, Hannover Medical School, 30625 Hannover, Germany; (R.H.); (R.L.)
| | - Hans Worthmann
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany; (T.v.G.); (G.M.G.); (J.F.); (M.M.G.); (H.W.); (K.W.)
| | - Ralf Lichtinghagen
- Institute of Clinical Chemistry, Hannover Medical School, 30625 Hannover, Germany; (R.H.); (R.L.)
| | - Karin Weissenborn
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany; (T.v.G.); (G.M.G.); (J.F.); (M.M.G.); (H.W.); (K.W.)
| |
Collapse
|
22
|
Zhang Z, Wang Q, Yao J, Zhou X, Zhao J, Zhang X, Dong J, Liao L. Chemokine Receptor 5, a Double-Edged Sword in Metabolic Syndrome and Cardiovascular Disease. Front Pharmacol 2020; 11:146. [PMID: 32194402 PMCID: PMC7063056 DOI: 10.3389/fphar.2020.00146] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 02/04/2020] [Indexed: 01/01/2023] Open
Abstract
The key characteristic of cardiovascular disease (CVD) is endothelial dysfunction, which is likely the consequence of inflammation. It is well demonstrated that chemokines and their receptors play a crucial role in regulating inflammatory responses, and recently, much attention has been paid to chemokine receptor 5 (CCR5) and its ligands. For example, CCR5 aggravates the inflammatory response in adipose tissue by regulating macrophage recruitment and M1/M2 phenotype switch, thus causing insulin resistance and obesity. Inhibition of CCR5 expression reduces the aggregation of pro-atherogenic cytokines to the site of arterial injury. However, targeting CCR5 is not always effective, and emerging evidence has shown that CCR5 facilitates progenitor cell recruitment and promotes vascular endothelial cell repair. In this paper, we provide recent insights into the role of CCR5 and its ligands in metabolic syndrome as related to cardiovascular disease and the opportunities and roadblocks in targeting CCR5 and its ligands.
Collapse
Affiliation(s)
- Zhongwen Zhang
- Department of Endocrinology, Shandong Provincial Qianfoshan Hospital, the First Hospital Affiliated with Shandong First Medical University, Jinan, China.,Division of Endocrinology, Department of Internal Medicine, Shandong Provincial QianFoShan Hospital, Shandong University, Jinan, China
| | - Qiannan Wang
- Department of Endocrinology, Shandong Provincial Qianfoshan Hospital, the First Hospital Affiliated with Shandong First Medical University, Jinan, China.,Division of Endocrinology, Department of Internal Medicine, Shandong Provincial QianFoShan Hospital, Shandong University, Jinan, China
| | - Jinming Yao
- Department of Endocrinology, Shandong Provincial Qianfoshan Hospital, the First Hospital Affiliated with Shandong First Medical University, Jinan, China.,Division of Endocrinology, Department of Internal Medicine, Shandong Provincial QianFoShan Hospital, Shandong University, Jinan, China
| | - Xiaojun Zhou
- Department of Endocrinology, Shandong Provincial Qianfoshan Hospital, the First Hospital Affiliated with Shandong First Medical University, Jinan, China.,Division of Endocrinology, Department of Internal Medicine, Shandong Provincial QianFoShan Hospital, Shandong University, Jinan, China
| | - Junyu Zhao
- Department of Endocrinology, Shandong Provincial Qianfoshan Hospital, the First Hospital Affiliated with Shandong First Medical University, Jinan, China.,Division of Endocrinology, Department of Internal Medicine, Shandong Provincial QianFoShan Hospital, Shandong University, Jinan, China
| | - Xiaoqian Zhang
- Department of Endocrinology, Shandong Provincial Qianfoshan Hospital, the First Hospital Affiliated with Shandong First Medical University, Jinan, China.,Division of Endocrinology, Department of Internal Medicine, Shandong Provincial QianFoShan Hospital, Shandong University, Jinan, China
| | - Jianjun Dong
- Division of Endocrinology, Department of Internal Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Lin Liao
- Department of Endocrinology, Shandong Provincial Qianfoshan Hospital, the First Hospital Affiliated with Shandong First Medical University, Jinan, China.,Division of Endocrinology, Department of Internal Medicine, Shandong Provincial QianFoShan Hospital, Shandong University, Jinan, China
| |
Collapse
|
23
|
Watanabe-Kusunoki K, Nakazawa D, Kusunoki Y, Kudo T, Hattanda F, Nishio S, Masuda S, Tomaru U, Kondo T, Atsumi T, Ishizu A. Recombinant thrombomodulin ameliorates autoimmune vasculitis via immune response regulation and tissue injury protection. J Autoimmun 2019; 108:102390. [PMID: 31883830 DOI: 10.1016/j.jaut.2019.102390] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/10/2019] [Accepted: 12/13/2019] [Indexed: 12/13/2022]
Abstract
Anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) is characterized by necrotizing vasculitis with the presence of pathogenic ANCA. ANCA can potentially cause neutrophil activation and induce neutrophil extracellular traps (NETs), resulting in endothelial damage as well as activation of autoreactive B cells and alternative complement pathway. Recombinant thrombomodulin (rTM) protects the endothelium from vascular injury during disseminated intravascular coagulation, thus we hypothesized that rTM ameliorates necrotizing vasculitis in AAV. In this study, rTM was administered in an experimental AAV rat model. Treatment of experimental AAV rats with rTM improved pulmonary hemorrhage and glomerulonephritis, with a suppression of ANCA production and NETs formation. In addition, in vitro experiments showed that rTM bound to neutrophils via Mac-1 (macrophage-1 antigen) and inhibited ANCA-induced NETs formation accompanied by a suppression of histone citrullination, leading to a protection of the endothelium from NETs toxicity. Additionally, rTM affected lymphocytes leading to the inhibition of pro-inflammatory cytokine/chemokin in PBMC during the antibody production process, which might indirectly be involved in the reduction of pathogenic ANCA. Our data revealed that the rTM could ameliorate autoimmune vasculitis through a combination of different biological mechanisms.
Collapse
Affiliation(s)
- Kanako Watanabe-Kusunoki
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Daigo Nakazawa
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan.
| | - Yoshihiro Kusunoki
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Takashi Kudo
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Fumihiko Hattanda
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Saori Nishio
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Sakiko Masuda
- Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Utano Tomaru
- Department of Pathology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Takeshi Kondo
- Department of Hematology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Tatsuya Atsumi
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Akihiro Ishizu
- Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| |
Collapse
|
24
|
Jordan LA, Erlandsson MC, Fenner BF, Davies R, Harvey AK, Choy EH, Errington R, Bokarewa MI, Williams AS. Inhibition of CCL3 abrogated precursor cell fusion and bone erosions in human osteoclast cultures and murine collagen-induced arthritis. Rheumatology (Oxford) 2019; 57:2042-2052. [PMID: 30053130 PMCID: PMC6199535 DOI: 10.1093/rheumatology/key196] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Indexed: 12/12/2022] Open
Abstract
Objective Macrophage inflammatory protein 1-alpha (CCL3) is a chemokine that regulates macrophage trafficking to the inflamed joint. The agonistic effect of CCL3 on osteolytic lesions in patients with multiple myeloma is recognized; however, its role in skeletal damage during inflammatory arthritis has not been established. The aim of the study was to explore the role of osteoclast-associated CCL3 upon bone resorption, and to test its pharmacological blockade for protecting against bone pathology during inflammatory arthritis. Methods CCL3 production was studied during osteoclast differentiation from osteoclast precursor cells: human CD14-positive mononuclear cells. Mice with CIA were treated with an anti-CCL3 antibody. The effect of CCL3 blockade through mAb was studied through osteoclast number, cytokine production and bone resorption on ivory disks, and in vivo through CIA progression (clinical score, paw diameter, synovial inflammation and bone damage). Results Over time, CCL3 increased in parallel with the number of osteoclasts in culture. Anti-CCL3 treatment achieved a concentration-dependent inhibition of osteoclast fusion and reduced pit formation on ivory disks (P ⩽ 0.05). In CIA, anti-CCL3 treatment reduced joint damage and significantly decreased multinucleated tartrate-resistant acid phosphatase-positive osteoclasts and erosions in the wrists (P < 0.05) and elbows (P < 0.05), while also reducing joint erosions in the hind (P < 0.01) and fore paws (P < 0.01) as confirmed by X-ray. Conclusion Inhibition of osteoclast-associated CCL3 reduced osteoclast formation and function whilst attenuating arthritis-associated bone loss and controlling development of erosion in murine joints, thus uncoupling bone damage from inflammation. Our findings may help future innovations for the diagnosis and treatment of inflammatory arthritis.
Collapse
Affiliation(s)
- Lauren A Jordan
- Division of Infection and Immunity, Cardiff, Wales, UK.,The Cardiff Regional Experimental Arthritis Treatment and Evaluation (CREATE) Centre, Cardiff, Wales, UK
| | - Malin C Erlandsson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, The University of Gothenburg, Göteborg, Sweden
| | | | - Ruth Davies
- Division of Infection and Immunity, Cardiff, Wales, UK.,The Cardiff Regional Experimental Arthritis Treatment and Evaluation (CREATE) Centre, Cardiff, Wales, UK
| | - Ann K Harvey
- Division of Infection and Immunity, Cardiff, Wales, UK
| | - Ernest H Choy
- Division of Infection and Immunity, Cardiff, Wales, UK.,The Cardiff Regional Experimental Arthritis Treatment and Evaluation (CREATE) Centre, Cardiff, Wales, UK
| | - Rachel Errington
- Division of Cancer and Genetics, Cardiff University, School of Medicine, Cardiff, UK
| | - Maria I Bokarewa
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, The University of Gothenburg, Göteborg, Sweden
| | - Anwen S Williams
- Division of Infection and Immunity, Cardiff, Wales, UK.,The Cardiff Regional Experimental Arthritis Treatment and Evaluation (CREATE) Centre, Cardiff, Wales, UK
| |
Collapse
|
25
|
Liberale L, Bertolotto M, Carbone F, Contini P, Wüst P, Spinella G, Pane B, Palombo D, Bonaventura A, Pende A, Mach F, Dallegri F, Camici GG, Montecucco F. Resistin exerts a beneficial role in atherosclerotic plaque inflammation by inhibiting neutrophil migration. Int J Cardiol 2018; 272:13-19. [PMID: 30075966 DOI: 10.1016/j.ijcard.2018.07.112] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 06/18/2018] [Accepted: 07/23/2018] [Indexed: 12/29/2022]
Abstract
BACKGROUND Neutrophil functions have been shown to be modulated by adipocytokines during atherogenesis. The immuno-regulatory role of resistin on neutrophil-mediated activities in atherosclerotic patients remains elusive. Here, we aimed at exploring the association between serum levels of resistin and neutrophil products either in the systemic circulation or within plaques in a cohort of patients with severe carotid plaque stenosis undergoing endarterectomy. In addition, we assessed the effects of resistin on neutrophil pro-atherosclerotic functions in vitro. METHODS Inflammatory biomarkers, neutrophil products and resistin levels were assessed in patients' sera and carotid plaques by ELISA and immunohistochemistry analysis. In vitro, human primary neutrophils isolated from healthy donors were assessed on different substrate cultures for: degranulation (by ELISA), migration (by microchemotaxis Boyden chamber), F-actin polymerization (by fluorescent assay), integrin and chemokine receptor expression (by flow cytometry) and apoptosis (by both morphologic analysis and flow cytometry). RESULTS Serum resistin was positively correlated with serum levels of neutrophil granule products, but inversely with intraplaque neutrophil and MMP-9 contents. In vitro, resistin was detected in supernatants of degranulating neutrophils and positively correlated with other granule products. Although resistin did not affect neutrophil degranulation, apoptosis and integrin or chemokine receptor expression, pre-incubation with human recombinant resistin abrogated CXCL8-induced neutrophil migration and F-actin polymerization by inhibiting ERK2 phosphorylation. CONCLUSION Resistin can be released by degranulating neutrophils and blunts neutrophil plaque infiltration by modulating their migration towards known atherosclerotic mediators. These results suggest a potential immunoregulatory role of resistin in inhibiting neutrophil-mediated atherosclerotic activities.
Collapse
Affiliation(s)
- Luca Liberale
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, 8952 Schlieren, Switzerland; First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy
| | - Maria Bertolotto
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy
| | - Paola Contini
- Clinical Immunology, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Patricia Wüst
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Giovanni Spinella
- Vascular and Endovascular Surgery Unit, Department of Surgery, Ospedale Policlinico San Martino, 10 Largo Benzi, 16132 Genoa, Italy
| | - Bianca Pane
- Vascular and Endovascular Surgery Unit, Department of Surgery, Ospedale Policlinico San Martino, 10 Largo Benzi, 16132 Genoa, Italy
| | - Domenico Palombo
- Vascular and Endovascular Surgery Unit, Department of Surgery, Ospedale Policlinico San Martino, 10 Largo Benzi, 16132 Genoa, Italy
| | - Aldo Bonaventura
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy
| | - Aldo Pende
- Clinic of Emergency Medicine, Department of Emergency Medicine, University of Genoa and Ospedale Policlinico San Martino, 10 Largo Rosanna Benzi, 16132 Genoa, Italy
| | - François Mach
- Division of Cardiology, Department of Medical Specialties, Geneva University Hospitals, 4 rue Gabrielle-Perret-Gentil, 1211 Geneva, Switzerland
| | - Franco Dallegri
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy; Ospedale Policlinico San Martino, 10 Largo Benzi, 16132 Genoa, Italy
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy; Ospedale Policlinico San Martino, 10 Largo Benzi, 16132 Genoa, Italy; Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 9 Viale Benedetto XV, 16132 Genoa, Italy.
| |
Collapse
|
26
|
Sharma D, Malik A, Guy C, Vogel P, Kanneganti TD. TNF/TNFR axis promotes pyrin inflammasome activation and distinctly modulates pyrin inflammasomopathy. J Clin Invest 2018; 129:150-162. [PMID: 30457980 DOI: 10.1172/jci121372] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 10/09/2018] [Indexed: 12/22/2022] Open
Abstract
Pyrin is an inflammasome sensor that promotes caspase-1-mediated pyroptotic cell death and maturation of proinflammatory cytokines IL-1β and IL-18. Familial Mediterranean fever (FMF), an autoinflammatory disorder, is associated with mutations in the gene encoding pyrin (MEFV). FMF-knockin (FMF-KI) mice that express chimeric pyrin protein with FMF mutation (MefvV726A/V726A) exhibit an autoinflammatory disorder mediated by autoactivation of the pyrin inflammasome. Increase in the levels of TNF are observed in FMF-KI mice, and many features of FMF overlap with the autoinflammatory disorder associated with TNF receptor signaling. In this study, we assessed the contribution of TNF signaling to pyrin inflammasome activation and its consequent role in distinct FMF pathologies. TNF signaling promoted the expression of pyrin in response to multiple stimuli and was required for inflammasome activation in response to canonical pyrin stimuli and in myeloid cells from FMF-KI mice. TNF signaling promoted systemic wasting, anemia, and neutrophilia in the FMF-KI mice. Further, TNF-induced pathology was induced specifically through the TNFR1 receptor, while TNFR2-mediated signaling was distinctly protective in colitis and ankle joint inflammation. Overall, our data show that TNF is a critical modulator of pyrin expression, inflammasome activation, and pyrin-inflammasomopathy. Further, specific blockade of TNFR1 or activation of TNFR2 could provide substantial protection against FMF pathologies.
Collapse
Affiliation(s)
| | | | | | - Peter Vogel
- Animal Resources Center and the Veterinary Pathology Core, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | |
Collapse
|
27
|
Morales N, Henriquez C, Sarmiento J, Uberti B, Moran G. Tamoxifen inhibits chemokinesis in equine neutrophils. Ir Vet J 2018; 71:22. [PMID: 30386589 PMCID: PMC6199699 DOI: 10.1186/s13620-018-0133-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 10/17/2018] [Indexed: 11/21/2022] Open
Abstract
Neutrophils are terminally differentiated innate effector cells at the first line of host defense. Neutrophil migration within tissues is complex and involves several steps, during which these cells must be able to interpret a variety of chemical and physical signals. Exacerbated neutrophil activity can be harmful to surrounding tissues; this is important in a range of diseases, including equine asthma. Tamoxifen (TX) is a non-steroidal estrogen receptor modulator with effects on cell growth and survival. Previous studies showed that TX treatment in horses with induced acute pulmonary inflammation promoted early apoptosis of blood and bronchoalveolar lavage fluid (BALF) neutrophils, reduction of BALF neutrophil content, and improvement in animals’ clinical status. Further, TX dampens chemotactic index and respiratory burst production in vitro. The aim of this study was to provide information on the effect of TX on chemokinesis in peripheral blood neutrophils from five healthy horses. Results showed that neutrophils increased migration and travelled distance in response to IL-8; but in the presence of TX, IL-8 did not produce neutrophil migration. This suggests that TX has an inhibitory effect on the kinesis of equine peripheral blood neutrophils stimulated with IL-8. However, further studies are required to fully understand the signaling pathways of TX on neutrophil chemokinesis.
Collapse
Affiliation(s)
- Natalia Morales
- 1Department of Pharmacology, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Claudio Henriquez
- 1Department of Pharmacology, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Jose Sarmiento
- 2Department of Physiology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Benjamin Uberti
- 3Department of Veterinary Clinical Sciences, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Gabriel Moran
- 1Department of Pharmacology, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia, Chile
| |
Collapse
|
28
|
Antoniellis Silveira AA, Dominical VM, Morelli Vital D, Alves Ferreira W, Trindade Maranhão Costa F, Werneck CC, Ferreira Costa F, Conran N. Attenuation of TNF-induced neutrophil adhesion by simvastatin is associated with the inhibition of Rho-GTPase activity, p50 activity and morphological changes. Int Immunopharmacol 2018; 58:160-165. [PMID: 29604489 DOI: 10.1016/j.intimp.2018.03.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 03/22/2018] [Accepted: 03/24/2018] [Indexed: 12/19/2022]
Abstract
Neutrophil adhesion to the vasculature in response to potent inflammatory stimuli, such as TNF-α (TNF), can contribute to atheroprogression amongst other pathophysiological mechanisms. Previous studies have shown that simvastatin, a statin with known pleiotropic anti-inflammatory properties, can partially abrogate the effects of TNF-induced neutrophil adhesion, in association with the modulation of β2-integrin expression. We aimed to further characterize the effects of this statin on neutrophil and leukocyte adhesive mechanisms in vitro and in vivo. A microfluidic assay confirmed the ability of simvastatin to inhibit TNF-induced human neutrophil adhesion to fibronectin ligand under conditions of shear stress, while intravital imaging microscopy demonstrated an abrogation of leukocyte recruitment by simvastatin in the microvasculature of mice that had received a TNF stimulus. This inhibition of neutrophil adhesion was accompanied by the inhibition of TNF-induced RhoA activity in human neutrophils, and alterations in cell morphology and β2-integrin activity. Additionally, TNF augmented the activity of the p50 NFκB subunit in human neutrophils and TNF-induced neutrophil adhesion and β2-integrin activity could be abolished using pharmacological inhibitors of NFκB translocation, BAY11-7082 and SC514. Accordingly, the TNF-induced elevation of neutrophil p50 activity was abolished by simvastatin. In conclusion, our data provide further evidence of the ability of simvastatin to inhibit neutrophil adhesive interactions in response to inflammatory stimuli, both in vivo and in vitro. Simvastatin appears to inhibit neutrophil adhesion by interfering in TNF-induced cytoskeletal rearrangements, in association with the inhibition of Rho A activity, NFκB translocation and, consequently, β2-integrin activity.
Collapse
Affiliation(s)
| | - Venina Marcela Dominical
- Hematology and Hemotherapy Center, School of Medicine, University of Campinas-UNICAMP, Campinas, Sao Paulo, Brazil
| | - Daiana Morelli Vital
- Hematology and Hemotherapy Center, School of Medicine, University of Campinas-UNICAMP, Campinas, Sao Paulo, Brazil
| | - Wilson Alves Ferreira
- Hematology and Hemotherapy Center, School of Medicine, University of Campinas-UNICAMP, Campinas, Sao Paulo, Brazil
| | - Fabio Trindade Maranhão Costa
- Laboratory of Tropical Diseases - Prof. Dr. Luiz Jacintho da Silva, Department of Genetics, Evolution and Bioagents, Institute of Biology (IB), University of Campinas-UNICAMP, Brazil
| | - Claudio C Werneck
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas-UNICAMP, Campinas, Brazil
| | - Fernando Ferreira Costa
- Hematology and Hemotherapy Center, School of Medicine, University of Campinas-UNICAMP, Campinas, Sao Paulo, Brazil
| | - Nicola Conran
- Hematology and Hemotherapy Center, School of Medicine, University of Campinas-UNICAMP, Campinas, Sao Paulo, Brazil.
| |
Collapse
|
29
|
Souza-Almeida G, D'Avila H, Almeida PE, Luna-Gomes T, Liechocki S, Walzog B, Hepper I, Castro-Faria-Neto HC, Bozza PT, Bandeira-Melo C, Maya-Monteiro CM. Leptin Mediates In Vivo Neutrophil Migration: Involvement of Tumor Necrosis Factor-Alpha and CXCL1. Front Immunol 2018; 9:111. [PMID: 29467755 PMCID: PMC5808117 DOI: 10.3389/fimmu.2018.00111] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 01/15/2018] [Indexed: 11/13/2022] Open
Abstract
Leptin directly activates macrophages and lymphocytes, but the role of leptin in neutrophil activation and migration is still controversial. Here, we investigate the in vivo mechanisms of neutrophil migration induced by leptin. The intraperitoneal injection of leptin (1 mg/kg) induces a time- and concentration-dependent neutrophil influx. We did not observe the enhancement of lipid bodies/droplets in neutrophils, after leptin treatment, as we had observed previously in peritoneal macrophages. The participation of leukotriene B4 (LTB4) in neutrophil recruitment triggered by leptin was investigated using different strategies. Leptin-induced neutrophil recruitment occurs both in the absence of 5-lipoxygenase activity in 5-lipoxygenase (5-LO)-/- mice and after the administration of either 5-LO inhibitor (Zileuton) or the LTB4 receptor antagonist (U-75302). Moreover, no direct induction of LTB4 by leptin could be observed. Neutrophil influx could not be prevented by the mammalian target of rapamycin (mTOR) inhibitor, rapamycin, contrasting with the leptin-induced signaling for lipid body formation in macrophage that is mTOR-dependent. Leptin administration led to tumor necrosis factor-alpha (TNFα) production by the peritoneal cells both in vivo and in vitro. In addition, neutrophil recruitment was inhibited in tumor necrosis factor receptor 1 (TNFR1-/-) mice, indicating a role for TNF in leptin-induced neutrophil recruitment to the peritoneal cavity. Leptin-induced neutrophil influx was PI3Kγ-dependent, as it was absent in PI3Kγ-/- mice. Accordingly, leptin induced the peritoneal cells to produce CXCL1, both in vivo and in vitro, and the neutrophil influx was ablated after using an antibody against CXCL1. Our results establish TNFα/TNFR1- and CXCL1-dependent signaling as important pathways for leptin-induced neutrophil migration in vivo.
Collapse
Affiliation(s)
- Glaucia Souza-Almeida
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Heloisa D'Avila
- Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Juiz de Fora, Brazil
| | - Patricia E Almeida
- Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Juiz de Fora, Brazil
| | - Tatiana Luna-Gomes
- Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Juiz de Fora, Brazil.,Departamento de Ciências da Natureza, Instituto de Aplicação Fernando Rodrigues da Silveira, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sally Liechocki
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Barbara Walzog
- Walter Brendel Centre of Experimental Medicine, Department of Cardiovascular Physiology and Pathophysiology, Biomedical Center, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Ingrid Hepper
- Walter Brendel Centre of Experimental Medicine, Department of Cardiovascular Physiology and Pathophysiology, Biomedical Center, Ludwig-Maximilians-Universität München, Munich, Germany
| | | | - Patricia T Bozza
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Christianne Bandeira-Melo
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Clarissa M Maya-Monteiro
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| |
Collapse
|
30
|
Neutrophil Phenotype Correlates With Postoperative Inflammatory Outcomes in Infants Undergoing Cardiopulmonary Bypass. Pediatr Crit Care Med 2017; 18:1145-1152. [PMID: 29068910 DOI: 10.1097/pcc.0000000000001361] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVES Infants with congenital heart disease frequently require cardiopulmonary bypass, which causes systemic inflammation. The goal of this study was to determine if neutrophil phenotype and activation status predicts the development of inflammatory complications following cardiopulmonary bypass. DESIGN Prospective cohort study. SETTING Tertiary care PICU with postoperative cardiac care. PATIENTS Thirty-seven patients 5 days to 10 months old with congenital heart disease requiring cardiopulmonary bypass. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Laboratory and clinical data collected included length of mechanical ventilation, acute kidney injury, and fluid overload. Neutrophils were isolated from whole blood at three time points surrounding cardiopulmonary bypass. Functional analyses included measurement of cell surface protein expression and nicotinamide adenine dinucleotide phosphate oxidase activity. Of all patients studied, 40.5% displayed priming of nicotinamide adenine dinucleotide phosphate oxidase activity in response to N-formyl-Met-Leu-Phe stimulation 24 hours post cardiopulmonary bypass as compared to pre bypass. Neonates who received steroids prior to bypass demonstrated enhanced priming of nicotinamide adenine dinucleotide phosphate oxidase activity at 48 hours. Patients who displayed priming post cardiopulmonary bypass were 8.8 times more likely to develop severe acute kidney injury as compared to nonprimers. Up-regulation of neutrophil surface CD11b levels pre- to postbypass occurred in 51.4% of patients, but this measure of neutrophil priming was not associated with acute kidney injury. Subsequent analyses of the basal neutrophil phenotype revealed that those with higher basal CD11b expression were significantly less likely to develop acute kidney injury. CONCLUSIONS Neutrophil priming occurs in a subset of infants undergoing cardiopulmonary bypass. Acute kidney injury was more frequent in those patients who displayed priming of nicotinamide adenine dinucleotide phosphate oxidase activity after cardiopulmonary bypass. This pilot study suggests that neutrophil phenotypic signature could be used to predict inflammatory organ dysfunction.
Collapse
|
31
|
Yang G, Chen X, Yan Z, Zhu Q, Yang C. CD11b promotes the differentiation of osteoclasts induced by RANKL through the spleen tyrosine kinase signalling pathway. J Cell Mol Med 2017; 21:3445-3452. [PMID: 28661042 PMCID: PMC5706498 DOI: 10.1111/jcmm.13254] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Accepted: 04/24/2017] [Indexed: 12/28/2022] Open
Abstract
Macrophage surface antigen‐1 (Mac‐1, CD11b/CD18) has been implicated in the regulation of osteoclastogenesis. In the synovial tissues of patients with aseptic loosening after total hip replacement, CD11b was up‐regulated, which indicated that CD11b is closely involved in osteolysis around the prosthesis. We found that CD11b, but not CD18, promoted osteoclast (OC) maturation. Here, we show CD11b up‐regulated the levels of spleen tyrosine kinase (Syk), c‐Fos and nuclear factor of activated T cells, cytoplasmic‐1 (NFATc1), as well as the activity of extracellular‐regulated kinase (Erk), and as a result, osteoclast precursors (OCPs) differentiated and became tartrate‐resistant acid phosphatase (TRAP)‐positive. In addition, increased tumour necrosis factor‐α (TNF‐α) induced by ultra‐high molecular weight polyethylene (UHMWPE) particles up‐regulated the level of CD11b. Taken together, these findings suggest that CD11b is a positive regulator of osteoclastogenesis and that it functions by activating the Syk signalling pathway, while CD18 does not have the same effect.
Collapse
Affiliation(s)
- Guoxi Yang
- Institute of Orthopaedic Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xiaoyong Chen
- Institute of Orthopaedic Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zhao Yan
- Institute of Orthopaedic Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Qingsheng Zhu
- Institute of Orthopaedic Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Chongfei Yang
- Institute of Orthopaedic Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
32
|
Miralda I, Uriarte SM, McLeish KR. Multiple Phenotypic Changes Define Neutrophil Priming. Front Cell Infect Microbiol 2017; 7:217. [PMID: 28611952 PMCID: PMC5447094 DOI: 10.3389/fcimb.2017.00217] [Citation(s) in RCA: 143] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 05/11/2017] [Indexed: 12/15/2022] Open
Abstract
Exposure to pro-inflammatory cytokines, chemokines, mitochondrial contents, and bacterial and viral products induces neutrophils to transition from a basal state into a primed one, which is currently defined as an enhanced response to activating stimuli. Although, typically associated with enhanced generation of reactive oxygen species (ROS) by the NADPH oxidase, primed neutrophils show enhanced responsiveness of exocytosis, NET formation, and chemotaxis. Phenotypic changes associated with priming also include activation of a subset of functions, including adhesion, transcription, metabolism, and rate of apoptosis. This review summarizes the breadth of phenotypic changes associated with priming and reviews current knowledge of the molecular mechanisms behind those changes. We conclude that the current definition of priming is too restrictive. Priming represents a combination of enhanced responsiveness and activated functions that regulate both adaptive and innate immune responses.
Collapse
Affiliation(s)
- Irina Miralda
- Department of Microbiology, University of Louisville School of MedicineLouisville, KY, United States
| | - Silvia M Uriarte
- Department of Microbiology, University of Louisville School of MedicineLouisville, KY, United States.,Department of Medicine, University of Louisville School of MedicineLouisville, KY, United States
| | - Kenneth R McLeish
- Department of Medicine, University of Louisville School of MedicineLouisville, KY, United States.,Robley Rex VA Medical CenterLouisville, KY, United States
| |
Collapse
|
33
|
Ampofo E, Lachnitt N, Rudzitis-Auth J, Schmitt BM, Menger MD, Laschke MW. Indole-3-carbinol is a potent inhibitor of ischemia-reperfusion-induced inflammation. J Surg Res 2017; 215:34-46. [PMID: 28688659 DOI: 10.1016/j.jss.2017.03.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 02/21/2017] [Accepted: 03/23/2017] [Indexed: 01/11/2023]
Abstract
BACKGROUND Ischemia-reperfusion (I/R) induces tissue inflammation, which is characterized by an increased leukocyte-endothelial cell interaction and leukocyte transmigration. These processes are mediated by the activation of the nuclear factor (NF)κB signaling pathway, resulting in an elevated expression of specific adhesion molecules. The phytochemical indole-3-carbinol (I3C) has been shown to exert anti-inflammatory effects by interfering with NFκB signal transduction. The aim of the present study was to investigate whether I3C is capable of counteracting the pathogenesis of I/R injury. MATERIALS AND METHODS We investigated the inhibitory effect of I3C on endothelial surface protein expression during hypoxia and reoxygenation by flow cytometry. Moreover, the subcellular localization of NFκB was analyzed by immunofluorescence and Western blot. Adhesion protein levels on leukocytes after tumor necrosis factor-α stimulation were determined using flow cytometry. Finally, leukocyte-endothelial cell interaction and leukocyte transmigration during I/R was investigated in dorsal skinfold chambers of BALB/c mice by means of repetitive intravital fluorescence microscopy and immunohistochemistry. RESULTS I3C suppressed the expression of E-selectin and intercellular adhesion molecule-1 on human dermal microvascular endothelial cells by reducing the transcriptional activity of NFκB. Furthermore, surface protein levels of macrophage-1 antigen as well as activated lymphocyte function-associated antigen-1 were markedly reduced on I3C-treated leukocytes. In vivo, I3C treatment decreased the numbers of adherent and transmigrated leukocytes. This was associated with a reduced macromolecular leakage when compared with vehicle-treated controls. CONCLUSIONS These novel results indicate that I3C reduces the expression of endothelial and leukocytic adhesion proteins, resulting in attenuated leukocyte-endothelial cell interactions during I/R. Accordingly, dietary supplements containing I3C may be beneficial for the treatment of I/R-induced inflammation.
Collapse
Affiliation(s)
- Emmanuel Ampofo
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg/Saar, Germany.
| | - Nico Lachnitt
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | | | - Beate M Schmitt
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Michael D Menger
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Matthias W Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg/Saar, Germany
| |
Collapse
|
34
|
Potera RM, Jensen MJ, Hilkin BM, South GK, Hook JS, Gross EA, Moreland JG. Neutrophil azurophilic granule exocytosis is primed by TNF-α and partially regulated by NADPH oxidase. Innate Immun 2016; 22:635-646. [PMID: 27655046 DOI: 10.1177/1753425916668980] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Neutrophil (polymorphonuclear leukocyte) activation with release of granule contents plays an important role in the pathogenesis of acute lung injury, prompting clinical trials of inhibitors of neutrophil elastase. Despite mounting evidence for neutrophil-mediated host tissue damage in a variety of disease processes, mechanisms regulating azurophilic granule exocytosis at the plasma membrane, and thus release of elastase and other proteases, are poorly characterized. We hypothesized that azurophilic granule exocytosis would be enhanced under priming conditions similar to those seen during acute inflammatory events and during chronic inflammatory disease, and selected the cytokine TNF-α to model this in vitro. Neutrophils stimulated with TNF-α alone elicited intracellular reactive oxygen species (ROS) generation and mobilization of secretory vesicles, specific, and gelatinase granules. p38 and ERK1/2 MAPK were involved in these components of priming. TNF-α priming alone did not mobilize azurophilic granules to the cell surface, but did markedly increase elastase release into the extracellular space in response to secondary stimulation with N-formyl-Met-Leu-Phe (fMLF). Priming of fMLF-stimulated elastase release was further augmented in the absence of NADPH oxidase-derived ROS. Our findings provide a mechanism for host tissue damage during neutrophil-mediated inflammation and suggest a novel anti-inflammatory role for the NADPH oxidase.
Collapse
Affiliation(s)
- Renee M Potera
- 1 Department of Pediatrics, University of Texas Southwestern Medical Center, USA
| | - Melissa J Jensen
- 2 Department of Pediatrics, The University of Iowa, USA.,3 The Inflammation Program, The University of Iowa and Veterans Affairs Medical Center, USA
| | - Brieanna M Hilkin
- 2 Department of Pediatrics, The University of Iowa, USA.,3 The Inflammation Program, The University of Iowa and Veterans Affairs Medical Center, USA
| | - Gina K South
- 2 Department of Pediatrics, The University of Iowa, USA
| | - Jessica S Hook
- 1 Department of Pediatrics, University of Texas Southwestern Medical Center, USA.,4 Department of Microbiology, University of Texas Southwestern Medical Center, USA
| | - Emily A Gross
- 2 Department of Pediatrics, The University of Iowa, USA.,3 The Inflammation Program, The University of Iowa and Veterans Affairs Medical Center, USA
| | - Jessica G Moreland
- 1 Department of Pediatrics, University of Texas Southwestern Medical Center, USA.,4 Department of Microbiology, University of Texas Southwestern Medical Center, USA
| |
Collapse
|
35
|
Sándor N, Lukácsi S, Ungai-Salánki R, Orgován N, Szabó B, Horváth R, Erdei A, Bajtay Z. CD11c/CD18 Dominates Adhesion of Human Monocytes, Macrophages and Dendritic Cells over CD11b/CD18. PLoS One 2016; 11:e0163120. [PMID: 27658051 PMCID: PMC5033469 DOI: 10.1371/journal.pone.0163120] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 09/03/2016] [Indexed: 12/13/2022] Open
Abstract
Complement receptors CR3 (CD11b/CD18) and CR4 (CD11c/CD18) belong to the family of beta2 integrins and are expressed mainly by myeloid cell types in humans. Previously, we proved that CR3 rather than CR4 plays a key role in phagocytosis. Here we analysed how CD11b and CD11c participate in cell adhesion to fibrinogen, a common ligand of CR3 and CR4, employing human monocytes, monocyte-derived macrophages (MDMs) and monocyte-derived dendritic cells (MDDCs) highly expressing CD11b as well as CD11c. We determined the exact numbers of CD11b and CD11c on these cell types by a bead-based technique, and found that the ratio of CD11b/CD11c is 1.2 for MDDCs, 1.7 for MDMs and 7.1 for monocytes, suggesting that the function of CD11c is preponderant in MDDCs and less pronounced in monocytes. Applying state-of-the-art biophysical techniques, we proved that cellular adherence to fibrinogen is dominated by CD11c. Furthermore, we found that blocking CD11b significantly enhances the attachment of MDDCs and MDMs to fibrinogen, demonstrating a competition between CD11b and CD11c for this ligand. On the basis of the cell surface receptor numbers and the measured adhesion strength we set up a model, which explains the different behavior of the three cell types.
Collapse
Affiliation(s)
- Noémi Sándor
- MTA-ELTE Immunology Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Szilvia Lukácsi
- Department of Immunology, Institute of Biology, Faculty of Science, Eötvös Loránd University, Budapest, Hungary
| | - Rita Ungai-Salánki
- Department of Biological Physics, Institute of Physics, Faculty of Science, Eötvös Loránd University, Budapest, Hungary
| | - Norbert Orgován
- Nanobiosensorics “Lendület” Group, Institute of Technical Physics and Material Sciences, Centre for Energy Research, Hungarian Academy of Sciences, Budapest, Hungary
| | - Bálint Szabó
- Department of Biological Physics, Institute of Physics, Faculty of Science, Eötvös Loránd University, Budapest, Hungary
| | - Róbert Horváth
- Nanobiosensorics “Lendület” Group, Institute of Technical Physics and Material Sciences, Centre for Energy Research, Hungarian Academy of Sciences, Budapest, Hungary
| | - Anna Erdei
- MTA-ELTE Immunology Research Group, Hungarian Academy of Sciences, Budapest, Hungary
- Department of Immunology, Institute of Biology, Faculty of Science, Eötvös Loránd University, Budapest, Hungary
| | - Zsuzsa Bajtay
- Department of Immunology, Institute of Biology, Faculty of Science, Eötvös Loránd University, Budapest, Hungary
- * E-mail:
| |
Collapse
|
36
|
Habitual physical activity is associated with the maintenance of neutrophil migratory dynamics in healthy older adults. Brain Behav Immun 2016; 56:12-20. [PMID: 26928196 PMCID: PMC4929133 DOI: 10.1016/j.bbi.2016.02.024] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 02/14/2016] [Accepted: 02/25/2016] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Dysfunctional neutrophils with advanced age are a hallmark of immunosenescence. Reduced migration and bactericidal activity increase the risk of infection. It remains unclear why neutrophil dysfunction occurs with age. Physical activity and structured exercise have been suggested to improve immune function in the elderly. The aim of this study was to assess a comprehensive range of neutrophil functions and determine their association with habitual physical activity. METHOD Physical activity levels were determined in 211 elderly (67±5years) individuals by 7-days of accelerometry wear. Twenty of the most physically active men and women were matched for age and gender to twenty of the least physically active individuals. Groups were compared for neutrophil migration, phagocytosis, oxidative burst, cell surface receptor expression, metabolic health parameters and systemic inflammation. Groups were also compared against ten young participants (23±4years). RESULTS The most active group completed over twice as many steps/day as the least active group (p<0.001), had lower BMI's (p=0.007) and body fat percentages (p=0.029). Neutrophils migrated towards IL-8 better in the most active group compared to the least active (p<0.05) and was comparable to that of the young (p>0.05). These differences remained after adjusting for BMI, body fat and plasma metabolic markers which were different between groups. Correlations revealed that steps/day, higher adiponectin and lower insulin were positively associated with migratory ability (p<0.05). There was no difference in expression of the chemokine receptors CXCR1 or CXCR2 (p>0.05 for both). CD11b was higher in the most active group compared to the least active (p=0.048). No differences between activity groups or young controls were observed for neutrophil phagocytosis or oxidative burst in response to Escherichia coli (p>0.05). The young group had lower concentrations of IL-6, IL-8, MCP-1, CRP, IL-10 and IL-13 (p<0.05 for all) with no differences between the two older groups. CONCLUSION These data suggest that impaired neutrophil migration, but not bactericidal function, in older adults may be, in part, the result of reduced physical activity. A 2-fold difference in physical activity is associated with better preserved neutrophil migratory dynamics in healthy older people. As a consequence increasing habitual physical activity may be beneficial for neutrophil mediated immunity.
Collapse
|
37
|
|
38
|
Zou J, Chen J, Yan Q, Guo Q, Bao C. Serum IL8 and mRNA level of CD11b in circulating neutrophils are increased in clinically amyopathic dermatomyositis with active interstitial lung disease. Clin Rheumatol 2015; 35:117-25. [DOI: 10.1007/s10067-015-3080-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Revised: 08/13/2015] [Accepted: 09/19/2015] [Indexed: 01/29/2023]
|
39
|
Zhang PP, Meng ZT, Wang LC, Guo LM, Li K. Astragalus polysaccharide promotes the release of mature granulocytes through the L-selectin signaling pathway. Chin Med 2015; 10:17. [PMID: 26161135 PMCID: PMC4497426 DOI: 10.1186/s13020-015-0043-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 06/23/2015] [Indexed: 01/03/2023] Open
Abstract
Background This study aims to investigate the leukogenic effect of astragalus polysaccharide (APS), to compare its effect of increasing the numbers of mature granulocytes with that of granulocyte colony-stimulating factor (G-CSF), and to investigate the mechanism. Methods Rats were arbitrarily grouped into four groups (control, cyclophosphamide (CTX), CTX + APS, and CTX + G-CSF groups), and each group was then arbitrarily divided into five subgroups according to the time period since CTX infusion (0, 4, 7, 10, and 14 days). The expression of leukocyte selectin (L-selectin), its ligand, and shedding-related protease on granulocytes was analyzed. Leukocyte counts were obtained. Chemotactic capacity of polymorphonuclear leukocytes (PMNLs) was assessed. Results Both APS and G-CSF restored the expression of L-selectin, P-selectin glycoprotein ligand-1 (PSGL-1), CD11b/CD18, and ADAM17 to normal levels (P > 0.05 vs. control group on each time point), with APS eliciting a greater effect than G-CSF (P = 0.005 on day 7, P < 0.001 on day 10 and 14 for L-selectin; P = 0.038 on day 7, P = 0.001 on day 10, P < 0.001 on day 14 for PSGL-1; P < 0.001 on day 7, 10 and 14 for ADAM17; P < 0.001 on day 7, 10, and 14 for CD11b/CD18). The percentages of the bands and segmented bone marrow (BM) cells in myeloid neutrophils were higher in the CTX + APS group than in the CTX group on day 7 (P = 0.030) and reached normal levels on day 10 (P = 0.547) and 14 (P = 0.431) vs. control group. The ability of APS to increase numbers of PMNLs in peripheral blood after chemotherapy was significantly superior to that of G-CSF 7 days after chemotherapy (P = 0.029 on day 10, P = 0.006 on day 14). Moreover, APS more significantly improved the chemotactic ability of PMNLs among mature BM granulocytes and peripheral blood neutrophils after chemotherapy than did G-CSF (P < 0.001 on day 7, P = 0.001 on day 10 and P = 0.005 on day 14). Conclusions APS promoted the differentiation and chemotactic ability of BM granulocytes via the L-selectin signaling pathway. Electronic supplementary material The online version of this article (doi:10.1186/s13020-015-0043-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ping-Ping Zhang
- Department of Thoracic Medical Oncology, Lung Cancer Diagnosis and Treatment Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060 China.,Department of Radiotherapy, Hubei Cancer Hospital, Wuhan, 430079 China
| | - Zhao-Ting Meng
- Department of Thoracic Medical Oncology, Lung Cancer Diagnosis and Treatment Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060 China
| | - Liu-Chun Wang
- Department of Thoracic Medical Oncology, Lung Cancer Diagnosis and Treatment Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060 China
| | - Lei-Ming Guo
- Tianjin Institute for Biomedicinal Research, Tianjin, 300050 China
| | - Kai Li
- Department of Thoracic Medical Oncology, Lung Cancer Diagnosis and Treatment Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060 China
| |
Collapse
|
40
|
Abstract
OBJECTIVE Iron participates in several mechanisms involving inflammation and innate immunity, yet the dysregulation of its homeostasis is a major cause of metabolic syndrome. Adipocytes should play a major role in iron metabolism, as an impairment in iron turnover is closely related to insulin resistance, obesity, and type 2 diabetes. The aim of this study was to investigate the role of iron in an in vitro-inflamed adipocyte model. METHODS Gene expression of tumor necrosis factor-α, interleukin-6, inflammatory chemokines (CCL3, CCL4, and CXCL12), and molecules involved in iron metabolism were evaluated in an in vitro mouse 3T3-L1 cell model. Cells underwent treatment with FeSO4 heptahydrate and lipopolysaccharide (LPS) stimulation. Toll-like receptor 4 (TLR4) membrane expression, lipid droplet immunohystochemistry, and lipolysis were also evaluated. RESULTS Iron sulphate heptahydrate elicited gene expression of hepcidin, hemojuvelin, and ferroportin at different time courses. Additionally, it activated lipolysis but did not trigger any adipokine gene expression. When cells treated with physiological doses of iron were also stimulated with LPS, an enhancement in the LPS-induced gene expression of cytokines and chemokines was observed. The enhancement occurred with different patterns depending on different time courses and investigated genes, showing its maximal effect for IL-6 gene expression. CONCLUSIONS FeSO4 heptahydrate at a relatively physiological dose, induced gene expression of iron modulatory proteins and also enhanced RNA transcripts of several inflammatory cytokines and chemokines through a priming/synergistic mechanism involving membrane TLR4.
Collapse
|
41
|
Carbone F, Vuilleumier N, Bertolotto M, Burger F, Galan K, Roversi G, Tamborino C, Casetta I, Seraceni S, Trentini A, Dallegri F, da Silva AR, Pende A, Artom N, Mach F, Coen M, Fainardi E, Montecucco F. Treatment with recombinant tissue plasminogen activator (r-TPA) induces neutrophil degranulation in vitro via defined pathways. Vascul Pharmacol 2015; 64:16-27. [PMID: 25530154 DOI: 10.1016/j.vph.2014.11.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 10/21/2014] [Accepted: 11/29/2014] [Indexed: 12/21/2022]
Abstract
Thrombolysis is recommended for reperfusion following acute ischemic stroke (AIS), but its effects on stroke-associated injury remain to be clarified. Here, we investigated the effects of recombinant tissue plasminogen activator (r-tPA) on neutrophil pathophysiology in vitro and in a case-control study with AIS patients submitted (n=60) or not (n=30) to thrombolysis. Patients underwent radiological and clinical examination as well as blood sampling at admission and after 1, 7 and 90days. In vitro, 30-min incubation with 0.1-1 mg/ml r-tPA induced neutrophil degranulation in different substrate cultures. Pre-incubation with kinase inhibitors and Western blot documented that degranulation was associated with activation of PI3K/Akt and ERK1/2 pathways in Teflon dishes and PI3K/Akt in polystyrene. In thrombolysed patients, a peak of neutrophil degranulation products (matrix metalloproteinase [MMP]-9, MMP-8, neutrophil elastase and myeloperoxidase), was shown during the first hours from drug administration. This was accompanied by serum augmentation of protective tissue inhibitor of metalloproteinase (TIMP)-1 and TIMP-2. An increased rate of haemorrhagic transformations on day 1 after AIS was shown in thrombolysed patients as compared to non-thrombolysed controls. In conclusion, r-tPA treatment was associated with in vitro neutrophil degranulation, indicating these cells as potential determinants in early haemorrhagic complications after thrombolysis in AIS patients.
Collapse
Affiliation(s)
- Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa School of Medicine. IRCCS Azienda Ospedaliera Universitaria San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, 6 viale Benedetto XV, 16132 Genoa, Italy; Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva, 64 avenue de la Roseraie, 1211 Geneva, Switzerland
| | - Nicolas Vuilleumier
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, 4 rue Gabrielle-Perret-Gentil, 1205 Geneva, Switzerland
| | - Maria Bertolotto
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa School of Medicine. IRCCS Azienda Ospedaliera Universitaria San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Fabienne Burger
- Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva, 64 avenue de la Roseraie, 1211 Geneva, Switzerland
| | - Katia Galan
- Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva, 64 avenue de la Roseraie, 1211 Geneva, Switzerland
| | - Gloria Roversi
- Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Italy
| | - Carmine Tamborino
- Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Italy
| | - Ilaria Casetta
- Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Italy
| | - Silva Seraceni
- Section of Infectious Diseases, Department of Medical Sciences, University of Ferrara, Italy
| | - Alessandro Trentini
- Section of Medical Biochemistry, Molecular Biology and Genetics, Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Italy
| | - Franco Dallegri
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa School of Medicine. IRCCS Azienda Ospedaliera Universitaria San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Analina Raquel da Silva
- Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva, 64 avenue de la Roseraie, 1211 Geneva, Switzerland
| | - Aldo Pende
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa School of Medicine. IRCCS Azienda Ospedaliera Universitaria San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Nathan Artom
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa School of Medicine. IRCCS Azienda Ospedaliera Universitaria San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - François Mach
- Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva, 64 avenue de la Roseraie, 1211 Geneva, Switzerland
| | - Matteo Coen
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Enrico Fainardi
- Neuroradiology Unit, Department of Neurosciences and Rehabilitation, Azienda Ospedaliera-Universitaria, Arcispedale S. Anna, Via Aldo Moro 8, Cona, Ferrara I-44124, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa School of Medicine. IRCCS Azienda Ospedaliera Universitaria San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, 6 viale Benedetto XV, 16132 Genoa, Italy; Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva, 64 avenue de la Roseraie, 1211 Geneva, Switzerland; Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva, 64 avenue de la Roseraie, 1211 Geneva, Switzerland.
| |
Collapse
|
42
|
Amsalem H, Kwan M, Hazan A, Zhang J, Jones RL, Whittle W, Kingdom JCP, Croy BA, Lye SJ, Dunk CE. Identification of a novel neutrophil population: proangiogenic granulocytes in second-trimester human decidua. THE JOURNAL OF IMMUNOLOGY 2014; 193:3070-9. [PMID: 25135830 DOI: 10.4049/jimmunol.1303117] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The maternal leukocytes of the first-trimester decidua play a fundamental role in implantation and early development of the fetus and placenta, yet little is known regarding the second-trimester decidual environment. Our multicolor flow cytometric analyses of human decidual leukocytes detected an elevation in tissue resident neutrophils in the second trimester. These cells in both human and murine samples were spatially restricted to decidua basalis. In comparison with peripheral blood neutrophils (PMNs), the decidual neutrophils expressed high levels of neutrophil activation markers and the angiogenesis-related proteins: vascular endothelial growth factor-A, Arginase-1, and CCL2, similarly shown in tumor-associated neutrophils. Functional in vitro assays showed that second-trimester human decidua conditioned medium stimulated transendothelial PMN invasion, upregulated VEGFA, ARG1, CCL2, and ICAM1 mRNA levels, and increased PMN-driven in vitro angiogenesis in a CXCL8-dependent manner. This study identified a novel neutrophil population with a physiological, angiogenic role in human decidua.
Collapse
Affiliation(s)
- Hagai Amsalem
- Research Centre for Women's and Infants' Health, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5T 3H7, Canada; Department of Obstetrics and Gynecology, Hadassah-Hebrew University Medical Center, Mt. Scopus, Jerusalem 91120, Israel
| | - Melissa Kwan
- Research Centre for Women's and Infants' Health, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5T 3H7, Canada; Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Aleah Hazan
- Research Centre for Women's and Infants' Health, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5T 3H7, Canada; Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Jianhong Zhang
- Research Centre for Women's and Infants' Health, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5T 3H7, Canada
| | - Rebecca L Jones
- Maternal and Fetal Health Research Centre, St. Mary's Hospital, The University of Manchester, Manchester M13 9WL, United Kingdom
| | - Wendy Whittle
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5G 1E2, Canada; and
| | - John C P Kingdom
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5G 1E2, Canada; and
| | - B Anne Croy
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Stephen J Lye
- Research Centre for Women's and Infants' Health, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5T 3H7, Canada; Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada; Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5G 1E2, Canada; and
| | - Caroline E Dunk
- Research Centre for Women's and Infants' Health, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5T 3H7, Canada; Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5G 1E2, Canada; and
| |
Collapse
|
43
|
Bertolotto M, Contini P, Ottonello L, Pende A, Dallegri F, Montecucco F. Neutrophil migration towards C5a and CXCL8 is prevented by non-steroidal anti-inflammatory drugs via inhibition of different pathways. Br J Pharmacol 2014; 171:3376-3393. [PMID: 24597536 PMCID: PMC4105927 DOI: 10.1111/bph.12670] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 02/25/2014] [Accepted: 02/28/2014] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND AND PURPOSE Non-steroidal anti-inflammatory drugs (NSAIDs) have been shown to induce PG-independent anti-inflammatory actions. Here, we investigated the role of three different NSAIDs (naproxen, ibuprofen and oxaprozin) on neutrophil responses to CXCL8 and C5a. EXPERIMENTAL APPROACH Human neutrophils were isolated from healthy volunteers by dextran and Ficoll-Hypaque density gradients. Neutrophils were pre-incubated with different concentrations (1-100 µM) of NSAIDs or kinase inhibitors. Neutrophil degranulation into supernatants was tested by elisa and zymography. Neutrophil chemotaxis was determined using Boyden chambers. F-actin polymerization was determined by Alexa-Fluor 488-conjugated phalloidin fluorescent assay. Integrin expression was assessed by flow cytometry. The phosphorylation of intracellular kinases was studied by Western blot. KEY RESULTS Pretreatment with NSAIDs did not affect neutrophil degranulation, but inhibited neutrophil migration and polymerization of F-actin, in response to CXCL8 and C5a. Pretreatment with different NSAIDs prevented C5a-induced integrin (CD11b) up-regulation, while only ibuprofen reduced CXCL8-induced CD11b up-regulation. Pre-incubation with naproxen or oxaprozin, but not ibuprofen, inhibited the PI3K/Akt-dependent chemotactic pathways. Both endogenous (released in cell supernatants) or exogenous (added to cell cultures) PGE2 did not affect C5a- or CXCL8-induced activities. Short-term incubation with NSAIDs did not affect neutrophil PGE2 release. CONCLUSION AND IMPLICATIONS Treatment with NSAIDs reduced C5a- and CXCL8-induced neutrophil migration and F-actin polymerization via different mechanisms. Inhibition by ibuprofen was associated with integrin down-regulation, while naproxen and oxaprozin blocked the PI3K/Akt pathway. Both NSAID actions were independent of COX inhibition and PGE2 release.
Collapse
Affiliation(s)
- Maria Bertolotto
- First Clinic of Internal Medicine, Department of Internal Medicine, University of GenoaGenoa, Italy
| | - Paola Contini
- First Clinic of Internal Medicine, Department of Internal Medicine, University of GenoaGenoa, Italy
| | - Luciano Ottonello
- First Clinic of Internal Medicine, Department of Internal Medicine, University of GenoaGenoa, Italy
| | - Aldo Pende
- First Clinic of Internal Medicine, Department of Internal Medicine, University of GenoaGenoa, Italy
| | - Franco Dallegri
- First Clinic of Internal Medicine, Department of Internal Medicine, University of GenoaGenoa, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of GenoaGenoa, Italy
- Division of Cardiology, Foundation for Medical Researches, University of GenevaGeneva, Switzerland
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University HospitalsGeneva, Switzerland
| |
Collapse
|
44
|
Cox N, Pilling D, Gomer RH. Serum amyloid P: a systemic regulator of the innate immune response. J Leukoc Biol 2014; 96:739-43. [PMID: 24804675 DOI: 10.1189/jlb.1mr0114-068r] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The pentraxin SAP reduces neutrophil adhesion to ECM proteins, inhibits the differentiation of monocytes into fibrocytes, attenuates profibrotic macrophages, activates the complement pathway, and promotes phagocytosis of cell debris. Together, these effects of SAP regulate key aspects of inflammation and set a threshold for immune cell activation. Here, we present a review of SAP biology with an emphasis on SAP receptor interactions and how the effect of SAP on monocytes and macrophages has been explored to develop this protein as a therapeutic for renal and lung injuries. We also discuss how there remain many unanswered questions about the role of SAP in innate immunity.
Collapse
Affiliation(s)
- Nehemiah Cox
- Department of Biology, Texas A&M University, College Station, Texas, USA
| | - Darrell Pilling
- Department of Biology, Texas A&M University, College Station, Texas, USA
| | - Richard H Gomer
- Department of Biology, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
45
|
Chen CC, Chen LL, Hsu YT, Liu KJ, Fan CS, Huang TS. The endothelin-integrin axis is involved in macrophage-induced breast cancer cell chemotactic interactions with endothelial cells. J Biol Chem 2014; 289:10029-44. [PMID: 24550382 DOI: 10.1074/jbc.m113.528406] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Elevated macrophage infiltration in tumor tissues is associated with breast cancer metastasis. Cancer cell migration/invasion toward angiogenic microvasculature is a key step in metastatic spread. We therefore studied how macrophages stimulated breast cancer cell interactions with endothelial cells. Macrophages produced cytokines, such as interleukin-8 and tumor necrosis factor-α, to stimulate endothelin (ET) and ET receptor (ETR) expression in breast cancer cells and human umbilical vascular endothelial cells (HUVECs). ET-1 was induced to a greater extent from HUVECs than from breast cancer cells, resulting in a density difference that facilitated cancer cell chemotaxis toward HUVECs. Macrophages also stimulated breast cancer cell adhesion to HUVECs and transendothelial migration, which were repressed by ET-1 antibody or ETR inhibitors. The ET axis induced integrins, such as αV and β1, and their counterligands, such as intercellular adhesion molecule-2 and P-selectin, in breast cancer cells and HUVECs, and antibodies against these integrins efficiently suppressed macrophage-stimulated breast cancer cell interactions with HUVECs. ET-1 induced Ets-like kinase-1 (Elk-1), signal transducer and activator of transcription-3 (STAT-3), and nuclear factor-κB (NF-κB) phosphorylation in breast cancer cells. The use of inhibitors to prevent their phosphorylation or ectopic overexpression of dominant-negative IκBα perturbed ET-1-induced integrin αV and integrin β1 expression. The physical associations of these three transcriptional factors with the gene promoters of the two integrins were furthermore evidenced by a chromatin immunoprecipitation assay. Finally, our mouse orthotopic tumor model revealed an ET axis-mediated lung metastasis of macrophage-stimulated breast cancer cells, suggesting that the ET axis was involved in macrophage-enhanced breast cancer cell endothelial interactions.
Collapse
Affiliation(s)
- Chia-Chi Chen
- From the National Institute of Cancer Research, National Health Research Institutes, No. 35, Keyan Road, Zhunan Town, Miaoli 350, Taiwan
| | | | | | | | | | | |
Collapse
|
46
|
Djokic J, Ninkov M, Mirkov I, Popov Aleksandrov A, Zolotarevski L, Kataranovski D, Kataranovski M. Differential effects of cadmium administration on peripheral blood granulocytes in rats. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2014; 37:210-219. [PMID: 24361699 DOI: 10.1016/j.etap.2013.11.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 11/22/2013] [Accepted: 11/28/2013] [Indexed: 06/03/2023]
Abstract
Infiltration of circulatory inflammatory cells is a common histopathological finding in target organs following cadmium administration, but there is paucity of data concerning their activity. In this study, the effects of sublethal (1 mg/kg) cadmium on peripheral blood polymorphonuclear (PMN) cells were examined 48 h following administration in rats, when tissue (liver and lung) infiltration of these cells was observed. Cadmium administration resulted in systemic inflammatory cytokine and acute phase response with an increase in circulatory neutrophil numbers and cells that express CD11b molecules. Rise in basic aspects of oxidative activity including intracellular myeloperoxidase (MPO), reactive oxygen (nitroblue tetrazolium/NBT cytochemical assay) and nitrogen (Griess assay) species production was observed in PMNs from cadmium-administered rats. A decrease in levels of mRNA for IL-1β, TNF-α and IL-6 was noted, but production of these cytokines was affected differentially. Described effects of cadmium on PMNs add further to the understanding of inflammatory potential of this environmental contaminant.
Collapse
Affiliation(s)
- J Djokic
- Department of Ecology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Bulevar Despota Stefana 142, 11000 Belgrade, Serbia
| | - M Ninkov
- Department of Ecology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Bulevar Despota Stefana 142, 11000 Belgrade, Serbia
| | - I Mirkov
- Department of Ecology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Bulevar Despota Stefana 142, 11000 Belgrade, Serbia
| | - A Popov Aleksandrov
- Department of Ecology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Bulevar Despota Stefana 142, 11000 Belgrade, Serbia
| | - L Zolotarevski
- Institute of Pathology, Military Medical Academy, University of Belgrade, Crnotravska 17, 11000 Belgrade, Serbia
| | - D Kataranovski
- Department of Ecology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Bulevar Despota Stefana 142, 11000 Belgrade, Serbia; Institute of Zoology, Faculty of Biology, University of Belgrade, Studentski trg 16, 11000 Belgrade, Serbia
| | - M Kataranovski
- Department of Ecology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Bulevar Despota Stefana 142, 11000 Belgrade, Serbia; Institute of Physiology and Biochemistry, Faculty of Biology, University of Belgrade, Studentski trg 16, 11000 Belgrade, Serbia.
| |
Collapse
|
47
|
Wang J, Tian Y, Phillips KLE, Chiverton N, Haddock G, Bunning RA, Cross AK, Shapiro IM, Le Maitre CL, Risbud MV. Tumor necrosis factor α- and interleukin-1β-dependent induction of CCL3 expression by nucleus pulposus cells promotes macrophage migration through CCR1. ACTA ACUST UNITED AC 2013; 65:832-42. [PMID: 23233369 DOI: 10.1002/art.37819] [Citation(s) in RCA: 138] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Accepted: 11/29/2012] [Indexed: 11/10/2022]
Abstract
OBJECTIVE To investigate tumor necrosis factor α (TNFα) and interleukin-1β (IL-1β) regulation of CCL3 expression in nucleus pulposus (NP) cells and in macrophage migration. METHODS Quantitative reverse transcription-polymerase chain reaction and immunohistochemistry were used to measure CCL3 expression in NP cells. Transfections were used to determine the role of NF-κB, CCAAT/enhancer binding protein (C/EBPβ), and MAPK on cytokine-mediated CCL3 promoter activity. The effect of NP-conditioned medium on macrophage migration was measured using a Transwell system. RESULTS An increase in CCL3 expression and promoter activity was observed in NP cells after TNFα or IL-1β treatment. Treatment of cells with NF-κB and MAPK inhibitors abolished the effect of the cytokines on CCL3 expression. The inductive effect of p65 and C/EBPβ on the CCL3 promoter was confirmed through gain-of-function and loss-of-function studies. Notably, cotransfection with p50 completely blocked cytokine- and p65-dependent induction. In contrast, c-Rel and RelB had little effect on promoter activity. Lentiviral transduction with short hairpin RNA for p65 (shp65) and shIKKβ significantly decreased the TNFα-dependent increase in CCL3 expression. Analysis of degenerated human NP tissue samples showed that CCL3, but not CCL4, expression correlated positively with the grade of tissue degeneration. Importantly, treatment of macrophages with conditioned medium of NP cells treated with TNFα or IL-1β promoted their migration. Pretreatment of macrophages with an antagonist of CCR1, the primary receptor for CCL3 and CCL4, blocked cytokine-mediated migration. CONCLUSION Our findings indicate that TNFα and IL-1β modulate the expression of CCL3 in NP cells by controlling the activation of MAPK, NF-κB, and C/EBPβ signaling. The CCL3-CCR1 axis may play an important role in promoting macrophage infiltration in degenerated, herniated discs.
Collapse
Affiliation(s)
- Jianru Wang
- Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Mechanisms of adhesion and subsequent actions of a haematopoietic stem cell line, HPC-7, in the injured murine intestinal microcirculation in vivo. PLoS One 2013; 8:e59150. [PMID: 23554986 PMCID: PMC3595270 DOI: 10.1371/journal.pone.0059150] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 02/11/2013] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES Although haematopoietic stem cells (HSCs) migrate to injured gut, therapeutic success clinically remains poor. This has been partially attributed to limited local HSC recruitment following systemic injection. Identifying site specific adhesive mechanisms underpinning HSC-endothelial interactions may provide important information on how to enhance their recruitment and thus potentially improve therapeutic efficacy. This study determined (i) the integrins and inflammatory cyto/chemokines governing HSC adhesion to injured gut and muscle (ii) whether pre-treating HSCs with these cyto/chemokines enhanced their adhesion and (iii) whether the degree of HSC adhesion influenced their ability to modulate leukocyte recruitment. METHODS Adhesion of HPC-7, a murine HSC line, to ischaemia-reperfused (IR) injured mouse gut or cremaster muscle was monitored intravitally. Critical adhesion molecules were identified by pre-treating HPC-7 with blocking antibodies to CD18 and CD49d. To identify cyto/chemokines capable of recruiting HPC-7, adhesion was monitored following tissue exposure to TNF-α, IL-1β or CXCL12. The effects of pre-treating HPC-7 with these cyto/chemokines on surface integrin expression/clustering, adhesion to ICAM-1/VCAM-1 and recruitment in vivo was also investigated. Endogenous leukocyte adhesion following HPC-7 injection was again determined intravitally. RESULTS IR injury increased HPC-7 adhesion in vivo, with intestinal adhesion dependent upon CD18 and muscle adhesion predominantly relying on CD49d. Only CXCL12 pre-treatment enhanced HPC-7 adhesion within injured gut, likely by increasing CD18 binding to ICAM-1 and/or CD18 surface clustering on HPC-7. Leukocyte adhesion was reduced at 4 hours post-reperfusion, but only when local HPC-7 adhesion was enhanced using CXCL12. CONCLUSION This data provides evidence that site-specific molecular mechanisms govern HPC-7 adhesion to injured tissue. Importantly, we show that HPC-7 adhesion is a modulatable event in IR injury and further demonstrate that adhesion instigated by injury alone is not sufficient for mediating anti-inflammatory effects. Enhancing local HSC presence may therefore be essential to realising their clinical potential.
Collapse
|
49
|
Pharmacological inhibition of p38 mitogen-activated protein kinases affects KC/CXCL1-induced intraluminal crawling, transendothelial migration, and chemotaxis of neutrophils in vivo. Mediators Inflamm 2013; 2013:290565. [PMID: 23533303 PMCID: PMC3603207 DOI: 10.1155/2013/290565] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Revised: 01/15/2013] [Accepted: 01/29/2013] [Indexed: 01/26/2023] Open
Abstract
p38 mitogen-activated protein kinase (MAPK) signalling is critical in the pathophysiology of a variety of inflammatory processes. Leukocyte recruitment to the site of inflammation is a multistep process governed by specific signalling cascades. After adhesion in the lumen, many leukocytes crawl to optimal sites at endothelial junctions and transmigrate to extravascular tissue in a Mac-1-dependent manner. The signalling mechanisms that regulate postadhesion steps of intraluminal crawling, transmigration, and chemotaxis in tissue remain incompletely understood. The present study explored the effect of p38 MAPK inhibitor SB203580 on various parameters of neutrophil recruitment triggered by chemokine KC (CXCL1) gradient. Neutrophil-endothelial interactions in microvasculature of murine cremaster muscle were determined using intravital microscopy and time-lapsed video analysis. SB203580 (100 nM) did not change leukocyte rolling but significantly attenuated neutrophil adhesion, emigration, and transmigration and impaired the initiation of neutrophil crawling and transmigration. In response to KC chemotactic gradient, SB203580 significantly reduced the velocity of migration and chemotaxis index of neutrophils in tissue. The upregulation of Mac-1 expression in neutrophils stimulated by KC was significantly blunted by SB203580 in vitro. Collectively, our findings demonstrate that pharmacological suppression of p38 MAPK significantly impairs multiple steps of neutrophil recruitment in vivo.
Collapse
|
50
|
Zachman AL, Page JM, Prabhakar G, Guelcher SA, Sung HJ. Elucidation of adhesion-dependent spontaneous apoptosis in macrophages using phase separated PEG/polyurethane films. Acta Biomater 2013; 9:4964-75. [PMID: 23128157 DOI: 10.1016/j.actbio.2012.10.038] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 10/03/2012] [Accepted: 10/29/2012] [Indexed: 01/24/2023]
Abstract
Circulating monocytes undergo spontaneous apoptosis when there is no activation stimulus, which is critical to population control for proper host response to implants. As activation and apoptosis of monocytes/macrophages are regulated by cell-cell and cell-matrix interactions, their regulatory mechanism was investigated in this study using polyethylene glycol (PEG)-containing polyurethane films in which PEG-rich and polyester-rich domains were phase separated. Human blood monocyte-derived macrophages (HBMs) preferentially adhered to PEG domains (cell-matrix interaction) due to the low molecular weight (600 g mol⁻¹), resulting in increased HBM density (cell-cell interaction). As both cell-cell and cell-matrix interactions were promoted, HBM apoptosis increased, while their activation as measured by phagocytosis, intracellular reactive oxygen species (ROS) level and matrix metalloproteinase-9 production decreased compared to PEG-free films. When cell seeding density and cell-adhesive gelatin coating on silicone films were controlled, a cooperative role of cell-matrix (adhesion) and cell-cell (density) interactions in inducing HBM apoptosis was observed. Expression of the macrophage adhesion molecule CD11b caused apoptosis in this context, which was mediated by tissue necrosis factor-α signaling but down-regulated by the ROS inhibitor diphenylene iodonium and the anti-inflammatory peptide Ac-SDKP, suggesting a new concept for the design of biomaterials that allows for cell adhesion without excessive inflammatory activation.
Collapse
|