1
|
Razim A, Zabłocka A, Schmid A, Thaler M, Černý V, Weinmayer T, Whitehead B, Martens A, Skalska M, Morandi M, Schmidt K, Wysmołek ME, Végvári A, Srutkova D, Schwarzer M, Neuninger L, Nejsum P, Hrdý J, Palmfeldt J, Brucale M, Valle F, Górska S, Wisgrill L, Inic‐Kanada A, Wiedermann U, Schabussova I. Bacterial extracellular vesicles as intranasal postbiotics: Detailed characterization and interaction with airway cells. J Extracell Vesicles 2024; 13:e70004. [PMID: 39429019 PMCID: PMC11491762 DOI: 10.1002/jev2.70004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 08/23/2024] [Accepted: 09/29/2024] [Indexed: 10/22/2024] Open
Abstract
Escherichia coli A0 34/86 (EcO83) is a probiotic strain used in newborns to prevent nosocomial infections and diarrhoea. This bacterium stimulates both pro- and anti-inflammatory cytokine production and its intranasal administration reduces allergic airway inflammation in mice. Despite its benefits, there are concerns about the use of live probiotic bacteria due to potential systemic infections and gene transfer. Extracellular vesicles (EVs) derived from EcO83 (EcO83-EVs) might offer a safer alternative to live bacteria. This study characterizes EcO83-EVs and investigates their interaction with host cells, highlighting their potential as postbiotic therapeutics. EcO83-EVs were isolated, purified, and characterised following the Minimal Information of Studies of Extracellular Vesicles (MISEV) guidelines. Ex vivo studies conducted in human nasal epithelial cells showed that EcO83-EVs increased the expression of proteins linked to oxidative stress and inflammation, indicating an effective interaction between EVs and the host cells. Further in vivo studies in mice demonstrated that EcO83-EVs interact with nasal-associated lymphoid tissue, are internalised by airway macrophages, and stimulate neutrophil recruitment in the lung. Mechanistically, EcO83-EVs activate the NF-κΒ signalling pathway, resulting in the nitric oxide production. EcO83-EVs demonstrate significant potential as a postbiotic alternative to live bacteria, offering a safer option for therapeutic applications. Further research is required to explore their clinical use, particularly in mucosal vaccination and targeted immunotherapy strategies.
Collapse
Affiliation(s)
- Agnieszka Razim
- Institute of Specific Prophylaxis and Tropical Medicine, Centre for PathophysiologyInfectiology and Immunology, Medical University of ViennaViennaAustria
| | - Agnieszka Zabłocka
- Hirszfeld Institute of Immunology and Experimental TherapyPolish Academy of SciencesWroclawPoland
| | - Anna Schmid
- Institute of Specific Prophylaxis and Tropical Medicine, Centre for PathophysiologyInfectiology and Immunology, Medical University of ViennaViennaAustria
| | - Michael Thaler
- Institute of Specific Prophylaxis and Tropical Medicine, Centre for PathophysiologyInfectiology and Immunology, Medical University of ViennaViennaAustria
| | - Viktor Černý
- Institute of Specific Prophylaxis and Tropical Medicine, Centre for PathophysiologyInfectiology and Immunology, Medical University of ViennaViennaAustria
| | - Tamara Weinmayer
- Institute of Specific Prophylaxis and Tropical Medicine, Centre for PathophysiologyInfectiology and Immunology, Medical University of ViennaViennaAustria
| | - Bradley Whitehead
- Department of Infectious DiseasesAarhus University HospitalAarhusDenmark
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Anke Martens
- Division of Neonatology, Paediatric Intensive Care and Neuropediatric, Department of Paediatrics and Adolescent Medicine, Comprehensive Centre for PaediatricsMedical University of ViennaViennaAustria
| | - Magdalena Skalska
- Department of Medical Physics, M. Smoluchowski Institute of Physics, Faculty of PhysicsAstronomy and Applied Computer Science, Jagiellonian UniversityKrakowPoland
| | - Mattia Morandi
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of SciencePragueCzech Republic
| | - Katy Schmidt
- Research Support Facilities, Imaging Unit CIUS, Faculty of Life SciencesUniversity of ViennaViennaAustria
| | - Magdalena E. Wysmołek
- Institute of Specific Prophylaxis and Tropical Medicine, Centre for PathophysiologyInfectiology and Immunology, Medical University of ViennaViennaAustria
| | - Akos Végvári
- Proteomics Biomedicum, Division of Chemistry I, Department of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSweden
- Division of Chemistry I, Department of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSweden
| | - Dagmar Srutkova
- Laboratory of GnotobiologyInstitute of Microbiology of the Czech Academy of SciencesNovy HradekCzech Republic
| | - Martin Schwarzer
- Laboratory of GnotobiologyInstitute of Microbiology of the Czech Academy of SciencesNovy HradekCzech Republic
| | - Lukas Neuninger
- Institute of Specific Prophylaxis and Tropical Medicine, Centre for PathophysiologyInfectiology and Immunology, Medical University of ViennaViennaAustria
| | - Peter Nejsum
- Department of Infectious DiseasesAarhus University HospitalAarhusDenmark
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Jiri Hrdý
- Institute of Immunology and Microbiology, First Faculty of MedicineCharles University and General University HospitalPragueCzech Republic
| | - Johan Palmfeldt
- Research Unit for Molecular Medicine, Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Marco Brucale
- Institute of Nanostructured MaterialsCNR‐ISMNBolognaItaly
| | | | - Sabina Górska
- Hirszfeld Institute of Immunology and Experimental TherapyPolish Academy of SciencesWroclawPoland
| | - Lukas Wisgrill
- Division of Neonatology, Paediatric Intensive Care and Neuropediatric, Department of Paediatrics and Adolescent Medicine, Comprehensive Centre for PaediatricsMedical University of ViennaViennaAustria
| | - Aleksandra Inic‐Kanada
- Institute of Specific Prophylaxis and Tropical Medicine, Centre for PathophysiologyInfectiology and Immunology, Medical University of ViennaViennaAustria
| | - Ursula Wiedermann
- Institute of Specific Prophylaxis and Tropical Medicine, Centre for PathophysiologyInfectiology and Immunology, Medical University of ViennaViennaAustria
| | - Irma Schabussova
- Institute of Specific Prophylaxis and Tropical Medicine, Centre for PathophysiologyInfectiology and Immunology, Medical University of ViennaViennaAustria
| |
Collapse
|
2
|
Yang PY, Tsaur ML. NS5806 reduces carrageenan-evoked inflammation by suppressing extracellular signal-regulated kinase activation in primary sensory neurons and immune cells. Eur J Pain 2023; 27:927-939. [PMID: 37172202 DOI: 10.1002/ejp.2123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 02/23/2023] [Accepted: 04/27/2023] [Indexed: 05/14/2023]
Abstract
BACKGROUND The compound NS5806 attenuates neuropathic pain via inhibiting extracellular signal-regulated kinase (ERK) activation in neuronal somata located at the dorsal root ganglion (DRG) and superficial spinal dorsal horn. NS5806 also reduces the expansion of DRG macrophages and spinal microglia several days after peripheral nerve injury, implying an anti-inflammatory effect. METHODS To test whether NS5806 inhibits inflammation, as a model we intraplantarly injected carrageenan into a hind paw of the rat. To examine whether NS5806 reduces carrageenan-evoked mechanical allodynia, thermal hyperalgesia, and edema, as well as ERK activation in the nerve fibres, mast cells, and macrophages in the hind paw skin, we used behavioural, immunohistochemical, and cytological methods. RESULTS NS5806 did not impair motor function, affect basal nociception, or cause edema in naive rats. Six hours after carrageenan injection, mechanical allodynia, thermal hyperalgesia, and edema appeared in the rat's ipsilateral hind paw, and all were reduced by intraplantar co-injection of NS5806. NS5806 suppressed carrageenan-evoked ERK activation in the peripheral axons and somata of L4 DRG neurons, as well as mast cells and macrophages in the paw skin. NS5806 also reduced carrageenan-evoked mast cell degranulation and macrophage proliferation. NS5806 and the ERK pathway inhibitor PD98059 had a similar effect in inhibiting the proliferation of cultured RAW264.7 macrophages. Furthermore, all the in vivo anti-inflammatory effects of NS5806 were similar to those of PD98059. CONCLUSIONS Acting like an ERK pathway inhibitor, NS5806 reduces inflammation-evoked mechanical allodynia, thermal hyperalgesia, and edema by suppressing ERK activation in primary sensory neurons, mast cells, and macrophages. SIGNIFICANCE Previous studies show that NS5806 only acts on neurons. This report unveils that NS5806 also acts on immune cells in the skin to exert its anti-inflammatory effects. Since NS5806 is lipid soluble for skin penetration, it suggests that NS5806 could also be developed into an anti-inflammatory drug for external use.
Collapse
Affiliation(s)
- Po-Yu Yang
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Meei-Ling Tsaur
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
3
|
Arpa L, Batlle C, Jiang P, Caelles C, Lloberas J, Celada A. Distinct Responses to IL4 in Macrophages Mediated by JNK. Cells 2023; 12:cells12081127. [PMID: 37190036 DOI: 10.3390/cells12081127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/20/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
IL(Interleukin)-4 is the main macrophage M2-type activator and induces an anti-inflammatory phenotype called alternative activation. The IL-4 signaling pathway involves the activation of STAT (Signal Transducer and Activator of Transcription)-6 and members of the MAPK (Mitogen-activated protein kinase) family. In primary-bone-marrow-derived macrophages, we observed a strong activation of JNK (Jun N-terminal kinase)-1 at early time points of IL-4 stimulation. Using selective inhibitors and a knockout model, we explored the contribution of JNK-1 activation to macrophages' response to IL-4. Our findings indicate that JNK-1 regulates the IL-4-mediated expression of genes typically involved in alternative activation, such as Arginase 1 or Mannose receptor, but not others, such as SOCS (suppressor of cytokine signaling) 1 or p21Waf-1 (cyclin dependent kinase inhibitor 1A). Interestingly, we have observed that after macrophages are stimulated with IL-4, JNK-1 has the capacity to phosphorylate STAT-6 on serine but not on tyrosine. Chromatin immunoprecipitation assays revealed that functional JNK-1 is required for the recruitment of co-activators such as CBP (CREB-binding protein)/p300 on the promoter of Arginase 1 but not on p21Waf-1. Taken together, these data demonstrate the critical role of STAT-6 serine phosphorylation by JNK-1 in distinct macrophage responses to IL-4.
Collapse
Affiliation(s)
- Luís Arpa
- Biology of Macrophages Group, Department of Cellular Biology, Physiology and Immunology, Universitat de Barcelona, 08007 Barcelona, Spain
| | - Carlos Batlle
- Biology of Macrophages Group, Department of Cellular Biology, Physiology and Immunology, Universitat de Barcelona, 08007 Barcelona, Spain
| | - Peijin Jiang
- Biology of Macrophages Group, Department of Cellular Biology, Physiology and Immunology, Universitat de Barcelona, 08007 Barcelona, Spain
| | - Carme Caelles
- Institute of Biomedicine, Universitat de Barcelona (IBUB), 08028 Barcelona, Spain
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Jorge Lloberas
- Biology of Macrophages Group, Department of Cellular Biology, Physiology and Immunology, Universitat de Barcelona, 08007 Barcelona, Spain
| | - Antonio Celada
- Biology of Macrophages Group, Department of Cellular Biology, Physiology and Immunology, Universitat de Barcelona, 08007 Barcelona, Spain
| |
Collapse
|
4
|
Filiberti S, Russo M, Lonardi S, Bugatti M, Vermi W, Tournier C, Giurisato E. Self-Renewal of Macrophages: Tumor-Released Factors and Signaling Pathways. Biomedicines 2022; 10:2709. [PMID: 36359228 PMCID: PMC9687165 DOI: 10.3390/biomedicines10112709] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/14/2022] [Accepted: 10/21/2022] [Indexed: 04/11/2024] Open
Abstract
Macrophages are the most abundant immune cells of the tumor microenvironment (TME) and have multiple important functions in cancer. During tumor growth, both tissue-resident macrophages and newly recruited monocyte-derived macrophages can give rise to tumor-associated macrophages (TAMs), which have been associated with poor prognosis in most cancers. Compelling evidence indicate that the high degree of plasticity of macrophages and their ability to self-renew majorly impact tumor progression and resistance to therapy. In addition, the microenvironmental factors largely affect the metabolism of macrophages and may have a major influence on TAMs proliferation and subsets functions. Thus, understanding the signaling pathways regulating TAMs self-renewal capacity may help to identify promising targets for the development of novel anticancer agents. In this review, we focus on the environmental factors that promote the capacity of macrophages to self-renew and the molecular mechanisms that govern TAMs proliferation. We also highlight the impact of tumor-derived factors on macrophages metabolism and how distinct metabolic pathways affect macrophage self-renewal.
Collapse
Affiliation(s)
- Serena Filiberti
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Mariapia Russo
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Silvia Lonardi
- Department of Molecular and Translational Medicine, University of Brescia, 25100 Brescia, Italy
| | - Mattia Bugatti
- Department of Molecular and Translational Medicine, University of Brescia, 25100 Brescia, Italy
| | - William Vermi
- Department of Molecular and Translational Medicine, University of Brescia, 25100 Brescia, Italy
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Cathy Tournier
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK
| | - Emanuele Giurisato
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK
| |
Collapse
|
5
|
Micallef P, Vujičić M, Wu Y, Peris E, Wang Y, Chanclón B, Ståhlberg A, Cardell SL, Wernstedt Asterholm I. C1QTNF3 is Upregulated During Subcutaneous Adipose Tissue Remodeling and Stimulates Macrophage Chemotaxis and M1-Like Polarization. Front Immunol 2022; 13:914956. [PMID: 35720277 PMCID: PMC9202579 DOI: 10.3389/fimmu.2022.914956] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/06/2022] [Indexed: 01/08/2023] Open
Abstract
The adipose tissue undergoes substantial tissue remodeling during weight gain-induced expansion as well as in response to the mechanical and immunological stresses from a growing tumor. We identified the C1q/TNF-related protein family member C1qtnf3 as one of the most upregulated genes that encode secreted proteins in tumor-associated inguinal adipose tissue - especially in high fat diet-induced obese mice that displayed 3-fold larger tumors than their lean controls. Interestingly, inguinal adipose tissue C1qtnf3 was co-regulated with several macrophage markers and chemokines and was primarily expressed in fibroblasts while only low levels were detected in adipocytes and macrophages. Administration of C1QTNF3 neutralizing antibodies inhibited macrophage accumulation in tumor-associated inguinal adipose tissue while tumor growth was unaffected. In line with this finding, C1QTNF3 exerted chemotactic actions on both M1- and M2-polarized macrophages in vitro. Moreover, C1QTNF3 treatment of M2-type macrophages stimulated the ERK and Akt pathway associated with increased M1-like polarization as judged by increased expression of M1-macrophage markers, increased production of nitric oxide, reduced oxygen consumption and increased glycolysis. Based on these results, we propose that macrophages are recruited to adipose tissue sites with increased C1QTNF3 production. However, the impact of the immunomodulatory effects of C1QTNF3 in adipose tissue remodeling warrants future investigations.
Collapse
Affiliation(s)
- Peter Micallef
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Göteborg, Sweden
| | - Milica Vujičić
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Göteborg, Sweden
| | - Yanling Wu
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Göteborg, Sweden
| | - Eduard Peris
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Göteborg, Sweden
| | - Ying Wang
- Department of Microbiology and Immunology, Institute of Biomedicine, The Sahlgrenska Academy at University of Gothenburg, Göteborg, Sweden
| | - Belén Chanclón
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Göteborg, Sweden
| | - Anders Ståhlberg
- Sahlgrenska Center for Cancer Research, Department of Laboratory Medicine, Institute of Biomedicine, The Sahlgrenska Academy at University of Gothenburg, Göteborg, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Göteborg, Sweden.,Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Göteborg, Sweden
| | - Susanna L Cardell
- Department of Microbiology and Immunology, Institute of Biomedicine, The Sahlgrenska Academy at University of Gothenburg, Göteborg, Sweden
| | - Ingrid Wernstedt Asterholm
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Göteborg, Sweden
| |
Collapse
|
6
|
Balestrieri B, Granata F, Loffredo S, Petraroli A, Scalia G, Morabito P, Cardamone C, Varricchi G, Triggiani M. Phenotypic and Functional Heterogeneity of Low-Density and High-Density Human Lung Macrophages. Biomedicines 2021; 9:biomedicines9050505. [PMID: 34064389 PMCID: PMC8147777 DOI: 10.3390/biomedicines9050505] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Pulmonary macrophages are a highly heterogeneous cell population distributed in different lung compartments. METHODS We separated two subpopulations of macrophages from human lung parenchyma according to flotation over density gradients. RESULTS Two-thirds 65.4% of the lung macrophages have a density between 1.065 and 1.078 (high-density macrophages: HDMs), and the remaining one-third (34.6) had a density between 1.039 and 1.052 (low-density macrophages: LDMs). LDMs had a larger area (691 vs. 462 μm2) and cell perimeter (94 vs. 77 μm) compared to HDMs. A significantly higher percentage of HDMs expressed CD40, CD45, and CD86 compared to LDMs. In contrast, a higher percentage of LDMs expressed the activation markers CD63 and CD64. The release of TNF-α, IL-6, IL-10 and IL-12 induced by lipopolysaccharide (LPS) was significantly higher in HDMs than in LDMs. CONCLUSION The human lung contains two subpopulations of macrophages that differ in buoyancy, morphometric parameters, surface marker expression and response to LPS. These subpopulations of macrophages probably play distinct roles in lung inflammation and immune responses.
Collapse
Affiliation(s)
- Barbara Balestrieri
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (S.L.); (A.P.); (G.V.)
- Correspondence: (B.B.); (F.G.)
| | - Francescopaolo Granata
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (S.L.); (A.P.); (G.V.)
- Center of Excellence, World Allergy Organization (WAO), 80131 Naples, Italy
- Correspondence: (B.B.); (F.G.)
| | - Stefania Loffredo
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (S.L.); (A.P.); (G.V.)
- Center of Excellence, World Allergy Organization (WAO), 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, 80131 Naples, Italy
| | - Angelica Petraroli
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (S.L.); (A.P.); (G.V.)
- Center of Excellence, World Allergy Organization (WAO), 80131 Naples, Italy
| | - Giulia Scalia
- Clinical and Experimental Cytometry Unit, CEINGE-Biotecnologie Avanzate, 80131 Naples, Italy;
| | - Paolo Morabito
- Laboratory of Clinical Pathology, A. Cardarelli Hospital, 80131 Naples, Italy;
| | - Chiara Cardamone
- Division of Allergy and Clinical Immunology, University of Salerno, 84084 Fisciano, Italy; (C.C.); (M.T.)
| | - Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (S.L.); (A.P.); (G.V.)
- Center of Excellence, World Allergy Organization (WAO), 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, 80131 Naples, Italy
| | - Massimo Triggiani
- Division of Allergy and Clinical Immunology, University of Salerno, 84084 Fisciano, Italy; (C.C.); (M.T.)
| |
Collapse
|
7
|
Carbó JM, León TE, Font-Díaz J, De la Rosa JV, Castrillo A, Picard FR, Staudenraus D, Huber M, Cedó L, Escolà-Gil JC, Campos L, Bakiri L, Wagner EF, Caelles C, Stratmann T, Van Ginderachter JA, Valledor AF. Pharmacologic Activation of LXR Alters the Expression Profile of Tumor-Associated Macrophages and the Abundance of Regulatory T Cells in the Tumor Microenvironment. Cancer Res 2020; 81:968-985. [PMID: 33361391 DOI: 10.1158/0008-5472.can-19-3360] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 10/29/2020] [Accepted: 12/18/2020] [Indexed: 11/16/2022]
Abstract
Liver X receptors (LXR) are transcription factors from the nuclear receptor family that are activated by oxysterols and synthetic high-affinity agonists. In this study, we assessed the antitumor effects of synthetic LXR agonist TO901317 in a murine model of syngeneic Lewis Lung carcinoma. Treatment with TO901317 inhibited tumor growth in wild-type, but not in LXR-deficient mice, indicating that the antitumor effects of the agonist depends on functional LXR activity in host cells. Pharmacologic activation of the LXR pathway reduced the intratumoral abundance of regulatory T cells (Treg) and the expression of the Treg-attracting chemokine Ccl17 by MHCIIhigh tumor-associated macrophages (TAM). Moreover, gene expression profiling indicated a broad negative impact of the LXR agonist on other mechanisms used by TAM for the maintenance of an immunosuppressive environment. In studies exploring the macrophage response to GM-CSF or IL4, activated LXR repressed IRF4 expression, resulting in subsequent downregulation of IRF4-dependent genes including Ccl17. Taken together, this work reveals the combined actions of the LXR pathway in the control of TAM responses that contribute to the antitumoral effects of pharmacologic LXR activation. Moreover, these data provide new insights for the development of novel therapeutic options for the treatment of cancer. SIGNIFICANCE: This study reveals unrecognized roles of LXR in the transcriptional control of the tumor microenvironment and suggests use of a synthetic LXR agonist as a novel therapeutic strategy to stimulate antitumor activity.
Collapse
Affiliation(s)
- José M Carbó
- Department of Cell Biology, Physiology and Immunology, School of Biology, University of Barcelona, Barcelona, Spain.,Leukaemia Stem Cell Group, Josep Carreras Leukemia Research Institute, Badalona, Spain
| | - Theresa E León
- Department of Cell Biology, Physiology and Immunology, School of Biology, University of Barcelona, Barcelona, Spain.,Department of Haematology, UCL Cancer Institute, University College London, London, United Kingdom
| | - Joan Font-Díaz
- Department of Cell Biology, Physiology and Immunology, School of Biology, University of Barcelona, Barcelona, Spain.,Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
| | - Juan Vladimir De la Rosa
- Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Grupo de Investigación Medio Ambiente y Salud (GIMAS, ULPGC), Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Antonio Castrillo
- Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Grupo de Investigación Medio Ambiente y Salud (GIMAS, ULPGC), Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain.,Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-Universidad Autónoma de Madrid, Madrid, Spain
| | - Felix R Picard
- Institute for Medical Microbiology and Hospital Hygiene, University of Marburg, Marburg, Germany
| | - Daniel Staudenraus
- Institute for Medical Microbiology and Hospital Hygiene, University of Marburg, Marburg, Germany
| | - Magdalena Huber
- Institute for Medical Microbiology and Hospital Hygiene, University of Marburg, Marburg, Germany
| | - Lídia Cedó
- Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Barcelona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Hospitalet de Llobregat, Spain
| | - Joan Carles Escolà-Gil
- Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Barcelona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Hospitalet de Llobregat, Spain
| | - Lucía Campos
- Institute for Medical Microbiology and Hospital Hygiene, University of Marburg, Marburg, Germany.,Departments of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Latifa Bakiri
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Erwin F Wagner
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria.,Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Carme Caelles
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain.,Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
| | - Thomas Stratmann
- Department of Cell Biology, Physiology and Immunology, School of Biology, University of Barcelona, Barcelona, Spain
| | - Jo A Van Ginderachter
- Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium.,Lab of Myeloid Cell Immunology, VIB Center for Inflammation Research, Brussels, Belgium
| | - Annabel F Valledor
- Department of Cell Biology, Physiology and Immunology, School of Biology, University of Barcelona, Barcelona, Spain. .,Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
| |
Collapse
|
8
|
Lee HH, Jang E, Kang SY, Shin JS, Han HS, Kim TW, Lee DH, Lee JH, Jang DS, Lee KT. Anti-inflammatory potential of Patrineolignan B isolated from Patrinia scabra in LPS-stimulated macrophages via inhibition of NF-κB, AP-1, and JAK/STAT pathways. Int Immunopharmacol 2020; 86:106726. [DOI: 10.1016/j.intimp.2020.106726] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 05/31/2020] [Accepted: 06/17/2020] [Indexed: 12/27/2022]
|
9
|
Hüttener M, Prieto A, Aznar S, Bernabeu M, Glaría E, Valledor AF, Paytubi S, Merino S, Tomás J, Juárez A. Expression of a novel class of bacterial Ig-like proteins is required for IncHI plasmid conjugation. PLoS Genet 2019; 15:e1008399. [PMID: 31527905 PMCID: PMC6764697 DOI: 10.1371/journal.pgen.1008399] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 09/27/2019] [Accepted: 09/04/2019] [Indexed: 01/10/2023] Open
Abstract
Antimicrobial resistance (AMR) is currently one of the most important challenges to the treatment of bacterial infections. A critical issue to combat AMR is to restrict its spread. In several instances, bacterial plasmids are involved in the global spread of AMR. Plasmids belonging to the incompatibility group (Inc)HI are widespread in Enterobacteriaceae and most of them express multiple antibiotic resistance determinants. They play a relevant role in the recent spread of colistin resistance. We present in this report novel findings regarding IncHI plasmid conjugation. Conjugative transfer in liquid medium of an IncHI plasmid requires expression of a plasmid-encoded, large-molecular-mass protein that contains an Ig-like domain. The protein, termed RSP, is encoded by a gene (ORF R0009) that maps in the Tra2 region of the IncHI1 R27 plasmid. The RSP protein is exported outside the cell by using the plasmid-encoded type IV secretion system that is also used for its transmission to new cells. Expression of the protein reduces cell motility and enables plasmid conjugation. Flagella are one of the cellular targets of the RSP protein. The RSP protein is required for a high rate of plasmid transfer in both flagellated and nonflagellated Salmonella cells. This effect suggests that RSP interacts with other cellular structures as well as with flagella. These unidentified interactions must facilitate mating pair formation and, hence, facilitate IncHI plasmid conjugation. Due to its location on the outer surfaces of the bacterial cell, targeting the RSP protein could be a means of controlling IncHI plasmid conjugation in natural environments or of combatting infections caused by AMR enterobacteria that harbor IncHI plasmids. Dissemination of antimicrobial resistance (AMR) among different bacterial populations occurs due to mainly the presence of plasmids that encode AMR determinants. IncHI plasmids are one of the groups of bacterial plasmids that confer AMR to several enterobacteria. Recently, resistance to one of the last-resort antibiotics (colistin) for some multidrug-resistant infections has spread very rapidly. IncHI plasmids represent 20% of all plasmids transmitting colistin resistance worldwide and 40% in Europe. When analyzing the interactions of the IncHI1 plasmid R27 with Salmonella, we identified a large-molecular-mass protein that is encoded by this plasmid and is exported to the external medium. The R27 plasmid gene coding for that protein (R0009) is widespread among IncHI plasmids. In this report, we characterize the protein, termed RSP. The presented data show that RSP plays a relevant role in IncHI plasmid conjugation and suggest that the protein is retained on the outer surface of the bacterial cells and facilitates cell-to-cell contact before plasmid DNA transfer. Considering that IncHI plasmids significantly contribute to AMR dissemination within enterobacteria, the findings reported in this paper suggest that the identified protein can be a target to control both IncHI-mediated AMR dissemination and infections caused by AMR enterobacteria that harbor these plasmids.
Collapse
Affiliation(s)
- Mário Hüttener
- Department of Genetics, Microbiology and Statistics, University of Barcelona, Barcelona, Spain
| | - Alejandro Prieto
- Department of Genetics, Microbiology and Statistics, University of Barcelona, Barcelona, Spain
| | - Sonia Aznar
- Department of Genetics, Microbiology and Statistics, University of Barcelona, Barcelona, Spain
| | - Manuel Bernabeu
- Department of Genetics, Microbiology and Statistics, University of Barcelona, Barcelona, Spain
| | - Estibaliz Glaría
- Department of Cell Biology, Physiology and Immunology, University of Barcelona, Barcelona, Spain
| | - Annabel F. Valledor
- Department of Cell Biology, Physiology and Immunology, University of Barcelona, Barcelona, Spain
| | - Sonia Paytubi
- Department of Genetics, Microbiology and Statistics, University of Barcelona, Barcelona, Spain
| | - Susana Merino
- Department of Genetics, Microbiology and Statistics, University of Barcelona, Barcelona, Spain
| | - Joan Tomás
- Department of Genetics, Microbiology and Statistics, University of Barcelona, Barcelona, Spain
| | - Antonio Juárez
- Department of Genetics, Microbiology and Statistics, University of Barcelona, Barcelona, Spain
- Institute for Bioengineering of Catalonia, The Barcelona Institute of Science and Technology, Barcelona, Spain
- * E-mail:
| |
Collapse
|
10
|
An J, Naruse TK, Hinohara K, Soejima Y, Sawabe M, Nakagawa Y, Kuwahara K, Kimura A. MRTF-A regulates proliferation and survival properties of pro-atherogenic macrophages. J Mol Cell Cardiol 2019; 133:26-35. [DOI: 10.1016/j.yjmcc.2019.05.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 04/01/2019] [Accepted: 05/16/2019] [Indexed: 12/12/2022]
|
11
|
Neamatallah T, Jabbar S, Tate R, Schroeder J, Shweash M, Alexander J, Plevin R. Whole Genome Microarray Analysis of DUSP4-Deletion Reveals A Novel Role for MAP Kinase Phosphatase-2 (MKP-2) in Macrophage Gene Expression and Function. Int J Mol Sci 2019; 20:ijms20143434. [PMID: 31336892 PMCID: PMC6679025 DOI: 10.3390/ijms20143434] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 07/08/2019] [Accepted: 07/09/2019] [Indexed: 12/22/2022] Open
Abstract
Background: Mitogen-activated protein kinase phosphatase-2 (MKP-2) is a type 1 nuclear dual specific phosphatase (DUSP-4). It plays an important role in macrophage inflammatory responses through the negative regulation of Mitogen activated protein kinase (MAPK) signalling. However, information on the effect of MKP-2 on other aspect of macrophage function is limited. Methods: We investigated the impact of MKP-2 in the regulation of several genes that are involved in function while using comparative whole genome microarray analysis in macrophages from MKP-2 wild type (wt) and knock out (ko) mice. Results: Our data showed that the lack of MKP-2 caused a significant down-regulation of colony-stimulating factor-2 (Csf2) and monocyte to macrophage-associated differentiation (Mmd) genes, suggesting a role of MKP-2 in macrophage development. When treated with macrophage colony stimulating factor (M-CSF), Mmd and Csf2 mRNA levels increased but significantly reduced in ko cells in comparison to wt counterparts. This effect of MKP-2 deletion on macrophage function was also observed by cell counting and DNA measurements. On the signalling level, M-CSF stimulation induced extracellular signal-regulated kinases (ERK) phosphorylation, which was significantly enhanced in the absence of MKP-2. Pharmacological inhibition of ERK reduced both Csf2 and Mmd genes in both wild type and ko cultures, which suggested that enhanced ERK activation in ko cultures may not explain effects on gene expression. Interestingly other functional markers were also shown to be reduced in ko macrophages in comparison to wt mice; the expression of CD115, which is a receptor for M-CSF, and CD34, a stem/progenitor cell marker, suggesting global regulation of gene expression by MKP-2. Conclusions: Transcriptome profiling reveals that MKP-2 regulates macrophage development showing candidate targets from monocyte-to-macrophage differentiation and macrophage proliferation. However, it is unclear whether effects upon ERK signalling are able to explain the effects of DUSP-4 deletion on macrophage function.
Collapse
Affiliation(s)
- Thikryat Neamatallah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, P.O. Box 80260, Jeddah 21589, Saudi Arabia.
| | - Shilan Jabbar
- Strathclyde Institute for Pharmacy & Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Rothwelle Tate
- Strathclyde Institute for Pharmacy & Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Juliane Schroeder
- Strathclyde Institute for Pharmacy & Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Muhannad Shweash
- Strathclyde Institute for Pharmacy & Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - James Alexander
- Strathclyde Institute for Pharmacy & Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Robin Plevin
- Strathclyde Institute for Pharmacy & Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| |
Collapse
|
12
|
Pfänder P, Fidan M, Burret U, Lipinski L, Vettorazzi S. Cdk5 Deletion Enhances the Anti-inflammatory Potential of GC-Mediated GR Activation During Inflammation. Front Immunol 2019; 10:1554. [PMID: 31354714 PMCID: PMC6635475 DOI: 10.3389/fimmu.2019.01554] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 06/21/2019] [Indexed: 11/13/2022] Open
Abstract
The suppression of activated pro-inflammatory macrophages during immune response has a major impact on the outcome of many inflammatory diseases including sepsis and rheumatoid arthritis. The pro- and anti-inflammatory functions of macrophages have been widely studied, whereas their regulation under immunosuppressive treatments such as glucocorticoid (GC) therapy is less well-understood. GC-mediated glucocorticoid receptor (GR) activation is crucial to mediate anti-inflammatory effects. In addition, the anti-cancer drug roscovitine, that is currently being tested in clinical trials, was recently described to regulate inflammatory processes by inhibiting different Cdks such as cyclin-dependent kinase 5 (Cdk5). Cdk5 was identified as a modulator of inflammatory processes in different immune cells and furthermore described to influence GR gene expression in the brain. Whether roscovitine can enhance the immunosuppressive effects of GCs and if the inhibition of Cdk5 affects GR gene regulatory function in innate immune cells, such as macrophages, has not yet been investigated. Here, we report that roscovitine enhances the immunosuppressive Dexamethasone (Dex) effect on the inducible nitric oxide synthase (iNos) expression, which is essential for immune regulation. Cdk5 deletion in macrophages prevented iNos protein and nitric oxide (NO) generation after a combinatory treatment with inflammatory stimuli and Dex. Cdk5 deletion in macrophages attenuated the GR phosphorylation on serine 211 after Dex treatment alone and in combination with inflammatory stimuli, but interestingly increased the GR-dependent anti-inflammatory target gene dual-specificity phosphatase 1 (Dusp1, Mkp1). Mkp1 phosphatase activity decreases the activation of its direct target p38Mapk, reduced iNos expression and NO production upon inflammatory stimuli and Dex treatment in the absence of Cdk5. Taken together, we identified Cdk5 as a potential novel regulator of NO generation in inflammatory macrophages under GC treatment. Our data suggest that GC treatment in combination with specific Cdk5 inhibtior(s) provides a stronger suppression of inflammation and could thus replace high-dose GC therapy which has severe side effects in the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Pauline Pfänder
- Institute of Comparative Molecular Endocrinology (CME), University of Ulm, Ulm, Germany
| | - Miray Fidan
- Institute of Comparative Molecular Endocrinology (CME), University of Ulm, Ulm, Germany
| | - Ute Burret
- Institute of Comparative Molecular Endocrinology (CME), University of Ulm, Ulm, Germany
| | - Lena Lipinski
- Institute of Comparative Molecular Endocrinology (CME), University of Ulm, Ulm, Germany
| | - Sabine Vettorazzi
- Institute of Comparative Molecular Endocrinology (CME), University of Ulm, Ulm, Germany
| |
Collapse
|
13
|
LIF regulates CXCL9 in tumor-associated macrophages and prevents CD8 + T cell tumor-infiltration impairing anti-PD1 therapy. Nat Commun 2019; 10:2416. [PMID: 31186412 PMCID: PMC6559950 DOI: 10.1038/s41467-019-10369-9] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 05/08/2019] [Indexed: 12/20/2022] Open
Abstract
Cancer response to immunotherapy depends on the infiltration of CD8+ T cells and the presence of tumor-associated macrophages within tumors. Still, little is known about the determinants of these factors. We show that LIF assumes a crucial role in the regulation of CD8+ T cell tumor infiltration, while promoting the presence of protumoral tumor-associated macrophages. We observe that the blockade of LIF in tumors expressing high levels of LIF decreases CD206, CD163 and CCL2 and induces CXCL9 expression in tumor-associated macrophages. The blockade of LIF releases the epigenetic silencing of CXCL9 triggering CD8+ T cell tumor infiltration. The combination of LIF neutralizing antibodies with the inhibition of the PD1 immune checkpoint promotes tumor regression, immunological memory and an increase in overall survival. LIF is a pleiotropic cytokine that promotes an immunosuppressive microenvironment and has critical functions in embryonic development. Here, the authors show that LIF regulates CD8+ T cell tumor infiltration in cancer by repressing CXCL19 and promoting the presence of protumoral macrophages and thatLIF inhibition, via neutralizing antibodies, promotes T cell infiltration and synergizes with immune checkpoint inhbitors resulting in tumor regression and immunological memory.
Collapse
|
14
|
Sulaiman M, Mahadevan RK, Kurup MG. Effect of Ascophyllan from Brown Algae Padina tetrastromatica on Cell Migration and Extracellular Matrix Stabilisation in Burn Wounds. ACTA ACUST UNITED AC 2019. [DOI: 10.2174/1573407214666180327123118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
Burn injuries are the most common injuries and a major health problem affecting
communities worldwide. Many alternative therapies are used as treatment for burns. The healing
efficacy of sulphated polysaccharide ascophyllan was evaluated and studied its mechanism of action on
experimental burn wounds in rats.
Methods:
Ascophyllan fractions were extracted from marine brown algae Padina tetrastromatica (Dictyotaceae)
and evaluated its burn wound healing potential. Full thickness burn wounds induced in male
albino rats were used for in vivo study and evaluated wound healing parameters.
Results:
The results showed that Ascophyllan Fraction 3 (AF3) had no cytotoxic effect and it increases
cell migration and production of VEGF in fibroblasts. AF3 significantly reduced in vitro secretion of
cytokines in blood mononuclear cells treated with Lipopolysaccharide (LPS). In vivo study showed that
AF3 (5%) has significant wound healing activity in albino rats and this dose was used for studying the
healing mechanism. The reference control used for the study was povidone-iodine ointment. Wound
area contraction and reepithelialisation was faster in AF3 (5%) administered group. When applied topically,
AF3 (5%) increased hydroxyproline and hexosamine content at the wound site. Uronic acid, DNA
and proteins levels were also increased. Compared to the control groups, AF3 (5%) treatment showed an
increase in neovascularization and fibroblast proliferation as evidenced by histopathology of granulation
tissue.
Conclusion:
Sulphated polysaccharide ascophyllan is beneficial for the wound environment as it
enhances the healing process and suggested the safe usage of this algal polysaccharide as an alternative
for replacing current synthetic wound healing agents in medicine.
Collapse
Affiliation(s)
- Mohsin Sulaiman
- Department of Aridland Agriculture, College of Food and Agriculture, United Arab Emirates University, Al Ain -15551, United Arab Emirates
| | | | - Muraleedhara G. Kurup
- Department of Biochemistry, University of Kerala, Thiruvananthapuram- 695581, Kerala, India
| |
Collapse
|
15
|
Lee K, Seo I, Choi MH, Jeong D. Roles of Mitogen-Activated Protein Kinases in Osteoclast Biology. Int J Mol Sci 2018; 19:ijms19103004. [PMID: 30275408 PMCID: PMC6213329 DOI: 10.3390/ijms19103004] [Citation(s) in RCA: 170] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 09/20/2018] [Accepted: 09/27/2018] [Indexed: 01/20/2023] Open
Abstract
Bone undergoes continuous remodeling, which is homeostatically regulated by concerted communication between bone-forming osteoblasts and bone-degrading osteoclasts. Multinucleated giant osteoclasts are the only specialized cells that degrade or resorb the organic and inorganic bone components. They secrete proteases (e.g., cathepsin K) that degrade the organic collagenous matrix and establish localized acidosis at the bone-resorbing site through proton-pumping to facilitate the dissolution of inorganic mineral. Osteoporosis, the most common bone disease, is caused by excessive bone resorption, highlighting the crucial role of osteoclasts in intact bone remodeling. Signaling mediated by mitogen-activated protein kinases (MAPKs), including extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and p38, has been recognized to be critical for normal osteoclast differentiation and activation. Various exogenous (e.g., toll-like receptor agonists) and endogenous (e.g., growth factors and inflammatory cytokines) stimuli contribute to determining whether MAPKs positively or negatively regulate osteoclast adhesion, migration, fusion and survival, and osteoclastic bone resorption. In this review, we delineate the unique roles of MAPKs in osteoclast metabolism and provide an overview of the upstream regulators that activate or inhibit MAPKs and their downstream targets. Furthermore, we discuss the current knowledge about the differential kinetics of ERK, JNK, and p38, and the crosstalk between MAPKs in osteoclast metabolism.
Collapse
Affiliation(s)
- Kyunghee Lee
- Department of Microbiology, Laboratory of Bone Metabolism and Control, Yeungnam University College of Medicine, Daegu 42415, Korea.
| | - Incheol Seo
- Department of Microbiology, Laboratory of Bone Metabolism and Control, Yeungnam University College of Medicine, Daegu 42415, Korea.
| | - Mun Hwan Choi
- Department of Microbiology, Laboratory of Bone Metabolism and Control, Yeungnam University College of Medicine, Daegu 42415, Korea.
| | - Daewon Jeong
- Department of Microbiology, Laboratory of Bone Metabolism and Control, Yeungnam University College of Medicine, Daegu 42415, Korea.
| |
Collapse
|
16
|
Zhang P, Ha N, Dai Q, Zhou S, Yu C, Jiang L. Hypoxia suppresses osteogenesis of bone mesenchymal stem cells via the extracellular signal‑regulated 1/2 and p38‑mitogen activated protein kinase signaling pathways. Mol Med Rep 2017; 16:5515-5522. [PMID: 28849067 DOI: 10.3892/mmr.2017.7276] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 05/25/2017] [Indexed: 11/05/2022] Open
Abstract
There is a growing body of evidence indicating an association between osteoporosis and vascular diseases, which are associated with reduced blood supply. Decreased vascular flow results in a hypoxic gradient in the local microenvironment, affecting local bone remodeling. Bone mesenchymal stem cells (BMSCs) have been demonstrated to be the key to bone remodeling. To elucidate the molecular mechanisms involved in vascular supply and osteoporosis, the present study investigated the effect of hypoxia on BMSCs in vitro during osteogenesis. The BMSC osteogenesis process was evaluated by alkaline phosphatase (ALP) activity assay and the mRNA expression of the osteogenic markers runt‑related transcription factor 2 (Runx2), ALP and osteocalcin. The function of extracellular signal‑regulated kinase (ERK)1/2 and p38 kinase were studied under hypoxia using specific inhibitors. The results demonstrated that hypoxia reduces the osteogenic differentiation of BMSCs by inactivating Runx2, followed by decreased ALP activity and mRNA expression levels of ALP, collagen type I and osteocalcin. Furthermore, these data suggested that the ERK1/2 and p38‑mitogen activated protein kinase signaling pathways might participate in hypoxia‑induced differentiation of BMSCs toward the osteogenic phenotype. Compared with ERK1/2, the p38‑Runx2 signaling pathway might exert a relatively more prominent effect in the above process. These findings may help to elucidate the pathophysiology of osteoporosis caused by decreased vascular supply.
Collapse
Affiliation(s)
- Peng Zhang
- Second Dental Center, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory of Stomatology, Shanghai 200011, P.R. China
| | - Nayoung Ha
- Center of Craniofacial Orthodontics, Department of Oral and Cranio‑Maxillofacial Science, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory of Stomatology, Shanghai 200011, P.R. China
| | - Qinggang Dai
- Center of Craniofacial Orthodontics, Department of Oral and Cranio‑Maxillofacial Science, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory of Stomatology, Shanghai 200011, P.R. China
| | - Siru Zhou
- Center of Craniofacial Orthodontics, Department of Oral and Cranio‑Maxillofacial Science, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory of Stomatology, Shanghai 200011, P.R. China
| | - Chuangqi Yu
- Department of Oral Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory of Stomatology, Shanghai 200011, P.R. China
| | - Lingyong Jiang
- Center of Craniofacial Orthodontics, Department of Oral and Cranio‑Maxillofacial Science, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory of Stomatology, Shanghai 200011, P.R. China
| |
Collapse
|
17
|
Brutkiewicz RR. Cell Signaling Pathways That Regulate Antigen Presentation. THE JOURNAL OF IMMUNOLOGY 2017; 197:2971-2979. [PMID: 27824592 DOI: 10.4049/jimmunol.1600460] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 07/08/2016] [Indexed: 12/11/2022]
Abstract
Cell signaling pathways regulate much in the life of a cell: from shuttling cargo through intracellular compartments and onto the cell surface, how it should respond to stress, protecting itself from harm (environmental insults or infections), to ultimately, death by apoptosis. These signaling pathways are important for various aspects of the immune response as well. However, not much is known in terms of the participation of cell signaling pathways in Ag presentation, a necessary first step in the activation of innate and adaptive T cells. In this brief review, I discuss the known signaling molecules (and pathways) that regulate how Ags are presented to T cells and the mechanism(s), if identified. Studies in this area have important implications in vaccine development and new treatment paradigms against infectious diseases, autoimmunity, and cancer.
Collapse
Affiliation(s)
- Randy R Brutkiewicz
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202
| |
Collapse
|
18
|
|
19
|
Yamaguchi R, Sakamoto A, Yamamoto T, Ishimaru Y, Narahara S, Sugiuchi H, Yamaguchi Y. Surfactant Protein D Inhibits Interleukin-12p40 Production by Macrophages Through the SIRPα/ROCK/ERK Signaling Pathway. Am J Med Sci 2017. [DOI: 10.1016/j.amjms.2017.03.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
20
|
Hammer A, Stegbauer J, Linker RA. Macrophages in neuroinflammation: role of the renin-angiotensin-system. Pflugers Arch 2017; 469:431-444. [PMID: 28190090 DOI: 10.1007/s00424-017-1942-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 01/18/2017] [Accepted: 01/23/2017] [Indexed: 12/12/2022]
Abstract
Macrophages are essential players of the innate immune system which are involved in the initiation and progression of various inflammatory and autoimmune diseases including neuroinflammation. In the past few years, it has become increasingly clear that the regulation of macrophage responses by the local tissue milieu is also influenced by mediators which were first discovered as regulators in the nervous or also cardiovascular system. Here, the renin-angiotensin system (RAS) is a major focus of current research. Besides its classical role in blood pressure control, body fluid, and electrolyte homeostasis, the RAS may influence (auto)immune responses, modulate T cells, and particularly act on macrophages via different signaling pathways. Activation of classical RAS pathways including angiotensin (Ang) II and AngII type 1 (AT1R) receptors may drive pro-inflammatory macrophage responses in neuroinflammation via regulation of chemokines. More recently, alternative RAS pathways were described, such as binding of Ang-(1-7) to its receptor Mas. Signaling via Mas pathways may counteract some of the AngII/AT1R-mediated effects. In macrophages, the Ang-(1-7)/Mas exerts beneficial effects on neuroinflammation via modulating macrophage polarization, migration, and T cell activation in vitro and in vivo. These data delineate a pivotal role of the RAS in inflammation of the nervous system and identify RAS modulation as a potential new target for immunotherapy with a special focus on macrophages.
Collapse
Affiliation(s)
- Anna Hammer
- Department of Neurology, University Hospital, Friedrich-Alexander University Erlangen-Nuremberg, Schwabachanlage 6, 91054 Erlangen, Germany
| | - Johannes Stegbauer
- Department of Nephrology, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Ralf A Linker
- Department of Neurology, University Hospital, Friedrich-Alexander University Erlangen-Nuremberg, Schwabachanlage 6, 91054 Erlangen, Germany.
| |
Collapse
|
21
|
Seoane PI, Rückerl D, Casaravilla C, Barrios AA, Pittini Á, MacDonald AS, Allen JE, Díaz A. Particles from the Echinococcus granulosus laminated layer inhibit IL-4 and growth factor-driven Akt phosphorylation and proliferative responses in macrophages. Sci Rep 2016; 6:39204. [PMID: 27966637 PMCID: PMC5155279 DOI: 10.1038/srep39204] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 11/11/2016] [Indexed: 12/23/2022] Open
Abstract
Proliferation of macrophages is a hallmark of inflammation in many type 2 settings including helminth infections. The cellular expansion is driven by the type 2 cytokine interleukin-4 (IL-4), as well as by M-CSF, which also controls homeostatic levels of tissue resident macrophages. Cystic echinococcosis, caused by the tissue-dwelling larval stage of the cestode Echinococcus granulosus, is characterised by normally subdued local inflammation. Infiltrating host cells make contact only with the acellular protective coat of the parasite, called laminated layer, particles of which can be ingested by phagocytic cells. Here we report that a particulate preparation from this layer (pLL) strongly inhibits the proliferation of macrophages in response to IL-4 or M-CSF. In addition, pLL also inhibits IL-4-driven up-regulation of Relm-α, without similarly affecting Chitinase-like 3 (Chil3/Ym1). IL-4-driven cell proliferation and up-regulation of Relm-α are both known to depend on the phosphatidylinositol (PI3K)/Akt pathway, which is dispensable for induction of Chil3/Ym1. Exposure to pLL in vitro inhibited Akt activation in response to proliferative stimuli, providing a potential mechanism for its activities. Our results suggest that the E. granulosus laminated layer exerts some of its anti-inflammatory properties through inhibition of PI3K/Akt activation and consequent limitation of macrophage proliferation.
Collapse
Affiliation(s)
- Paula I Seoane
- Cátedra de Inmunología, Departamento de Biociencias (Facultad de Química) e Instituto de Química Biológica (Facultad de Ciencias), Universidad de la República, Montevideo, Uruguay
| | - Dominik Rückerl
- Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, UK
| | - Cecilia Casaravilla
- Cátedra de Inmunología, Departamento de Biociencias (Facultad de Química) e Instituto de Química Biológica (Facultad de Ciencias), Universidad de la República, Montevideo, Uruguay
| | - Anabella A Barrios
- Cátedra de Inmunología, Departamento de Biociencias (Facultad de Química) e Instituto de Química Biológica (Facultad de Ciencias), Universidad de la República, Montevideo, Uruguay
| | - Álvaro Pittini
- Cátedra de Inmunología, Departamento de Biociencias (Facultad de Química) e Instituto de Química Biológica (Facultad de Ciencias), Universidad de la República, Montevideo, Uruguay
| | - Andrew S MacDonald
- Manchester Collaborative Centre for Inflammation Research (MCCIR), University of Manchester, Manchester, UK
| | - Judith E Allen
- Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, UK
| | - Alvaro Díaz
- Cátedra de Inmunología, Departamento de Biociencias (Facultad de Química) e Instituto de Química Biológica (Facultad de Ciencias), Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
22
|
Yang L, Guo H, Li Y, Meng X, Yan L, Dan Zhang, Wu S, Zhou H, Peng L, Xie Q, Jin X. Oleoylethanolamide exerts anti-inflammatory effects on LPS-induced THP-1 cells by enhancing PPARα signaling and inhibiting the NF-κB and ERK1/2/AP-1/STAT3 pathways. Sci Rep 2016; 6:34611. [PMID: 27721381 PMCID: PMC5056375 DOI: 10.1038/srep34611] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 09/09/2016] [Indexed: 01/01/2023] Open
Abstract
The present study aimed to examine the anti-inflammatory actions of oleoylethanolamide (OEA) in lipopolysaccharide (LPS)-induced THP-1 cells. The cells were stimulated with LPS (1 μg/ml) in the presence or absence of OEA (10, 20 and 40 μM). The pro-inflammatory cytokines were evaluated by qRT-PCR and ELISA. The THP-1 cells were transiently transfected with PPARα small-interfering RNA, and TLR4 activity was determined with a blocking test using anti-TLR4 antibody. Additionally, a special inhibitor was used to analyse the intracellular signaling pathway. OEA exerted a potent anti-inflammatory effect by reducing the production of pro-inflammatory cytokines and TLR4 expression, and by enhancing PPARα expression. The modulatory effects of OEA on LPS-induced inflammation depended on PPARα and TLR4. Importantly, OEA inhibited LPS-induced NF-κB activation, IκBα degradation, expression of AP-1, and the phosphorylation of ERK1/2 and STAT3. In summary, our results demonstrated that OEA exerts anti-inflammatory effects by enhancing PPARα signaling, inhibiting the TLR4-mediated NF-κB signaling pathway, and interfering with the ERK1/2-dependent signaling cascade (TLR4/ERK1/2/AP-1/STAT3), which suggests that OEA may be a therapeutic agent for inflammatory diseases.
Collapse
Affiliation(s)
- Lichao Yang
- Xiamen Key Laboratory of Chiral Drugs, Medical College, Xiamen University, Xiamen, Fujian, 361102, P. R. China
| | - Han Guo
- Xiamen Key Laboratory of Chiral Drugs, Medical College, Xiamen University, Xiamen, Fujian, 361102, P. R. China
| | - Ying Li
- Department of Pharmacology, Xiamen Medical College, Xiamen, Fujian, 361008, P. R. China
| | - Xianglan Meng
- Xiamen Key Laboratory of Chiral Drugs, Medical College, Xiamen University, Xiamen, Fujian, 361102, P. R. China
| | - Lu Yan
- Xiamen Key Laboratory of Chiral Drugs, Medical College, Xiamen University, Xiamen, Fujian, 361102, P. R. China
| | - Dan Zhang
- Department of Cardiology, the First Affiliated Hospital of Xiamen University, Xiamen, Fujian, 361003, P. R. China
| | - Sangang Wu
- Xiamen Cancer Center, Department of Radiation Oncology, the First Affiliated Hospital of Xiamen University, Xiamen, Fujian, 361003, P. R. China
| | - Hao Zhou
- Xiamen Key Laboratory of Chiral Drugs, Medical College, Xiamen University, Xiamen, Fujian, 361102, P. R. China
| | - Lu Peng
- Xiamen Key Laboratory of Chiral Drugs, Medical College, Xiamen University, Xiamen, Fujian, 361102, P. R. China
| | - Qiang Xie
- Department of Cardiology, the First Affiliated Hospital of Xiamen University, Xiamen, Fujian, 361003, P. R. China
| | - Xin Jin
- Xiamen Key Laboratory of Chiral Drugs, Medical College, Xiamen University, Xiamen, Fujian, 361102, P. R. China
| |
Collapse
|
23
|
Song L, Papaioannou G, Zhao H, Luderer HF, Miller C, Dall'Osso C, Nazarian RM, Wagers AJ, Demay MB. The Vitamin D Receptor Regulates Tissue Resident Macrophage Response to Injury. Endocrinology 2016; 157:4066-4075. [PMID: 27526034 PMCID: PMC5045513 DOI: 10.1210/en.2016-1474] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Ligand-dependent actions of the vitamin D receptor (VDR) play a pleiotropic role in the regulation of innate and adaptive immunity. The liganded VDR is required for recruitment of macrophages during the inflammatory phase of cutaneous wound healing. Although the number of macrophages in the granulation tissue 2 days after wounding is markedly reduced in VDR knockout (KO) compared with wild-type mice, VDR ablation does not alter macrophage polarization. Parabiosis studies demonstrate that circulatory chimerism with wild-type mice is unable to rescue the macrophage defect in the wounds of VDR KO mice and reveal that wound macrophages are of local origin, regardless of VDR status. Wound cytokine analyses demonstrated a decrease in macrophage colony-stimulating factor (M-CSF) protein levels in VDR KO mice. Consistent with this, induction of M-CSF gene expression by TGFβ and 1,25-dihydroxyvitamin D was impaired in dermal fibroblasts isolated from VDR KO mice. Because M-CSF is important for macrophage self-renewal, studies were performed to evaluate the response of tissue resident macrophages to this cytokine. A decrease in M-CSF induced proliferation and cyclin D1 expression was observed in peritoneal resident macrophages isolated from VDR KO mice, suggesting an intrinsic macrophage abnormality. Consistent with this, wound-healing assays in mice with macrophage-specific VDR ablation demonstrate that a normal wound microenvironment cannot compensate for the absence of the VDR in macrophages and thus confirm a critical role for the macrophage VDR in the inflammatory response to injury.
Collapse
Affiliation(s)
- Lige Song
- Endocrine Unit (L.S., G.P., H.Z., H.F.L., M.B.D.) and Department of Pathology (R.M.N.), Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114; and Department of Stem Cell and Regenerative Biology (C.M., C.D., A.J.W.), Harvard University and Harvard Stem Cell Institute, Cambridge, Massachusetts 02138
| | - Garyfallia Papaioannou
- Endocrine Unit (L.S., G.P., H.Z., H.F.L., M.B.D.) and Department of Pathology (R.M.N.), Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114; and Department of Stem Cell and Regenerative Biology (C.M., C.D., A.J.W.), Harvard University and Harvard Stem Cell Institute, Cambridge, Massachusetts 02138
| | - Hengguang Zhao
- Endocrine Unit (L.S., G.P., H.Z., H.F.L., M.B.D.) and Department of Pathology (R.M.N.), Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114; and Department of Stem Cell and Regenerative Biology (C.M., C.D., A.J.W.), Harvard University and Harvard Stem Cell Institute, Cambridge, Massachusetts 02138
| | - Hilary F Luderer
- Endocrine Unit (L.S., G.P., H.Z., H.F.L., M.B.D.) and Department of Pathology (R.M.N.), Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114; and Department of Stem Cell and Regenerative Biology (C.M., C.D., A.J.W.), Harvard University and Harvard Stem Cell Institute, Cambridge, Massachusetts 02138
| | - Christine Miller
- Endocrine Unit (L.S., G.P., H.Z., H.F.L., M.B.D.) and Department of Pathology (R.M.N.), Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114; and Department of Stem Cell and Regenerative Biology (C.M., C.D., A.J.W.), Harvard University and Harvard Stem Cell Institute, Cambridge, Massachusetts 02138
| | - Claudia Dall'Osso
- Endocrine Unit (L.S., G.P., H.Z., H.F.L., M.B.D.) and Department of Pathology (R.M.N.), Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114; and Department of Stem Cell and Regenerative Biology (C.M., C.D., A.J.W.), Harvard University and Harvard Stem Cell Institute, Cambridge, Massachusetts 02138
| | - Rosalynn M Nazarian
- Endocrine Unit (L.S., G.P., H.Z., H.F.L., M.B.D.) and Department of Pathology (R.M.N.), Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114; and Department of Stem Cell and Regenerative Biology (C.M., C.D., A.J.W.), Harvard University and Harvard Stem Cell Institute, Cambridge, Massachusetts 02138
| | - Amy J Wagers
- Endocrine Unit (L.S., G.P., H.Z., H.F.L., M.B.D.) and Department of Pathology (R.M.N.), Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114; and Department of Stem Cell and Regenerative Biology (C.M., C.D., A.J.W.), Harvard University and Harvard Stem Cell Institute, Cambridge, Massachusetts 02138
| | - Marie B Demay
- Endocrine Unit (L.S., G.P., H.Z., H.F.L., M.B.D.) and Department of Pathology (R.M.N.), Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114; and Department of Stem Cell and Regenerative Biology (C.M., C.D., A.J.W.), Harvard University and Harvard Stem Cell Institute, Cambridge, Massachusetts 02138
| |
Collapse
|
24
|
Lloberas J, Valverde-Estrella L, Tur J, Vico T, Celada A. Mitogen-Activated Protein Kinases and Mitogen Kinase Phosphatase 1: A Critical Interplay in Macrophage Biology. Front Mol Biosci 2016; 3:28. [PMID: 27446931 PMCID: PMC4923182 DOI: 10.3389/fmolb.2016.00028] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 06/13/2016] [Indexed: 12/21/2022] Open
Abstract
Macrophages are necessary in multiple processes during the immune response or inflammation. This review emphasizes the critical role of the mitogen-activated protein kinases (MAPKs) and mitogen kinase phosphatase-1 (MKP-1) in the functional activities of macrophages. While the phosphorylation of MAPKs is required for macrophage activation or proliferation, MKP-1 dephosphorylates these kinases, thus playing a balancing role in the control of macrophage behavior. MKP-1 is a nuclear-localized dual-specificity phosphatase whose expression is regulated at multiple levels, including at the transcriptional and post-transcriptional level. The regulatory role of MKP-1 in the interplay between MAPK phosphorylation/dephosphorylation makes this molecule a critical regulator of macrophage biology and inflammation.
Collapse
Affiliation(s)
- Jorge Lloberas
- Departament of Cell Biology, Physiology and Immunology, Universitat de Barcelona Barcelona, Spain
| | - Lorena Valverde-Estrella
- Departament of Cell Biology, Physiology and Immunology, Universitat de Barcelona Barcelona, Spain
| | - Juan Tur
- Departament of Cell Biology, Physiology and Immunology, Universitat de Barcelona Barcelona, Spain
| | - Tania Vico
- Departament of Cell Biology, Physiology and Immunology, Universitat de Barcelona Barcelona, Spain
| | - Antonio Celada
- Departament of Cell Biology, Physiology and Immunology, Universitat de Barcelona Barcelona, Spain
| |
Collapse
|
25
|
Kim HJ, Ohk B, Kang WY, Seong SJ, Suk K, Lim MS, Kim SY, Yoon YR. Deficiency of Lipocalin-2 Promotes Proliferation and Differentiation of Osteoclast Precursors via Regulation of c-Fms Expression and Nuclear Factor-kappa B Activation. J Bone Metab 2016; 23:8-15. [PMID: 26981515 PMCID: PMC4791440 DOI: 10.11005/jbm.2016.23.1.8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 02/02/2016] [Accepted: 02/02/2016] [Indexed: 01/27/2023] Open
Abstract
Background Lipocalin-2 (LCN2), a small glycoprotein, has a pivotal role in diverse biological processes such as cellular proliferation and differentiation. We previously reported that LCN2 is implicated in osteoclast formation induced by receptor activator of nuclear factor-kappa B ligand (RANKL) and macrophage colony-stimulating factor (M-CSF). In the present study, we used a knockout mouse model to further investigate the role of LCN2 in osteoclast development. Methods Osteoclastogenesis was assessed using primary bone marrow-derived macrophages. RANKL and M-CSF signaling was determined by immunoblotting, cell proliferation by bromodeoxyuridine (BrdU) enzyme-linked immunosorbent assay (ELISA), and apoptosis by cell death detection ELISA. Bone morphometric parameters were determined using a micro-computed tomography system. Results Our results showed that LCN2 deficiency increases tartrate-resistant acid phosphatase (TRAP)-positive multinucleated osteoclast formation in vitro, a finding that reflects enhanced proliferation and differentiation of osteoclast lineage cells. LCN2 deficiency promotes M-CSF-induced proliferation of bone marrow macrophages (BMMs), osteoclast precursors, without altering their survival. The accelerated proliferation of LCN2-deficient precursors is associated with enhanced expression and activation of the M-CSF receptor, c-Fms. Furthermore, LCN2 deficiency stimulates the induction of c-Fos and nuclear factor of activated T cells c1 (NFATc1), key transcription factors for osteoclastogenesis, and promotes RANKL-induced inhibitor of kappa B (IκBα) phosphorylation. Interestingly, LCN2 deficiency does not affect basal osteoclast formation in vivo, suggesting that LCN2 might play a role in the enhanced osteoclast development that occurs under some pathological conditions. Conclusions Our study establishes LCN2 as a negative modulator of osteoclast formation, results that are in accordance with our previous findings.
Collapse
Affiliation(s)
- Hyun-Ju Kim
- Department of Biomedical Science, Cell and Matrix Research Institute, BK21 Plus KNU Biomedical Convergence Program, Clinical Trial Center, School of Medicine, Kyungpook National University and Hospital, Daegu, Korea.; Skeletal Diseases Genome Research Center, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Boram Ohk
- Department of Biomedical Science, Cell and Matrix Research Institute, BK21 Plus KNU Biomedical Convergence Program, Clinical Trial Center, School of Medicine, Kyungpook National University and Hospital, Daegu, Korea
| | - Woo Youl Kang
- Department of Biomedical Science, Cell and Matrix Research Institute, BK21 Plus KNU Biomedical Convergence Program, Clinical Trial Center, School of Medicine, Kyungpook National University and Hospital, Daegu, Korea
| | - Sook Jin Seong
- Department of Biomedical Science, Cell and Matrix Research Institute, BK21 Plus KNU Biomedical Convergence Program, Clinical Trial Center, School of Medicine, Kyungpook National University and Hospital, Daegu, Korea
| | - Kyoungho Suk
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Mi-Sun Lim
- College of Pharmacy, Yeungnam University, Gyeongsan, Korea
| | - Shin-Yoon Kim
- Skeletal Diseases Genome Research Center, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Young-Ran Yoon
- Department of Biomedical Science, Cell and Matrix Research Institute, BK21 Plus KNU Biomedical Convergence Program, Clinical Trial Center, School of Medicine, Kyungpook National University and Hospital, Daegu, Korea
| |
Collapse
|
26
|
Hoppstädter J, Kessler SM, Bruscoli S, Huwer H, Riccardi C, Kiemer AK. Glucocorticoid-Induced Leucine Zipper: A Critical Factor in Macrophage Endotoxin Tolerance. THE JOURNAL OF IMMUNOLOGY 2015; 194:6057-6067. [DOI: 10.4049/jimmunol.1403207] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Abstract
Induction of glucocorticoid-induced leucine zipper (GILZ) by glucocorticoids plays a key role in their anti-inflammatory action. In activated macrophages, GILZ levels are downregulated via tristetraprolin-mediated GILZ mRNA destabilization. To assess the functional significance of GILZ downregulation, we generated myeloid-specific GILZ knockout (KO) mice. GILZ-deficient macrophages displayed a higher responsiveness toward LPS, as indicated by increased TNF-α and IL-1β expression. This effect was due to an activation of ERK, which was significantly amplified in GILZ KO cells. The LPS-induced activation of macrophages is attenuated upon pretreatment of macrophages with low-dose LPS, an effect termed endotoxin tolerance. In LPS-tolerant macrophages, GILZ mRNA was stabilized, whereas ERK activation was strongly decreased. In contrast, GILZ KO macrophages exhibited a strongly reduced desensitization. To explore the contribution of GILZ expression in macrophages to endotoxin tolerance in vivo, we treated GILZ KO mice with repeated i.p. injections of low-dose LPS followed by treatment with high-dose LPS. LPS pretreatment resulted in reduced proinflammatory mediator expression upon high-dose LPS treatment in serum and tissues. In contrast, cytokine induction was preserved in tolerized GILZ KO animals. In summary, our data suggest that GILZ is a key regulator of macrophage functions.
Collapse
Affiliation(s)
- Jessica Hoppstädter
- *Department of Pharmacy, Pharmaceutical Biology, Saarland University, 66041 Saarbrücken, Germany
| | - Sonja M. Kessler
- *Department of Pharmacy, Pharmaceutical Biology, Saarland University, 66041 Saarbrücken, Germany
| | - Stefano Bruscoli
- †Section of Pharmacology, Department of Medicine, University of Perugia, 06100 Perugia, Italy; and
| | - Hanno Huwer
- ‡Department of Cardiothoracic Surgery, Völklingen Heart Centre, 66333 Völklingen, Germany
| | - Carlo Riccardi
- †Section of Pharmacology, Department of Medicine, University of Perugia, 06100 Perugia, Italy; and
| | - Alexandra K. Kiemer
- *Department of Pharmacy, Pharmaceutical Biology, Saarland University, 66041 Saarbrücken, Germany
| |
Collapse
|
27
|
Park JH, Jun JG, Kim JK. (E)-3-(3,4-dihydroxy-2-methoxyphenyl)-1-(2,4-dihydroxyphenyl)prop-2-en-1-one, a novel licochalcone B derivative compound, suppresses lipopolysaccharide-stimulated inflammatory reactions in RAW264.7 cells and endotoxin shock in mice. Chem Biol Interact 2014; 224:142-8. [DOI: 10.1016/j.cbi.2014.10.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 10/09/2014] [Accepted: 10/15/2014] [Indexed: 12/22/2022]
|
28
|
Mateos MV, Kamerbeek CB, Giusto NM, Salvador GA. The phospholipase D pathway mediates the inflammatory response of the retinal pigment epithelium. Int J Biochem Cell Biol 2014; 55:119-28. [DOI: 10.1016/j.biocel.2014.08.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 08/12/2014] [Accepted: 08/18/2014] [Indexed: 12/25/2022]
|
29
|
Abstract
Mitogen-activated protein kinases (MAPKs) mediate a wide variety of cellular behaviors in response to extracellular stimuli. One of the main subgroups, the p38 MAP kinases, has been implicated in a wide range of complex biologic processes, such as cell proliferation, cell differentiation, cell death, cell migration, and invasion. Dysregulation of p38 MAPK levels in patients are associated with advanced stages and short survival in cancer patients (e.g., prostate, breast, bladder, liver, and lung cancer). p38 MAPK plays a dual role as a regulator of cell death, and it can either mediate cell survival or cell death depending not only on the type of stimulus but also in a cell type specific manner. In addition to modulating cell survival, an essential role of p38 MAPK in modulation of cell migration and invasion offers a distinct opportunity to target this pathway with respect to tumor metastasis. The specific function of p38 MAPK appears to depend not only on the cell type but also on the stimuli and/or the isoform that is activated. p38 MAPK signaling pathway is activated in response to diverse stimuli and mediates its function by components downstream of p38. Extrapolation of the knowledge gained from laboratory findings is essential to address the clinical significance of p38 MAPK signaling pathways. The goal of this review is to provide an overview on recent progress made in defining the functions of p38 MAPK pathways with respect to solid tumor biology and generate testable hypothesis with respect to the role of p38 MAPK as an attractive target for intervention of solid tumors.
Collapse
Affiliation(s)
- Hari K Koul
- Department of Biochemistry & Molecular Biology, LSU Health Sciences Center, Shreveport, LA, USA ; Feist-Weiller Cancer Center, Shreveport, LA, USA ; Veterans Administration Medical Center, Shreveport, LA, USA
| | - Mantu Pal
- Department of Biochemistry & Molecular Biology, LSU Health Sciences Center, Shreveport, LA, USA ; Veterans Administration Medical Center, Shreveport, LA, USA
| | - Sweaty Koul
- Feist-Weiller Cancer Center, Shreveport, LA, USA ; Department of Urology, LSU Health Sciences Center, Shreveport, LA, USA
| |
Collapse
|
30
|
Pascual-García M, Rué L, León T, Julve J, Carbó JM, Matalonga J, Auer H, Celada A, Escolà-Gil JC, Steffensen KR, Pérez-Navarro E, Valledor AF. Reciprocal negative cross-talk between liver X receptors (LXRs) and STAT1: effects on IFN-γ-induced inflammatory responses and LXR-dependent gene expression. THE JOURNAL OF IMMUNOLOGY 2013; 190:6520-32. [PMID: 23686490 DOI: 10.4049/jimmunol.1201393] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Liver X receptors (LXRs) exert key functions in lipid homeostasis and in control of inflammation. In this study we have explored the impact of LXR activation on the macrophage response to the endogenous inflammatory cytokine IFN-γ. Transcriptional profiling studies demonstrate that ∼38% of the IFN-γ-induced transcriptional response is repressed by LXR activation in macrophages. LXRs also mediated inhibitory effects on selected IFN-γ-induced genes in primary microglia and in a model of IFN-γ-induced neuroinflammation in vivo. LXR activation resulted in reduced STAT1 recruitment to the promoters tested in this study without affecting STAT1 phosphorylation. A closer look into the mechanism revealed that SUMOylation of LXRs, but not the presence of nuclear receptor corepressor 1, was required for repression of the NO synthase 2 promoter. We have also analyzed whether IFN-γ signaling exerts reciprocal effects on LXR targets. Treatment with IFN-γ inhibited, in a STAT1-dependent manner, the LXR-dependent upregulation of selective targets, including ATP-binding cassette A1 (ABCA1) and sterol response element binding protein 1c. Downregulation of ABCA1 expression correlated with decreased cholesterol efflux to apolipoprotein A1 in macrophages stimulated with IFN-γ. The inhibitory effects of IFN-γ on LXR signaling did not involve reduced binding of LXR/retinoid X receptor heterodimers to target gene promoters. However, overexpression of the coactivator CREB-binding protein/p300 reduced the inhibitory actions of IFN-γ on the Abca1 promoter, suggesting that competition for CREB-binding protein may contribute to STAT1-dependent downregulation of LXR targets. The results from this study suggest an important level of bidirectional negative cross-talk between IFN-γ/STAT1 and LXRs with implications both in the control of IFN-γ-mediated immune responses and in the regulation of lipid metabolism.
Collapse
Affiliation(s)
- Mónica Pascual-García
- Nuclear Receptor Group, Department of Physiology and Immunology, School of Biology, University of Barcelona, 08028 Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Kim CH, Yoo YM. Fluid shear stress and melatonin in combination activate anabolic proteins in MC3T3-E1 osteoblast cells. J Pineal Res 2013; 54:453-61. [PMID: 23397978 DOI: 10.1111/jpi.12043] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2012] [Accepted: 01/11/2013] [Indexed: 12/19/2022]
Abstract
In this study, we investigated whether fluid shear stress and melatonin in combination stimulate the anabolic proteins through the phosphorylation of extracellular signal-regulated kinase (p-ERK) in MC3T3-E1 osteoblast cells. First, we researched why fluid shear stress and melatonin in combination influence cell survival. Fluid shear stress (1 hr) and melatonin (1 mM) in combination reduced autophagic marker LC3-II compared with fluid shear stress (1 hr) and/or melatonin (0.1 mM). Under the same conditions for fluid shear stress, markers of cell survival signaling pathway p-ERK, phosphorylation of serine-threonine protein kinase (p-Akt), phosphorylation of mammalian target of rapamycin (p-mTOR), and p85-S6K were investigated. p-Akt, p-mTOR (Ser 2481) expressions increased with the addition of 1 mM melatonin prior to 0.1 mM melatonin treatment. However, p-S6K expression did not change significantly. Next, mitochondria activity including Bcl-2, Bax, catalase, and Mn-superoxide dismutase (Mn-SOD) were studied. Expressions of Bcl-2, Bax, and catalase proteins were low under fluid shear stress plus 1 mM melatonin compared with only fluid shear stress alone, whereas Mn-SOD expression was high compared with conditions of no fluid shear stress. Finally, the anabolic proteins of bone, osteoprotegerin, type I collagen (collagen I), and bone sialoprotein II (BSP II) were checked. These proteins increased with combined fluid shear stress (1, 4 hr) and melatonin (0.1, 1 mM). Together, these results suggest that fluid shear stress and melatonin in combination may increase the expression of anabolic proteins through the p-ERK in MC3T3-E1 osteoblast cells. Therefore, fluid shear stress in combination with melatonin may promote the anabolic response of osteoblasts.
Collapse
Affiliation(s)
- Chi Hyun Kim
- Department of Biomedical Engineering, College of Health Science, Yonsei University, Wonju, Gangwon-do, Korea
| | | |
Collapse
|
32
|
You R, Long W, Lai Z, Sha L, Wu K, Yu X, Lai Y, Ji H, Huang Z, Zhang Y. Discovery of a potential anti-inflammatory agent: 3-oxo-29-noroleana-1,9(11),12-trien-2,20-dicarbonitrile. J Med Chem 2013; 56:1984-95. [PMID: 23373965 DOI: 10.1021/jm301652t] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Fifteen novel derivatives of glycyrrhetinic acid (GA) were synthesized and evaluated for anti-inflammatory activities. It was found that the introduction of 1-en-3-one and 9(11),12-diene and 2,20-dinitrile functionalities into the scaffold of GA led to the discovery of potent compound 19 for inhibition of LPS-induced NO production. Furthermore, 19 effectively inhibited the protein and mRNA expression of inducible NO synthase (iNOS) and the mRNA expression of TNF-α, IL-6, and IL-1β in LPS-stimulated RAW 264.7 macrophages. Mechanistically, 19 exerted inhibitory effects on the activation of the three main MAPKs and phosphorylation and degradation of IκB-α, as well as the ratio of nuclear/cytosolic content of p65. Importantly, 19 significantly decreased the mortality rate in the mouse model of LPS-induced sepsis shock. It is noteworthy that inhibitory effect of 19 on NO production was not blocked by the glucocorticoid receptor antagonist mifepristone, indicating that it does not act through the glucocorticoid receptor.
Collapse
Affiliation(s)
- Ran You
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Functional components in Luffa cylindrica and their effects on anti-inflammation of macrophage cells. Food Chem 2012; 135:386-95. [DOI: 10.1016/j.foodchem.2012.04.128] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Revised: 03/26/2012] [Accepted: 04/23/2012] [Indexed: 11/23/2022]
|
34
|
Differential Ly-6C expression identifies the recruited macrophage phenotype, which orchestrates the regression of murine liver fibrosis. Proc Natl Acad Sci U S A 2012; 109:E3186-95. [PMID: 23100531 DOI: 10.1073/pnas.1119964109] [Citation(s) in RCA: 724] [Impact Index Per Article: 55.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Although macrophages are widely recognized to have a profibrotic role in inflammation, we have used a highly tractable CCl(4)-induced model of reversible hepatic fibrosis to identify and characterize the macrophage phenotype responsible for tissue remodeling: the hitherto elusive restorative macrophage. This CD11B(hi) F4/80(int) Ly-6C(lo) macrophage subset was most abundant in livers during maximal fibrosis resolution and represented the principle matrix metalloproteinase (MMP) -expressing subset. Depletion of this population in CD11B promoter-diphtheria toxin receptor (CD11B-DTR) transgenic mice caused a failure of scar remodeling. Adoptive transfer and in situ labeling experiments showed that these restorative macrophages derive from recruited Ly-6C(hi) monocytes, a common origin with profibrotic Ly-6C(hi) macrophages, indicative of a phenotypic switch in vivo conferring proresolution properties. Microarray profiling of the Ly-6C(lo) subset, compared with Ly-6C(hi) macrophages, showed a phenotype outside the M1/M2 classification, with increased expression of MMPs, growth factors, and phagocytosis-related genes, including Mmp9, Mmp12, insulin-like growth factor 1 (Igf1), and Glycoprotein (transmembrane) nmb (Gpnmb). Confocal microscopy confirmed the postphagocytic nature of restorative macrophages. Furthermore, the restorative macrophage phenotype was recapitulated in vitro by the phagocytosis of cellular debris with associated activation of the ERK signaling cascade. Critically, induced phagocytic behavior in vivo, through administration of liposomes, increased restorative macrophage number and accelerated fibrosis resolution, offering a therapeutic strategy to this orphan pathological process.
Collapse
|
35
|
Comalada M, Lloberas J, Celada A. MKP-1: A critical phosphatase in the biology of macrophages controlling the switch between proliferation and activation. Eur J Immunol 2012; 42:1938-48. [DOI: 10.1002/eji.201242441] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Mònica Comalada
- Macrophage Biology Group; Institute for Research in Biomedicine (IRB Barcelona); Barcelona; Spain
| | | | | |
Collapse
|
36
|
Yu W, Chen J, Xiong Y, Pixley FJ, Yeung YG, Stanley ER. Macrophage proliferation is regulated through CSF-1 receptor tyrosines 544, 559, and 807. J Biol Chem 2012; 287:13694-704. [PMID: 22375015 DOI: 10.1074/jbc.m112.355610] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Colony-stimulating factor-1 (CSF-1)-stimulated CSF-1 receptor (CSF-1R) tyrosine phosphorylation initiates survival, proliferation, and differentiation signaling pathways in macrophages. Either activation loop Y807F or juxtamembrane domain (JMD) Y559F mutations severely compromise CSF-1-regulated proliferation and differentiation. YEF, a CSF-1R in which all eight tyrosines phosphorylated in the activated receptor were mutated to phenylalanine, lacks in vitro kinase activity and in vivo CSF-1-regulated tyrosine phosphorylation. The addition of Tyr-807 alone to the YEF backbone (Y807AB) led to CSF-1-independent but receptor kinase-dependent proliferation, without detectable activation loop Tyr-807 phosphorylation. The addition of Tyr-559 alone (Y559AB) supported a low level of CSF-1-independent proliferation that was slightly enhanced by CSF-1, indicating that Tyr-559 has a positive Tyr-807-independent effect. Consistent with the postulated autoinhibitory role of the JMD Tyr-559 and its relief by ligand-induced Tyr-559 phosphorylation, the addition of Tyr-559 to the Y807AB background suppressed proliferation in the absence of CSF-1, but restored most of the CSF-1-stimulated proliferation. Full restoration of kinase activation and proliferation required the additional add back of JMD Tyr-544. Inhibitor experiments indicate that the constitutive proliferation of Y807AB macrophages is mediated by the phosphatidylinositol 3-kinase (PI3K) and ERK1/2 pathways, whereas proliferation of WT and Y559,807AB macrophages is, in addition, contributed to by Src family kinase (SFK)-dependent pathways. Thus Tyr-807 confers sufficient kinase activity for strong CSF-1-independent proliferation, whereas Tyr-559 maintains the receptor in an inactive state. Tyr-559 phosphorylation releases this restraint and may also contribute to the CSF-1-regulated proliferative response by activating Src family kinase.
Collapse
Affiliation(s)
- Wenfeng Yu
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | | | | | | | | | |
Collapse
|
37
|
Li H, Zhang L, Yin D, Zhang Y, Miao J. Targeting phosphatidylcholine-specific phospholipase C for atherogenesis therapy. Trends Cardiovasc Med 2011; 20:172-6. [PMID: 21742273 DOI: 10.1016/j.tcm.2011.02.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Atherosclerosis, a dynamic and progressive vascular disease arising from the combination of endothelial dysfunction and inflammation, is becoming a major killer in the 21st century. Accumulating evidence implicates phosphatidylcholine-specific phospholipase C (PC-PLC) in endothelial dysfunction and several inflammation processes. In addition, in a recent study, we demonstrated that PC-PLC contributed to the progression of atherosclerosis. Considering the important roles of PC-PLC in vascular endothelial cell dysfunction and its proinflammatory properties, we propose that a pharmacological blockade of PC-PLC represents a rational approach to atherosclerosis therapy.
Collapse
Affiliation(s)
- HaiYing Li
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, Institute of Developmental Biology, School of Life Science, Shandong University, Jinan 250100, China
| | | | | | | | | |
Collapse
|
38
|
Nikolic D, Calderon L, Du L, Post SR. SR-A ligand and M-CSF dynamically regulate SR-A expression and function in primary macrophages via p38 MAPK activation. BMC Immunol 2011; 12:37. [PMID: 21736734 PMCID: PMC3141791 DOI: 10.1186/1471-2172-12-37] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Accepted: 07/07/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Inflammation is characterized by dynamic changes in the expression of cytokines, such as M-CSF, and modifications of lipids and proteins that result in the formation of ligands for Class A Scavenger Receptors (SR-A). These changes are associated with altered SR-A expression in macrophages; however, the intracellular signal pathways involved and the extent to which SR-A ligands regulate SR-A expression are not well defined. To address these questions, SR-A expression and function were examined in resident mouse peritoneal macrophages incubated with M-CSF or the selective SR-A ligand acetylated-LDL (AcLDL). RESULTS M-CSF increased SR-A expression and function, and required the specific activation of p38 MAPK, but not ERK1/2 or JNK. Increased SR-A expression and function returned to basal levels 72 hours after removing M-CSF. We next determined whether prolonged incubation of macrophages with SR-A ligand alters SR-A expression. In contrast to most receptors, which are down-regulated by chronic exposure to ligand, SR-A expression was reversibly increased by incubating macrophages with AcLDL. AcLDL activated p38 in wild-type macrophages but not in SR-A-/- macrophages, and p38 activation was specifically required for AcLDL-induced SR-A expression. CONCLUSIONS These results demonstrate that in resident macrophages SR-A expression and function can be dynamically regulated by changes in the macrophage microenvironment that are typical of inflammatory processes. In particular, our results indicate a previously unrecognized role for ligand binding to SR-A in up-regulating SR-A expression and activating p38 MAPK. In this way, SR-A may modulate inflammatory responses by enhancing macrophage uptake of modified protein/lipid, bacteria, and cell debris; and by regulating the production of inflammatory cytokines, growth factors, and proteolytic enzymes.
Collapse
Affiliation(s)
- Dejan Nikolic
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, USA
| | | | | | | |
Collapse
|
39
|
Pascual-García M, Carbó JM, León T, Matalonga J, Out R, Van Berkel T, Sarrias MR, Lozano F, Celada A, Valledor AF. Liver X receptors inhibit macrophage proliferation through downregulation of cyclins D1 and B1 and cyclin-dependent kinases 2 and 4. THE JOURNAL OF IMMUNOLOGY 2011; 186:4656-67. [PMID: 21398609 DOI: 10.4049/jimmunol.1000585] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Macrophages serve essential functions as regulators of immunity and homeostasis, and their proliferation contributes to pathogenesis of certain disorders. In this report, we show that induction of macrophage proliferation by the growth factor M-CSF is negatively modulated by agonists that activate the nuclear receptor liver X receptor (LXR), both in vitro and in vivo. Both isoforms LXR α and β are involved in the antiproliferative actions of LXR ligands in macrophages. In contrast, M-CSF does not exert negative effects on LXR-mediated gene expression. Treatment with LXR agonists results in the accumulation of macrophages in the G(0)/G(1) phase of the cell cycle without affecting ERK-1/2 phosphorylation. The use of small interfering RNA or genetically modified mice revealed that, in contrast to other cellular models, functional expression of either the cyclin-dependent kinase inhibitor p27KIP1 or the cholesterol transporters ATP-binding cassette A1 or ATP-binding cassette G1 was not required for the antiproliferative effects of LXR agonists in macrophages. Western blot analysis revealed that protein expression of key molecules that regulate progression through the cell cycle, such as cyclins D1 and B1 and cyclin-dependent kinases 2 and 4, was downregulated upon LXR activation. These observations suggest a role for LXR agonists in limiting macrophage proliferative responses associated to pathogenic disorders.
Collapse
Affiliation(s)
- Mónica Pascual-García
- Nuclear Receptor Group, Department of Physiology, School of Biology, University of Barcelona, 08028 Barcelona, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Viji V, Shobha B, Kavitha SK, Ratheesh M, Kripa K, Helen A. Betulinic acid isolated from Bacopa monniera (L.) Wettst suppresses lipopolysaccharide stimulated interleukin-6 production through modulation of nuclear factor-kappaB in peripheral blood mononuclear cells. Int Immunopharmacol 2010; 10:843-9. [PMID: 20430119 DOI: 10.1016/j.intimp.2010.04.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2009] [Revised: 04/20/2010] [Accepted: 04/20/2010] [Indexed: 10/19/2022]
Abstract
The purpose of this study was to investigate the anti-inflammatory function and mechanism(s) of action of an active component-betulinic acid isolated from Bacopa monniera. Betulinic acid, a pentacyclic triterpenoid markedly suppressed lipopolysaccharide (LPS) induced IL-6 production in blood mononuclear cells both in vivo and in vitro. Betulinic acid also prevented LPS induced nuclear translocation of p65 NF-kappaB in hPBMCs. LPS induced nuclear translocation of NF-kappaB and IL-6 production was also abolished by p38 and ERK MAPK inhibitors PD98059 and SB203580. Addition of each of these inhibitors to cell cultures along with betulinic acid caused significant downregulation of IL-6 production and inhibition of p65 NF-kappaB nuclear translocation. The inhibitory effect of both betulinic acid and the inhibitors was higher than that of cells treated with inhibitors alone. These results suggest that betulinic acid inhibited IL-6 production by preventing p65 NF-kappaB nuclear translocation and there is a possibility that this prevention of p65 nuclear translocation may involve p38 and ERK MAPKs as cross talks occur between MAPK and NF-kappaB pathways. This study provides an insight into the probable mechanism(s) underlying the anti-inflammatory and therapeutic properties of betulinic acid.
Collapse
Affiliation(s)
- V Viji
- Department of Biochemistry, University of Kerala, Kariavattom Campus, Thiruvanthapuram, Kerala 695 581, India
| | | | | | | | | | | |
Collapse
|
41
|
Kim HJ, Zou W, Ito Y, Kim SY, Chappel J, Ross FP, Teitelbaum SL. Src-like adaptor protein regulates osteoclast generation and survival. J Cell Biochem 2010; 110:201-9. [PMID: 20225239 DOI: 10.1002/jcb.22527] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Src-like adaptor protein (SLAP) is a hematopoietic adaptor containing Src homology (SH)3 and SH2 motifs and a unique carboxy terminus. Unlike c-Src, SLAP lacks a tyrosine kinase domain. We investigated the role of SLAP in osteoclast development and resorptive function. Employing SLAP-deficient mice, we find lack of the adaptor enhances in vitro proliferation of osteoclast precursors in the form of bone marrow macrophages (BMMs), without altering their survival. Furthermore, osteoclastogenic markers appear more rapidly in SLAP-/- BMMs exposed to RANK ligand (RANKL). The accelerated proliferation of M-CSF-treated, SLAP-deficient precursors is associated with enhanced ERK activation. SLAP's role as a mediator of M-CSF signaling, in osteoclastic cells, is buttressed by complexing of the adaptor protein and c-Fms in lipid rafts. Unlike c-Src, SLAP does not impact resorptive function of mature osteoclasts but induces their early apoptosis. Thus, SLAP negatively regulates differentiation of osteoclasts and proliferation of their precursors. Conversely, SLAP decreases osteoclast death by inhibiting activation of caspase 3. These counterbalancing events yield indistinguishable bones of WT and SLAP-/- mice which contain equal numbers of osteoclasts in basal and stimulated conditions.
Collapse
Affiliation(s)
- Hyun-Ju Kim
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, Missouri 63110, USA.
| | | | | | | | | | | | | |
Collapse
|
42
|
Gueller S, Goodridge HS, Niebuhr B, Xing H, Koren-Michowitz M, Serve H, Underhill DM, Brandts CH, Koeffler HP. Adaptor protein Lnk inhibits c-Fms-mediated macrophage function. J Leukoc Biol 2010; 88:699-706. [PMID: 20571037 DOI: 10.1189/jlb.0309185] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The M-CSFR (c-Fms) participates in proliferation, differentiation, and survival of macrophages and is involved in the regulation of distinct macrophage functions. Interaction with the ligand M-CSF results in phosphorylation of tyrosine residues on c-Fms, thereby creating binding sites for molecules containing SH2 domains. Lnk is a SH2 domain adaptor protein that negatively regulates hematopoietic cytokine receptors. Here, we show that Lnk binds to c-Fms. Biological and functional effects of this interaction were examined in macrophages from Lnk-deficient (KO) and WT mice. Clonogenic assays demonstrated an elevated number of M-CFUs in the bone marrow of Lnk KO mice. Furthermore, the M-CSF-induced phosphorylation of Akt in Lnk KO macrophages was increased and prolonged, whereas phosphorylation of Erk was diminished. Zymosan-stimulated production of ROS was increased dramatically in a M-CSF-dependent manner in Lnk KO macrophages. Lastly, Lnk inhibited M-CSF-induced migration of macrophages. In summary, we show that Lnk binds to c-Fms and can blunt M-CSF stimulation. Modulation of levels of Lnk in macrophages may provide a unique therapeutic approach to increase innate host defenses.
Collapse
Affiliation(s)
- Saskia Gueller
- Department of Hematology and Oncology, Johann Wolfgang Goethe University, Frankfurt, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Spallarossa P, Altieri P, Aloi C, Garibaldi S, Barisione C, Ghigliotti G, Fugazza G, Barsotti A, Brunelli C. Doxorubicin induces senescence or apoptosis in rat neonatal cardiomyocytes by regulating the expression levels of the telomere binding factors 1 and 2. Am J Physiol Heart Circ Physiol 2009; 297:H2169-81. [PMID: 19801496 DOI: 10.1152/ajpheart.00068.2009] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Low or high doses of doxorubicin induce either senescence or apoptosis, respectively, in cardiomyocytes. The mechanism by which different doses of doxorubicin may induce different stress-response cellular programs is not well understood. A recent study showed that the level of telomere dysfunction may induce senescence or apoptosis. We investigated the pathways to both apoptosis and senescence in neonatal rat cardiomyocytes and in H9c2 cells exposed to a single pulsed incubation with low or high doses of doxorubicin. High-dose doxorubicin strongly reduces TRF2 expression while enhancing TRF1 expression, and it determines early apoptosis. Low-dose doxorubicin induces downregulation of both TRF2 and TRF1, and it also increases the senescence-associated-beta-galactosidase activity, downregulates the checkpoint kinase Chk2, induces chromosomal abnormalities, and alters the cell cycle. The involvement of TRF1 and TRF2 with apoptosis and senescence was assessed by short interfering RNA interference. The cells maintain telomere dysfunction and a senescent phenotype over time and undergo late death. The increase in the phase>4N and the presence of micronuclei and anaphase bridges indicate that cells die by mitotic catastrophe. p38 modulates TRF2 expression, whereas JNK and cytoplasmic p53 regulate TRF1. Pretreatment with specific inhibitors of MAPKs and p53 may either attenuate the damage induced by doxorubicin or shift the cellular response to stress from senescence to apoptosis. In conclusion, various doses of doxorubicin induce differential regulation of TRF1 and TRF2 through p53 and MAPK, which is responsible for inducing either early apoptosis or senescence and late death due to mitotic catastrophe.
Collapse
Affiliation(s)
- Paolo Spallarossa
- Research Center of Cardiovascular Biology, Division of Cardiology, University of Genoa, Viale Benedetto XV, 6, 16132 Genoa, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Lattin JE, Greenwood KP, Daly NL, Kelly G, Zidar DA, Clark RJ, Thomas WG, Kellie S, Craik DJ, Hume DA, Sweet MJ. Beta-arrestin 2 is required for complement C1q expression in macrophages and constrains factor-independent survival. Mol Immunol 2009; 47:340-7. [PMID: 19783052 DOI: 10.1016/j.molimm.2009.09.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Revised: 09/03/2009] [Accepted: 09/03/2009] [Indexed: 12/22/2022]
Abstract
The beta-arrestins (ARRB1 and ARRB2) regulate G-protein coupled receptor (GPCR) dependent- and independent-signaling pathways and are ubiquitously expressed. Here we show that ARRB2 mRNA and protein expression is enriched in macrophages, and that it regulates complement C1q expression and cell survival. Basal and Toll-like receptor (TLR) inducible expression of mRNAs encoding the complement subcomponents C1qa, C1qb and C1qc was greatly reduced in bone marrow-derived macrophages (BMM) from ARRB2-deficient, but not ARRB1-deficient mice, while factor-independent survival of ARRB2(-/-) BMM was enhanced compared to wildtype BMM. TatARRB2(23), a cell-permeable peptide that contains the MAPK JNK-binding motif from within the ARRB2 C-domain, impaired ARRB2 interaction with JNK3, down-regulated C1q expression and permitted factor-independent survival in BMM, thus suggesting that this peptide antagonises ARRB2 function in macrophages. In addition, TatARRB2(23) transiently activated the phosphorylation of JNK and ERK, but not p38 in BMM. These data imply that ARRB2 acts to limit JNK/ERK activation and survival in macrophages, but is required for basal and TLR-inducible complement C1q expression. Given that loss of C1q function is strongly associated with the development of systemic lupus erythematosus, ARRB2 may act to limit the development of autoimmune disease.
Collapse
Affiliation(s)
- Jane E Lattin
- The University of Queensland, Institute for Molecular Bioscience, QLD 4072, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Casals-Casas C, Alvarez E, Serra M, de la Torre C, Farrera C, Sánchez-Tilló E, Caelles C, Lloberas J, Celada A. CREB and AP-1 activation regulates MKP-1 induction by LPS or M-CSF and their kinetics correlate with macrophage activation versus proliferation. Eur J Immunol 2009; 39:1902-13. [PMID: 19585511 DOI: 10.1002/eji.200839037] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
MAPK phosphatase-1 (MKP-1) is a protein phosphatase that plays a crucial role in innate immunity. This phosphatase inactivates ERK1/2, which are involved in two opposite functional activities of the macrophage, namely proliferation and activation. Here we found that although macrophage proliferation and activation induce MKP-1 with different kinetics, gene expression is mediated by the proximal promoter sequences localized between -380 and -180 bp. Mutagenesis experiments of the proximal element determined that CRE/AP-1 is required for LPS- or M-CSF-induced activation of the MKP-1 gene. Moreover, the results from gel shift analysis and chromatin immunoprecipitation indicated that c-Jun and CREB bind to the CRE/AP-1 box. The distinct kinetics shown by M-CSF and LPS correlates with the induction of JNK and c-jun, as well as the requirement for Raf-1. The signal transduction pathways that activate the induction of MKP-1 correlate kinetically with induction by M-CSF and LPS.
Collapse
|
46
|
Feng D, Ling WH, Duan RD. Lycopene suppresses LPS-induced NO and IL-6 production by inhibiting the activation of ERK, p38MAPK, and NF-κB in macrophages. Inflamm Res 2009; 59:115-21. [DOI: 10.1007/s00011-009-0077-8] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2009] [Accepted: 07/27/2009] [Indexed: 11/29/2022] Open
|
47
|
Sebastián C, Herrero C, Serra M, Lloberas J, Blasco MA, Celada A. Telomere shortening and oxidative stress in aged macrophages results in impaired STAT5a phosphorylation. THE JOURNAL OF IMMUNOLOGY 2009; 183:2356-64. [PMID: 19605693 DOI: 10.4049/jimmunol.0901131] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Macrophages are an essential component of both innate and adaptive immunity, and altered function of these cells with aging may play a key role in immunosenescence. To determine the effect of aging on macrophages, we produced bone marrow-derived macrophages in vitro. In these conditions, we analyzed the effect of aging on macrophages without the influence of other cell types that may be affected by aging. We showed that telomeres shorten with age in macrophages leading to a decreased GM-CSF but not M-CSF-dependent proliferation of these cells as a result of decreased phosphorylation of STAT5a. Macrophages from aged mice showed increased susceptibility to oxidants and an accumulation of intracellular reactive oxygen species. In these macrophages STAT5a oxidation was reduced, which led to the decreased phosphorylation observed. Interestingly, the same cellular defects were found in macrophages from telomerase knockout (Terc(-/-)) mice suggesting that telomere loss is the cause for the enhanced oxidative stress, the reduced Stat5a oxidation and phosphorylation and, ultimately, for the impaired GM-CSF-dependent macrophage proliferation.
Collapse
Affiliation(s)
- Carlos Sebastián
- Institute for Research in Biomedicine and University of Barcelona, Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
48
|
Wilms H, Schwabedissen B, Sievers J, Lucius R. Erythropoietin does not attenuate cytokine production and inflammation in microglia--implications for the neuroprotective effect of erythropoietin in neurological diseases. J Neuroimmunol 2009; 212:106-11. [PMID: 19464738 DOI: 10.1016/j.jneuroim.2009.04.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2008] [Revised: 03/31/2009] [Accepted: 04/23/2009] [Indexed: 12/18/2022]
Abstract
Erythropoietin is a hematopoietic cytokine which is also produced in the brain under hypoxia. Since this pathology is associated with glial cell activation and release of cytotoxic molecules, we investigated the expression of EPO receptors (EPO-R) and effects of erythropoietin on microglial cell functions in vitro using RT-PCR, Western immunoblotting, nitric oxide measurement, tumor necrosis factor-alpha-(TNF-alpha)-ELISA and gel shift assay analyses. Furthermore, we examined if erythropoietin could modulate proliferation of microglia. As shown by reverse transcription-polymerase chain reaction and immunocytochemistry, rat microglial cells and the murine microglia cell line BV-2 express the EPO-R. However, EPO showed no effect on the release of the proinflammatory mediators' nitric oxide and TNF-alpha. Moreover, EPO was not able to reduce the LPS (lipopolysaccharide) stimulated translocation of the proinflammatory transcription factor NF-kappaB into the nucleus of murine microglia, but induced (3)H-thymidine incorporation into DNA of microglial cells. These results show that microglia are target cells for erythropoietin which possesses mitogenic, but not anti-inflammatory effects on microglia. Therefore, the well-documented neuroprotective effects of erythropoietin could not be ascribed to an anti-inflammatory effect on microglia.
Collapse
Affiliation(s)
- Henrik Wilms
- Department of Neurology, University of Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany.
| | | | | | | |
Collapse
|
49
|
Guo LY, Hung TM, Bae K, Jang S, Shin EM, Chung JW, Kang SS, Kim HP, Kim YS. Effects of schisandrin on transcriptional factors in lipopolysaccharide-pretreated macrophages. Arch Pharm Res 2009; 32:399-405. [DOI: 10.1007/s12272-009-1313-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2008] [Revised: 03/04/2009] [Accepted: 03/06/2009] [Indexed: 11/30/2022]
|
50
|
Corsaro A, Thellung S, Chiovitti K, Villa V, Simi A, Raggi F, Paludi D, Russo C, Aceto A, Florio T. Dual modulation of ERK1/2 and p38 MAP kinase activities induced by minocycline reverses the neurotoxic effects of the prion protein fragment 90-231. Neurotox Res 2009; 15:138-54. [PMID: 19384577 DOI: 10.1007/s12640-009-9015-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2008] [Revised: 08/15/2008] [Accepted: 09/18/2008] [Indexed: 11/27/2022]
Abstract
Several in vitro and in vivo studies addressed the identification of molecular determinants of the neuronal death induced by PrP(Sc) or related peptides. We developed an experimental model to assess PrP(Sc) neurotoxicity using a recombinant polypeptide encompassing amino acids 90-231 of human PrP (hPrP90-231) that corresponds to the protease-resistant core of PrP(Sc) identified in prion-infected brains. By means of mild thermal denaturation, we can convert hPrP90-231 from a PrP(C)-like conformation into a PrP(Sc)-like structure. In virtue of these structural changes, hPrP90-231 powerfully affected the survival of SH-SY5Y cells, inducing caspase 3 and p38-dependent apoptosis, while in the native alpha-helix-rich conformation, hPrP90-231 did not induce cell toxicity. The aim of this study was to identify drugs able to block hPrP90-231 neurotoxic effects, focusing on minocycline, a tetracycline with known neuroprotective activity. hPrP90-231 caused a caspase 3-dependent apoptosis via the blockade of ERK1/2 activation and the subsequent activation of p38 MAP kinase. We propose that hPrP90-231-induced apoptosis is dependent on the inhibition of ERK1/2 responsiveness to neurotrophic factors, removing a tonic inhibition of p38 activity and resulting in caspase 3 activation. Minocycline prevented hPrP90-231-induced toxicity interfering with this mechanism: the pretreatment with this tetracycline restored ERK1/2 activity and reverted p38 and caspase 3 activities. The effects of minocycline were not mediated by the prevention of hPrP90-231 structural changes or cell internalization (differently from Congo Red). In conclusion, minocycline elicits anti-apoptotic effects against the neurotoxic activity of hPrP90-231 and these effects are mediated by opposite modulation of ERK1/2 and p38 MAP kinase activities.
Collapse
Affiliation(s)
- Alessandro Corsaro
- Laboratory of Pharmacology and Neuroscience, Department of Oncology Biology and Genetics, University of Genova, Viale Benedetto XV, 2, Genova 16132, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|