1
|
Khot S, Tackley G, Choy E. How to Distinguish Non-Inflammatory from Inflammatory Pain in RA? Curr Rheumatol Rep 2024; 26:403-413. [PMID: 39120749 PMCID: PMC11527911 DOI: 10.1007/s11926-024-01159-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2024] [Indexed: 08/10/2024]
Abstract
PURPOSE OF THE REVIEW Managing non-inflammatory pain in rheumatoid arthritis (RA) can be a huge burden for the rheumatologist. Pain that persists despite optimal RA treatment is extremely challenging for patient and physician alike. Here, we outline the latest research relevant to distinguishing non-inflammatory from inflammatory RA pain and review the current understanding of its neurobiology and management. RECENT FINDINGS Nociplastic pain is a recently introduced term by the international pain community. Its definition encompasses the non-inflammatory pain of RA and describes pain that is not driven by inflamed joints or compromised nerves, but that is instead driven by a functional reorganisation of the central nervous system (CNS). Insights from all areas of nociplastic pain research, including fibromyalgia, support a personalised pain management approach for non-inflammatory pain of RA, with evidence-based guidelines favouring use of non-pharmacological interventions. Future developments include novel CNS targeting pharmacotherapeutic approaches to treat nociplastic pain.
Collapse
Affiliation(s)
- Sharmila Khot
- Department of Anaesthesia, Intensive Care and Pain Medicine, Cardiff and Vale University Health Board, Cardiff CF14 4XW and Cardiff University Brain Research Imaging Centre (CUBRIC), Cardiff University, Maindy Road, Cardiff, Wales, CF24 4HQ, UK.
| | - George Tackley
- Cardiff University Brain Research Imaging Centre (CUBRIC), Cardiff University, Maindy Road, Cardiff, Wales, CF24 4HQ, UK
| | - Ernest Choy
- Head of Rheumatology and Translational Research at the Division of Infection and Immunity and Director of the Cardiff Regional Experimental Arthritis Treatment and Evaluation (CREATE) Centre at Cardiff University School of Medicine, Cardiff, Wales, UK, CF14 4YS
| |
Collapse
|
2
|
Lossavaro PKDMB, Felipe JL, Lencina JDS, Bonfá IS, de Souza KFS, Machado LL, Fernandes MML, Ferreira JV, Souza MIL, Candeloro L, Kassuya CAL, Paredes-Gamero EJ, Parisotto EB, Toffoli-Kadri MC, Silva-Filho SE. Antiarthritic and Antinociceptive Properties of Ylang-Ylang ( Cananga odorata) Essential Oil in Experimental Models. Curr Issues Mol Biol 2024; 46:9033-9046. [PMID: 39194751 DOI: 10.3390/cimb46080534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/14/2024] [Accepted: 08/16/2024] [Indexed: 08/29/2024] Open
Abstract
The aim of this study was to evaluate the effect of ylang-ylang (Cananga odorata) essential oil (YEO) on models of experimental arthritis, persistent inflammation, and nociception in mice. YEO treatment at doses of 100 and 200 mg/kg reduced the infiltration of leukocytes into the joint cavities of mice submitted to zymosan-induced arthritis 6 h and 7 days after arthritis induction. At these doses, YEO treatment reduced the formation of joint edema 4 and 6 h after arthritis induction, and at a dose of 200 mg/kg, YEO treatment reduced mechanical hyperalgesia 3 and 4 h after arthritis induction. At the dose of 200 mg/kg, YEO treatment reduced interleukin-6 (IL-6) levels and cartilage destruction in the zymosan-induced arthritis model, and reduced edema formation and mechanical hyperalgesia in the model of persistent inflammation (21 days) induced by complete Freund's adjuvant (CFA) in mice. YEO treatment at a dose of 200 mg/kg reduced the nociceptive response in experimental models of nociception induced by acetic acid and formalin. The YEO treatment reduced inflammatory parameters in the experimental arthritis model, and presented antiarthritic, anti-hyperalgesic, antinociceptive, and anti-inflammatory properties.
Collapse
Affiliation(s)
| | - Josyelen Lousada Felipe
- Pharmaceutical Sciences, Food and Nutrition College, Federal University of Mato Grosso do Sul, Campo Grande 79070-900, MS, Brazil
| | - Joyce Dos Santos Lencina
- Pharmaceutical Sciences, Food and Nutrition College, Federal University of Mato Grosso do Sul, Campo Grande 79070-900, MS, Brazil
| | - Iluska Senna Bonfá
- Pharmaceutical Sciences, Food and Nutrition College, Federal University of Mato Grosso do Sul, Campo Grande 79070-900, MS, Brazil
| | - Kamylla Fernanda Souza de Souza
- Pharmaceutical Sciences, Food and Nutrition College, Federal University of Mato Grosso do Sul, Campo Grande 79070-900, MS, Brazil
| | - Lucas Luiz Machado
- Pharmaceutical Sciences, Food and Nutrition College, Federal University of Mato Grosso do Sul, Campo Grande 79070-900, MS, Brazil
| | - Mila Marluce Lima Fernandes
- Pharmaceutical Sciences, Food and Nutrition College, Federal University of Mato Grosso do Sul, Campo Grande 79070-900, MS, Brazil
| | - João Victor Ferreira
- Pharmaceutical Sciences, Food and Nutrition College, Federal University of Mato Grosso do Sul, Campo Grande 79070-900, MS, Brazil
| | - Maria Inês Lenz Souza
- Biosciences Institute, Federal University of Mato Grosso do Sul, Campo Grande 79070-900, MS, Brazil
| | - Luciane Candeloro
- Biosciences Institute, Federal University of Mato Grosso do Sul, Campo Grande 79070-900, MS, Brazil
| | | | - Edgar Julian Paredes-Gamero
- Pharmaceutical Sciences, Food and Nutrition College, Federal University of Mato Grosso do Sul, Campo Grande 79070-900, MS, Brazil
| | - Eduardo Benedetti Parisotto
- Pharmaceutical Sciences, Food and Nutrition College, Federal University of Mato Grosso do Sul, Campo Grande 79070-900, MS, Brazil
| | - Mônica Cristina Toffoli-Kadri
- Pharmaceutical Sciences, Food and Nutrition College, Federal University of Mato Grosso do Sul, Campo Grande 79070-900, MS, Brazil
| | - Saulo Euclides Silva-Filho
- Pharmaceutical Sciences, Food and Nutrition College, Federal University of Mato Grosso do Sul, Campo Grande 79070-900, MS, Brazil
| |
Collapse
|
3
|
Daniele-Silva A, Parente AMES, de Sousa Ferreira S, Pontes da Silva D, Torres-Rêgo M, Cavalcanti FF, Assunção Ferreira MR, de Freitas Fernandes-Pedrosa M, Lira Soares LA. In vitro and in vivo anti-inflammatory and antiophidic effects of the extract and fraction of Eugenia uniflora. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117223. [PMID: 37748636 DOI: 10.1016/j.jep.2023.117223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/19/2023] [Accepted: 09/22/2023] [Indexed: 09/27/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Eugenia uniflora Linn, popularly known as 'pitanga', is a native plant endemic to Brazil that belongs to the Myrtaceae family. Its traditional use (leaves infusion) has been reported for the treatment of different diseases, including hypertension, inflammation, and as a diuretic agent. Considering the snakebite problem and the rich molecule repertoire of this herbal species, studies that evaluate its antiophidic potential are relevant for a broad social impact. AIM OF THE STUDY This approach aims to evaluate the anti-inflammatory and antiophidic potential in vitro and in vivo of the extract (aqueous) and a fraction (ethyl acetate) of E. uniflora leaves against Bothrops leucurus and Bothrops brazili venoms. MATERIALS AND METHODS Extract and fraction from E. uniflora leaves were obtained by turbo-extraction and partitioning. The cytotoxicity was assayed on normal cell lines (Vero E6 and 3T3) using the 3-methyl-[4-5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide method. The anti-inflammatory activity of the aqueous extract was analyzed in vivo in the zymosan-induced air pouch model, and the leukocytes migration and other molecular inflammatory mediators quantified (myeloperoxidase, total protein, pro-inflammatory cytokine, malondialdehyde, and glutathione). In vitro, the antiophidic effect was evaluated by the ability of the E. uniflora extract and fraction to inhibit the enzymatic action (proteolytic, phospholipase A2, and hyaluronidase) of B. leucurus and B. brazili venoms. In addition, the antiophidic action in vivo was investigated after treatment with E. uniflora extract and fraction (50, 100, and 200 mg/kg) in the B. leucurus venom-induced paw edema with an evaluation of the antiedematogenic effect and quantification of myeloperoxidase (MPO) and pro-inflammatory cytokine levels. RESULTS The E. uniflora leaves extract (7.8-125 mg/mL) revealed no toxicity in cell culture, but reduced MTT by 47% at the highest concentration (250 mg/mL) in Vero E6 cells. In contrast, the E. uniflora fraction (7.8-250 mg/mL) showed no cytotoxicity for both cell lines. In the air pouch model, E. uniflora leaves extract demonstrated anti-inflammatory activity, reducing cell migration, MPO activity, protein, malondialdehyde, and proinflammatory cytokines, and increased glutathione levels. Evaluating the antiophidic action in vitro, E. uniflora extract and fraction inhibited the proteolytic, phospholipase, and hyaluronidase effects of B. leucurus and B. brazili venoms at low concentrations. In addition, the extract and fraction also demonstrated in vivo antiophidic activity by reducing edema in the first 0.5 h after treatment, besides reducing MPO and pro-inflammatory cytokines levels. CONCLUSION E. uniflora leaves extract showed cytotoxicity only at the highest concentration while the fraction revealed no toxic effect in vitro. This approach showed for the first time that the aqueous extract and ethyl acetate fraction of E. uniflora leaves has similar antiophidic action in vitro and in vivo, with antiedematogenic and anti-inflammatory effects and the ability to inhibit the enzymatic action of B. leucurus and B. brazili venoms. Therefore, this study points to the presence of bioactive components in the leaves of E. uniflora useful for the treatment of inflammatory disorders and ophidian accidents, expanding the therapeutic potential of this herbal species.
Collapse
Affiliation(s)
- Alessandra Daniele-Silva
- Laboratory of Pharmacognosy, Department of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, Brazil; Laboratory of Technology and Pharmaceutical Biotechnology, Faculty of Pharmacy, Federal University of Rio Grande do Norte, Brazil
| | - Adriana Marina E Silva Parente
- Laboratory of Pharmacognosy, Department of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, Brazil; Laboratory of Technology and Pharmaceutical Biotechnology, Faculty of Pharmacy, Federal University of Rio Grande do Norte, Brazil
| | - Sarah de Sousa Ferreira
- Laboratory of Technology and Pharmaceutical Biotechnology, Faculty of Pharmacy, Federal University of Rio Grande do Norte, Brazil
| | - Diana Pontes da Silva
- Laboratory of Technology and Pharmaceutical Biotechnology, Faculty of Pharmacy, Federal University of Rio Grande do Norte, Brazil
| | - Manoela Torres-Rêgo
- Laboratory of Technology and Pharmaceutical Biotechnology, Faculty of Pharmacy, Federal University of Rio Grande do Norte, Brazil; Graduate Program of Chemistry, Chemistry Institute, Federal University of Rio Grande do Norte, Brazil
| | - Felipe França Cavalcanti
- Laboratory of Technology and Pharmaceutical Biotechnology, Faculty of Pharmacy, Federal University of Rio Grande do Norte, Brazil; Graduate Program of Chemistry, Chemistry Institute, Federal University of Rio Grande do Norte, Brazil
| | | | | | - Luiz Alberto Lira Soares
- Laboratory of Pharmacognosy, Department of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, Brazil.
| |
Collapse
|
4
|
Felipe JL, Bonfá IS, Lossavaro PKMB, Lencina JS, B Carvalho D, Candeloro L, Ferreira GIS, das Neves AR, Souza MIL, Silva-Filho SE, Baroni ACM, Toffoli-Kadri MC. 1,4-Diaryl-1,2,3-triazole neolignan-celecoxib hybrids inhibit experimental arthritis induced by zymosan. Inflammopharmacology 2023; 31:3227-3241. [PMID: 37806984 DOI: 10.1007/s10787-023-01345-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 09/14/2023] [Indexed: 10/10/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that causes cartilage damage. Anti-inflammatories are widely used in the management of RA, but they can have side effects such as gastrointestinal and/or cardiovascular disorders. Studies published by our group showed that the synthesis of hybrid triazole analogs neolignan-celecoxib containing the substituent groups sulfonamide (L15) or carboxylic acid (L18) exhibited anti-inflammatory activity in an acute model of inflammation, inhibited expression of P-selectin related to platelet activation and did not induce gastric ulcer, minimizing the related side effects. In continuation, the present study evaluated the anti-inflammatory effects of these analogs in an experimental model of arthritis and on the functions of one of the important cells in this process, macrophages. Mechanical hyperalgesia, joint edema, leukocyte recruitment to the joint and damage to cartilage in experimental arthritis and cytotoxicity, spread of disease, phagocytic activity and nitric oxide (NO) and hydrogen peroxide production by macrophages were evaluated. Pre-treatment with L15 and L18 reduced mechanical hyperalgesia, joint edema and the influx of leukocytes into the joint cavity after different periods of the stimulus. The histological evaluation of the joint showed that L15 and L18 reduced cartilage damage and there was no formation of rheumatoid pannus. Furthermore, L15 and L18 were non-cytotoxic. The analogs inhibited the spreading, the production of NO and hydrogen peroxide. L15 decreased the phagocytosis. Therefore, L15 and L18 may be potential therapeutic prototypes to treat chronic inflammatory diseases such as RA.
Collapse
Affiliation(s)
- Josyelen L Felipe
- Laboratory of Pharmacology and Inflammation, Faculty of Pharmaceutical Sciences, Food and Nutrition, Federal University of Mato Grosso Do Sul, UFMS, Campo Grande, MS, 79070-900, Brazil
| | - Iluska S Bonfá
- Laboratory of Pharmacology and Inflammation, Faculty of Pharmaceutical Sciences, Food and Nutrition, Federal University of Mato Grosso Do Sul, UFMS, Campo Grande, MS, 79070-900, Brazil
| | - Paloma K M B Lossavaro
- Laboratory of Pharmacology and Inflammation, Faculty of Pharmaceutical Sciences, Food and Nutrition, Federal University of Mato Grosso Do Sul, UFMS, Campo Grande, MS, 79070-900, Brazil
| | - Joyce S Lencina
- Laboratory of Pharmacology and Inflammation, Faculty of Pharmaceutical Sciences, Food and Nutrition, Federal University of Mato Grosso Do Sul, UFMS, Campo Grande, MS, 79070-900, Brazil
| | - Diego B Carvalho
- Laboratory of Synthesis and Medicinal Chemistry, Faculty of Pharmaceutical Sciences, Food and Nutrition, Federal University of Mato Grosso Do Sul, UFMS, Campo Grande, MS, Brazil
| | - Luciane Candeloro
- Laboratory of Hystology, Institute of Biosciences, Federal University of Mato Grosso Do Sul, Campo Grande, MS, Brazil
| | - Giovanni I S Ferreira
- Laboratory of Hystology, Institute of Biosciences, Federal University of Mato Grosso Do Sul, Campo Grande, MS, Brazil
| | - Amarith R das Neves
- Laboratory of Synthesis and Medicinal Chemistry, Faculty of Pharmaceutical Sciences, Food and Nutrition, Federal University of Mato Grosso Do Sul, UFMS, Campo Grande, MS, Brazil
| | - Maria Inês L Souza
- Department of Biophysiopharmacology, Institute of Biosciences, Federal University of Mato Grosso Do Sul, Campo Grande, MS, Brazil
| | - Saulo E Silva-Filho
- Laboratory of Pharmacology and Inflammation, Faculty of Pharmaceutical Sciences, Food and Nutrition, Federal University of Mato Grosso Do Sul, UFMS, Campo Grande, MS, 79070-900, Brazil
| | - Adriano C M Baroni
- Laboratory of Synthesis and Medicinal Chemistry, Faculty of Pharmaceutical Sciences, Food and Nutrition, Federal University of Mato Grosso Do Sul, UFMS, Campo Grande, MS, Brazil.
| | - Mônica C Toffoli-Kadri
- Laboratory of Pharmacology and Inflammation, Faculty of Pharmaceutical Sciences, Food and Nutrition, Federal University of Mato Grosso Do Sul, UFMS, Campo Grande, MS, 79070-900, Brazil.
| |
Collapse
|
5
|
Rodrigues FF, Lino CI, Oliveira VLS, Zaidan I, Melo ISF, Braga AV, Costa SOAM, Morais MI, Barbosa BCM, da Costa YFG, Moreira NF, Alves MS, Braga AD, Carneiro FS, Carvalho AFS, Queiroz-Junior CM, Sousa LP, Amaral FA, Oliveira RB, Coelho MM, Machado RR. A clindamycin acetylated derivative with reduced antibacterial activity inhibits articular hyperalgesia and edema by attenuating neutrophil recruitment, NF-κB activation and tumor necrosis factor-α production. Int Immunopharmacol 2023; 122:110609. [PMID: 37429145 DOI: 10.1016/j.intimp.2023.110609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 06/22/2023] [Accepted: 07/03/2023] [Indexed: 07/12/2023]
Abstract
We recently demonstrated that clindamycin exhibits activities in acute and chronic models of pain and inflammation. In the present study, we investigated the effects of clindamycin and a clindamycin acetylated derivative (CAD) in models of acute joint inflammation and in a microbiological assay. Joint inflammation was induced in mice by intraarticular (i.a.) injection of zymosan or lipopolysaccharide (LPS). Clindamycin or CAD were administered via the intraperitoneal route 1 h before zymosan or LPS. Paw withdrawal threshold, joint diameter, histological changes, neutrophil recruitment, tumor necrosis factor-α (TNF-α) production and phosphorylation of the IκBα and NF-κB/p65 were evaluated. In vitro assays were used to measure the antibacterial activity of clindamycin and CAD and also their effects on zymosan-induced TNF-α production by RAW264.7 macrophages. Clindamycin exhibited activity against Staphylococcus aureus and Salmonella Typhimurium ATCC® strains at much lower concentrations than CAD. Intraarticular injection of zymosan or LPS induced articular hyperalgesia, edema and neutrophil infiltration in the joints. Zymosan also induced histological changes, NF-κB activation and TNF-α production. Responses induced by zymosan and LPS were inhibited by clindamycin (200 and 400 mg/kg) or CAD (436 mg/kg). Both clindamycin and CAD inhibited in vitro TNF-α production by macrophages. In summary, we provided additional insights of the clindamycin immunomodulatory effects, whose mechanism was associated with NF-κB inhibition and reduced TNF-α production. Such effects were extended to a clindamycin derivative with reduced antibacterial activity, indicating that clindamycin derivatives should be investigated as candidates to drugs that could be useful in the management of inflammatory and painful conditions.
Collapse
Affiliation(s)
- Felipe F Rodrigues
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, Belo Horizonte, MG, CEP 31270-901, Brasil
| | - Cleudiomar I Lino
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, Belo Horizonte, MG, CEP 31270-901, Brasil
| | - Vívian L S Oliveira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, Belo Horizonte, MG, CEP 31270-901, Brasil
| | - Isabella Zaidan
- Laboratório de Sinalização na Inflamação, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais. Avenida Antônio Carlos, 6627, Belo Horizonte, MG, CEP 31270-901, Brasil
| | - Ivo S F Melo
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, Belo Horizonte, MG, CEP 31270-901, Brasil
| | - Alysson V Braga
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, Belo Horizonte, MG, CEP 31270-901, Brasil
| | - Sarah O A M Costa
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, Belo Horizonte, MG, CEP 31270-901, Brasil
| | - Marcela I Morais
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, Belo Horizonte, MG, CEP 31270-901, Brasil
| | - Bárbara C M Barbosa
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, Belo Horizonte, MG, CEP 31270-901, Brasil
| | - Ygor F G da Costa
- Laboratório de Bioatividade Celular e Molecular, Centro de Pesquisas Farmacêuticas, Faculdade de Farmácia, Universidade Federal de Juiz de Fora, Rua José Lourenço Kelmer s/n°, Juiz de Fora, MG, CEP 36036-900, Brasil
| | - Nicole F Moreira
- Laboratório de Bioatividade Celular e Molecular, Centro de Pesquisas Farmacêuticas, Faculdade de Farmácia, Universidade Federal de Juiz de Fora, Rua José Lourenço Kelmer s/n°, Juiz de Fora, MG, CEP 36036-900, Brasil
| | - Maria S Alves
- Laboratório de Bioatividade Celular e Molecular, Centro de Pesquisas Farmacêuticas, Faculdade de Farmácia, Universidade Federal de Juiz de Fora, Rua José Lourenço Kelmer s/n°, Juiz de Fora, MG, CEP 36036-900, Brasil
| | - Amanda D Braga
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, Belo Horizonte, MG, CEP 31270-901, Brasil
| | - Fernanda S Carneiro
- Laboratório de Sinalização na Inflamação, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais. Avenida Antônio Carlos, 6627, Belo Horizonte, MG, CEP 31270-901, Brasil
| | - Antônio F S Carvalho
- Laboratório de Sinalização na Inflamação, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais. Avenida Antônio Carlos, 6627, Belo Horizonte, MG, CEP 31270-901, Brasil
| | - Celso M Queiroz-Junior
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, Belo Horizonte, MG, CEP 31270-901, Brasil
| | - Lirlândia P Sousa
- Laboratório de Sinalização na Inflamação, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais. Avenida Antônio Carlos, 6627, Belo Horizonte, MG, CEP 31270-901, Brasil
| | - Flávio A Amaral
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, Belo Horizonte, MG, CEP 31270-901, Brasil
| | - Renata B Oliveira
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, Belo Horizonte, MG, CEP 31270-901, Brasil
| | - Márcio M Coelho
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, Belo Horizonte, MG, CEP 31270-901, Brasil
| | - Renes R Machado
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, Belo Horizonte, MG, CEP 31270-901, Brasil.
| |
Collapse
|
6
|
Staurengo-Ferrari L, Deng L, Chiu IM. Interactions between nociceptor sensory neurons and microbial pathogens in pain. Pain 2022; 163:S57-S68. [PMID: 36252233 PMCID: PMC9586460 DOI: 10.1097/j.pain.0000000000002721] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/21/2022] [Indexed: 02/07/2023]
Affiliation(s)
- Larissa Staurengo-Ferrari
- Harvard Medical School, Blavatnik Institute, Department of Immunology, Boston, Massachusetts, United States of America
| | - Liwen Deng
- Harvard Medical School, Blavatnik Institute, Department of Immunology, Boston, Massachusetts, United States of America
| | - Isaac M. Chiu
- Harvard Medical School, Blavatnik Institute, Department of Immunology, Boston, Massachusetts, United States of America
| |
Collapse
|
7
|
Torres-Rêgo M, Aquino-Vital AKSD, Cavalcanti FF, Rocha EEA, Daniele-Silva A, Furtado AA, Silva DPD, Ururahy MAG, Silveira ER, Fernandes-Pedrosa MDF, Araújo RM. Phytochemical analysis and preclinical toxicological, antioxidant, and anti-inflammatory evaluation of hydroethanol extract from the roots of Harpalyce brasiliana Benth (Leguminosae). JOURNAL OF ETHNOPHARMACOLOGY 2022; 294:115364. [PMID: 35551979 DOI: 10.1016/j.jep.2022.115364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 04/28/2022] [Accepted: 05/06/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Harpalyce brasiliana Benth (Leguminosae) is a shrub endemic to Brazil, popularly known as "snake's root." This species is used in folk medicine for the treatment of inflammation and snakebites. However, up to now there is no scientific research to justify its popular use. The study aimed to characterize the phytochemical profile of the hydroethanol extract from the roots of H. brasiliana (Hb), to evaluate its antioxidant and anti-inflammatory potential, as well as to investigate its cytotoxicity and acute toxicity. MATERIALS AND METHODS The extract was obtained by maceration method using a solution of ethanol:water (70: 30, v/v). The phytochemical profile was obtained by liquid chromatography coupled to mass spectrometry. The cytotoxicity of extract (31-2000 μg/mL) was evaluated in vitro, by the 3-methyl-[4-5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) method using murine macrophage and fibroblast cell lines (RAW 247.6 and 3T3, respectively) and by the hemolytic assay. For the in vivo acute toxicity, the extract (2000 mg/kg) was administered and after 14 days the weight (body and organs) and hematological and biochemical parameters were analyzed. Chemical free radical scavenging effect of the extract (125-2000 μg/mL) was investigated through diphenylpicryl hydrazine reduction, total antioxidant capacity, reducing power, hydroxyl radical scavenging, and iron and copper chelating assays. In vitro anti-inflammatory effect of the extract (125, 500, and 2000 μg/mL) was demonstrated through of nitric oxide (NO) analyzed in lipopolysaccharides stimulated RAW 264.7 cells. In vivo anti-inflammatory activities were evaluated in carrageenan-induced paw edema and zymosan-air-pouch models, with gavage administration (post-treatment) of extract at 100, 200, and 400 mg/kg. For the first animal model, the anti-edematogenic activity and myeloperoxidase (MPO) levels were investigated, while in the zymosan-air-pouch model the leukocyte number, MPO, total protein and pro-inflammatory cytokine (IL-1β, IL-6, and TNF-α) levels were quantified. In addition, the oxidative parameters such as malondialdehyde (MDA) and reduced glutathione (GSH) were determined. RESULTS The phytochemical profile revealed the presence of 20 compounds, mainly prenylated and geranylated pterocarpans. The extract demonstrated no cytotoxicity in erythrocytes, macrophages and fibroblasts cells at the tested concentrations, as well as no sign of toxicity and mortality or significant alterations on the hematological and biochemical parameters in the acute toxicity model. The extract was also able to neutralize chemical free radicals, with copper and iron chelating effect. For the NO dosage, the extract evidenced the reduction of expression of NO after the administration of the extract (500 and 2000 μg/mL). The edematogenic model revealed a decrease in paw edema and MPO level, while the zymosan-air-pouch model evidenced a reduction of leukocyte number (especially of polymorphornuclears), MPO production, and total protein and cytokine levels, and demonstrated the antioxidant effect through a decrease in MDA and increase in GSH parameters. CONCLUSION This approach demonstrates for the first time that Hb is not cytotoxic, has low acute toxicity, and possesses antioxidant and anti-inflammatory properties in preclinical analyses, corroborating its popular use.
Collapse
Affiliation(s)
- Manoela Torres-Rêgo
- Graduate Program of Chemistry, Chemistry Institute, Federal University of Rio Grande do Norte, Senador Salgado Filho Avenue, 3000, Lagoa Nova, Natal, 59072-970, Brazil; Laboratory of Technology and Pharmaceutical Biotechnology (Tecbiofar), College of Pharmacy, Federal University of Rio Grande do Norte, General Gustavo Cordeiro de Farias Street, S/N, Petrópolis, Natal, 59012-570, Brazil.
| | - Ana Karoline Silva de Aquino-Vital
- Graduate Program of Chemistry, Chemistry Institute, Federal University of Rio Grande do Norte, Senador Salgado Filho Avenue, 3000, Lagoa Nova, Natal, 59072-970, Brazil.
| | - Felipe França Cavalcanti
- Graduate Program of Chemistry, Chemistry Institute, Federal University of Rio Grande do Norte, Senador Salgado Filho Avenue, 3000, Lagoa Nova, Natal, 59072-970, Brazil.
| | - Enos Emanuel Azevedo Rocha
- Graduate Program of Chemistry, Chemistry Institute, Federal University of Rio Grande do Norte, Senador Salgado Filho Avenue, 3000, Lagoa Nova, Natal, 59072-970, Brazil.
| | - Alessandra Daniele-Silva
- Laboratory of Technology and Pharmaceutical Biotechnology (Tecbiofar), College of Pharmacy, Federal University of Rio Grande do Norte, General Gustavo Cordeiro de Farias Street, S/N, Petrópolis, Natal, 59012-570, Brazil.
| | - Allanny Alves Furtado
- Laboratory of Technology and Pharmaceutical Biotechnology (Tecbiofar), College of Pharmacy, Federal University of Rio Grande do Norte, General Gustavo Cordeiro de Farias Street, S/N, Petrópolis, Natal, 59012-570, Brazil.
| | - Diana Pontes da Silva
- Laboratory of Technology and Pharmaceutical Biotechnology (Tecbiofar), College of Pharmacy, Federal University of Rio Grande do Norte, General Gustavo Cordeiro de Farias Street, S/N, Petrópolis, Natal, 59012-570, Brazil.
| | - Marcela Abbott Galvão Ururahy
- Department of Clinical Analysis and Toxicology, College of Pharmacy, Federal University of Rio Grande do Norte, General Gustavo Cordeiro de Farias Street, S/N, Petrópolis, Natal, 59012-570, Brazil.
| | - Edilberto Rocha Silveira
- Department of Organic and Inorganic Chemistry, Federal University of Ceará, Fortaleza, Humberto Monte Street, S/N, Campus Pici, Pici, Fortaleza, 60021-970, Brazil.
| | - Matheus de Freitas Fernandes-Pedrosa
- Laboratory of Technology and Pharmaceutical Biotechnology (Tecbiofar), College of Pharmacy, Federal University of Rio Grande do Norte, General Gustavo Cordeiro de Farias Street, S/N, Petrópolis, Natal, 59012-570, Brazil.
| | - Renata Mendonça Araújo
- Graduate Program of Chemistry, Chemistry Institute, Federal University of Rio Grande do Norte, Senador Salgado Filho Avenue, 3000, Lagoa Nova, Natal, 59072-970, Brazil.
| |
Collapse
|
8
|
Brandolini L, Aramini A, Bianchini G, Ruocco A, Bertini R, Novelli R, Angelico P, Valsecchi AE, Russo R, Castelli V, Cimini A, Allegretti M. Inflammation-Independent Antinociceptive Effects of DF2755A, a CXCR1/2 Selective Inhibitor: A New Potential Therapeutic Treatment for Peripheral Neuropathy Associated to Non-Ulcerative Interstitial Cystitis/Bladder Pain Syndrome. Front Pharmacol 2022; 13:854238. [PMID: 35571079 PMCID: PMC9096165 DOI: 10.3389/fphar.2022.854238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 03/29/2022] [Indexed: 11/23/2022] Open
Abstract
Interstitial cystitis (IC)/bladder pain syndrome (BPS) is a chronic bladder disease of unknown etiology characterized by urinary frequency and episodic and chronic pain. Analgesic treatments for IC/BPS are limited, especially for patients with non-Hunner (non-ulcerative) type IC who usually have poor overall outcomes. Here, we demonstrate that oral treatment with DF2755A, a potent and selective inhibitor of chemokine receptors CXCR1/2, can prevent and reverse peripheral neuropathy associated to non-Hunner IC/BPS by directly inhibiting chemokine-induced excitation of sensory neurons. We tested DF2755A antinociceptive effects in a cyclophosphamide (CYP)-induced non-ulcerative IC rat model characterized by severe peripheral neuropathy in the absence of bladder inflammatory infiltrate, urothelial hyperplasia, and hemorrhage. Treatment with DF2755A prevented the onset of peripheral neuropathy and reversed its development in CYP-induced IC rats, showing a strong and long-lasting anti-hyperalgesic effect. Ex vivo and in vitro studies showed that DF2755A treatment strongly inhibited the expression of CXCR2 agonists, CXCL1/KC, and CXCL5 and of transient receptor potential vanilloid 1 (TRPV1) compared to vehicle, suggesting that its effects can be due to the inhibition of the nociceptive signaling passing through the CXCL1/CXCR1-2 axis and TRPV1. In conclusion, our results highlight the key pathophysiological role played by the CXCL1/CXCR1-2 axis and TRPV1 in the onset and development of peripheral neuropathy in non-Hunner IC and propose DF2755A as a potential therapeutic approach for the treatment of not only inflammatory painful conditions but also neuropathic ones and in particular non-Hunner IC/BPS.
Collapse
Affiliation(s)
- Laura Brandolini
- Research and Early Development, Dompé Farmaceutici S.p.A., L’Aquila, Italy
| | - Andrea Aramini
- Research and Early Development, Dompé Farmaceutici S.p.A., L’Aquila, Italy
| | - Gianluca Bianchini
- Research and Early Development, Dompé Farmaceutici S.p.A., L’Aquila, Italy
| | - Anna Ruocco
- Research and Early Development, Dompé Farmaceutici S.p.A., Naples, Italy
| | | | - Rubina Novelli
- Research and Early Development, Dompé Farmaceutici S.p.A., Milan, Italy
| | | | | | - Roberto Russo
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
| | - Marcello Allegretti
- Research and Early Development, Dompé Farmaceutici S.p.A., L’Aquila, Italy
- *Correspondence: Marcello Allegretti,
| |
Collapse
|
9
|
Corrêa LB, de Oliveira Henriques MDGM, Rosas EC, Santos-Oliveira R. Intra-articular use of radium dichloride ([ 223Ra] RaCl 2) showed relevant anti-inflammatory response on experimental arthritis model. Eur J Nucl Med Mol Imaging 2021; 49:336-344. [PMID: 34370060 DOI: 10.1007/s00259-021-05515-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 07/28/2021] [Indexed: 01/15/2023]
Abstract
Rheumatoid arthritis (RA) is an inflammatory chronic autoimmune disease. The treatment of RA is difficult and, in many cases, ineffective, and the arsenal of drugs is limited. Due the longevity of the disease, RA may cause extreme musculoskeletal disorders with a high impact on quality of life. Also, RA is related with severe comorbidities decreasing the life expectancy. Finally, RA has been reported to impact in economy and healthy public. In this direction, the necessity to discover new strategies to efficiently treat RA is immediate. In this direction, we have reported the use of low doses of [223Ra] RaCl2 (radium dichloride) as intra-articular injection to treat RA. Mice were post-treated with [223Ra] RaCl2 (1.48 µCi; i.a.) 24 h after zymosan stimulus. Zymosan-induced arthrithis is responsible for leucocyte recruitment (total leukocytes, neutrophils, and mononuclear cells), which were inhibited by intra-articular injection of [223Ra] RaCl2 (69%, 77%, and 66%, respectively).
Collapse
Affiliation(s)
- Luana Barbosa Corrêa
- Laboratory of Nanoradiopharmaceticals and Synthesis of Novel Radiopharmaceuticals, Brazilian Nuclear Energy Commission, Nuclear Engineering Institute, Rio de Janeiro, 21941906, Brazil
- National Institute for Science and Technology On Innovation On Diseases of Neglected Populations (INCT/IDPN), Oswaldo Cruz Foundation, Rio de Janeiro, 21041361, Brazil
- Laboratory of Applied Pharmacology, FarmanguinhosRio de Janeiro, Oswaldo Cruz Foundation, 21041361, Brazil
| | - Maria das Graças Muller de Oliveira Henriques
- National Institute for Science and Technology On Innovation On Diseases of Neglected Populations (INCT/IDPN), Oswaldo Cruz Foundation, Rio de Janeiro, 21041361, Brazil
- Laboratory of Applied Pharmacology, FarmanguinhosRio de Janeiro, Oswaldo Cruz Foundation, 21041361, Brazil
| | - Elaine Cruz Rosas
- National Institute for Science and Technology On Innovation On Diseases of Neglected Populations (INCT/IDPN), Oswaldo Cruz Foundation, Rio de Janeiro, 21041361, Brazil
- Laboratory of Applied Pharmacology, FarmanguinhosRio de Janeiro, Oswaldo Cruz Foundation, 21041361, Brazil
| | - Ralph Santos-Oliveira
- Laboratory of Nanoradiopharmaceticals and Synthesis of Novel Radiopharmaceuticals, Brazilian Nuclear Energy Commission, Nuclear Engineering Institute, Rio de Janeiro, 21941906, Brazil.
- Laboratory of Radiopharmacy and Nanoradiopharmaceuticals, Zona Oeste State University, Rio de Janeiro, 23070200, Brazil.
| |
Collapse
|
10
|
de Andrade RCLC, de Araújo NK, Torres-Rêgo M, Furtado AA, Daniele-Silva A, de Souza Paiva W, de Medeiros Dantas JM, da Silva NS, da Silva-Júnior AA, Ururahy MAG, de Assis CF, De Santis Ferreira L, Rocha HAO, de Freitas Fernandes-Pedrosa M. Production and Characterization of Chitooligosaccharides: Evaluation of Acute Toxicity, Healing, and Anti-Inflammatory Actions. Int J Mol Sci 2021; 22:ijms221910631. [PMID: 34638973 PMCID: PMC8508594 DOI: 10.3390/ijms221910631] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 08/27/2021] [Accepted: 09/01/2021] [Indexed: 01/21/2023] Open
Abstract
The search for promising biomolecules such as chitooligosaccharides (COS) has increased due to the need for healing products that act efficiently, avoiding complications resulting from exacerbated inflammation. Therefore, this study aimed to produce COS in two stages of hydrolysis using chitosanases derived from Bacillus toyonensis. Additionally, this study aimed to structurally characterize the COS via mass spectrometry, to analyze their biocompatibility in acute toxicity models in vivo, to evaluate their healing action in a cell migration model in vitro, to analyze the anti-inflammatory activity in in vivo models of xylol-induced ear edema and zymosan-induced air pouch, and to assess the wound repair action in vivo. The structural characterization process pointed out the presence of hexamers. The in vitro and in vivo biocompatibility of COS was reaffirmed. The COS stimulated the fibroblast migration. In the in vivo inflammatory assays, COS showed an antiedematogenic response and significant reductions in leukocyte migration, cytokine release, and protein exudate. The COS healing effect in vivo was confirmed by the significant wound reduction after seven days of the experiment. These results indicated that the presence of hexamers influences the COS biological properties, which have potential uses in the pharmaceutical field due to their healing and anti-inflammatory action.
Collapse
Affiliation(s)
- Rafael Caetano Lisbôa Castro de Andrade
- Laboratory of Technology and Pharmaceutical Biotechnology (Tecbiofar), College of Pharmacy, Federal University of Rio Grande do Norte, Natal 59012-570, Brazil; (R.C.L.C.d.A.); (N.K.d.A.); (A.A.F.); (A.D.-S.); (N.S.d.S.); (A.A.d.S.-J.)
| | - Nathália Kelly de Araújo
- Laboratory of Technology and Pharmaceutical Biotechnology (Tecbiofar), College of Pharmacy, Federal University of Rio Grande do Norte, Natal 59012-570, Brazil; (R.C.L.C.d.A.); (N.K.d.A.); (A.A.F.); (A.D.-S.); (N.S.d.S.); (A.A.d.S.-J.)
| | - Manoela Torres-Rêgo
- Laboratory of Technology and Pharmaceutical Biotechnology (Tecbiofar), College of Pharmacy, Federal University of Rio Grande do Norte, Natal 59012-570, Brazil; (R.C.L.C.d.A.); (N.K.d.A.); (A.A.F.); (A.D.-S.); (N.S.d.S.); (A.A.d.S.-J.)
- Graduate Program of Chemistry, Chemistry Institute, Federal University of Rio Grande do Norte, Natal 59072-970, Brazil
- Correspondence: (M.T.-R.); (M.d.F.F.-P.)
| | - Allanny Alves Furtado
- Laboratory of Technology and Pharmaceutical Biotechnology (Tecbiofar), College of Pharmacy, Federal University of Rio Grande do Norte, Natal 59012-570, Brazil; (R.C.L.C.d.A.); (N.K.d.A.); (A.A.F.); (A.D.-S.); (N.S.d.S.); (A.A.d.S.-J.)
| | - Alessandra Daniele-Silva
- Laboratory of Technology and Pharmaceutical Biotechnology (Tecbiofar), College of Pharmacy, Federal University of Rio Grande do Norte, Natal 59012-570, Brazil; (R.C.L.C.d.A.); (N.K.d.A.); (A.A.F.); (A.D.-S.); (N.S.d.S.); (A.A.d.S.-J.)
| | - Weslley de Souza Paiva
- Laboratory of Biotechnology of Natural Biopolymers, Department of Biochemistry, Bioscience Center, Federal University of Rio Grande do Norte, Natal 59072-970, Brazil; (W.d.S.P.); (H.A.O.R.)
| | - Julia Maria de Medeiros Dantas
- Postgraduate Program in Chemical Engineering, Technology Center, Federal University of Rio Grande do Norte, Natal 59072-970, Brazil;
| | - Nayara Sousa da Silva
- Laboratory of Technology and Pharmaceutical Biotechnology (Tecbiofar), College of Pharmacy, Federal University of Rio Grande do Norte, Natal 59012-570, Brazil; (R.C.L.C.d.A.); (N.K.d.A.); (A.A.F.); (A.D.-S.); (N.S.d.S.); (A.A.d.S.-J.)
| | - Arnóbio Antônio da Silva-Júnior
- Laboratory of Technology and Pharmaceutical Biotechnology (Tecbiofar), College of Pharmacy, Federal University of Rio Grande do Norte, Natal 59012-570, Brazil; (R.C.L.C.d.A.); (N.K.d.A.); (A.A.F.); (A.D.-S.); (N.S.d.S.); (A.A.d.S.-J.)
| | - Marcela Abbott Galvão Ururahy
- Department of Clinical Analysis and Toxicology, College of Pharmacy, Federal University of Rio Grande do Norte, Natal 59012-570, Brazil; (M.A.G.U.); (C.F.d.A.)
| | - Cristiane Fernandes de Assis
- Department of Clinical Analysis and Toxicology, College of Pharmacy, Federal University of Rio Grande do Norte, Natal 59012-570, Brazil; (M.A.G.U.); (C.F.d.A.)
| | - Leandro De Santis Ferreira
- Department of Pharmacy, College of Pharmacy, Federal University of Rio Grande do Norte, Natal 59012-570, Brazil;
| | - Hugo Alexandre Oliveira Rocha
- Laboratory of Biotechnology of Natural Biopolymers, Department of Biochemistry, Bioscience Center, Federal University of Rio Grande do Norte, Natal 59072-970, Brazil; (W.d.S.P.); (H.A.O.R.)
| | - Matheus de Freitas Fernandes-Pedrosa
- Laboratory of Technology and Pharmaceutical Biotechnology (Tecbiofar), College of Pharmacy, Federal University of Rio Grande do Norte, Natal 59012-570, Brazil; (R.C.L.C.d.A.); (N.K.d.A.); (A.A.F.); (A.D.-S.); (N.S.d.S.); (A.A.d.S.-J.)
- Correspondence: (M.T.-R.); (M.d.F.F.-P.)
| |
Collapse
|
11
|
Jiang ZJ, Li QY, Zhang YY, Zeng MX, Hu H, Zhang FM, Bi LB, Gu JH, Liu XJ. Deletion of MyD88 adaptor in nociceptor alleviates low-dose formalin-induced acute pain and persistent pain in mice. Neuroreport 2021; 32:378-385. [PMID: 33661805 DOI: 10.1097/wnr.0000000000001608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The myeloid differentiation factor 88 (MyD88) adaptor mediates signaling by Toll-like receptors and some interleukins (ILs) in neural and non-neuronal cells. Recently, MyD88 protein was found to express in primary sensory neurons and be involved in the maintenance of persistent pain induced by complete Freund's adjuvant, chronic constriction injury and chemotherapy treatment in rodents. However, whether MyD88 in nociceptive neurons contributes to persistent pain induced by intraplantar injection of formalin remains elusive. Here, using conditional knockout (CKO) mice, we found that selective deletion of Myd88 in Nav1.8-expressing primary nociceptive neurons led to reduced pain response in the recovery phase of 1% formalin-induced mechanical pain and impaired the persistent thermal pain. Moreover, CKO mice exhibited reduced phase II pain response in 1%, but not 5%, formalin-induced acute inflammatory pain. Finally, nociceptor MyD88 deletion resulted in less neuronal c-Fos activation in spinal dorsal horns following 1% formalin stimulation. These data suggest that MyD88 in nociceptive neurons is not only involved in persistent mechanical pain but also promotes the transition from acute inflammatory pain to persistent thermal hyperalgesia induced by low-dose formalin stimulation.
Collapse
Affiliation(s)
- Zuo-Jie Jiang
- Pain and Related Disease Research Laboratory, Shantou University Medical College, Shantou, Guangdong Province
| | - Qing-Yi Li
- Pain and Related Disease Research Laboratory, Shantou University Medical College, Shantou, Guangdong Province
| | - Ying-Ying Zhang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing
| | - Mei-Xing Zeng
- Pain and Related Disease Research Laboratory, Shantou University Medical College, Shantou, Guangdong Province
| | - Han Hu
- Institute of Apicultural Research/Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture, Chinese Academy of Agricultural Sciences, No. 1 Beigou Xiangshan, Beijing, China
| | - Feng-Ming Zhang
- Pain and Related Disease Research Laboratory, Shantou University Medical College, Shantou, Guangdong Province
| | - Ling-Bo Bi
- Pain and Related Disease Research Laboratory, Shantou University Medical College, Shantou, Guangdong Province
| | - Jia-Hui Gu
- Pain and Related Disease Research Laboratory, Shantou University Medical College, Shantou, Guangdong Province
| | - Xing-Jun Liu
- Pain and Related Disease Research Laboratory, Shantou University Medical College, Shantou, Guangdong Province
| |
Collapse
|
12
|
Kassuya RM, Dos Santos E, Bosso FH, Pedroso TF, Marinho JVN, Salvador MJ, Kassuya CAL, Gasparotto Junior A. Anti-inflammatory Properties of Ethanolic Extract and 2″-O-β-D-Glucopyranosyl-vitexin Obtained from Alternanthera tenella Colla Whole Plant. Inflammation 2021; 44:1540-1552. [PMID: 33646486 DOI: 10.1007/s10753-021-01438-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/04/2021] [Accepted: 02/10/2021] [Indexed: 11/25/2022]
Abstract
Six flavonoids were identified and isolated from the ethanolic extract of Alternanthera tenella Colla (Amaranthaceae) whole plant (EEAT) including 2″-O-β-D-glucopyranosyl-vitexin (A19). Flavonoids have anti-inflammatory activity; however, the 2″-O-β-D-glucopyranosyl-vitexin anti-inflammatory property was not totally explored. The aim of the present study was to investigate the anti-inflammatory effects of ethanolic extract from A. tenella whole plant and isolated flavone C-glycoside A19 in models of inflammation. The oral administration (p.o.) of EEAT (30, 100, and 300 mg/kg), A19 (0.1, 1, and 10 mg/kg), and prednisolone (3 mg/kg) were tested against the carrageenan-induced paw edema in Swiss mice. The EEAT (100 mg/kg, p.o.), A19 (1 mg/kg, p.o.), and prednisolone (3 mg/kg, p.o.) were tested in the zymosan-articular inflammation, carrageenan-pleurisy, and complete Freund's adjuvant (CFA) models in Swiss mice. In silico analysis and search for structural relationships between vitexin derivatives flavones present in the EEAT and TNF-α inhibitors were performed. EEAT, A19, and prednisolone significantly inhibited (i) edema, mechanical hyperalgesia in carrageenan-induced paw inflammation; (ii) leukocyte migration and protein extravasation in carrageenan-induced pleurisy; (iii) knee edema, mechanical hyperalgesia, and leukocyte migration in articular inflammation induced by zymosan. Still the CFA induced the increase in myeloperoxidase and N-acetylglucosaminidase activities, EEAT, A19, and prednisolone significantly inhibited these enzymes. The in silico analysis showed that molecules with similar structure to apigenin and vitexin have a potential inhibition on the TNF system. This study confirms the anti-inflammatory properties of EEAT and A19. The C-glycosylated flavone A19 could be responsible for the EEAT anti-edematogenic and anti-hyperalgesic effects and a potential TNF-α inhibitor.
Collapse
Affiliation(s)
- Roberto Mikio Kassuya
- College of Health Science, Federal University of Grande Dourados, Dourados, MS, Brazil
| | - Elisangela Dos Santos
- College of Health Science, Federal University of Grande Dourados, Dourados, MS, Brazil
| | - Fernando Henrique Bosso
- Institute of Biology, Department of Plant Biology, PPG BTPB, PPG BV and PPG BCE, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Taíse Fonseca Pedroso
- Institute of Biology, Department of Plant Biology, PPG BTPB, PPG BV and PPG BCE, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Jane Vasconcelos Neves Marinho
- Institute of Biology, Department of Plant Biology, PPG BTPB, PPG BV and PPG BCE, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Marcos José Salvador
- Institute of Biology, Department of Plant Biology, PPG BTPB, PPG BV and PPG BCE, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | | | | |
Collapse
|
13
|
Verdan M, Branquinho L, Silva-Filho S, Oliveira R, Cardoso CL, Arena A, Kassuya CL. Antiarthritic and antinociceptive potential of ethanolic extract from leaves of Doliocarpus dentatus (aubl.) standl. in mouse model. Pharmacognosy Res 2021. [DOI: 10.4103/pr.pr_79_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
14
|
Lagomarsino VN, Kostic AD, Chiu IM. Mechanisms of microbial-neuronal interactions in pain and nociception. NEUROBIOLOGY OF PAIN 2020; 9:100056. [PMID: 33392418 PMCID: PMC7772816 DOI: 10.1016/j.ynpai.2020.100056] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 11/18/2020] [Accepted: 11/27/2020] [Indexed: 02/06/2023]
Abstract
Molecular mechanisms of how microorganisms communicate with sensory afferent neurons. How pathogenic microorganisms directly communicate with nociceptor neurons to inflict pain on the host. Symbiotic bacterial communication with gut-extrinsic sensory afferent neurons. Plausible roles on how gut symbionts directly mediate pain and nociception.
Nociceptor sensory neurons innervate barrier tissues that are constantly exposed to microbial stimuli. During infection, pathogenic microorganisms can breach barrier surfaces and produce pain by directly activating nociceptors. Microorganisms that live in symbiotic relationships with their hosts, commensals and mutualists, have also been associated with pain, but the molecular mechanisms of how symbionts act on nociceptor neurons to modulate pain remain largely unknown. In this review, we will discuss the known molecular mechanisms of how microbes directly interact with sensory afferent neurons affecting nociception in the gut, skin and lungs. We will touch on how bacterial, viral and fungal pathogens signal to the host to inflict or suppress pain. We will also discuss recent studies examining how gut symbionts affect pain. Specifically, we will discuss how gut symbionts may interact with sensory afferent neurons either directly, through secretion of metabolites or neurotransmitters, or indirectly,through first signaling to epithelial cells or immune cells, to regulate visceral, neuropathic and inflammatory pain. While this area of research is still in its infancy, more mechanistic studies to examine microbial-sensory neuron crosstalk in nociception may allow us to develop new therapies for the treatment of acute and chronic pain.
Collapse
Affiliation(s)
- Valentina N Lagomarsino
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA.,Joslin Diabetes Center, Boston, MA 02115, USA.,Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA
| | - Aleksandar D Kostic
- Joslin Diabetes Center, Boston, MA 02115, USA.,Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA
| | - Isaac M Chiu
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
15
|
Fattori V, Ferraz CR, Rasquel-Oliveira FS, Verri WA. Neuroimmune communication in infection and pain: Friends or foes? Immunol Lett 2020; 229:32-43. [PMID: 33248166 DOI: 10.1016/j.imlet.2020.11.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 10/02/2020] [Accepted: 11/20/2020] [Indexed: 12/13/2022]
Abstract
Clinically, a variety of micro-organisms cause painful infections. Before seen as bystanders in the context of infections, recent studies have demonstrated that, as immune cells, nociceptors can sense pathogen-derived products. Nociceptors and immune cells, therefore, have evolved to communicate with each other to control inflammatory and host responses against pathogens in a complementary way. This interaction is named as neuroimmune communication (or axon-axon immune reflex) and initiates after the release of neuropeptides, such as CGRP and VIP by neurons. By this neurogenic response, nociceptors orchestrate the activity of innate and adaptive immune cells in a context-dependent manner. In this review, we focus on how nociceptors sense pathogen-derived products to shape the host response. We also highlight the new concept involving the resolution of inflammation, which is related to an active and time-dependent biosynthetic shift from pro-inflammatory to pro-resolution mediators, the so-called specialized pro-resolving lipid mediators (SPMs). At very low doses, SPMs act on specific receptors to silence nociceptors, limit pain and neurogenic responses, and resolve infections. Furthermore, stimulation of the vagus nerve induces SPMs production to regulate immune responses in infections. Therefore, harnessing the current understanding of neuro-immune communication and neurogenic responses might provide the bases for reprogramming host responses against infections through well balanced and effective immune response and inflammation resolution.
Collapse
Affiliation(s)
- Victor Fattori
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Londrina State University, Londrina, PR, Brazil; Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Camila R Ferraz
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Londrina State University, Londrina, PR, Brazil
| | - Fernanda S Rasquel-Oliveira
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Londrina State University, Londrina, PR, Brazil
| | - Waldiceu A Verri
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Londrina State University, Londrina, PR, Brazil.
| |
Collapse
|
16
|
Rasquel-Oliveira FS, Manchope MF, Staurengo-Ferrari L, Ferraz CR, Saraiva-Santos T, Zaninelli TH, Fattori V, Artero NA, Badaro-Garcia S, de Freitas A, Casagrande R, Verri WA. Hesperidin methyl chalcone interacts with NFκB Ser276 and inhibits zymosan-induced joint pain and inflammation, and RAW 264.7 macrophage activation. Inflammopharmacology 2020; 28:979-992. [PMID: 32048121 DOI: 10.1007/s10787-020-00686-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 01/27/2020] [Indexed: 01/29/2023]
Abstract
Arthritis can be defined as a painful musculoskeletal disorder that affects the joints. Hesperidin methyl chalcone (HMC) is a flavonoid with analgesic, anti-inflammatory, and antioxidant effects. However, its effects on a specific cell type and in the zymosan-induced inflammation are unknown. We aimed at evaluating the effects of HMC in a zymosan-induced arthritis model. A dose-response curve of HMC (10, 30, or 100 mg/kg) was performed to determine the most effective analgesic dose after intra-articular zymosan stimuli. Knee joint oedema was determined using a calliper. Leukocyte recruitment was performed by cell counting on knee joint wash as well as histopathological analysis. Oxidative stress was measured by colorimetric assays (GSH, FRAP, ABTS and NBT) and RT-qPCR (gp91phox and HO-1 mRNA expression) performed. In vitro, oxidative stress was assessed by DCFDA assay using RAW 264.7 macrophages. Cytokine production was evaluated in vivo and in vitro by ELISA. In vitro NF-κB activation was analysed by immunofluorescence. We observed HMC reduced mechanical hypersensitivity and knee joint oedema, leukocyte recruitment, and pro-inflammatory cytokine levels. We also observed a reduction in zymosan-induced oxidative stress as per increase in total antioxidant capacity and reduction in gp91phox and increase in HO-1 mRNA expression. Accordingly, total ROS production and macrophage NFκB activation were diminished. HMC interaction with NFκB p65 at Ser276 was revealed using molecular docking analysis. Thus, data presented in this work suggest the usefulness of HMC as an analgesic and anti-inflammatory in a zymosan-induced arthritis model, possibly by targeting NFκB activation in macrophages.
Collapse
Affiliation(s)
- Fernanda S Rasquel-Oliveira
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Londrina State University, Londrina, PR, Brazil
| | - Marilia F Manchope
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Londrina State University, Londrina, PR, Brazil
| | - Larissa Staurengo-Ferrari
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Londrina State University, Londrina, PR, Brazil
| | - Camila R Ferraz
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Londrina State University, Londrina, PR, Brazil
| | - Telma Saraiva-Santos
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Londrina State University, Londrina, PR, Brazil
| | - Tiago H Zaninelli
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Londrina State University, Londrina, PR, Brazil
| | - Victor Fattori
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Londrina State University, Londrina, PR, Brazil
| | - Nayara A Artero
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Londrina State University, Londrina, PR, Brazil
| | - Stephanie Badaro-Garcia
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Londrina State University, Londrina, PR, Brazil
| | - Andressa de Freitas
- Departament of Physiological Sciences, Centre of Biological Sciences, Londrina State University, Londrina, PR, Brazil
| | - Rubia Casagrande
- Departament of Pharmaceutical Sciences, Centre of Health Sciences, Londrina State University, Londrina, PR, Brazil
| | - Waldiceu A Verri
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Londrina State University, Londrina, PR, Brazil.
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rod. Celso Garcia Cid PR 445, KM 380, PO Box 10.011, Londrina, Parana, 86057-970, Brazil.
| |
Collapse
|
17
|
Bussmann AJC, Borghi SM, Zaninelli TH, Dos Santos TS, Guazelli CFS, Fattori V, Domiciano TP, Pinho-Ribeiro FA, Ruiz-Miyazawa KW, Casella AMB, Vignoli JA, Camilios-Neto D, Casagrande R, Verri WA. The citrus flavanone naringenin attenuates zymosan-induced mouse joint inflammation: induction of Nrf2 expression in recruited CD45 + hematopoietic cells. Inflammopharmacology 2019; 27:1229-1242. [PMID: 30612217 DOI: 10.1007/s10787-018-00561-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 12/31/2018] [Indexed: 01/28/2023]
Abstract
BACKGROUND Naringenin is a biologically active analgesic, anti-inflammatory, and antioxidant flavonoid. Naringenin targets in inflammation-induced articular pain remain poorly explored. METHODS The present study investigated the cellular and molecular mechanisms involved in the analgesic/anti-inflammatory effects of naringenin in zymosan-induced arthritis. Mice were pre-treated orally with naringenin (16.7-150 mg/kg), followed by intra-articular injection of zymosan. Articular mechanical hyperalgesia and oedema, leucocyte recruitment to synovial cavity, histopathology, expression/production of pro- and anti-inflammatory mediators and NFκB activation, inflammasome component expression, and oxidative stress were evaluated. RESULTS Naringenin inhibited articular pain and oedema in a dose-dependent manner. The dose of 50 mg/kg inhibited leucocyte recruitment, histopathological alterations, NFκB activation, and NFκB-dependent pro-inflammatory cytokines (TNF-α, IL-1β, and IL-33), and preproET-1 mRNA expression, but increased anti-inflammatory IL-10. Naringenin also inhibited inflammasome upregulation (reduced Nlrp3, ASC, caspase-1, and pro-IL-1β mRNA expression) and oxidative stress (reduced gp91phox mRNA expression and superoxide anion production, increased GSH levels, induced Nrf2 protein in CD45+ hematopoietic recruited cells, and induced Nrf2 and HO-1 mRNA expression). CONCLUSIONS Naringenin presents analgesic and anti-inflammatory effects in zymosan-induced arthritis by targeting its main physiopathological mechanisms. These data highlight this flavonoid as an interesting therapeutic compound to treat joint inflammation, deserving additional pre-clinical and clinical studies.
Collapse
Affiliation(s)
- Allan J C Bussmann
- Department of Pathology, Biological Science Center, Londrina State University, Rod. Celso Garcia Cid, PR 445, Km 380, Londrina, Paraná State, 86051-990, Brazil
| | - Sergio M Borghi
- Department of Pathology, Biological Science Center, Londrina State University, Rod. Celso Garcia Cid, PR 445, Km 380, Londrina, Paraná State, 86051-990, Brazil
| | - Tiago H Zaninelli
- Department of Pathology, Biological Science Center, Londrina State University, Rod. Celso Garcia Cid, PR 445, Km 380, Londrina, Paraná State, 86051-990, Brazil
| | - Telma S Dos Santos
- Department of Pathology, Biological Science Center, Londrina State University, Rod. Celso Garcia Cid, PR 445, Km 380, Londrina, Paraná State, 86051-990, Brazil
| | - Carla F S Guazelli
- Department of Pathology, Biological Science Center, Londrina State University, Rod. Celso Garcia Cid, PR 445, Km 380, Londrina, Paraná State, 86051-990, Brazil
| | - Victor Fattori
- Department of Pathology, Biological Science Center, Londrina State University, Rod. Celso Garcia Cid, PR 445, Km 380, Londrina, Paraná State, 86051-990, Brazil
| | - Talita P Domiciano
- Department of Pathology, Biological Science Center, Londrina State University, Rod. Celso Garcia Cid, PR 445, Km 380, Londrina, Paraná State, 86051-990, Brazil
| | - Felipe A Pinho-Ribeiro
- Department of Pathology, Biological Science Center, Londrina State University, Rod. Celso Garcia Cid, PR 445, Km 380, Londrina, Paraná State, 86051-990, Brazil
| | - Kenji W Ruiz-Miyazawa
- Department of Pathology, Biological Science Center, Londrina State University, Rod. Celso Garcia Cid, PR 445, Km 380, Londrina, Paraná State, 86051-990, Brazil
| | - Antonio M B Casella
- Department of Clinical Medicine, Health Science Center, Londrina State University, University Hospital, 86039-440, Londrina, Paraná State, Brazil
| | - Josiane A Vignoli
- Department of Biochemistry and Biotechnology, Exact Sciences Center, Londrina State University, Londrina, 86057-970, Brazil
| | - Doumit Camilios-Neto
- Department of Biochemistry and Biotechnology, Exact Sciences Center, Londrina State University, Londrina, 86057-970, Brazil
| | - Rubia Casagrande
- Department of Pharmaceutical Sciences, Health Science Center, Londrina State University, University Hospital, Londrina, Paraná State, 86039-440, Brazil
| | - Waldiceu A Verri
- Department of Pathology, Biological Science Center, Londrina State University, Rod. Celso Garcia Cid, PR 445, Km 380, Londrina, Paraná State, 86051-990, Brazil.
| |
Collapse
|
18
|
Yin B, Liu H, Tan B, Dong X, Chi S, Yang Q, Zhang S, Chen L. Cottonseed protein concentrate (CPC) suppresses immune function in different intestinal segments of hybrid grouper ♀Epinephelus fuscoguttatus×♂Epinephelus lanceolatu via TLR-2/MyD88 signaling pathways. FISH & SHELLFISH IMMUNOLOGY 2018; 81:318-328. [PMID: 30030116 DOI: 10.1016/j.fsi.2018.07.038] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 06/11/2018] [Accepted: 07/17/2018] [Indexed: 06/08/2023]
Abstract
Cottonseed protein concentrate (CPC) has similar amino acid composition compared with fish meal, and has the characteristics of low gossypol and low toxicity. The present study was conducted to investigate the growth performance, antioxidant capacity and different intestinal segments immune responses of hybrid grouper to replacement dietary fish meal ofCPC. Six iso-nitrogenous (50% crude protein) and iso-lipidic (10% crude lipid) diets were formulated: a reference diet (FM) containing 60% fishmeal and five experimental diets (12%, 24%, 36%, 48 and 60%) in which fishmeal protein was substituted at different levels by CPC to feed fish (initial body weight: 11 ± 0.23 g) for 8 weeks. Thena challenge test with injection of Vibrio parahaemolyticus was conducted for 7 days until the fish stabilized. The results showed that specific growth rate (SGR) was the highest with 24% replacement level and feed conversion ratio (FCR)was significantly increased when the replacement level reached 48% (P < 0.05). The content of malonaldehyde (MDA) in the serum was significantly increased when the replacement level reached 36% (P < 0.05). The plica height in the proximal, mid and distal intestine were significantly decreased with the replacement level up to 48% (P < 0.05). Hepatic fat deposition wasaggravatedwhen the replacement level reached 36% (P < 0.05). The expression of IL-6, TNF-α, and IL-1β mRNAs were significantly up-regulated (P < 0.05). The hepcidin mRNA expression was significantly down-regulated (P < 0.05). In proximal intestine (PI) and mid intestine (MI), IFN-γ mRNA expression was significantly up-regulated (P < 0.05). These results suggested that the CPC decreased hybrid grouper growth performance and inflammation function, and different inflammation function responses in PI,MI, and distal intestine (DI) were mediated partly by the TLR-2/MyD88 signaling pathway. According to the analysis of specific growth rate, the dietary optimum replacement level and maximum replacement level were estimated to be 17% and 34%, respectively.
Collapse
Affiliation(s)
- Bin Yin
- Laboratory of Aquatic Animal Nutrition and Feed, Fisheries College, Guangdong Ocean University, Zhanjiang, 524025, PR China; Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang, 524025, PR China
| | - Hongyu Liu
- Laboratory of Aquatic Animal Nutrition and Feed, Fisheries College, Guangdong Ocean University, Zhanjiang, 524025, PR China; Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang, 524025, PR China
| | - Beiping Tan
- Laboratory of Aquatic Animal Nutrition and Feed, Fisheries College, Guangdong Ocean University, Zhanjiang, 524025, PR China; Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang, 524025, PR China.
| | - Xiaohui Dong
- Laboratory of Aquatic Animal Nutrition and Feed, Fisheries College, Guangdong Ocean University, Zhanjiang, 524025, PR China; Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang, 524025, PR China
| | - Shuyan Chi
- Laboratory of Aquatic Animal Nutrition and Feed, Fisheries College, Guangdong Ocean University, Zhanjiang, 524025, PR China; Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang, 524025, PR China
| | - Qihui Yang
- Laboratory of Aquatic Animal Nutrition and Feed, Fisheries College, Guangdong Ocean University, Zhanjiang, 524025, PR China; Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang, 524025, PR China
| | - Shuang Zhang
- Laboratory of Aquatic Animal Nutrition and Feed, Fisheries College, Guangdong Ocean University, Zhanjiang, 524025, PR China; Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang, 524025, PR China
| | - Liqiao Chen
- College of Life Sciences, East China Normal University, Shanghai, 200062, PR China
| |
Collapse
|
19
|
Maruyama K, Takayama Y, Sugisawa E, Yamanoi Y, Yokawa T, Kondo T, Ishibashi KI, Sahoo BR, Takemura N, Mori Y, Kanemaru H, Kumagai Y, Martino MM, Yoshioka Y, Nishijo H, Tanaka H, Sasaki A, Ohno N, Iwakura Y, Moriyama Y, Nomura M, Akira S, Tominaga M. The ATP Transporter VNUT Mediates Induction of Dectin-1-Triggered Candida Nociception. iScience 2018; 6:306-318. [PMID: 30240621 PMCID: PMC6137711 DOI: 10.1016/j.isci.2018.08.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 07/09/2018] [Accepted: 08/09/2018] [Indexed: 12/02/2022] Open
Abstract
Candida albicans infection can cause skin, vulvar, or oral pain. Despite the obvious algesic activity of C. albicans, the molecular mechanisms of fungal nociception remain largely unknown. Here we show that the C. albicans-specific signaling pathway led to severe mechanical allodynia. We discovered that C. albicans-derived β-glucan stimulated nociceptors depending on Dectin-1, and two pathways in inflammatory pain. The major pathway operates via the Dectin-1-mediated ATP-P2X3/P2X2/3 axis through intercellular relationships between keratinocytes and primary sensory neurons, which depends on the ATP transporter vesicular nucleotide transporter (VNUT). The other pathway operates via the Dectin-1-mediated PLC-TRPV1/TRPA1 axis in primary sensory neurons. Intriguingly, C. albicans-derived β-glucan has the ability to enhance histamine-independent pruritus, and VNUT inhibitor clodronate can be used to treat unpleasant feelings induced by β-glucan. Collectively, this is the first report to indicate that Dectin-1 and VNUT mediated innate sensory mechanisms that detect fungal infection. β-Glucan-induced pain is abolished in Dectin-1- and TRPV1/TRPA1-deficient mice β-–Glucan stimulates nociceptors via Dectin-1-PLC axis, activating neurons β-Glucan-induced allodynia is dependent on extracellular ATP and VNUT VNUT inhibitor clodronate can be used to treat β-glucan-induced allodynia
Collapse
Affiliation(s)
- Kenta Maruyama
- Laboratory of Host Defense, Osaka University, Osaka 565-0871, Japan; WPI Immunology Frontier Research Center (IFReC), Osaka University, Osaka 565-0871, Japan.
| | - Yasunori Takayama
- Thermal Biology group, Exploratory Research Center on Life and Living Systems National Institutes of Natural Sciences, Okazaki Aichi 444-8787, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan; Department of Physiological Sciences, the Graduate University for Advanced Studies, Aichi 444-8787, Japan
| | - Erika Sugisawa
- Laboratory of Host Defense, Osaka University, Osaka 565-0871, Japan; WPI Immunology Frontier Research Center (IFReC), Osaka University, Osaka 565-0871, Japan
| | - Yu Yamanoi
- Thermal Biology group, Exploratory Research Center on Life and Living Systems National Institutes of Natural Sciences, Okazaki Aichi 444-8787, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan; Department of Physiological Sciences, the Graduate University for Advanced Studies, Aichi 444-8787, Japan; Research Laboratory, Ikedamohando Co., Ltd., 2-16-16 Iwamoto-cho, Chiyoda-ku, Tokyo 101-0032, Japan
| | - Takashi Yokawa
- BioView Corporation, 2-16-16 Iwamoto-cho, Chiyoda-ku, Tokyo 101-0032, Japan
| | - Takeshi Kondo
- Laboratory of Host Defense, Osaka University, Osaka 565-0871, Japan; WPI Immunology Frontier Research Center (IFReC), Osaka University, Osaka 565-0871, Japan
| | - Ken-Ichi Ishibashi
- Laboratory for Immunopharmacology of Microbial Products, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Bikash Ranjan Sahoo
- Laboratory of Host Defense, Osaka University, Osaka 565-0871, Japan; WPI Immunology Frontier Research Center (IFReC), Osaka University, Osaka 565-0871, Japan
| | - Naoki Takemura
- Department of Mucosal Immunology, School of Medicine, Chiba University, 1-8-1 Inohana, Chuou-ku, Chiba 260-8670, Japan
| | - Yuki Mori
- Laboratory of Biofunctional Imaging, Osaka University, Osaka 565-0871, Japan; WPI Immunology Frontier Research Center (IFReC), Osaka University, Osaka 565-0871, Japan
| | - Hisashi Kanemaru
- Laboratory of Host Defense, Osaka University, Osaka 565-0871, Japan; WPI Immunology Frontier Research Center (IFReC), Osaka University, Osaka 565-0871, Japan
| | - Yutaro Kumagai
- Laboratory of Host Defense, Osaka University, Osaka 565-0871, Japan; WPI Immunology Frontier Research Center (IFReC), Osaka University, Osaka 565-0871, Japan; Biotechnology Research Institute for Drug Discovery National Institute of Advanced Industrial Science and Technology Central 5-41, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan
| | - Mikaël M Martino
- Laboratory of Host Defense, Osaka University, Osaka 565-0871, Japan; WPI Immunology Frontier Research Center (IFReC), Osaka University, Osaka 565-0871, Japan; European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Innovation Walk, Monash University, Wellington Road, Clayton, VIC 3800, Australia
| | - Yoshichika Yoshioka
- Laboratory of Biofunctional Imaging, Osaka University, Osaka 565-0871, Japan; WPI Immunology Frontier Research Center (IFReC), Osaka University, Osaka 565-0871, Japan
| | - Hisao Nishijo
- System Emotional Science (Physiology), Graduate School of Medicine and Pharmaceutical Sciences for Research, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Hiroki Tanaka
- Laboratory of Host Defense, Osaka University, Osaka 565-0871, Japan; WPI Immunology Frontier Research Center (IFReC), Osaka University, Osaka 565-0871, Japan
| | - Atsushi Sasaki
- Research Unit/Neuroscience Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000, Kamoshida-cho, Aoba-ku, Yokohama 227-0033, Japan
| | - Naohito Ohno
- Laboratory for Immunopharmacology of Microbial Products, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Yoichiro Iwakura
- Research Institute for Biomedical Sciences, Tokyo University of Science, 2669 Yamazaki, Noda, Chiba 278-0022, Japan
| | - Yoshinori Moriyama
- Department of Membrane Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8530, Japan
| | - Masatoshi Nomura
- Department of Medicine and Bioregulatory Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Shizuo Akira
- Laboratory of Host Defense, Osaka University, Osaka 565-0871, Japan; WPI Immunology Frontier Research Center (IFReC), Osaka University, Osaka 565-0871, Japan
| | - Makoto Tominaga
- Thermal Biology group, Exploratory Research Center on Life and Living Systems National Institutes of Natural Sciences, Okazaki Aichi 444-8787, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan; Department of Physiological Sciences, the Graduate University for Advanced Studies, Aichi 444-8787, Japan; Institute for Environmental and Gender Specific Medicine, Juntendo University, 2-1-1 Tomioka, Urayasu, Chiba 279-0021, Japan.
| |
Collapse
|
20
|
Cook AD, Lee MC, Saleh R, Khiew HW, Christensen AD, Achuthan A, Fleetwood AJ, Lacey DC, Smith JE, Förster I, Hamilton JA. TNF and granulocyte macrophage-colony stimulating factor interdependence mediates inflammation via CCL17. JCI Insight 2018; 3:99249. [PMID: 29563337 DOI: 10.1172/jci.insight.99249] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 02/20/2018] [Indexed: 12/15/2022] Open
Abstract
TNF and granulocyte macrophage-colony stimulating factor (GM-CSF) have proinflammatory activity and both contribute, for example, to rheumatoid arthritis pathogenesis. We previously identified a new GM-CSF→JMJD3 demethylase→interferon regulatory factor 4 (IRF4)→CCL17 pathway that is active in monocytes/macrophages in vitro and important for inflammatory pain, as well as for arthritic pain and disease. Here we provide evidence for a nexus between TNF and this pathway, and for TNF and GM-CSF interdependency. We report that the initiation of zymosan-induced inflammatory pain and zymosan-induced arthritic pain and disease are TNF dependent. Once arthritic pain and disease are established, blockade of GM-CSF or CCL17, but not of TNF, is still able to ameliorate them. TNF is required for GM-CSF-driven inflammatory pain and for initiation of GM-CSF-driven arthritic pain and disease, but not once they are established. TNF-driven inflammatory pain and TNF-driven arthritic pain and disease are dependent on GM-CSF and mechanistically require the same downstream pathway involving GM-CSF→CCL17 formation via JMJD3-regulated IRF4 production, indicating that GM-CSF and CCL17 can mediate some of the proinflammatory and algesic actions of TNF. Given we found that TNF appears important only early in arthritic pain and disease progression, targeting a downstream mediator, such as CCL17, which appears to act throughout the course of disease, could be effective at ameliorating chronic inflammatory conditions where TNF is implicated.
Collapse
Affiliation(s)
- Andrew D Cook
- University of Melbourne, Department of Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Ming-Chin Lee
- University of Melbourne, Department of Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Reem Saleh
- University of Melbourne, Department of Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Hsu-Wei Khiew
- University of Melbourne, Department of Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Anne D Christensen
- University of Melbourne, Department of Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Adrian Achuthan
- University of Melbourne, Department of Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Andrew J Fleetwood
- University of Melbourne, Department of Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Derek C Lacey
- University of Melbourne, Department of Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Julia E Smith
- Cytokine, Chemokine and Complement DPU, Immunoinflammation TA, GSK Medicines Research Centre, Stevenage, Hertfordshire, United Kingdom
| | - Irmgard Förster
- Immunology and Environment, Life and Medical Sciences Institute University of Bonn, Bonn, Germany
| | - John A Hamilton
- University of Melbourne, Department of Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia
| |
Collapse
|
21
|
Guazelli CFS, Staurengo-Ferrari L, Zarpelon AC, Pinho-Ribeiro FA, Ruiz-Miyazawa KW, Vicentini FTMC, Vignoli JA, Camilios-Neto D, Georgetti SR, Baracat MM, Casagrande R, Verri WA. Quercetin attenuates zymosan-induced arthritis in mice. Biomed Pharmacother 2018; 102:175-184. [PMID: 29554596 DOI: 10.1016/j.biopha.2018.03.057] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 03/09/2018] [Accepted: 03/11/2018] [Indexed: 12/24/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by articular lesions, recruitment of inflammatory cells and increased levels of pro-inflammatory cytokine. The intra-articular administration of zymosan is an experimental model that promotes inflammatory parameters resembling RA. Therefore, this model was used to investigate the efficacy of quercetin as a treatment of articular inflammation. Treatment with quercetin dose-dependently reduced zymosan-induced hyperalgesia, articular edema and the recruitment of neutrophils to the knee joint cavity. Histological analysis confirmed that quercetin inhibited zymosan-induced arthritis. The treatment with quercetin also inhibited zymosan-induced depletion of reduced glutathione (GSH) levels, TNFα and IL-1β production, and gp91phox, prepro-endothelin-1 (preproET-1), and cyclooxygenase-2 mRNA expression. These molecular effects of quercetin were related to the inhibition of the nuclear factor kappa-B and induction of Nuclear factor erythroid 2- related factor (Nrf2)/home oxygenase (HO-1) pathway. Thus, quercetin exerted anti-inflammatory, analgesic and antioxidant effects in experimental arthritis, suggesting quercetin is a possible candidate for arthritis treatment.
Collapse
Affiliation(s)
- Carla F S Guazelli
- Departamento de Ciências Patológicas - Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina 86051-990, Brazil
| | - Larissa Staurengo-Ferrari
- Departamento de Ciências Patológicas - Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina 86051-990, Brazil
| | - Ana C Zarpelon
- Departamento de Ciências Patológicas - Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina 86051-990, Brazil
| | - Felipe A Pinho-Ribeiro
- Departamento de Ciências Patológicas - Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina 86051-990, Brazil
| | - Kenji W Ruiz-Miyazawa
- Departamento de Ciências Patológicas - Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina 86051-990, Brazil
| | - Fabiana T M C Vicentini
- Farmacore Biotecnologia LTDA, Rua Edson Souto, 728, Lagoinha, 14095-250 Ribeirão Preto, Brazil
| | - Josiane A Vignoli
- Departamento de Bioquímica e Biotecnologia - Centro de Ciências Exatas, Universidade Estadual de Londrina 86057-970, Brazil
| | - Doumit Camilios-Neto
- Departamento de Bioquímica e Biotecnologia - Centro de Ciências Exatas, Universidade Estadual de Londrina 86057-970, Brazil
| | - Sandra R Georgetti
- Departamento de Ciências Farmacêuticas - Centro de Ciências de Saúde, Universidade Estadual de Londrina, Londrina 86038-350, Brazil
| | - Marcela M Baracat
- Departamento de Ciências Farmacêuticas - Centro de Ciências de Saúde, Universidade Estadual de Londrina, Londrina 86038-350, Brazil
| | - Rubia Casagrande
- Departamento de Ciências Farmacêuticas - Centro de Ciências de Saúde, Universidade Estadual de Londrina, Londrina 86038-350, Brazil.
| | - Waldiceu A Verri
- Departamento de Ciências Patológicas - Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina 86051-990, Brazil.
| |
Collapse
|
22
|
Kang MH, Jin YH, Kim BS. Effects of Keratinocyte-Derived Cytokine (CXCL-1) on the Development of Theiler's Virus-Induced Demyelinating Disease. Front Cell Infect Microbiol 2018; 8:9. [PMID: 29410948 PMCID: PMC5787060 DOI: 10.3389/fcimb.2018.00009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/09/2018] [Indexed: 12/31/2022] Open
Abstract
CXCL-1, also called keratinocyte-derived cytokine (KC), is a predominant chemokine produced in glial cells upon infection with Theiler's murine encephalomyelitis virus (TMEV). In this study, we assessed the role of KC in the development of TMEV-induced demyelinating disease by utilizing polyclonal anti-KC antibodies as well as KC-expressing recombinant TMEV. Our results indicate that the level of KC produced after infection with TMEV or stimulation with various TLRs is significantly higher in various cells from susceptible SJL mice compared to those in cells from resistant B6 mice. SJL mice treated with rabbit anti-KC antibodies displayed accelerated development of TMEV-induced demyelinating disease, elevated viral loads in the CNS and decreased antiviral T cell responses. In addition, infection of susceptible SJL mice with recombinant KC-TMEV produced biologically active KC, which resulted in the accelerated pathogenesis of demyelinating disease and elevated T cell responses to viral antigens compared to mice infected with control recombinant HEL-TMEV. These results strongly suggest that both the lack of KC during TMEV infection and the excessive presence of the chemokine promote the pathogenesis of demyelinating disease. Therefore, a balance in the level of KC during TMEV infection appears to be critically important in controlling the pathogenesis of demyelinating disease.
Collapse
Affiliation(s)
- Min H Kang
- Department of Microbiology-Immunology, Northwestern University Medical School, Chicago, IL, United States
| | - Young H Jin
- Department of Microbiology-Immunology, Northwestern University Medical School, Chicago, IL, United States
| | - Byung S Kim
- Department of Microbiology-Immunology, Northwestern University Medical School, Chicago, IL, United States
| |
Collapse
|
23
|
de Araújo JCB, Gondim DV, Cavalcante ALC, Lisboa MRP, de Castro Brito GA, Vale ML. Inflammatory pain assessment in the arthritis of the temporomandibular joint in rats: A comparison between two phlogistic agents. J Pharmacol Toxicol Methods 2017; 88:100-108. [PMID: 28797764 DOI: 10.1016/j.vascn.2017.08.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 07/21/2017] [Accepted: 08/05/2017] [Indexed: 12/01/2022]
Abstract
Temporomandibular joint (TMJ) disorders are a group of conditions that result in TMJ pain, which frequently limits basic daily activities. Experimental models that allow the study of the mechanisms underlying these inflammatory and pain conditions are of great clinical relevance. The aim of this study was to evaluate nociception, inflammation and participation of the macrophage/microglia cells in the arthritis of the TMJ induced by two phlogistic agents. 84 rats were divided into 2 groups: Zy, which received zymosan intra-articularly, or Cg, which received carrageenan intra-articularly. Mechanical nociception, total leukocyte influx to the synovial fluid and histopathological analyses were evaluated in the TMJ. The participation of macrophage/microglia located in trigeminal ganglia (TG) and in the subnucleus caudalis (V-SnC) was assessed immunohistochemically. Both agents induced mechanical hyperalgesia 6h after the induction, but a more persistent algesic state was perceived in the Cg group, which lasted for 120h. Even though both groups presented increased leukocyte influx, the Zy-group presented a more intense influx. Zymosan recruited resident macrophage in the trigeminal ganglia 24h after the injection. In the V-SnC, the group Cg presented a more prolonged immunolabeling pattern in comparison with the group Zy. It can be concluded that zymosan induced a more intense infiltrate and peripheral nervous changes, while Cg lead to a moderate TMJ inflammation with prominent changes in the V-SnC.
Collapse
Affiliation(s)
| | - Delane Viana Gondim
- Morphofunctional Sciences Post Graduation Program, Department of Morphology, Federal University of Ceará, Brazil
| | - André Luiz Cunha Cavalcante
- Medical Sciences Post Graduation Program, Department of Clinical Medicine, Federal University of Ceará, Brazil
| | - Mario Roberto Pontes Lisboa
- Morphofunctional Sciences Post Graduation Program, Department of Morphology, Federal University of Ceará, Brazil
| | - Gerly Anne de Castro Brito
- Morphofunctional Sciences Post Graduation Program, Department of Morphology, Federal University of Ceará, Brazil
| | - Mariana Lima Vale
- Pharmacology Post Graduation Program, Department of Physiology and Pharmacology, Federal University of Ceará, Brazil; Morphofunctional Sciences Post Graduation Program, Department of Morphology, Federal University of Ceará, Brazil.
| |
Collapse
|
24
|
Lee MC, McCubbin JA, Christensen AD, Poole DP, Rajasekhar P, Lieu T, Bunnett NW, Garcia-Caraballo S, Erickson A, Brierley SM, Saleh R, Achuthan A, Fleetwood AJ, Anderson RL, Hamilton JA, Cook AD. G-CSF Receptor Blockade Ameliorates Arthritic Pain and Disease. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 198:3565-3575. [PMID: 28320832 PMCID: PMC10069442 DOI: 10.4049/jimmunol.1602127] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 02/20/2017] [Indexed: 01/08/2023]
Abstract
G-CSF or CSF-3, originally defined as a regulator of granulocyte lineage development via its cell surface receptor (G-CSFR), can play a role in inflammation, and hence in many pathologies, due to its effects on mature lineage populations. Given this, and because pain is an extremely important arthritis symptom, the efficacy of an anti-G-CSFR mAb for arthritic pain and disease was compared with that of a neutrophil-depleting mAb, anti-Ly6G, in both adaptive and innate immune-mediated murine models. Pain and disease were ameliorated in Ag-induced arthritis, zymosan-induced arthritis, and methylated BSA/IL-1 arthritis by both prophylactic and therapeutic anti-G-CSFR mAb treatment, whereas only prophylactic anti-Ly6G mAb treatment was effective. Efficacy for pain and disease correlated with reduced joint neutrophil numbers and, importantly, benefits were noted without necessarily the concomitant reduction in circulating neutrophils. Anti-G-CSFR mAb also suppressed zymosan-induced inflammatory pain. A new G-CSF-driven (methylated BSA/G-CSF) arthritis model was established enabling us to demonstrate that pain was blocked by a cyclooxygenase-2 inhibitor, suggesting an indirect effect on neurons. Correspondingly, dorsal root ganglion neurons cultured in G-CSF failed to respond to G-CSF in vitro, and Csf3r gene expression could not be detected in dorsal root ganglion neurons by single-cell RT-PCR. These data suggest that G-CSFR/G-CSF targeting may be a safe therapeutic strategy for arthritis and other inflammatory conditions, particularly those in which pain is important, as well as for inflammatory pain per se.
Collapse
Affiliation(s)
- Ming-Chin Lee
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria 3050, Australia
| | - James A McCubbin
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria 3050, Australia
| | - Anne D Christensen
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria 3050, Australia
| | - Daniel P Poole
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia.,Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Pradeep Rajasekhar
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - TinaMarie Lieu
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Nigel W Bunnett
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia.,Department of Surgery, Columbia University, New York, NY 10032
| | - Sonia Garcia-Caraballo
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of Adelaide, Adelaide, South Australia 5000, Australia.,South Australian Health and Medical Research Institute, Adelaide, South Australia 5000, Australia.,Visceral Pain Group, Department of Human Physiology, Flinders University, Bedford Park, South Australia 5042, Australia
| | - Andelain Erickson
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of Adelaide, Adelaide, South Australia 5000, Australia.,South Australian Health and Medical Research Institute, Adelaide, South Australia 5000, Australia.,Visceral Pain Group, Department of Human Physiology, Flinders University, Bedford Park, South Australia 5042, Australia
| | - Stuart M Brierley
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of Adelaide, Adelaide, South Australia 5000, Australia.,South Australian Health and Medical Research Institute, Adelaide, South Australia 5000, Australia.,Visceral Pain Group, Department of Human Physiology, Flinders University, Bedford Park, South Australia 5042, Australia
| | - Reem Saleh
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria 3050, Australia
| | - Adrian Achuthan
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria 3050, Australia
| | - Andrew J Fleetwood
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria 3050, Australia
| | - Robin L Anderson
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria 3084, Australia; and.,School of Cancer Medicine, La Trobe University, Heidelberg, Victoria 3084, Australia
| | - John A Hamilton
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria 3050, Australia;
| | - Andrew D Cook
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria 3050, Australia
| |
Collapse
|
25
|
Abstract
Pain and itch are unpleasant sensations that often accompany infections caused by viral, bacterial, parasitic, and fungal pathogens. Recent studies show that sensory neurons are able to directly detect pathogens to mediate pain and itch. Nociceptor and pruriceptor neurons respond to pathogen-associated molecular patterns, including Toll-like receptor ligands, N-formyl peptides, and bacterial toxins. Other pathogens are able to silence neuronal activity to produce analgesia during infection. Pain and itch could lead to neuronal modulation of the immune system or behavioral avoidance of future pathogen exposure. Conversely, pathogens could modulate neuronal signaling to potentiate their pathogenesis and facilitate their spread to other hosts. Defining how pathogens modulate pain and itch has critical implications for sensory neurobiology and our understanding of host-microbe interactions.
Collapse
Affiliation(s)
- Isaac M Chiu
- Department of Microbiology and Immunobiology, Division of Immunology, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
26
|
Mechanisms leading from systemic autoimmunity to joint-specific disease in rheumatoid arthritis. Nat Rev Rheumatol 2016; 13:79-86. [PMID: 27974851 DOI: 10.1038/nrrheum.2016.200] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A key unanswered question in the pathophysiology of rheumatoid arthritis (RA) is how systemic autoimmunity progresses to joint-specific inflammation. In patients with seropositive RA (that is, characterized by the presence of autoantibodies) evidence is accumulating that immunity against post-translationally modified (such as citrullinated) autoantigens might be triggered in mucosal organs, such as the lung, long before the first signs of inflammation are seen in the joints. However, the mechanism by which systemic autoimmunity specifically homes to the joint and bone compartment, thereby triggering inflammation, remains elusive. This Review summarizes potential pathways involved in this joint-homing mechanism, focusing particularly on osteoclasts as the primary targets of anti-citrullinated protein antibodies (ACPAs) in the bone and joint compartment. Osteoclasts are dependent on citrullinating enzymes for their normal differentiation and are unique in displaying citrullinated antigens on their cell surface in a non-inflamed state. The binding of ACPAs to osteoclasts releases the chemokine IL-8, leading to bone erosion and pain. This process initiates a chain of events that could lead to attraction and activation of neutrophils, resulting in a complex series of proinflammatory processes in the synovium, eventually leading to RA.
Collapse
|
27
|
Malmström V, Catrina AI, Klareskog L. The immunopathogenesis of seropositive rheumatoid arthritis: from triggering to targeting. Nat Rev Immunol 2016; 17:60-75. [PMID: 27916980 DOI: 10.1038/nri.2016.124] [Citation(s) in RCA: 285] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Patients with rheumatoid arthritis can be divided into two major subsets characterized by the presence versus absence of antibodies to citrullinated protein antigens (ACPAs) and of rheumatoid factor (RF). The antibody-positive subset of disease, also known as seropositive rheumatoid arthritis, constitutes approximately two-thirds of all cases of rheumatoid arthritis and generally has a more severe disease course. ACPAs and RF are often present in the blood long before any signs of joint inflammation, which suggests that the triggering of autoimmunity may occur at sites other than the joints (for example, in the lung). This Review summarizes recent progress in our understanding of this gradual disease development in seropositive patients. We also emphasize the implications of this new understanding for the development of preventive and therapeutic strategies. Similar temporal and spatial separation of immune triggering and clinical manifestations, with novel opportunities for early intervention, may also occur in other immune-mediated diseases.
Collapse
Affiliation(s)
- Vivianne Malmström
- Rheumatology Unit, Department of Medicine at Solna, Karolinska University Hospital, Karolinska Institute, 171 76 Stockholm, Sweden
| | - Anca I Catrina
- Rheumatology Unit, Department of Medicine at Solna, Karolinska University Hospital, Karolinska Institute, 171 76 Stockholm, Sweden
| | - Lars Klareskog
- Rheumatology Unit, Department of Medicine at Solna, Karolinska University Hospital, Karolinska Institute, 171 76 Stockholm, Sweden
| |
Collapse
|
28
|
Zhou J, Lin W, Chen H, Fan Y, Yang C. TRESK contributes to pain threshold changes by mediating apoptosis via MAPK pathway in the spinal cord. Neuroscience 2016; 339:622-633. [PMID: 27789381 DOI: 10.1016/j.neuroscience.2016.10.039] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 10/10/2016] [Accepted: 10/17/2016] [Indexed: 01/05/2023]
Abstract
The mechanism underlying neuropathic pain (NP) is complex and has not been fully elucidated. The TWIK-related spinal cord K+ (TRESK) is the major background potassium current in dorsal root ganglia (DRG), we found that mitogen-activated protein kinase (MAPK) signal pathway were activated in spinal cord accompanied by TRESK down regulation in response to NP. Therefore, we investigated whether TRESK mediates inflammation and apoptosis by MAPK pathway in the spinal cord of NP rats. SNI rats exhibited reduced TRESK expression in DRG and spinal cord and higher sensitivity to mechanical stimuli but no effect on thermal stimuli. Intrathecal injections of TRESK overexpressing adenovirus alleviated mechanical allodynia, inhibited phosphorylation of extracellular signal-regulated kinase (ERK) and p38, and decreased inflammatory reactions and apoptosis in the spinal cords of SNI rats. Down regulation of TRESK in DRG and spinal cord was detected in normal rats after intrathecal TRESK shRNA lentivirus injection, which induced mechanical allodynia but had no effect on pain thresholds for heat stimulation. Phosphorylated ERK and p38 were increased in the spinal cord. Intrathecal injection of an ERK antagonist (PD98059) and p38 antagonist (SB203580) prevented ERK and p38 activation in the spinal cord and mechanical allodynia induced by TRESK shRNA lentivirus. In conclusion, our study clearly demonstrated an important role for TRESK in NP and that TRESK regulation contributes to pain sensitivity mediates inflammation and apoptosis by ERK and p38 MAPK signaling in the spinal cord.
Collapse
Affiliation(s)
- Jun Zhou
- Department of Anesthesiology, First People's Hospital of Foshan, Foshan, Guangdong Province 528000, China.
| | - Wenjing Lin
- Department of Anesthesiology, First People's Hospital of Foshan, Foshan, Guangdong Province 528000, China
| | - Hongtao Chen
- Department of Anesthesiology, Eighth People's Hospital of Guangzhou, Guangzhou, Guangdong Province 510060, China
| | - Youling Fan
- Department of Anesthesiology, Central Hospital of Panyu District, Guangzhou 511400, Guangdong Province, China
| | - Chengxiang Yang
- Department of Anesthesiology, First People's Hospital of Foshan, Foshan, Guangdong Province 528000, China
| |
Collapse
|
29
|
Guerrero ATG, Pinto LG, Cunha FQ, Ferreira SH, Alves-Filho JC, Verri WA, Cunha TM. Mechanisms underlying the hyperalgesic responses triggered by joint activation of TLR4. Pharmacol Rep 2016; 68:1293-1300. [PMID: 27689757 DOI: 10.1016/j.pharep.2016.08.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 08/15/2016] [Accepted: 08/23/2016] [Indexed: 01/04/2023]
Abstract
BACKGROUND Toll-like receptors (TLRs) including TLR4 and their signal pathways contribute to the pathogenesis of arthritis. Herein, we evaluated the mechanisms underlying the hyperalgesic response caused by TLR4 activation in the tibio-tarsal joint in mice. METHODS Joint inflammatory hyperalgesia was induced by intra-articular (ia) injection of LPS (lipopolysaccharide- TLR4 agonist) in C57BL/6, TLR4, TLR2, MyD88, TRIF, TNFR1/2 and IL-1R1 knockout (-/-) mice. Joint hyperalgesia was evaluated using an electronic von Frey. Neutrophil recruitment was assessed by MPO activity. Joint levels of cytokines were measured by ELISA. RESULTS Firstly, it was shown that LPS injected into the joints causes a dose- and time-dependent reduction in the mechanical nociceptive threshold. The TLR4 activation in the joint triggers mechanical hyperalgesia and neutrophil migration, which was abolished in TLR4 -/- and MyD88-/-, but not in TLR2-/- and TRIF-/- mice. Besides, joint administration of LPS increased the release of TNF-α, IL-1β, and KC/CXCL1, which were reduced in TLR4-/- and MyD88-/-, but not in TRIF-/- mice. In agreement, the LPS-induced joint nociceptive effect was decreased in TNFR1/2-/- and IL-1R1-/- mice or in mice pre-treated with a CXCR1/2 selective antagonist (DF2156A). CONCLUSIONS These results suggest that TLR4 activation in the joint produces articular hyperalgesia via MyD88 signaling pathway. Moreover, this pathway is involved in the cascade of events of articular hyperalgesia through mechanisms dependent on cytokines and chemokines production. Thus, TLR4/MyD88 signaling pathway inhibitors might be useful for the treatment of inflammatory joint pain.
Collapse
Affiliation(s)
- Ana T G Guerrero
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Avenida Bandeirantes, 3900, 14049-900 Ribeirão Preto, São Paulo, Brazil; Fundação Oswaldo Cruz-FIOCRUZ Mato Grosso do Sul, Rua Gabriel Abrão 92, 79081-746 Campo Grande, Mato Grosso do Sul, Brazil.
| | - Larissa G Pinto
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Avenida Bandeirantes, 3900, 14049-900 Ribeirão Preto, São Paulo, Brazil.
| | - Fernando Q Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Avenida Bandeirantes, 3900, 14049-900 Ribeirão Preto, São Paulo, Brazil.
| | - Sérgio H Ferreira
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Avenida Bandeirantes, 3900, 14049-900 Ribeirão Preto, São Paulo, Brazil.
| | - Jose C Alves-Filho
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Avenida Bandeirantes, 3900, 14049-900 Ribeirão Preto, São Paulo, Brazil.
| | - Waldiceu A Verri
- Departamento de Patologia, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Paraná, Rod. Celso Garcia Cid Km380 PR445, 86057-970 Londrina, Paraná, Brazil.
| | - Thiago M Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Avenida Bandeirantes, 3900, 14049-900 Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
30
|
Torres-Rêgo M, Furtado AA, Bitencourt MAO, Lima MCJDS, Andrade RCLCD, Azevedo EPD, Soares TDC, Tomaz JC, Lopes NP, da Silva-Júnior AA, Zucolotto SM, Fernandes-Pedrosa MDF. Anti-inflammatory activity of aqueous extract and bioactive compounds identified from the fruits of Hancornia speciosa Gomes (Apocynaceae). Altern Ther Health Med 2016; 16:275. [PMID: 27496015 PMCID: PMC4974780 DOI: 10.1186/s12906-016-1259-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 08/03/2016] [Indexed: 11/10/2022]
Abstract
Background Hancornia speciosa Gomes (Apocynaceae), popularly known as “mangabeira,” has been used in folk medicine to treat inflammatory disorders, hypertension, dermatitis, diabetes, liver diseases and gastric disorders. Although the ethnobotany indicates that its fruits can be used for the treatment of ulcers and inflammatory disorders, only few studies have been conducted to prove such biological activities. This study investigated the anti-inflammatory properties of the aqueous extract of the fruits of H. speciosa Gomes as well as its bioactive compounds using in vivo experimental models. Methods The bioactive compounds were identified by High Performance Liquid Chromatography coupled with diode array detector (HPLC-DAD) and Liquid Chromatography coupled with Mass Spectrometry (LC-MS). The anti-inflammatory properties were investigated through in vivo tests, which comprised xylene-induced ear edema, carrageenan-induced peritonitis and zymosan-induced air pouch. The levels of IL-1β, IL-6, IL-12 and TNF-α were determined using ELISA. Results Rutin and chlorogenic acid were identified in the extract as the main secondary metabolites. In addition, the extract as well as rutin and chlorogenic acid significantly inhibited the xilol-induced ear edema and also reduced the cell migration in both carrageenan-induced peritonitis and zymosan-induced air pouch models. Reduced levels of cytokines were also observed. Conclusion This is the first study that demonstrated the anti-inflammatory activity of the extract of H. speciosa fruits against different inflammatory agents in animal models, suggesting that its bioactive molecules, especially rutin and chlorogenic acid are, at least in part, responsible for such activity. These findings support the widespread use of Hancornia speciosa in popular medicine and demonstrate that its aqueous extract has therapeutical potential for the development of herbal drugs with anti-inflammatory properties.
Collapse
|
31
|
Gaurilcikaite E, Renton T, Grant AD. The paradox of painless periodontal disease. Oral Dis 2016; 23:451-463. [PMID: 27397640 DOI: 10.1111/odi.12537] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 05/04/2016] [Accepted: 06/23/2016] [Indexed: 12/23/2022]
Abstract
Periodontal diseases, primarily gingivitis and periodontitis, are characterised by progressive inflammation and tissue destruction. However, they are unusual in that they are not also accompanied by the pain commonly seen in other inflammatory conditions. This suggests that interactions between periodontal bacteria and host cells create a unique environment in which the pro-algesic effects of inflammatory mediators and factors released during tissue damage are directly or indirectly inhibited. In this review, we summarise the evidence that periodontal disease is characterised by an accumulation of classically pro-algesic factors from bacteria and host cells. We then discuss several mechanisms by which inflammatory sensitisation of nociceptive fibres could be prevented through inactivation or inhibition of these factors. Further studies are necessary to fully understand the molecular processes underlying the endogenous localised hypoalgesia in human periodontal disease. This knowledge might provide a rational basis to develop future therapeutic interventions, such as host modulation therapies, against a wide variety of other human pain conditions.
Collapse
Affiliation(s)
- E Gaurilcikaite
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - T Renton
- Department of Oral Surgery, Dental Institute, King's College London, London, UK
| | - A D Grant
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
32
|
Correa LB, Pádua TA, Seito LN, Costa TEMM, Silva MA, Candéa ALP, Rosas EC, Henriques MG. Anti-inflammatory Effect of Methyl Gallate on Experimental Arthritis: Inhibition of Neutrophil Recruitment, Production of Inflammatory Mediators, and Activation of Macrophages. JOURNAL OF NATURAL PRODUCTS 2016; 79:1554-1566. [PMID: 27227459 DOI: 10.1021/acs.jnatprod.5b01115] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Methyl gallate (MG) is a prevalent phenolic acid in the plant kingdom, and its presence in herbal medicines might be related to its remarkable biological effects, such as its antioxidant, antitumor, and antimicrobial activities. Although some indirect evidence suggests anti-inflammatory activity for MG, there are no studies demonstrating this effect in animal models. Herein, we demonstrated that MG (0.7-70 mg/kg) inhibited zymosan-induced experimental arthritis in a dose-dependent manner. The oral administration of MG (7 mg/kg) attenuates arthritis induced by zymosan, affecting edema formation, leukocyte migration, and the production of inflammatory mediators (IL-1β, IL-6, TNF-α, CXCL-1, LTB4, and PGE2). Pretreatment with MG inhibited in vitro neutrophil chemotaxis elicited by CXCL-1, as well as the adhesion of these cells to TNF-α-primed endothelial cells. MG also impaired zymosan-stimulated macrophages by inhibiting IL-6 and NO production, COX-2 and iNOS expression, and intracellular calcium mobilization. Thus, MG is likely to present an anti-inflammatory effect by targeting multiple cellular events such as the production of various inflammatory mediators, as well as leukocyte activation and migration.
Collapse
Affiliation(s)
- Luana Barbosa Correa
- Laboratory of Applied Pharmacology, Farmanguinhos, and ‡National Institute for Science and Technology on Innovation on Neglected Diseases (INCT/IDN), Center for Technological Development in Health (CDTS), Oswaldo Cruz Foundation (Fiocruz) , Rio de Janeiro, RJ, Brazil
| | - Tatiana Almeida Pádua
- Laboratory of Applied Pharmacology, Farmanguinhos, and ‡National Institute for Science and Technology on Innovation on Neglected Diseases (INCT/IDN), Center for Technological Development in Health (CDTS), Oswaldo Cruz Foundation (Fiocruz) , Rio de Janeiro, RJ, Brazil
| | - Leonardo Noboru Seito
- Laboratory of Applied Pharmacology, Farmanguinhos, and ‡National Institute for Science and Technology on Innovation on Neglected Diseases (INCT/IDN), Center for Technological Development in Health (CDTS), Oswaldo Cruz Foundation (Fiocruz) , Rio de Janeiro, RJ, Brazil
| | - Thadeu Estevam Moreira Maramaldo Costa
- Laboratory of Applied Pharmacology, Farmanguinhos, and ‡National Institute for Science and Technology on Innovation on Neglected Diseases (INCT/IDN), Center for Technological Development in Health (CDTS), Oswaldo Cruz Foundation (Fiocruz) , Rio de Janeiro, RJ, Brazil
| | - Magaiver Andrade Silva
- Laboratory of Applied Pharmacology, Farmanguinhos, and ‡National Institute for Science and Technology on Innovation on Neglected Diseases (INCT/IDN), Center for Technological Development in Health (CDTS), Oswaldo Cruz Foundation (Fiocruz) , Rio de Janeiro, RJ, Brazil
| | - André Luis Peixoto Candéa
- Laboratory of Applied Pharmacology, Farmanguinhos, and ‡National Institute for Science and Technology on Innovation on Neglected Diseases (INCT/IDN), Center for Technological Development in Health (CDTS), Oswaldo Cruz Foundation (Fiocruz) , Rio de Janeiro, RJ, Brazil
| | - Elaine Cruz Rosas
- Laboratory of Applied Pharmacology, Farmanguinhos, and ‡National Institute for Science and Technology on Innovation on Neglected Diseases (INCT/IDN), Center for Technological Development in Health (CDTS), Oswaldo Cruz Foundation (Fiocruz) , Rio de Janeiro, RJ, Brazil
| | - Maria G Henriques
- Laboratory of Applied Pharmacology, Farmanguinhos, and ‡National Institute for Science and Technology on Innovation on Neglected Diseases (INCT/IDN), Center for Technological Development in Health (CDTS), Oswaldo Cruz Foundation (Fiocruz) , Rio de Janeiro, RJ, Brazil
| |
Collapse
|
33
|
Wigerblad G, Bas DB, Fernades-Cerqueira C, Krishnamurthy A, Nandakumar KS, Rogoz K, Kato J, Sandor K, Su J, Jimenez-Andrade JM, Finn A, Bersellini Farinotti A, Amara K, Lundberg K, Holmdahl R, Jakobsson PJ, Malmström V, Catrina AI, Klareskog L, Svensson CI. Autoantibodies to citrullinated proteins induce joint pain independent of inflammation via a chemokine-dependent mechanism. Ann Rheum Dis 2015; 75:730-8. [PMID: 26613766 PMCID: PMC4819624 DOI: 10.1136/annrheumdis-2015-208094] [Citation(s) in RCA: 195] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 10/18/2015] [Indexed: 01/30/2023]
Abstract
Objective An interesting and so far unexplained feature of chronic pain in autoimmune disease is the frequent disconnect between pain and inflammation. This is illustrated well in rheumatoid arthritis (RA) where pain in joints (arthralgia) may precede joint inflammation and persist even after successful anti-inflammatory treatment. In the present study, we have addressed the possibility that autoantibodies against citrullinated proteins (ACPA), present in RA, may be directly responsible for the induction of pain, independent of inflammation. Methods Antibodies purified from human patients with RA, healthy donors and murinised monoclonal ACPA were injected into mice. Pain-like behaviour was monitored for up to 28 days, and tissues were analysed for signs of pathology. Mouse osteoclasts were cultured and stimulated with antibodies, and supernatants analysed for release of factors. Mice were treated with CXCR1/2 (interleukin (IL) 8 receptor) antagonist reparixin. Results Mice injected with either human or murinised ACPA developed long-lasting pronounced pain-like behaviour in the absence of inflammation, while non-ACPA IgG from patients with RA or control monoclonal IgG were without pronociceptive effect. This effect was coupled to ACPA-mediated activation of osteoclasts and release of the nociceptive chemokine CXCL1 (analogue to human IL-8). ACPA-induced pain-like behaviour was reversed with reparixin. Conclusions The data suggest that CXCL1/IL-8, released from osteoclasts in an autoantibody-dependent manner, produces pain by activating sensory neurons. The identification of this new pain pathway may open new avenues for pain treatment in RA and also in other painful diseases associated with autoantibody production and/or osteoclast activation.
Collapse
Affiliation(s)
- Gustaf Wigerblad
- Molecular Pain Research, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Duygu B Bas
- Molecular Pain Research, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Cátia Fernades-Cerqueira
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, CMM, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Akilan Krishnamurthy
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, CMM, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Kutty Selva Nandakumar
- Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Katarzyna Rogoz
- Molecular Pain Research, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jungo Kato
- Molecular Pain Research, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Katalin Sandor
- Molecular Pain Research, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jie Su
- Molecular Pain Research, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Juan Miguel Jimenez-Andrade
- Department of Unidad Academica Multidisciplinaria Reynosa Aztlan, Universidad Autonoma de Tamaulipas, Reynosa, Tamaulipas, Mexico
| | - Anja Finn
- Molecular Pain Research, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Alex Bersellini Farinotti
- Molecular Pain Research, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Khaled Amara
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, CMM, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Karin Lundberg
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, CMM, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Rikard Holmdahl
- Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Per-Johan Jakobsson
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, CMM, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Vivianne Malmström
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, CMM, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Anca I Catrina
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, CMM, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Lars Klareskog
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, CMM, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Camilla I Svensson
- Molecular Pain Research, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
34
|
Fong FLY, Shah NP, Kirjavainen P, El-Nezami H. Mechanism of Action of Probiotic Bacteria on Intestinal and Systemic Immunities and Antigen-Presenting Cells. Int Rev Immunol 2015; 35:179-88. [PMID: 26606641 DOI: 10.3109/08830185.2015.1096937] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Immunomodulation has been shown to be one of the major functions of probiotic bacteria. This review is presented to provide detailed information on the immunomodulatory properties of probiotics in various animal models and clinical practices. Probiotics can regulate helper T (Th) responses and release of cytokines in a strain-specific manner. For example, Lactobacillus rhamnosus GG can induce beneficial Th1 immunomodulatory effect in infants with cow's milk allergy and relieve intestinal inflammation in atopic children by promoting IL-10 generation. Mechanism of action of probiotics on antigen-presenting cells at gastrointestinal tract is also postulated in this review. Probiotic bacterial cells and their soluble factors may activate dendritic cells, macrophages, and to certain extent monocytes via toll-like-receptor recognition and may further provoke specific Th responses. They are speculated to elicit immunomodulatory effects on intestinal and systemic immunities.
Collapse
Affiliation(s)
- Fiona Long Yan Fong
- a School of Biological Sciences , The University of Hong Kong , Hong Kong.,b Present address: School of Science and Technology , The Open University of Hong Kong >, Hong Kong
| | - Nagendra P Shah
- a School of Biological Sciences , The University of Hong Kong , Hong Kong
| | - Pirkka Kirjavainen
- b Present address: School of Science and Technology , The Open University of Hong Kong >, Hong Kong.,c Present address: Department of Environmental Health , National Institute for Health and Welfare , Kuopio , Finland
| | - Hani El-Nezami
- a School of Biological Sciences , The University of Hong Kong , Hong Kong.,d Institute of Public Health and Clinical Nutrition , University of Easter Finland , Kuopio , Finland
| |
Collapse
|
35
|
Xu H, Xu T, Ma XQ, Jiang W. Chronic morphine treatment increased the expression of myeloid differentiation primary response protein 88 in rat spinal cord. J Integr Neurosci 2015; 13:607-15. [PMID: 25182346 DOI: 10.1142/s0219635214500216] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Chronic morphine exposure leads to tolerance, which limits the clinical use of this potent analgesic in the treatment of severe and chronic pain. Compelling evidence suggest that neuro-immune activation (pro-inflammatory cytokines including IL-1β, IL-6 and TNF) as well as neuro-inflammation have been shown to mediate the development of morphine analgesic tolerance. Toll-like receptors (TLRs), especially TLR-4, have also been reported to contribute to the development of morphine analgesic tolerance. Besides, mitogen-activated protein kinases (MAPKs; especially p38 MAPK and c-Jun N -terminal kinase), as well as nuclear factor-κB (NF-κB) modulate the development of morphine antinociceptive tolerance. Hence, we hypothesis the possible involvement of myeloid differentiation primary response protein 88 (MyD88), a key adaptor protein for the TLR and IL-1R families, in the development of tolerance to morphine-induced analgesia. Our study demonstrated that chronic intrathecal morphine injection led to a robust increase of MyD88 expression in rat spinal cord. Sustained elevation of MyD88 may play a role in modulating the development of morphine antinociceptive tolerance.
Collapse
Affiliation(s)
- Hao Xu
- Department of Anesthesiology, Shanghai Jiaotong University, Affiliated Shanghai Sixth People's Hospital, Shanghai 200233, P. R. China
| | | | | | | |
Collapse
|
36
|
Melnik BC. Endoplasmic reticulum stress: key promoter of rosacea pathogenesis. Exp Dermatol 2014; 23:868-73. [DOI: 10.1111/exd.12517] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2014] [Indexed: 12/14/2022]
Affiliation(s)
- Bodo C. Melnik
- Department of Dermatology, Environmental Medicine and Health Theory; University of Osnabrück; Osnabrück Germany
| |
Collapse
|
37
|
Low-frequency high-magnitude mechanical strain of articular chondrocytes activates p38 MAPK and induces phenotypic changes associated with osteoarthritis and pain. Int J Mol Sci 2014; 15:14427-41. [PMID: 25196344 PMCID: PMC4159860 DOI: 10.3390/ijms150814427] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 08/12/2014] [Accepted: 08/14/2014] [Indexed: 01/07/2023] Open
Abstract
Osteoarthritis (OA) is a debilitating joint disorder resulting from an incompletely understood combination of mechanical, biological, and biochemical processes. OA is often accompanied by inflammation and pain, whereby cytokines associated with chronic OA can up-regulate expression of neurotrophic factors such as nerve growth factor (NGF). Several studies suggest a role for cytokines and NGF in OA pain, however the effects of changing mechanical properties in OA tissue on chondrocyte metabolism remain unclear. Here, we used high-extension silicone rubber membranes to examine if high mechanical strain (HMS) of primary articular chondrocytes increases inflammatory gene expression and promotes neurotrophic factor release. HMS cultured chondrocytes displayed up-regulated NGF, TNFα and ADAMTS4 gene expression while decreasing TLR2 expression, as compared to static controls. HMS culture increased p38 MAPK activity compared to static controls. Conditioned medium from HMS dynamic cultures, but not static cultures, induced significant neurite sprouting in PC12 cells. The increased neurite sprouting was accompanied by consistent increases in PC12 cell death. Low-frequency high-magnitude mechanical strain of primary articular chondrocytes in vitro drives factor secretion associated with degenerative joint disease and joint pain. This study provides evidence for a direct link between cellular strain, secretory factors, neo-innervation, and pain in OA pathology.
Collapse
|
38
|
Nociceptive neurons regulate innate and adaptive immunity and neuropathic pain through MyD88 adapter. Cell Res 2014; 24:1374-7. [PMID: 25112711 DOI: 10.1038/cr.2014.106] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|
39
|
Sun Y, Sahbaie P, Liang D, Li W, Clark JD. Opioids enhance CXCL1 expression and function after incision in mice. THE JOURNAL OF PAIN 2014; 15:856-66. [PMID: 24887006 PMCID: PMC4131856 DOI: 10.1016/j.jpain.2014.05.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 04/29/2014] [Accepted: 05/20/2014] [Indexed: 12/20/2022]
Abstract
UNLABELLED Chronic opioid consumption increases postoperative pain. Epigenetic changes related to chronic opioid use and surgical incision may be partially responsible for this enhancement. The CXCL1/CXCR2 signaling pathway, implicated in several pain models, is known to be epigenetically regulated via histone acetylation. The current study was designed to investigate the role of CXCL1/CXCR2 signaling in opioid-enhanced incisional sensitization and to elucidate the possible epigenetic mechanism underlying CXCL1/CXCR2 pathway-mediated regulation of nociceptive sensitization in mice. Chronic morphine treatment generated mechanical and thermal nociceptive sensitization and also significantly exacerbated incision-induced mechanical allodynia. Peripheral but not central messenger RNA levels of CXCL1 and CXCR2 were increased after incision. The source of peripheral CXCL1 appeared to be wound area neutrophils. Histone H3 subunit acetylated at the lysine 9 position (AcH3K9) was increased in infiltrating dermal neutrophils after incision and was further increased in mice with chronic morphine treatment. The association of AcH3K9 with the promoter region of CXCL1 was enhanced in mice after chronic morphine treatment. The increase in CXCL1 near wounds caused by chronic morphine pretreatment was mimicked by pharmacologic inhibition of histone deacetylation. Finally, local injection of CXCL1 induced mechanical sensitivity in naive mice, whereas blocking CXCR2 reversed mechanical hypersensitivity after hind paw incision. PERSPECTIVE Peripheral CXCL1/CXCR2 signaling helps to control nociceptive sensitization after incision, and epigenetic regulation of CXCL1 expression explains in part opioid-enhanced incisional allodynia in mice. These results suggest that targeting CXCL1/CXCR2 signaling may be useful in treating nociceptive sensitization, particularly for postoperative pain in chronic opioid-consuming patients.
Collapse
MESH Headings
- Analgesics, Opioid/administration & dosage
- Analgesics, Opioid/pharmacology
- Animals
- Chemokine CXCL1/genetics
- Chemokine CXCL1/metabolism
- Disease Models, Animal
- Drug Administration Schedule
- Gene Expression Regulation/drug effects
- Hyperalgesia/drug therapy
- Hyperalgesia/physiopathology
- Male
- Mice
- Mice, Inbred C57BL
- Morphine/administration & dosage
- Neutrophils/drug effects
- Neutrophils/metabolism
- Pain Measurement
- Pain Threshold/drug effects
- Pain, Postoperative/drug therapy
- Pain, Postoperative/metabolism
- Phenylurea Compounds/administration & dosage
- Phenylurea Compounds/pharmacology
- Receptors, Interleukin-8B/antagonists & inhibitors
- Receptors, Interleukin-8B/genetics
- Time Factors
- Wounds, Penetrating/complications
- Wounds, Penetrating/drug therapy
Collapse
Affiliation(s)
- Yuan Sun
- Department of Anesthesiology, Stanford University School of Medicine, Stanford, California; Department of Anesthesiology, Veterans Affairs Palo Alto Health Care System, Palo Alto, California
| | - Peyman Sahbaie
- Department of Anesthesiology, Stanford University School of Medicine, Stanford, California; Department of Anesthesiology, Veterans Affairs Palo Alto Health Care System, Palo Alto, California
| | - DeYong Liang
- Department of Anesthesiology, Stanford University School of Medicine, Stanford, California; Department of Anesthesiology, Veterans Affairs Palo Alto Health Care System, Palo Alto, California
| | - Wenwu Li
- Department of Anesthesiology, Stanford University School of Medicine, Stanford, California
| | - J David Clark
- Department of Anesthesiology, Stanford University School of Medicine, Stanford, California; Department of Anesthesiology, Veterans Affairs Palo Alto Health Care System, Palo Alto, California.
| |
Collapse
|
40
|
Mu H, Yao RB, Zhao LJ, Shen SY, Zhao ZM, Cai H. Sinomenine decreases MyD88 expression and improves inflammation-induced joint damage progression and symptoms in rat adjuvant-induced arthritis. Inflammation 2014; 36:1136-44. [PMID: 23605561 DOI: 10.1007/s10753-013-9648-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Sinomenine (SIN) is the active principle of the Chinese medical plant Sinomenium acutum which is widely used for the treatment of rheumatoid arthritis (RA) in China. Recently, several groups indicated that myeloid differentiation primary response protein 88 (MyD88) might be associated with disease progression of RA. Here, we observed the effect of SIN on MyD88 expression and showed its therapeutic role in RA. First, immunohistochemical staining in clinical specimens showed that MyD88 was mainly located in characteristic pathological structures of RA synovial tissues. Second, we found that MyD88 was overexpressed in the synovial tissues of the rats with adjuvant-induced arthritis (AIA). Treatment with SIN markedly decreased the expression of MyD88 in AIA rats. Finally, we provided evidences that SIN suppressed inflammation response and inflammation-induced joint destructive progression and arthritis symptoms in AIA rats. Therefore, SIN is an effective therapeutic agent for RA. Targeting MyD88 signaling may provide new methods for the treatment of RA.
Collapse
Affiliation(s)
- Hui Mu
- Department of Integrated Traditional and Western Medicine, Jinling Hospital, School of Medicine, Second Military Medical University (Shanghai), Nanjing, 210002, People's Republic of China
| | | | | | | | | | | |
Collapse
|
41
|
Pádua TA, de Abreu BSSC, Costa TEMM, Nakamura MJ, Valente LMM, Henriques MDG, Siani AC, Rosas EC. Anti-inflammatory effects of methyl ursolate obtained from a chemically derived crude extract of apple peels: potential use in rheumatoid arthritis. Arch Pharm Res 2014; 37:1487-95. [DOI: 10.1007/s12272-014-0345-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 01/26/2014] [Indexed: 01/16/2023]
|
42
|
Borghi SM, Zarpelon AC, Pinho-Ribeiro FA, Cardoso RD, Martins-Pinge MC, Tatakihara RI, Cunha TM, Ferreira SH, Cunha FQ, Casagrande R, Verri WA. Role of TNF-α/TNFR1 in intense acute swimming-induced delayed onset muscle soreness in mice. Physiol Behav 2014; 128:277-87. [DOI: 10.1016/j.physbeh.2014.01.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 11/27/2013] [Accepted: 01/26/2014] [Indexed: 12/18/2022]
|
43
|
Gawri R, Rosenzweig DH, Krock E, Ouellet JA, Stone LS, Quinn TM, Haglund L. High mechanical strain of primary intervertebral disc cells promotes secretion of inflammatory factors associated with disc degeneration and pain. Arthritis Res Ther 2014; 16:R21. [PMID: 24457003 PMCID: PMC3979109 DOI: 10.1186/ar4449] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 01/13/2014] [Indexed: 02/02/2023] Open
Abstract
Introduction Excessive mechanical loading of intervertebral discs (IVDs) is thought to alter matrix properties and influence disc cell metabolism, contributing to degenerative disc disease and development of discogenic pain. However, little is known about how mechanical strain induces these changes. This study investigated the cellular and molecular changes as well as which inflammatory receptors and cytokines were upregulated in human intervertebral disc cells exposed to high mechanical strain (HMS) at low frequency. The impact of these metabolic changes on neuronal differentiation was also explored to determine a role in the development of disc degeneration and discogenic pain. Methods Isolated human annulus fibrosus (AF) and nucleus pulposus (NP) cells were exposed to HMS (20% cyclical stretch at 0.001 Hz) on high-extension silicone rubber dishes coupled to a mechanical stretching apparatus and compared to static control cultures. Gene expression of Toll-like receptors (TLRs), neuronal growth factor (NGF) and tumour necrosis factor α (TNFα) was assessed. Collected conditioned media were analysed for cytokine content and applied to rat pheocromocytoma PC12 cells for neuronal differentiation assessment. Results HMS caused upregulation of TLR2, TLR4, NGF and TNFα gene expression in IVD cells. Medium from HMS cultures contained elevated levels of growth-related oncogene, interleukin 6 (IL-6), IL-8, IL-15, monocyte chemoattractant protein 1 (MCP-1), MCP-3, monokine induced by γ interferon, transforming growth factor β1, TNFα and NGF. Exposure of PC12 cells to HMS-conditioned media resulted in both increased neurite sprouting and cell death. Conclusions HMS culture of IVD cells in vitro drives cytokine and inflammatory responses associated with degenerative disc disease and low-back pain. This study provides evidence for a direct link between cellular strain, secretory factors, neoinnervation and potential degeneration and discogenic pain in vivo.
Collapse
|
44
|
Dawes JM, McMahon SB. Chemokines as peripheral pain mediators. Neurosci Lett 2013; 557 Pt A:1-8. [PMID: 24120432 DOI: 10.1016/j.neulet.2013.10.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 09/25/2013] [Accepted: 10/01/2013] [Indexed: 11/30/2022]
Abstract
Multiple lines of evidence support the notion that much if not most chronic pain is dependent on on-going peripheral activity in nociceptors. This is not to say that central changes are unimportant, only that much of the central change is supported by a peripheral drive. This begs the question of what causes this peripheral drive. In some instances, particularly in association with peripheral nerve injury, nociceptors may become spontaneously active because of alterations in ion channel function or expression. But in most cases nociceptor activity arises because of the actions of peripheral mediators released by injured or damaged tissue. Some of these mediators are well known, such as the prostanoids. Others have more recently been identified, such as nerve growth factor (NGF). However, the limited efficacy of existing analgesic therapies strongly suggests that other important pain mediators exist. Here we discuss the evidence that a family of secreted proteins, the chemokines - well known for their actions in regulating immune cell migration - also play an important role in sustaining abnormal nociceptor activity in persistent pain states.
Collapse
Affiliation(s)
- John M Dawes
- Nuffield Department of Clinical Neurosciences, West Wing, Level 6, John Radcliffe Hospital, Oxford OX3 9DU, UK.
| | | |
Collapse
|
45
|
Zaman M, Toth I. Immunostimulation by synthetic lipopeptide-based vaccine candidates: structure-activity relationships. Front Immunol 2013; 4:318. [PMID: 24130558 PMCID: PMC3793171 DOI: 10.3389/fimmu.2013.00318] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 09/19/2013] [Indexed: 12/23/2022] Open
Abstract
Peptide-based vaccines offer several advantages over conventional whole organism or protein approaches by offering improved purity and specificity in inducing immune response. However, peptides alone are generally non-immunogenic. Concerns remain about the toxicity of adjuvants which are critical for immunogenicity of synthetic peptides. The use of lipopeptides in peptide vaccines is currently under intensive investigation because potent immune responses can be generated without the use of adjuvant (thus are self-adjuvanting). Several lipopeptides derived from microbial origin, and their synthetic versions or simpler fatty acid moieties impart this self-adjuvanting activity by signaling via Toll-like receptor 2 (TLR2). Engagement of this innate immune receptor on antigen-presenting cell leads to the initiation and development of potent immune responses. Therefore optimization of lipopeptides to enhance TLR2-mediated activation is a promising strategy for vaccine development. Considerable structure-activity relationships that determine TLR2 binding and consequent stimulation of innate immune responses have been investigated for a range of lipopeptides. In this mini review we address the development of lipopeptide vaccines, mechanism of TLR2 recognition, and immune activation. An overview is provided of the best studied lipopeptide vaccine systems.
Collapse
Affiliation(s)
- Mehfuz Zaman
- School of Chemistry and Molecular Biosciences, The University of Queensland , St Lucia, QLD , Australia
| | | |
Collapse
|
46
|
Zhang ZJ, Cao DL, Zhang X, Ji RR, Gao YJ. Chemokine contribution to neuropathic pain: respective induction of CXCL1 and CXCR2 in spinal cord astrocytes and neurons. Pain 2013; 154:2185-2197. [PMID: 23831863 DOI: 10.1016/j.pain.2013.07.002] [Citation(s) in RCA: 191] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 06/28/2013] [Accepted: 07/01/2013] [Indexed: 12/18/2022]
Abstract
Recent studies have indicated an important role of chemokines such as CCL2 in the development of chronic pain. However, the distinct roles of different chemokines in the development and maintenance of neuropathic pain and in their interactions with neurons have not been clearly elucidated. We found that spinal nerve ligation (SNL) not only induced persistent neuropathic pain symptoms, including mechanical allodynia and heat hyperalgesia, but also produced sustained CXCL1 upregulation in the spinal cord. Double staining of immunofluorescence and in situ hybridization revealed that CXCL1 was primarily induced in spinal astrocytes. In cultured astrocytes, tumor necrosis factor-α induced robust CXCL1 expression via the activation of the c-jun N-terminal kinase. Intrathecal administration of CXCL1 neutralizing antibody transiently reduced SNL-induced pain hypersensitivity, suggesting an essential role of CXCL1 in neuropathic pain sensitization. In particular, intraspinal delivery of CXCL1 shRNA lentiviral vectors, either before or after SNL, persistently attenuated SNL-induced pain hypersensitivity. Spinal application of CXCL1 not only elicited pain hypersensitivity but also induced rapid neuronal activation, as indicated by the expression of phosphorylated extracellular signal-regulated kinase and cAMP response element binding protein, and c-Fos in spinal cord neurons. Interestingly, CXCR2, the primary receptor of CXCL1, was upregulated in dorsal horn neurons after SNL, and the CXCR2 antagonist SB225002 completely blocked the CXCL1-induced heat hyperalgesia. SB225002 also attenuated SNL-induced pain hypersensitivity. Collectively, our results have demonstrated a novel form of chemokine-mediated glial-neuronal interaction in the spinal cord that can drive neuropathic pain. Inhibition of the CXCL1-CXCR2 signaling may offer a new therapy for neuropathic pain management.
Collapse
Affiliation(s)
- Zhi-Jun Zhang
- Institute of Nautical Medicine, Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong 226001, China Department of Anatomy, Medical School of Nantong University, Nantong 226001, China Sensory Plasticity Laboratory, Department of Anesthesiology and Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | |
Collapse
|
47
|
Anti-inflammatory and Immunomodulatory Effect of an Extract of Coccidioides posadasii in Experimental Arthritis. Mycopathologia 2013; 175:193-206. [DOI: 10.1007/s11046-013-9621-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Accepted: 01/21/2013] [Indexed: 02/03/2023]
|
48
|
Petho G, Reeh PW. Sensory and signaling mechanisms of bradykinin, eicosanoids, platelet-activating factor, and nitric oxide in peripheral nociceptors. Physiol Rev 2013; 92:1699-775. [PMID: 23073630 DOI: 10.1152/physrev.00048.2010] [Citation(s) in RCA: 201] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Peripheral mediators can contribute to the development and maintenance of inflammatory and neuropathic pain and its concomitants (hyperalgesia and allodynia) via two mechanisms. Activation or excitation by these substances of nociceptive nerve endings or fibers implicates generation of action potentials which then travel to the central nervous system and may induce pain sensation. Sensitization of nociceptors refers to their increased responsiveness to either thermal, mechanical, or chemical stimuli that may be translated to corresponding hyperalgesias. This review aims to give an account of the excitatory and sensitizing actions of inflammatory mediators including bradykinin, prostaglandins, thromboxanes, leukotrienes, platelet-activating factor, and nitric oxide on nociceptive primary afferent neurons. Manifestations, receptor molecules, and intracellular signaling mechanisms of the effects of these mediators are discussed in detail. With regard to signaling, most data reported have been obtained from transfected nonneuronal cells and somata of cultured sensory neurons as these structures are more accessible to direct study of sensory and signal transduction. The peripheral processes of sensory neurons, where painful stimuli actually affect the nociceptors in vivo, show marked differences with respect to biophysics, ultrastructure, and equipment with receptors and ion channels compared with cellular models. Therefore, an effort was made to highlight signaling mechanisms for which supporting data from molecular, cellular, and behavioral models are consistent with findings that reflect properties of peripheral nociceptive nerve endings. Identified molecular elements of these signaling pathways may serve as validated targets for development of novel types of analgesic drugs.
Collapse
Affiliation(s)
- Gábor Petho
- Pharmacodynamics Unit, Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Pécs, Pécs, Hungary
| | | |
Collapse
|